1
|
Talaat KR, Porter CK, Chakraborty S, Feijoo BL, Brubaker J, Adjoodani BM, DeNearing B, Prouty MG, Poole ST, Bourgeois AL, Billingsley M, Sack DA, Eder-Lingelbach S, Taucher C. Validation of a Human Challenge Model Using an LT-Expressing Enterotoxigenic E. coli Strain (LSN03-016011) and Characterization of Potential Amelioration of Disease by an Investigational Oral Vaccine Candidate (VLA1701). Microorganisms 2024; 12:727. [PMID: 38674674 PMCID: PMC11051778 DOI: 10.3390/microorganisms12040727] [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: 02/07/2024] [Revised: 03/22/2024] [Accepted: 03/23/2024] [Indexed: 04/28/2024] Open
Abstract
Controlled human infection models are important tools for the evaluation of vaccines against diseases where an appropriate correlate of protection has not been identified. Enterotoxigenic Escherichia coli (ETEC) strain LSN03-016011/A (LSN03) is an LT enterotoxin and CS17-expressing ETEC strain useful for evaluating vaccine candidates targeting LT-expressing strains. We sought to confirm the ability of the LSN03 strain to induce moderate-to-severe diarrhea in a healthy American adult population, as well as the impact of immunization with an investigational cholera/ETEC vaccine (VLA-1701) on disease outcomes. A randomized, double-blinded pilot study was conducted in which participants received two doses of VLA1701 or placebo orally, one week apart; eight days after the second vaccination, 30 participants (15 vaccinees and 15 placebo recipients) were challenged with approximately 5 × 109 colony-forming units of LSN03. The vaccine was well tolerated, with no significant adverse events. The vaccine also induced serum IgA and IgG responses to LT. After challenge, 11 of the placebo recipients (73.3%; 95%CI: 48.0-89.1) and 7 of the VLA1701 recipients (46.7%; 95%CI: 24.8-68.8) had moderate-to-severe diarrhea (p = 0.26), while 14 placebo recipients (93%) and 8 vaccine recipients (53.3%) experienced diarrhea of any severity, resulting in a protective efficacy of 42.9% (p = 0.035). In addition, the vaccine also appeared to provide protection against more severe diarrhea (p = 0.054). Vaccinees also tended to shed lower levels of the LSN03 challenge strain compared to placebo recipients (p = 0.056). In addition, the disease severity score was lower for the vaccinees than for the placebo recipients (p = 0.046). In summary, the LSN03 ETEC challenge strain induced moderate-to-severe diarrhea in 73.3% of placebo recipients. VLA1701 vaccination ameliorated disease severity, as observed by several parameters, including the percentage of participants experiencing diarrhea, as well as stool frequency and ETEC severity scores. These data highlight the potential value of LSN03 as a suitable ETEC challenge strain to evaluate LT-based vaccine targets (NCT03576183).
Collapse
Affiliation(s)
- Kawsar R. Talaat
- Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA; (S.C.); (B.L.F.); (B.M.A.); (B.D.); (A.L.B.); (M.B.); (D.A.S.)
| | - Chad K. Porter
- Naval Medical Research Command, Silver Spring, MD 20910, USA; (C.K.P.); (M.G.P.); (S.T.P.)
| | - Subhra Chakraborty
- Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA; (S.C.); (B.L.F.); (B.M.A.); (B.D.); (A.L.B.); (M.B.); (D.A.S.)
| | - Brittany L. Feijoo
- Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA; (S.C.); (B.L.F.); (B.M.A.); (B.D.); (A.L.B.); (M.B.); (D.A.S.)
| | - Jessica Brubaker
- Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA; (S.C.); (B.L.F.); (B.M.A.); (B.D.); (A.L.B.); (M.B.); (D.A.S.)
| | - Brittany M. Adjoodani
- Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA; (S.C.); (B.L.F.); (B.M.A.); (B.D.); (A.L.B.); (M.B.); (D.A.S.)
| | - Barbara DeNearing
- Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA; (S.C.); (B.L.F.); (B.M.A.); (B.D.); (A.L.B.); (M.B.); (D.A.S.)
| | - Michael G. Prouty
- Naval Medical Research Command, Silver Spring, MD 20910, USA; (C.K.P.); (M.G.P.); (S.T.P.)
| | - Steven T. Poole
- Naval Medical Research Command, Silver Spring, MD 20910, USA; (C.K.P.); (M.G.P.); (S.T.P.)
| | - A. Louis Bourgeois
- Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA; (S.C.); (B.L.F.); (B.M.A.); (B.D.); (A.L.B.); (M.B.); (D.A.S.)
| | - Madison Billingsley
- Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA; (S.C.); (B.L.F.); (B.M.A.); (B.D.); (A.L.B.); (M.B.); (D.A.S.)
| | - David A. Sack
- Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA; (S.C.); (B.L.F.); (B.M.A.); (B.D.); (A.L.B.); (M.B.); (D.A.S.)
| | | | | |
Collapse
|
2
|
Fontenele ALA, de Oliveira Nóbrega CG, do Nascimento WRC, Santos PDA, de Lorena VMB, Peixoto DM, de Azevedo Albuquerque MCP, Solé D, Costa VMA, Sarinho ESC, de Souza VMO. Respiratory allergy symptoms and cytokine profiles in the presence of anti-Ascaris antibody in Giardia lamblia-infected children. Parasitol Res 2023; 122:3147-3158. [PMID: 37875615 DOI: 10.1007/s00436-023-08005-z] [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: 05/26/2023] [Accepted: 10/09/2023] [Indexed: 10/26/2023]
Abstract
Anti-Ascaris lumbricoides (Asc) IgE and IgG can immunomodulate the allergy; however, the influence of these isotypes has not been investigated in the giardiasis and allergy. Therefore, the frequency of respiratory allergy (RA) symptoms in Giardia lamblia-infected children, with or without anti-Asc IgE, IgG1, or IgG4 and Th1, Th2/Treg, and Th17 cytokine production, was evaluated. We performed a case-control study with children aged 2-10 years old selected by questionnaire and stool exams to form the groups: infected or uninfected with RA (G-RA, n = 55; nG-RA, n = 43); infected and uninfected without RA (G-nRA, n = 59; nG-nRA, n = 54). We performed blood leukocyte counts and in vitro culture. Cytokine levels in the supernatants (CBA), serum total IgE and anti-Asc IgE (ImmunoCAP), IgG1, IgG4, and total IgA (ELISA) were measured. Infection was not associated with allergy. Infected children showed increased levels of anti-Asc IgG1, IL-2, IFN-γ, IL-4, and IL-10. There was a lower frequency of allergy-related symptoms in anti-Asc IgG1-positive children than IgG1-negative (OR = 0.38; CI = 0.17-0.90, p = 0.027) and few eosinophils in G-RA than in G-nRA and more in G-nRA than in nG-nRA, whereas TNF-α levels were higher in the G-RA than in the nG-nRA group. For infected and positive anti-Asc IgG1, there was higher TNF-α and IL-10 production than G/-IgG1. IL-10 levels were lower in nG/ + IgG1 than in infected or non-infected, and both were negative for anti-Asc IgG1. Th1/Th2/IL-10 profiles were stimulated in the infected patients, and in those with circulating anti-Asc IgG1, the TNF-α production was strengthened with a lower risk for respiratory allergy symptoms.
Collapse
Affiliation(s)
- Ana Lúcia Arruda Fontenele
- Setor de Imunologia, Laboratório de Imunopatologia Keizo Asami, Universidade Federal de Pernambuco, Recife, Brazil
| | | | | | | | | | - Décio Medeiros Peixoto
- Centro de Pesquisa em Alergia e Imunologia Clínica, Hospital das Clínicas, Universidade Federal de Pernambuco, Recife, Brazil
| | | | - Dirceu Solé
- Divisão de Alergia, Imunologia Clínica e Reumatologia, Departamento de Pediatria da, Universidade Federal de São Paulo, São Paulo, Brazil
| | | | | | - Valdenia Maria Oliveira de Souza
- Setor de Imunologia, Laboratório de Imunopatologia Keizo Asami, Universidade Federal de Pernambuco, Recife, Brazil.
- Departamento de Ciências Farmacêuticas, Universidade Federal de Pernambuco, Av. Prof. Moraes Rego, S/N. Cidade Universitária, 50.670-901, Recife, PE, Brazil.
| |
Collapse
|
3
|
Andersen-Civil AIS, Thamsborg SM, Williams AR. Profiling of hepatic transcriptomes reveals modulatory effects of parasitic infection on the metabolic response to dietary polyphenols in pigs. J Nutr Biochem 2023; 116:109316. [PMID: 36940885 DOI: 10.1016/j.jnutbio.2023.109316] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2022] [Revised: 01/20/2023] [Accepted: 03/07/2023] [Indexed: 03/23/2023]
Abstract
Polyphenols are a class of bioactive plant compounds with health-promoting properties, however the interactions between polyphenols and pathogen infection and their cumulative impact on inflammation and metabolic health are not well understood. Here, we investigated if a sub-clinical parasitic infection modulates the hepatic response to dietary polyphenol supplementation in a porcine model. Pigs were fed a diet with or without 1% grape proanthocyanidin (PAC) for 28 days. During the final 14 days of the experiment, half the pigs in each dietary group were inoculated with the parasitic nematode Ascaris suum. Serum biochemistry was measured and hepatic transcriptional responses were determined by RNA-sequencing coupled with gene-set enrichment analysis. A. suum infection resulted in reduced serum phosphate, potassium and calcium, and increased serum iron concentrations. In uninfected pigs, PAC supplementation markedly changed the liver transcriptome including genes related to carbohydrate and lipid metabolism, insulin signaling, and bile acid synthesis. However, during A. suum infection, a separate set of genes were modulated by dietary PAC, indicating that the polyphenol-mediated effects were dependent on infection status. A. suum infection strongly influenced the expression of genes related to cellular metabolism, and, in contrast to the effects of PAC, these changes were mostly identical in both control-fed and PAC-fed pigs. Thus, the hepatic response to infection was mostly unaffected by concurrent polyphenol intake. We conclude that the presence of a commonly occurring parasite substantially influences the outcome of dietary polyphenol supplementation, which may have important relevance for nutritional interventions in populations where intestinal parasitism is widespread.
Collapse
Affiliation(s)
| | - Stig M Thamsborg
- Department of Veterinary and Animal Sciences, University of Copenhagen, Denmark
| | - Andrew R Williams
- Department of Veterinary and Animal Sciences, University of Copenhagen, Denmark.
| |
Collapse
|
4
|
Sauvaitre T, Van Landuyt J, Durif C, Roussel C, Sivignon A, Chalancon S, Uriot O, Van Herreweghen F, Van de Wiele T, Etienne-Mesmin L, Blanquet-Diot S. Role of mucus-bacteria interactions in Enterotoxigenic Escherichia coli (ETEC) H10407 virulence and interplay with human microbiome. NPJ Biofilms Microbiomes 2022; 8:86. [PMID: 36266277 PMCID: PMC9584927 DOI: 10.1038/s41522-022-00344-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Accepted: 10/04/2022] [Indexed: 11/09/2022] Open
Abstract
The intestinal mucus layer has a dual role in human health constituting a well-known microbial niche that supports gut microbiota maintenance but also acting as a physical barrier against enteric pathogens. Enterotoxigenic Escherichia coli (ETEC), the major agent responsible for traveler's diarrhea, is able to bind and degrade intestinal mucins, representing an important but understudied virulent trait of the pathogen. Using a set of complementary in vitro approaches simulating the human digestive environment, this study aimed to describe how the mucus microenvironment could shape different aspects of the human ETEC strain H10407 pathophysiology, namely its survival, adhesion, virulence gene expression, interleukin-8 induction and interactions with human fecal microbiota. Using the TNO gastrointestinal model (TIM-1) simulating the physicochemical conditions of the human upper gastrointestinal (GI) tract, we reported that mucus secretion and physical surface sustained ETEC survival, probably by helping it to face GI stresses. When integrating the host part in Caco2/HT29-MTX co-culture model, we demonstrated that mucus secreting-cells favored ETEC adhesion and virulence gene expression, but did not impede ETEC Interleukin-8 (IL-8) induction. Furthermore, we proved that mucosal surface did not favor ETEC colonization in a complex gut microbial background simulated in batch fecal experiments. However, the mucus-specific microbiota was widely modified upon the ETEC challenge suggesting its role in the pathogen infectious cycle. Using multi-targeted in vitro approaches, this study supports the major role played by mucus in ETEC pathophysiology, opening avenues in the design of new treatment strategies.
Collapse
Affiliation(s)
- Thomas Sauvaitre
- Université Clermont Auvergne, INRAE, UMR 454 MEDIS, Microbiologie Environnement Digestif et Santé (MEDIS), CRNH Auvergne, 63000, Clermont-Ferrand, France.,Ghent University, Faculty of Bioscience Engineering, Center for Microbial Ecology and Technology (CMET), Ghent, Belgium
| | - Josefien Van Landuyt
- Ghent University, Faculty of Bioscience Engineering, Center for Microbial Ecology and Technology (CMET), Ghent, Belgium
| | - Claude Durif
- Université Clermont Auvergne, INRAE, UMR 454 MEDIS, Microbiologie Environnement Digestif et Santé (MEDIS), CRNH Auvergne, 63000, Clermont-Ferrand, France
| | - Charlène Roussel
- Université Laval, Nutrition and Functional Foods Institute (INAF), 2440 Bd Hochelaga Suite 1710, Québec, QC, G1V 0A6, Canada
| | - Adeline Sivignon
- Université Clermont Auvergne, UMR 1071 Inserm, USC-INRAE 2018, Microbes, Intestin, Inflammation et Susceptibilité de l'Hôte (M2iSH), 63000, Clermont-Ferrand, France
| | - Sandrine Chalancon
- Université Clermont Auvergne, INRAE, UMR 454 MEDIS, Microbiologie Environnement Digestif et Santé (MEDIS), CRNH Auvergne, 63000, Clermont-Ferrand, France
| | - Ophélie Uriot
- Université Clermont Auvergne, INRAE, UMR 454 MEDIS, Microbiologie Environnement Digestif et Santé (MEDIS), CRNH Auvergne, 63000, Clermont-Ferrand, France
| | - Florence Van Herreweghen
- Ghent University, Faculty of Bioscience Engineering, Center for Microbial Ecology and Technology (CMET), Ghent, Belgium
| | - Tom Van de Wiele
- Ghent University, Faculty of Bioscience Engineering, Center for Microbial Ecology and Technology (CMET), Ghent, Belgium
| | - Lucie Etienne-Mesmin
- Université Clermont Auvergne, INRAE, UMR 454 MEDIS, Microbiologie Environnement Digestif et Santé (MEDIS), CRNH Auvergne, 63000, Clermont-Ferrand, France
| | - Stéphanie Blanquet-Diot
- Université Clermont Auvergne, INRAE, UMR 454 MEDIS, Microbiologie Environnement Digestif et Santé (MEDIS), CRNH Auvergne, 63000, Clermont-Ferrand, France.
| |
Collapse
|
5
|
Dougherty M, Bartelt LA. Giardia and growth impairment in children in high-prevalence settings: consequence or co-incidence? Curr Opin Infect Dis 2022; 35:417-423. [PMID: 35980005 PMCID: PMC10373467 DOI: 10.1097/qco.0000000000000877] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
PURPOSE OF REVIEW Giardia is a common intestinal parasite worldwide, and infection can be associated with clear, and sometimes persistent symptomatology. However, in children in high-prevalence settings, it is most often not associated with or is perhaps even protective against acute diarrhea. Nonetheless, recent longitudinal studies in high-prevalence settings increasingly identify an association with long-term outcomes that has been difficult to discern. RECENT FINDINGS Recent studies have made progress in disentangling this apparent paradox. First, prospective, well characterized cohort studies have repeatedly identified associations between Giardia infection, gut function, and child growth. Second, experimental animal and in-vitro models have further characterized the biological plausibility that Giardia could impair intestinal function and subsequently child development through different pathways, depending upon biological and environmental factors. Finally, new work has shed light on the potential for Giardia conspiring with specific other gut microbes, which may explain discrepant findings in the literature, help guide future higher resolution analyses of this pathogen, and inform new opportunities for intervention. SUMMARY Recent prospective studies have confirmed a high, if not universal, prevalence of persistent Giardia infections in low-and-middle income countries associated with child-growth shortfalls and altered gut permeability. However, the predominance of subclinical infections limits understanding of the true clinical impact of endemic pediatric giardiasis, and global disease burdens remain uncalculated. Integrating the role of Giardia in multipathogen enteropathies and how nutritional, microbial, metabolic, and pathogen-strain variables influence Giardia infection outcomes could sharpen delineations between pathogenic and potentially beneficial attributes of this enigmatic parasite.
Collapse
Affiliation(s)
- Michael Dougherty
- Division of Gastroenterology and Hepatology, Department of Medicine, University of North Carolina at Chapel Hill
- Rex Digestive Healthcare, UNC REX Healthcare, Raleigh
| | - Luther A. Bartelt
- Division of Infectious Diseases, Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| |
Collapse
|
6
|
Mucosal Immune Profiles Associated with Diarrheal Disease Severity in Shigella- and Enteropathogenic Escherichia coli-Infected Children Enrolled in the Global Enteric Multicenter Study. mBio 2022; 13:e0053822. [PMID: 35924851 PMCID: PMC9426439 DOI: 10.1128/mbio.00538-22] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Enteropathogenic Escherichia coli (EPEC) and Shigella are etiologic agents of diarrhea in children <5 years old living in resource-poor countries. Repeated bouts of infection lead to lifelong morbidity and even death. The goal of this study was to characterize local mucosal immune responses in Shigella- and EPEC-infected children <5 years of age with moderate to severe diarrhea (MSD) enrolled in the Global Enteric Multicenter Study (GEMS). We hypothesized that infection with each of these pathogens would induce distinct gut mucosal immune profiles indicative of disease etiology and severity. To test this hypothesis, innate and adaptive immune markers were measured in stools from children with diarrhea due to EPEC, Shigella, or other organisms and in children who had no diarrhea. Shigella-positive diarrhea evoked robust proinflammatory and TH1/TH2 cytokine responses compared to diarrhea caused by EPEC or other organisms, with the exception of interleukin 5 (IL-5), which was associated with EPEC infection. The presence of IL-1β, IL-4, IL-16, and tumor necrosis factor beta (TNF-β) was associated with the absence of dysentery. EPEC-positive diarrhea evoked high levels of IL-1β, vascular endothelial growth factor (VEGF), and IL-10. Granulocyte-macrophage colony-stimulating factor (GM-CSF) had opposing roles in disease severity, being associated with absence of diarrhea in EPEC-infected children and with dysenteric Shigella infection. High levels of antigen-specific antibodies were detected in the controls and children with Shigella without dysentery, which suggests a protective role against severe disease. In summary, this study identified distinct local immune responses associated with two clinically relevant diarrheagenic pathogens, Shigella and EPEC, in children and identified protective immune phenotypes that can inform the development of preventive measures.
Collapse
|
7
|
Sauvaitre T, Van Herreweghen F, Delbaere K, Durif C, Van Landuyt J, Fadhlaoui K, Huille S, Chaucheyras-Durand F, Etienne-Mesmin L, Blanquet-Diot S, Van de Wiele T. Lentils and Yeast Fibers: A New Strategy to Mitigate Enterotoxigenic Escherichia coli (ETEC) Strain H10407 Virulence? Nutrients 2022; 14:nu14102146. [PMID: 35631287 PMCID: PMC9144138 DOI: 10.3390/nu14102146] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Revised: 05/16/2022] [Accepted: 05/18/2022] [Indexed: 01/10/2023] Open
Abstract
Dietary fibers exhibit well-known beneficial effects on human health, but their anti-infectious properties against enteric pathogens have been poorly investigated. Enterotoxigenic Escherichia coli (ETEC) is a major food-borne pathogen that causes acute traveler’s diarrhea. Its virulence traits mainly rely on adhesion to an epithelial surface, mucus degradation, and the secretion of two enterotoxins associated with intestinal inflammation. With the increasing burden of antibiotic resistance worldwide, there is an imperious need to develop novel alternative strategies to control ETEC infections. This study aimed to investigate, using complementary in vitro approaches, the inhibitory potential of two dietary-fiber-containing products (a lentil extract and yeast cell walls) against the human ETEC reference strain H10407. We showed that the lentil extract decreased toxin production in a dose-dependent manner, reduced pro-inflammatory interleukin-8 production, and modulated mucus-related gene induction in ETEC-infected mucus-secreting intestinal cells. We also report that the yeast product reduced ETEC adhesion to mucin and Caco-2/HT29-MTX cells. Both fiber-containing products strengthened intestinal barrier function and modulated toxin-related gene expression. In a complex human gut microbial background, both products did not elicit a significant effect on ETEC colonization. These pioneering data demonstrate the promising role of dietary fibers in controlling different stages of the ETEC infection process.
Collapse
Affiliation(s)
- Thomas Sauvaitre
- UMR 454 INRAE, Microbiology, Digestive Environment and Health (MEDIS), Université Clermont Auvergne, 28 Place Henri Dunant, F-63000 Clermont-Ferrand, France; (T.S.); (C.D.); (K.F.); (F.C.-D.); (L.E.-M.)
- Center for Microbial Ecology and Technology (CMET), Faculty of Bioscience Engineering, Ghent University, Coupure Links 653, B-9000 Ghent, Belgium; (F.V.H.); (K.D.); (J.V.L.); (T.V.d.W.)
| | - Florence Van Herreweghen
- Center for Microbial Ecology and Technology (CMET), Faculty of Bioscience Engineering, Ghent University, Coupure Links 653, B-9000 Ghent, Belgium; (F.V.H.); (K.D.); (J.V.L.); (T.V.d.W.)
| | - Karen Delbaere
- Center for Microbial Ecology and Technology (CMET), Faculty of Bioscience Engineering, Ghent University, Coupure Links 653, B-9000 Ghent, Belgium; (F.V.H.); (K.D.); (J.V.L.); (T.V.d.W.)
| | - Claude Durif
- UMR 454 INRAE, Microbiology, Digestive Environment and Health (MEDIS), Université Clermont Auvergne, 28 Place Henri Dunant, F-63000 Clermont-Ferrand, France; (T.S.); (C.D.); (K.F.); (F.C.-D.); (L.E.-M.)
| | - Josefien Van Landuyt
- Center for Microbial Ecology and Technology (CMET), Faculty of Bioscience Engineering, Ghent University, Coupure Links 653, B-9000 Ghent, Belgium; (F.V.H.); (K.D.); (J.V.L.); (T.V.d.W.)
| | - Khaled Fadhlaoui
- UMR 454 INRAE, Microbiology, Digestive Environment and Health (MEDIS), Université Clermont Auvergne, 28 Place Henri Dunant, F-63000 Clermont-Ferrand, France; (T.S.); (C.D.); (K.F.); (F.C.-D.); (L.E.-M.)
| | | | - Frédérique Chaucheyras-Durand
- UMR 454 INRAE, Microbiology, Digestive Environment and Health (MEDIS), Université Clermont Auvergne, 28 Place Henri Dunant, F-63000 Clermont-Ferrand, France; (T.S.); (C.D.); (K.F.); (F.C.-D.); (L.E.-M.)
- Lallemand SAS, 19 Rue des Briquetiers, BP 59, CEDEX, F-31702 Blagnac, France
| | - Lucie Etienne-Mesmin
- UMR 454 INRAE, Microbiology, Digestive Environment and Health (MEDIS), Université Clermont Auvergne, 28 Place Henri Dunant, F-63000 Clermont-Ferrand, France; (T.S.); (C.D.); (K.F.); (F.C.-D.); (L.E.-M.)
| | - Stéphanie Blanquet-Diot
- UMR 454 INRAE, Microbiology, Digestive Environment and Health (MEDIS), Université Clermont Auvergne, 28 Place Henri Dunant, F-63000 Clermont-Ferrand, France; (T.S.); (C.D.); (K.F.); (F.C.-D.); (L.E.-M.)
- Correspondence: ; Tel.: +33-(0)4-73-17-83-90
| | - Tom Van de Wiele
- Center for Microbial Ecology and Technology (CMET), Faculty of Bioscience Engineering, Ghent University, Coupure Links 653, B-9000 Ghent, Belgium; (F.V.H.); (K.D.); (J.V.L.); (T.V.d.W.)
| |
Collapse
|
8
|
Sun M, Yan Z, Sun R, Tian W, Yi W, Zhang J. Dynamic monitoring and a clinical correlation analysis of the serum vitamin A, D, and E levels in children with recurrent respiratory tract infections. Am J Transl Res 2022; 14:3533-3538. [PMID: 35702083 PMCID: PMC9185048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2021] [Accepted: 06/17/2021] [Indexed: 06/15/2023]
Abstract
OBJECTIVE To investigate the correlation of the serum vitamin A, D, and E (VA, VD, and VE) levels with the occurrence and development of recurrent respiratory tract infections (RRTIs). METHODS A total of 129 children with respiratory tract infections (RTIs) treated in our hospital from January 2018 to February 2020 (the RTIs group) and 50 healthy children undergoing physical examinations (the control group) in our hospital were recruited as the study cohort. The serum VA, VD, and VE levels were measured upon admission (the active phase) and at two weeks after discharge (the stable phase). The serum VA, VD, and VE levels in the children with RRTIs were compared with the levels in the control group, and the correlation between these three vitamins and the occurrence and development of RRTIs was analyzed. RESULTS The RRTIs group and the RTIs group witnessed markedly lower serum VA, VD, VE, and humoral immunity index levels, including IgG, IgA, and IgM, compared to the control group, with an apparent lower outcome in the RRTIs group than in the RTIs group. The serum levels of the above indexes in the RRTIs children were reduced in the active phase compared with the stable phase. A Pearson correlation analysis showed a positive correlation between VA and IgA. A multivariate logistic regression analysis revealed that a low BMI (Body mass index), prematurity, VA deficiency, VD deficiency, and VE deficiency were the risk factors for RRTIs in children, and outdoor activity was the protective factor. CONCLUSION The VA, VD, and VE levels are closely related to RRTIs in children. It is important to determine and supplement the VA, VD, and VE levels to prevent RTIs in children.
Collapse
Affiliation(s)
- Mei Sun
- Department of Pediatrics, Cangzhou Central Hospital Cangzhou, China
| | - Zhixin Yan
- Department of Pediatrics, Cangzhou Central Hospital Cangzhou, China
| | - Rongrong Sun
- Department of Pediatrics, Cangzhou Central Hospital Cangzhou, China
| | - Wenqiu Tian
- Department of Pediatrics, Cangzhou Central Hospital Cangzhou, China
| | - Wenxia Yi
- Department of Pediatrics, Cangzhou Central Hospital Cangzhou, China
| | - Jing Zhang
- Department of Pediatrics, Cangzhou Central Hospital Cangzhou, China
| |
Collapse
|
9
|
Enterotoxigenic Escherichia coli enterotoxins regulate epithelial to immune relay of IL-33 and IL-1Ra cytokines. Infect Immun 2022; 90:e0063721. [PMID: 35191758 DOI: 10.1128/iai.00637-21] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Enterotoxigenic Escherichia coli (ETEC) remain a major cause of diarrheal mortality and morbidity in children in low-resource settings. Few studies have explored the consequences of simultaneous intoxication with heat-stable (ST) and heat-labile (LT) enterotoxins despite the increased prevalence of wild ETEC isolates expressing both toxins. We therefore used a combination of tissue culture and murine models to explore the impact of simultaneous ST+LT intoxication of epithelial and myeloid cell responses. We report that LT induces sustained IL-33 and IL-1Ra responses in T84 intestinal epithelial cells via cAMP-production and protein kinase A activation. We demonstrate that combined ST+LT intoxication hastens epithelial transcriptional responses induced more slowly by LT alone. ST- and LT-mediated luminal fluid accumulation in vivo correlates with significant increases in IL-33 and IL-1Ra in small intestinal mucosal scrapings. Additionally, IL-33 receptor (IL-33R)-deficient mice are less susceptible to ST-mediated secretion. In the immune compartment, IL-33 is sensed by myeloid cells, and LT suppresses IL-33-induced TNFα secretion from macrophages but amplifies IL-33-mediated induction of IL-6 from bone marrow-derived dendritic cells. In conclusion, our studies suggest that enterotoxin-induced IL-33 and IL-1Ra modulate intestinal inflammation and IL-1 receptor signaling in the intestinal mucosa in response to ETEC enterotoxins.
Collapse
|
10
|
Ledwaba SE, Costa DVS, Bolick DT, Giallourou N, Medeiros PHQS, Swann JR, Traore AN, Potgieter N, Nataro JP, Guerrant RL. Enteropathogenic Escherichia coli Infection Induces Diarrhea, Intestinal Damage, Metabolic Alterations, and Increased Intestinal Permeability in a Murine Model. Front Cell Infect Microbiol 2020; 10:595266. [PMID: 33392105 PMCID: PMC7773950 DOI: 10.3389/fcimb.2020.595266] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2020] [Accepted: 11/11/2020] [Indexed: 12/15/2022] Open
Abstract
Enteropathogenic E. coli (EPEC) are recognized as one of the leading bacterial causes of infantile diarrhea worldwide. Weaned C57BL/6 mice pretreated with antibiotics were challenged orally with wild-type EPEC or escN mutant (lacking type 3 secretion system) to determine colonization, inflammatory responses and clinical outcomes during infection. Antibiotic disruption of intestinal microbiota enabled efficient colonization by wild-type EPEC resulting in growth impairment and diarrhea. Increase in inflammatory biomarkers, chemokines, cellular recruitment and pro-inflammatory cytokines were observed in intestinal tissues. Metabolomic changes were also observed in EPEC infected mice with changes in tricarboxylic acid (TCA) cycle intermediates, increased creatine excretion and shifts in gut microbial metabolite levels. In addition, by 7 days after infection, although weights were recovering, EPEC-infected mice had increased intestinal permeability and decreased colonic claudin-1 levels. The escN mutant colonized the mice with no weight loss or increased inflammatory biomarkers, showing the importance of the T3SS in EPEC virulence in this model. In conclusion, a murine infection model treated with antibiotics has been developed to mimic clinical outcomes seen in children with EPEC infection and to examine potential roles of selected virulence traits. This model can help in further understanding mechanisms involved in the pathogenesis of EPEC infections and potential outcomes and thus assist in the development of potential preventive or therapeutic interventions.
Collapse
Affiliation(s)
- Solanka E. Ledwaba
- Department of Microbiology, University of Venda, Thohoyandou, South Africa
| | - Deiziane V. S. Costa
- Department of Physiology and Pharmacology, Federal University of Ceará, Fortaleza, Brazil
| | - David T. Bolick
- Center for Global Health, Division of Infectious Disease and International Health, University of Virginia School of Medicine, Charlottesville, VA, United States
| | - Natasa Giallourou
- Faculty of Medicine, Department of Metabolism, Digestion and Reproduction, Imperial College, London, England
| | | | - Jonathan R. Swann
- Faculty of Medicine, Department of Metabolism, Digestion and Reproduction, Imperial College, London, England
| | - Afsatou N. Traore
- Department of Microbiology, University of Venda, Thohoyandou, South Africa
| | - Natasha Potgieter
- Department of Microbiology, University of Venda, Thohoyandou, South Africa
| | - James P. Nataro
- Department of Pediatrics, University of Virginia School of Medicine, Charlottesville, VA, United States
| | - Richard L. Guerrant
- Center for Global Health, Division of Infectious Disease and International Health, University of Virginia School of Medicine, Charlottesville, VA, United States
| |
Collapse
|
11
|
Liang Q, Liu J, Wei J, Jia J, Shen H, Chen W, Liang W, Gao B, Xu Z, Zhang L. The effect of Clostridium tyrobutyricum Spo0A overexpression in the intestine of mice. Benef Microbes 2020; 11:573-589. [PMID: 33032473 DOI: 10.3920/bm2019.0131] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Clostridium tyrobutyricum shows probiotic properties and can affect the composition of gut microbiota and regulate the intestinal immune system. Compared with other probiotics, this spore-producing bacterium shows unparalleled advantages in commercial production. In addition to being resistant to extreme living environments for extended periods, its endophytic spores are implicated in inhibiting cancer cell growth. We speculated that C. tyrobutyricum spores can also promote gut health, which mean it can maintain intestinal homeostasis. To date, the beneficial effects of C. tyrobutyricum spores on gut health have not been reported. In this study, a Spo0A-overexpressing C. tyrobutyricum strain was developed to increase spore production, and its probiotic effects on the gut were assessed. Compared with the wild-type, the engineered strain showed significantly increased sporulation rates. Mice administered with the engineered strain exhibited enhanced intestinal villi and the villus height/crypt depth ratio, weight gain and improved Firmicutes/Bacteroidetes ratio to facilitate intestinal homeostasis. This study demonstrated for the first time that enhanced spore production in C. tyrobutyricum can improve intestinal homeostasis, which is advantageous for its commercial application in food and pharmaceutical industry.
Collapse
Affiliation(s)
- Q Liang
- Guangdong Provincial Key Laboratory of Protein Function and Regulation in Agricultural Organisms, College of Life Sciences, South China Agricultural University, Guangzhou, Guangdong 510642, China P.R
| | - J Liu
- Guangdong Provincial Key Laboratory of Protein Function and Regulation in Agricultural Organisms, College of Life Sciences, South China Agricultural University, Guangzhou, Guangdong 510642, China P.R.,Guangdong Provincial Key Laboratory of Livestock and Poultry Disease Control, Guangdong Provincial Institute of Veterinary Public Health, Public Health Laboratory, Institute of Animal Health, Guangdong Academy of Agricultural Sciences, Guangzhou, Guangdong 510642, China P.R
| | - J Wei
- Guangdong Provincial Key Laboratory of Protein Function and Regulation in Agricultural Organisms, College of Life Sciences, South China Agricultural University, Guangzhou, Guangdong 510642, China P.R
| | - J Jia
- Guangdong Provincial Key Laboratory of Protein Function and Regulation in Agricultural Organisms, College of Life Sciences, South China Agricultural University, Guangzhou, Guangdong 510642, China P.R
| | - H Shen
- Guangdong Provincial Key Laboratory of Protein Function and Regulation in Agricultural Organisms, College of Life Sciences, South China Agricultural University, Guangzhou, Guangdong 510642, China P.R
| | - W Chen
- Guangdong Provincial Key Laboratory of Livestock and Poultry Disease Control, Guangdong Provincial Institute of Veterinary Public Health, Public Health Laboratory, Institute of Animal Health, Guangdong Academy of Agricultural Sciences, Guangzhou, Guangdong 510642, China P.R
| | - W Liang
- Guangdong Provincial Key Laboratory of Livestock and Poultry Disease Control, Guangdong Provincial Institute of Veterinary Public Health, Public Health Laboratory, Institute of Animal Health, Guangdong Academy of Agricultural Sciences, Guangzhou, Guangdong 510642, China P.R
| | - B Gao
- Guangdong Provincial Key Laboratory of Livestock and Poultry Disease Control, Guangdong Provincial Institute of Veterinary Public Health, Public Health Laboratory, Institute of Animal Health, Guangdong Academy of Agricultural Sciences, Guangzhou, Guangdong 510642, China P.R
| | - Z Xu
- Guangdong Provincial Key Laboratory of Livestock and Poultry Disease Control, Guangdong Provincial Institute of Veterinary Public Health, Public Health Laboratory, Institute of Animal Health, Guangdong Academy of Agricultural Sciences, Guangzhou, Guangdong 510642, China P.R
| | - L Zhang
- Guangdong Provincial Key Laboratory of Protein Function and Regulation in Agricultural Organisms, College of Life Sciences, South China Agricultural University, Guangzhou, Guangdong 510642, China P.R.,Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou 510642, China P.R
| |
Collapse
|
12
|
Wang J, Han C, Lu Z, Ge P, Cui Y, Zhao D, Yang X, Wu B, Qiang L, Zhang Y, Chai Q, Lei Z, Li L, Hua Liu C, Zhang L. Simulated microgravity suppresses MAPK pathway-mediated innate immune response to bacterial infection and induces gut microbiota dysbiosis. FASEB J 2020; 34:14631-14644. [PMID: 32918764 DOI: 10.1096/fj.202001428r] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Revised: 08/10/2020] [Accepted: 08/19/2020] [Indexed: 12/12/2022]
Abstract
During spaceflight, astronauts are subjected to various physical stressors including microgravity, which could cause immune dysfunction and thus potentially predispose astronauts to infections and illness. However, the mechanisms by which microgravity affects innate immunity remain largely unclear. In this study, we conducted RNA-sequencing analysis to show that simulated microgravity (SMG) suppresses the production of inflammatory cytokines including tumor necrosis factor (TNF) and interleukin-6 (IL-6) as well as the activation of the innate immune signaling pathways including the p38 mitogen-activated protein kinase (MAPK) and the Erk1/2 MAPK pathways in the Enteropathogenic escherichia coli (EPEC)-infected macrophage cells. We then adopted hindlimb-unloading (HU) mice, a model mimicking the microgravity of a spaceflight environment, to demonstrate that microgravity suppresses proinflammatory cytokine-mediated intestinal immunity to Citrobacter rodentium infection and induces the disturbance of gut microbiota, both of which phenotypes could be largely corrected by the introduction of VSL#3, a high-concentration probiotic preparation of eight live freeze-dried bacterial species. Taken together, our study provides new insights into microgravity-mediated innate immune suppression and intestinal microbiota disturbance, and suggests that probiotic VSL#3 has great potential as a dietary supplement in protecting individuals from spaceflight mission-associated infections and gut microbiota dysbiosis.
Collapse
Affiliation(s)
- Jing Wang
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Beijing, China
| | - Conghui Han
- School of Public Health, Southern Medical University, Guangzhou, China
| | - Zhe Lu
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Beijing, China.,Savaid Medical School, University of Chinese Academy of Sciences, Beijing, China
| | - Pupu Ge
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Beijing, China.,Savaid Medical School, University of Chinese Academy of Sciences, Beijing, China
| | - Yu Cui
- State Key Laboratory of Proteomics, National Center of Protein Sciences, Beijing Institute of Lifeomics, Beijing, China
| | - Dongdong Zhao
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Beijing, China.,Savaid Medical School, University of Chinese Academy of Sciences, Beijing, China
| | - Xi Yang
- State Key Laboratory of Proteomics, National Center of Protein Sciences, Beijing Institute of Lifeomics, Beijing, China
| | - Bo Wu
- State Key Laboratory of Proteomics, National Center of Protein Sciences, Beijing Institute of Lifeomics, Beijing, China
| | - Lihua Qiang
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Beijing, China.,Savaid Medical School, University of Chinese Academy of Sciences, Beijing, China
| | - Yong Zhang
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Beijing, China
| | - Qiyao Chai
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Beijing, China.,Savaid Medical School, University of Chinese Academy of Sciences, Beijing, China
| | - Zehui Lei
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Beijing, China.,Savaid Medical School, University of Chinese Academy of Sciences, Beijing, China
| | - Ling Li
- School of Public Health, Southern Medical University, Guangzhou, China
| | - Cui Hua Liu
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Beijing, China.,Savaid Medical School, University of Chinese Academy of Sciences, Beijing, China
| | - Lingqiang Zhang
- State Key Laboratory of Proteomics, National Center of Protein Sciences, Beijing Institute of Lifeomics, Beijing, China
| |
Collapse
|
13
|
Nettleford SK, Zhao L, Qian F, Herold M, Arner B, Desai D, Amin S, Xiong N, Singh V, Carlson BA, Prabhu KS. The Essential Role of Selenoproteins in the Resolution of Citrobacter rodentium-Induced Intestinal Inflammation. Front Nutr 2020; 7:96. [PMID: 32775340 PMCID: PMC7381334 DOI: 10.3389/fnut.2020.00096] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Accepted: 05/26/2020] [Indexed: 12/13/2022] Open
Abstract
Enteropathogenic Escherichia coli (EPEC) leads to adverse colonic inflammation associated with poor resolution of inflammation and loss of epithelial integrity. Micronutrient trace element selenium (Se) is incorporated into selenoproteins as the 21st amino acid, selenocysteine (Sec). Previous studies have shown that such an incorporation of Sec into the selenoproteome is key for the anti-inflammatory functions of Se in macrophages and other immune cells. An intriguing mechanism underlying the anti-inflammatory and pro-resolving effects of Se stems from the ability of selenoproteins to skew arachidonic acid metabolism from pro-inflammatory mediators, prostaglandin E2 (PGE2) toward anti-inflammatory mediators derived from PGD2, such as 15-deoxy-Δ12, 14- prostaglandin J2 (15d-PGJ2), via eicosanoid class switching of bioactive lipids. The impact of Se and such an eicosanoid-class switching mechanism was tested in an enteric infection model of gut inflammation by C. rodentium, a murine equivalent of EPEC. C57BL/6 mice deficient in Se (Se-D) experienced higher mortality when compared to those on Se adequate (0.08 ppm Se) and Se supplemented (0.4 ppm Se) diets following infection. Decreased survival was associated with decreased group 3 innate lymphoid cells (ILC3s) and T helper 17 (Th17) cells in colonic lamina propria of Se-D mice along with deceased expression of epithelial barrier protein Zo-1. Inhibition of metabolic inactivation of PGE2 by 15-prostaglandin dehydrogenase blocked the Se-dependent increase in ILC3 and Th17 cells in addition to reducing epithelial barrier integrity, as seen by increased systemic levels of FITC-dextran following oral administration; while 15d-PGJ2 administration in Se-D mice alleviated the effects by increasing ILC3 and Th17 cells. Mice lacking selenoproteins in monocyte/macrophages via the conditional deletion of the tRNA[Sec] showed increased mortality post infection. Our studies indicate a crucial role for dietary Se in the protection against inflammation following enteric infection via immune mechanisms involving epithelial barrier integrity.
Collapse
Affiliation(s)
- Shaneice K Nettleford
- Department of Veterinary and Biomedical Sciences, Center for Molecular Immunology and Infectious Disease, The Pennsylvania State University, State College, PA, United States
| | - Luming Zhao
- Department of Veterinary and Biomedical Sciences, Center for Molecular Immunology and Infectious Disease, The Pennsylvania State University, State College, PA, United States
| | - Fenghua Qian
- Department of Veterinary and Biomedical Sciences, Center for Molecular Immunology and Infectious Disease, The Pennsylvania State University, State College, PA, United States
| | - Morgan Herold
- Department of Veterinary and Biomedical Sciences, Center for Molecular Immunology and Infectious Disease, The Pennsylvania State University, State College, PA, United States
| | - Brooke Arner
- Department of Veterinary and Biomedical Sciences, Center for Molecular Immunology and Infectious Disease, The Pennsylvania State University, State College, PA, United States
| | - Dhimant Desai
- Department of Pharmacology, Organic Synthesis Core Laboratory, Penn State Cancer Institute, Penn State College of Medicine, Hershey, PA, United States
| | - Shantu Amin
- Department of Pharmacology, Organic Synthesis Core Laboratory, Penn State Cancer Institute, Penn State College of Medicine, Hershey, PA, United States
| | - Na Xiong
- Department of Microbiology, Immunology & Molecular Genetics, Long School of Medicine, University of Texas Health San Antonio, San Antonio, TX, United States
| | - Vishal Singh
- Department of Nutritional Sciences, The Pennsylvania State University, State College, PA, United States
| | - Bradley A Carlson
- Molecular Biology of Selenium Section, Mouse Genetics Program, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| | - K Sandeep Prabhu
- Department of Veterinary and Biomedical Sciences, Center for Molecular Immunology and Infectious Disease, The Pennsylvania State University, State College, PA, United States
| |
Collapse
|
14
|
Williams AR, Andersen-Civil AIS, Zhu L, Blanchard A. Dietary phytonutrients and animal health: regulation of immune function during gastrointestinal infections. J Anim Sci 2020; 98:5718206. [PMID: 31999321 DOI: 10.1093/jas/skaa030] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2019] [Accepted: 01/26/2020] [Indexed: 12/13/2022] Open
Abstract
The composition of dietary macronutrients (proteins, carbohydrates, and fibers) and micronutrients (vitamins, phytochemicals) can markedly influence the development of immune responses to enteric infection. This has important implications for livestock production, where a significant challenge exists to ensure healthy and productive animals in an era of increasing drug resistance and concerns about the sector's environmental footprint. Nutritional intervention may ultimately be a sustainable method to prevent disease and improve efficiency of livestock enterprises, and it is now well established that certain phytonutrients can significantly improve animal performance during challenge with infectious pathogens. However, many questions remain unanswered concerning the complex interplay between diet, immunity, and infection. In this review, we examine the role of phytonutrients in regulating immune and inflammatory responses during enteric bacterial and parasitic infections in livestock, with a specific focus on some increasingly well-studied phytochemical classes-polyphenols (especially proanthocyanidins), essential oil components (cinnamaldehyde, eugenol, and carvacrol), and curcumin. Despite the contrasting chemical structures of these molecules, they appear to induce a number of similar immunological responses. These include promotion of mucosal antibody and antimicrobial peptide production, coupled with a strong suppression of inflammatory cytokines and reactive oxygen species. Although there have been some recent advances in our understanding of the mechanisms underlying their bioactivity, how these phytonutrients modulate immune responses in the intestine remains mostly unknown. We discuss the complex inter-relationships between metabolism of dietary phytonutrients, the gut microbiota, and the mucosal immune system, and propose that an increased understanding of the basic immunological mechanisms involved will allow the rational development of novel dietary additives to promote intestinal health in farmed animals.
Collapse
Affiliation(s)
- Andrew R Williams
- Department of Veterinary and Animal Sciences, University of Copenhagen, Frederiksberg, Denmark
| | | | - Ling Zhu
- Department of Veterinary and Animal Sciences, University of Copenhagen, Frederiksberg, Denmark
| | | |
Collapse
|
15
|
Prevalence of Giardia duodenalis infection, co-morbidities and associated risk factors in dogs admitted to a veterinary teaching hospital in Israel. Comp Immunol Microbiol Infect Dis 2019; 68:101401. [PMID: 31837597 DOI: 10.1016/j.cimid.2019.101401] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2019] [Revised: 11/22/2019] [Accepted: 11/25/2019] [Indexed: 11/21/2022]
Abstract
Giardia duodenalis causes gastro-intestinal (GI) disease and carries a zoonotic risk. The risk for infection depends on local prevalence and individual immunity, but clinical signs are inconsistent and the role of G. duodenalis in other GI diseases is unknown. The current study aims were to evaluate the prevalence of G. duodenalis in dogs presented to a university veterinary teaching hospital in Israel, investigate risk factors for infection and its associations with other diseases. Fecal samples from 163 dogs were tested for G. duodenalis by an antigen assay (FASTest® Giardia strip). Prevalence of G. duodenalis infection was 11.9 %. It was significantly associated with young age (≤1year) and canine parvoviral enteritis. Housing conditions, signs of GI disease or other systemic diseases were unassociated with infection. In conclusion, G. duodenalis infection prevalence is relatively low in the study population. Young age and diagnosis of co-infections are common, warranting screening for G. duodenalis.
Collapse
|
16
|
Sharba S, Navabi N, Padra M, Persson JA, Quintana-Hayashi MP, Gustafsson JK, Szeponik L, Venkatakrishnan V, Sjöling Å, Nilsson S, Quiding-Järbrink M, Johansson MEV, Linden SK. Interleukin 4 induces rapid mucin transport, increases mucus thickness and quality and decreases colitis and Citrobacter rodentium in contact with epithelial cells. Virulence 2019; 10:97-117. [PMID: 30665337 PMCID: PMC6363059 DOI: 10.1080/21505594.2019.1573050] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Citrobacter rodentium infection is a murine model for pathogenic intestinal Escherichia coli infection. C. rodentium infection causes an initial decrease in mucus layer thickness, followed by an increase during clearance. We aimed to identify the cause of these changes and to utilize this naturally occurring mucus stimulus to decrease pathogen impact and inflammation. We identified that mucin production and speed of transport from Golgi to secretory vesicles at the apical surface increased concomitantly with increased mucus thickness. Of the cytokines differentially expressed during increased mucus thickness, IFN-γ and TNF-α decreased the mucin production and transport speed, whereas IL-4, IL-13, C. rodentium and E. coli enhanced these aspects. IFN-γ and TNF-α treatment in combination with C. rodentium and pathogenic E. coli infection negatively affected mucus parameters in vitro, which was relieved by IL-4 treatment. The effect of IL-4 was more pronounced than that of IL-13, and in wild type mice, only IL-4 was present. Increased expression of Il-4, Il-4-receptor α, Stat6 and Spdef during clearance indicate that this pathway contributes to the increase in mucin production. In vivo IL-4 administration initiated 10 days after infection increased mucus thickness and quality and decreased colitis and pathogen contact with the epithelium. Thus, during clearance of infection, the concomitant increase in IL-4 protects and maintains goblet cell function against the increasing levels of TNF-α and IFN-γ. Furthermore, IL-4 affects intestinal mucus production, pathogen contact with the epithelium and colitis. IL-4 treatment may thus have therapeutic benefits for mucosal healing.
Collapse
Affiliation(s)
- S Sharba
- a Department of Medical Biochemistry and Cell Biology , Sahlgrenska Academy, University of Gothenburg , Gothenburg , Sweden
| | - N Navabi
- a Department of Medical Biochemistry and Cell Biology , Sahlgrenska Academy, University of Gothenburg , Gothenburg , Sweden
| | - M Padra
- a Department of Medical Biochemistry and Cell Biology , Sahlgrenska Academy, University of Gothenburg , Gothenburg , Sweden
| | - J A Persson
- a Department of Medical Biochemistry and Cell Biology , Sahlgrenska Academy, University of Gothenburg , Gothenburg , Sweden
| | - M P Quintana-Hayashi
- a Department of Medical Biochemistry and Cell Biology , Sahlgrenska Academy, University of Gothenburg , Gothenburg , Sweden
| | - J K Gustafsson
- a Department of Medical Biochemistry and Cell Biology , Sahlgrenska Academy, University of Gothenburg , Gothenburg , Sweden
| | - L Szeponik
- b Department of Microbiology and Immunology , Sahlgrenska Academy, University of Gothenburg , Gothenburg , Sweden
| | - V Venkatakrishnan
- a Department of Medical Biochemistry and Cell Biology , Sahlgrenska Academy, University of Gothenburg , Gothenburg , Sweden
| | - Å Sjöling
- c Department of Microbiology, Tumor and Cell Biology , Karolinska Institutet , Stockholm , Sweden
| | - S Nilsson
- d Department of Pathology & Genetics, Sahlgrenska Academy , University of Gothenburg , Sweden.,e Department of Mathematical Sciences , Chalmer University of Technology , Gothenburg , Sweden
| | - M Quiding-Järbrink
- b Department of Microbiology and Immunology , Sahlgrenska Academy, University of Gothenburg , Gothenburg , Sweden
| | - M E V Johansson
- a Department of Medical Biochemistry and Cell Biology , Sahlgrenska Academy, University of Gothenburg , Gothenburg , Sweden
| | - S K Linden
- a Department of Medical Biochemistry and Cell Biology , Sahlgrenska Academy, University of Gothenburg , Gothenburg , Sweden
| |
Collapse
|
17
|
Immune response markers in sera of children infected with Giardia duodenalis AI and AII subassemblages. Immunobiology 2019; 224:595-603. [PMID: 30962033 DOI: 10.1016/j.imbio.2019.04.001] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2019] [Revised: 04/01/2019] [Accepted: 04/01/2019] [Indexed: 12/29/2022]
Abstract
In this study, we evaluated serum markers of immune responses in children infected with G. duodenalis and compared them with the characterized parasite isolates. The reactivity indexes (RI) of IgG (1.503 ± 0.819) and IgA (2.308 ± 1.935) antibodies were significantly higher (P < 0.001) in infected children than in non-infected children. There were also statistically significantly higher serum levels (P < 0.05) of IFN-γ (393.10 ± 983.90 pg/mL) as well as serum (30.03 ± 10.92 μmol/L) and saliva nitric oxid derivatives (NOx) (192.4 ± 151.2 μmol/L) in children infected with G. duodenalis compared to the group of non-parasitized children (127.4 ± 274.30 pg/mL; 25.82 ± 7.74 μmol/L and 122.5 ± 105.90 μmol/L, respectively). Regarding the characterized genetic variants of G. duodenalis and the immune response profiles, no differences were observed in terms of antibody reactivity or levels of serum cytokine and NOx among children infected with AI or AII subassemblages. The elevated levels of IFN-γ and NOx indicate that G. duodenalis intestinal infection in humans induces a cellular immune response detectable at the systemic level. Moreover, no significant differences in the antibody reactivity profile or the cytokine and NOx production in the sera of children infected with AI or AII G. duodenalis variants were observed, suggesting that subtypes of the parasite do not influence the immune response profile.
Collapse
|
18
|
Liu Y, Ji P. Dietary Factors in Prevention of Pediatric Escherichia coli Infection: A Model Using Domestic Piglets. ILAR J 2018; 59:338-351. [PMID: 31095688 DOI: 10.1093/ilar/ilz005] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2017] [Revised: 02/04/2019] [Indexed: 01/16/2023] Open
Abstract
Enterotoxigenic Escherichia coli (ETEC) is the major etiological agent causing acute watery diarrhea that is most frequently seen in young children in lower-income countries. The duration of diarrheal symptom may be shortened by antibiotic treatment, but ETEC is relative refractory to common antibiotics. Burgeoning evidence suggests bioactive components that naturally occur in human milk (e.g., lysozyme and oligosaccharides) and plants (e.g., nondigestible carbohydrates and phytochemicals) contain antimicrobial functions are promising preventive measures to control ETEC infection. Although the exact protective mechanisms may vary for each compound and are still not completely understood, they generally act to (1) competitively inhibit the binding of pathogenic bacteria and toxins to gut epithelium; (2) directly kill pathogens; and (3) stimulate and/or enhance host mucosal and systemic immune defense against pathogenic microorganisms. An appropriate ETEC-challenge animal model is critical to evaluate the effect and unveil the mechanism of bioactive compounds in prevention of enteric infection. Despite wide application in biomedical research, rodents do not usually manifest typical clinical signs of enteric infections. The remarkable differences in digestive physiology, immune response, and gut microbiota between rodents and human beings necessitate the use of alternative animal models. Pigs are closely related to humans in terms of genomes, physiology, anatomy of gastrointestinal tracts, digestive enzymes, components of immune system, and gut microbiota. Like human infants and young children, nursing and nursery piglets are more susceptible to ETEC infection and reproduce the clinical signs as observed in humans. Hence, the ETEC-challenge piglet represents a valuable translational model to study pathogenesis and evaluate dietary factors (e.g., milk bioactive compounds, nondigestible carbohydrates, and phytochemicals) as preventive measures for ETEC infection in pediatrics.
Collapse
Affiliation(s)
| | - Peng Ji
- Department of Nutrition, University of California, Davis, California
| |
Collapse
|
19
|
Anti-infectious properties of the probiotic Saccharomyces cerevisiae CNCM I-3856 on enterotoxigenic E. coli (ETEC) strain H10407. Appl Microbiol Biotechnol 2018; 102:6175-6189. [DOI: 10.1007/s00253-018-9053-y] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2017] [Revised: 04/25/2018] [Accepted: 04/26/2018] [Indexed: 12/15/2022]
|
20
|
Cytokines, Antibodies, and Histopathological Profiles during Giardia Infection and Variant-Specific Surface Protein-Based Vaccination. Infect Immun 2018; 86:IAI.00773-17. [PMID: 29555679 DOI: 10.1128/iai.00773-17] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2017] [Accepted: 03/10/2018] [Indexed: 12/13/2022] Open
Abstract
Giardiasis is one of the most common human intestinal diseases worldwide. Several experimental animal models have been used to evaluate Giardia infections, with gerbils (Meriones unguiculatus) being the most valuable model due to their high susceptibility to Giardia infection, abundant shedding of cysts, and pathophysiological alterations and signs of disease similar to those observed in humans. Here, we report cytokine and antibody profiles both during the course of Giardia infection in gerbils and after immunization with a novel oral vaccine comprising a mixture of purified variant-specific surface proteins (VSPs). Transcript levels of representative cytokines of different immune profiles as well as macro- and microtissue alterations were assessed in Peyer's patches, mesenteric lymph nodes, and spleens. During infection, cytokine responses showed a biphasic profile: an early induction of Th1 (gamma interferon [IFN-γ], interleukin-1β [IL-1β], IL-6, and tumor necrosis factor [TNF]), Th17 (IL-17), and Th2 (IL-4) cytokines, together with intestinal alterations typical of inflammation, followed by a shift toward a predominant Th2 (IL-5) response, likely associated with a counterregulatory mechanism. Conversely, immunization with an oral vaccine comprising the entire repertoire of VSPs specifically showed high levels of IL-17, IL-6, IL-4, and IL-5, without obvious signs of inflammation. Both immunized and infected animals developed local (intestinal secretory IgA [S-IgA]) and systemic (serum IgG) humoral immune responses against VSPs; however, only infected animals showed evident signs of giardiasis. This is the first comprehensive report of cytokine expression and anti-Giardia antibody production during infection and VSP vaccination in gerbils, a reliable model of the human disease.
Collapse
|
21
|
Noel G, Doucet M, Nataro JP, Kaper JB, Zachos NC, Pasetti MF. Enterotoxigenic Escherichia coli is phagocytosed by macrophages underlying villus-like intestinal epithelial cells: modeling ex vivo innate immune defenses of the human gut. Gut Microbes 2017; 9:0. [PMID: 29087765 PMCID: PMC6219640 DOI: 10.1080/19490976.2017.1398871] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/18/2017] [Revised: 10/24/2017] [Accepted: 10/25/2017] [Indexed: 02/07/2023] Open
Abstract
There is a paucity of information on diarrheagenic enterotoxigenic Escherichia coli (ETEC)'s interaction with innate immune cells, in part due to the lack of reliable models that recapitulate infection in human gut. In a recent publication, we described the development of an ex vivo enteroid-macrophage co-culture model using human primary cells. We reported that macrophages residing underneath the epithelial monolayer acquired "resident macrophage" phenotype characterized by lower production of inflammatory cytokines and strong phagocytic activity. These macrophages extended projections across the epithelium, which captured ETEC applied to the apical side of the epithelium and reduced luminal bacterial load. Additional evidence presented in this addendum confirms these findings and further demonstrates that macrophage adaptation occurs regardless of the stage of differentiation of epithelial cells, and that ETEC uptake arises rapidly after infection. The enteroid-macrophage co-culture represents a novel and relevant tool to study host-cell interactions and pathogenesis of enteric infections in humans.
Collapse
Affiliation(s)
- Gaelle Noel
- Center for Vaccine Development, Department of Microbiology and Immunology. University of Maryland School of Medicine, Baltimore, MD, USA
| | - Michele Doucet
- Department of Medicine, Division of Gastroenterology and Hepatology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - James P. Nataro
- Department of Pediatrics, University of Virginia School of Medicine, Charlottesville, VA, USA
| | - James B. Kaper
- Department of Microbiology and Immunology. University of Maryland School of Medicine, Baltimore, MD, USA
| | - Nicholas C. Zachos
- Department of Medicine, Division of Gastroenterology and Hepatology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Marcela F. Pasetti
- Center for Vaccine Development, Department of Microbiology and Immunology. University of Maryland School of Medicine, Baltimore, MD, USA
| |
Collapse
|
22
|
Abstract
PURPOSE OF REVIEW Giardia is a common intestinal parasite worldwide, and infection can be associated with clear and sometimes persistent symptomatology. However, in children in high-prevalence settings, it is not associated with or is perhaps even protective against acute diarrhea, and the association with long-term outcomes has been difficult to discern. RECENT FINDINGS Recent studies have made progress in helping us disentangle this apparent paradox. First, prospective, well-characterized cohort studies have added to the data on the association between Giardia and diarrhea in these settings and have further characterized associations between Giardia infection and nutrition, gut function, and growth. Second, animal models have further characterized the host response to Giardia and helped elucidate mechanisms by which Giardia could impair child development. Finally, new work has shed light on the heterogeneity of human Giardia strains, which may both explain discrepant findings in the literature and help guide higher-resolution analyses of this pathogen in the future. SUMMARY The true clinical impact of endemic pediatric giardiasis remains unclear, but recent prospective studies have confirmed a high prevalence of persistent, subclinical Giardia infections and associated growth shortfalls. Integrating how nutritional, microbial, metabolic, and pathogen-strain variables influence these outcomes could sharpen delineations between pathogenic and potentially beneficial attributes of this enigmatic parasite.
Collapse
|
23
|
Mooney D, Edgar D, Einarsson G, Downey D, Elborn S, Tunney M. Chronic lung disease in common variable immune deficiency (CVID): A pathophysiological role for microbial and non-B cell immune factors. Crit Rev Microbiol 2017; 43:508-519. [PMID: 28068853 DOI: 10.1080/1040841x.2016.1268568] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
One of the most common and most severe forms of primary antibody deficiency encountered in the clinical setting is a heterogeneous group of syndromes termed common variable immune deficiency (CVID). This disorder is characterized by reduced immunoglobulin production and increased susceptibility to infection, particularly of the respiratory tract. Infection and subsequent immunological/inflammatory processes may contribute to the development of pulmonary complications such as bronchiectasis and interstitial lung disease. Immunoglobulin replacement and/or antibiotic therapy, to prevent infection, are routinely prescribed treatments. However, chronic lung disease, the major cause of morbidity and mortality in this patient cohort, may still progress. This clinical progression suggests that pathogens recalcitrant to currently prescribed treatments and other immunological defects may be contributing to the development of pulmonary disease. This review describes the potential role of microbiological and non-B cell immunological factors, including T-cells, neutrophils, complement, toll like receptors, and antimicrobial peptides, in the pathogenicity of chronic lung disease in patients with CVID.
Collapse
Affiliation(s)
- Denver Mooney
- a Halo Research Group, Queen's University Belfast , Belfast , United Kingdom
- b Centre for Experimental Medicine, School of Medicine , Dentistry and Biomedical Sciences. Queen's University Belfast , Belfast , United Kingdom
| | - David Edgar
- c T he Royal Hospitals, Belfast Health and Social Care Trust , Regional Immunology Service , Belfast , United Kingdom
| | - Gisli Einarsson
- a Halo Research Group, Queen's University Belfast , Belfast , United Kingdom
- b Centre for Experimental Medicine, School of Medicine , Dentistry and Biomedical Sciences. Queen's University Belfast , Belfast , United Kingdom
| | - Damian Downey
- d Belfast City Hospital, Belfast Health and Social Care Trust , Regional Respiratory Centre , Belfast , United Kingdom
| | - Stuart Elborn
- a Halo Research Group, Queen's University Belfast , Belfast , United Kingdom
- b Centre for Experimental Medicine, School of Medicine , Dentistry and Biomedical Sciences. Queen's University Belfast , Belfast , United Kingdom
| | - Michael Tunney
- a Halo Research Group, Queen's University Belfast , Belfast , United Kingdom
- e School of Pharmacy , Queen's University Belfast , Belfast , United Kingdom
| |
Collapse
|
24
|
Bartelt LA, Bolick DT, Mayneris-Perxachs J, Kolling GL, Medlock GL, Zaenker EI, Donowitz J, Thomas-Beckett RV, Rogala A, Carroll IM, Singer SM, Papin J, Swann JR, Guerrant RL. Cross-modulation of pathogen-specific pathways enhances malnutrition during enteric co-infection with Giardia lamblia and enteroaggregative Escherichia coli. PLoS Pathog 2017; 13:e1006471. [PMID: 28750066 PMCID: PMC5549954 DOI: 10.1371/journal.ppat.1006471] [Citation(s) in RCA: 60] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2017] [Accepted: 06/14/2017] [Indexed: 12/17/2022] Open
Abstract
Diverse enteropathogen exposures associate with childhood malnutrition. To
elucidate mechanistic pathways whereby enteric microbes interact during
malnutrition, we used protein deficiency in mice to develop a new model of
co-enteropathogen enteropathy. Focusing on common enteropathogens in
malnourished children, Giardia lamblia and enteroaggregative
Escherichia coli (EAEC), we provide new insights into
intersecting pathogen-specific mechanisms that enhance malnutrition. We show for
the first time that during protein malnutrition, the intestinal microbiota
permits persistent Giardia colonization and simultaneously
contributes to growth impairment. Despite signals of intestinal injury, such as
IL1α, Giardia-infected mice lack pro-inflammatory intestinal
responses, similar to endemic pediatric Giardia infections.
Rather, Giardia perturbs microbial host co-metabolites of
proteolysis during growth impairment, whereas host nicotinamide utilization
adaptations that correspond with growth recovery increase. EAEC promotes
intestinal inflammation and markers of myeloid cell activation. During
co-infection, intestinal inflammatory signaling and cellular recruitment
responses to EAEC are preserved together with a
Giardia-mediated diminishment in myeloid cell activation.
Conversely, EAEC extinguishes markers of host energy expenditure regulatory
responses to Giardia, as host metabolic adaptations appear
exhausted. Integrating immunologic and metabolic profiles during co-pathogen
infection and malnutrition, we develop a working mechanistic model of how
cumulative diet-induced and pathogen-triggered microbial perturbations result in
an increasingly wasted host. Malnourished children are exposed to multiple sequential, and oftentimes,
persistent enteropathogens. Intestinal microbial disruption and inflammation are
known to contribute to the pathogenesis of malnutrition, but how co-pathogens
interact with each other, with the resident microbiota, or with the host to
alter these pathways is unknown. Using a new model of enteric co-infection with
Giardia lamblia and enteroaggregative Escherichia
coli in mice fed a protein deficient diet, we identify host growth
and intestinal immune responses that are differentially mediated by
pathogen-microbe interactions, including parasite-mediated changes in intestinal
microbial host co-metabolism, and altered immune responses during co-infection.
Our data model how early life cumulative enteropathogen exposures progressively
disrupt intestinal immunity and host metabolism during crucial developmental
periods. Furthermore, studies in this co-infection model reveal new insights
into environmental and microbial determinants of pathogenicity for presently
common, but poorly understood enteropathogens like Giardia
lamblia, that may not conform to existing paradigms of microbial
pathogenesis based on single pathogen-designed models.
Collapse
Affiliation(s)
- Luther A. Bartelt
- Division of Infectious Diseases, Department of Medicine, University of
North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of
America
- Center for Gastrointestinal Biology and Disease, Department of Medicine,
University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United
States of America
- * E-mail:
| | - David T. Bolick
- Division of Infectious Diseases and International Health, Department of
Medicine, University of Virginia, Charlottesville, Virginia, United States of
America
| | - Jordi Mayneris-Perxachs
- Division of Computational and Systems Medicine, Department of Surgery and
Cancer, Imperial College London, United Kingdom
| | - Glynis L. Kolling
- Division of Infectious Diseases and International Health, Department of
Medicine, University of Virginia, Charlottesville, Virginia, United States of
America
| | - Gregory L. Medlock
- Department of Biomedical Engineering, University of Virginia,
Charlottesville, Virginia, United States of America
| | - Edna I. Zaenker
- Division of Infectious Diseases and International Health, Department of
Medicine, University of Virginia, Charlottesville, Virginia, United States of
America
| | - Jeffery Donowitz
- Division of Pediatric Infectious Diseases, Children’s Hospital of
Richmond at Virginia Commonwealth University, Richmond, Virginia, United States
of America
| | - Rose Viguna Thomas-Beckett
- Division of Infectious Diseases, Department of Medicine, University of
North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of
America
| | - Allison Rogala
- Center for Gastrointestinal Biology and Disease, Department of Medicine,
University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United
States of America
| | - Ian M. Carroll
- Center for Gastrointestinal Biology and Disease, Department of Medicine,
University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United
States of America
| | - Steven M. Singer
- Department of Biology, Georgetown University, Washington, DC, United
States of America
| | - Jason Papin
- Department of Biomedical Engineering, University of Virginia,
Charlottesville, Virginia, United States of America
| | - Jonathan R. Swann
- Division of Computational and Systems Medicine, Department of Surgery and
Cancer, Imperial College London, United Kingdom
| | - Richard L. Guerrant
- Division of Infectious Diseases and International Health, Department of
Medicine, University of Virginia, Charlottesville, Virginia, United States of
America
| |
Collapse
|
25
|
Papadopoulos GA, Poutahidis T, Tallarico N, Hardas A, Teliousis K, Arsenos G, Fortomaris PD. Dietary supplementation of encapsulated organic acids enhances performance and modulates immune regulation and morphology of jejunal mucosa in piglets. Res Vet Sci 2017; 115:174-182. [PMID: 28458106 DOI: 10.1016/j.rvsc.2017.04.014] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2016] [Revised: 03/23/2017] [Accepted: 04/21/2017] [Indexed: 01/22/2023]
Abstract
The aim of the study was to test two encapsulated regimens containing organic acids and/or zinc oxide (ZnO) on weaned piglet performance and jejunal mucosa morphology and immunity. For that, weaned piglets were allocated to treatments including control, supplemented with encapsulated organic acids (ACID group), and supplemented with organic acids and ZnO, both encapsulated (ACIDplus group). Antibiotics were used at similar concentrations in all groups during the first two weeks, but withdrawn from the ACIDplus group during the last three weeks of the experiment. ZnO was given with feed in the Control and ACID groups only during the first two weeks. The experimental period lasted 5 weeks. Piglets from the ACID group exhibited higher average daily gain compared to other groups during the last 3 weeks of the experiment (P<0.05). The ACIDplus group performed similarly with controls. The mucosal height of jejunum was higher in both ACID (P<0.01) and ACIDplus groups compared to controls (P<0.05). Immunohistochemical analysis of jejunal mucosa, showed higher numbers of neutrophils in ACID and ACIDplus groups compared to controls (P<0.01 and P<0.001, respectively). Treatments had the opposite effect on mucosal regulatory T-cells (Foxp3-positive cells) in jejunum, being higher (P<0.001) in control group compared to ACID and ACIDplus groups. The number of CD3-positive cells was higher (P<0.05) in the ACIDplus and control groups compared to the ACID group. In conclusion, the encapsulated products used had beneficial effects on growth performance coexisting with improvements on jejunal histomorphology and modulation of mucosal immunity.
Collapse
Affiliation(s)
- Georgios A Papadopoulos
- Laboratory of Animal Husbandry, Faculty of Veterinary Medicine, School of Health Sciences, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece
| | - Theofilos Poutahidis
- Laboratory of Pathology, Faculty of Veterinary Medicine, School of Health Sciences, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece
| | | | - Alexandros Hardas
- Laboratory of Pathology, Faculty of Veterinary Medicine, School of Health Sciences, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece
| | - Konstantinos Teliousis
- Laboratory of Pathology, Faculty of Veterinary Medicine, School of Health Sciences, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece
| | - Georgios Arsenos
- Laboratory of Animal Husbandry, Faculty of Veterinary Medicine, School of Health Sciences, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece
| | - Paschalis D Fortomaris
- Laboratory of Animal Husbandry, Faculty of Veterinary Medicine, School of Health Sciences, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece.
| |
Collapse
|
26
|
Li H, Zhang L, Chen L, Zhu Q, Wang W, Qiao J. Lactobacillus acidophilus alleviates the inflammatory response to enterotoxigenic Escherichia coli K88 via inhibition of the NF-κB and p38 mitogen-activated protein kinase signaling pathways in piglets. BMC Microbiol 2016; 16:273. [PMID: 27832756 PMCID: PMC5105324 DOI: 10.1186/s12866-016-0862-9] [Citation(s) in RCA: 56] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2016] [Accepted: 10/11/2016] [Indexed: 12/14/2022] Open
Abstract
Background A newly isolated L. acidophilus strain has been reported to have potential anti-inflammatory activities against lipopolysaccharide (LPS) challenge in piglet, while the details of the related inflammatory responses are limited. Here we aimed to analysis the ability of L. acidophilus to regulate inflammatory responses and to elucidate the mechanisms involved in its anti-inflammatory activity. Results The ETEC (enterotoxigenic Escherichia coli) K88-induced up-regulations of IL-1β, IL-8 and TNF-α were obviously inhibited by L. acidophilus while IL-10 was significantly increased. Moreover, L. acidophilus down-regulated pattern recognition receptors TLR (Toll-like receptor) 2 and TLR4 expression in both spleen and mesenteric lymph nodes of ETEC-challenged piglets, in accompanied with the reduced phosphorylation levels of nuclear factor kappa B (NF-κB) p65 and mitogen-activated protein kinase (MAPK) p38 as well in spleen of ETEC-infected piglets. Furthermore, L.acidophilus significantly increased the expression of the negative regulators of TLRs signaling, including Tollip, IRAK-M, A20 and Bcl-3 in spleen of ETEC-challenged piglets. Conclusions Our findings suggested that L. acidophilus regulated inflammatory response to ETEC via impairing both NF-κB and MAPK signaling pathways in piglets.
Collapse
Affiliation(s)
- Haihua Li
- Tianjin Institute of Animal Husbandry and Veterinary Medicine, Tianjin, China
| | - Lei Zhang
- Tianjin Institute of Animal Husbandry and Veterinary Medicine, Tianjin, China
| | - Longbin Chen
- Tianjin Institute of Animal Husbandry and Veterinary Medicine, Tianjin, China
| | - Qi Zhu
- Tianjin Institute of Animal Husbandry and Veterinary Medicine, Tianjin, China
| | - Wenjie Wang
- Tianjin Institute of Animal Husbandry and Veterinary Medicine, Tianjin, China
| | - Jiayun Qiao
- Tianjin Institute of Animal Husbandry and Veterinary Medicine, Tianjin, China.
| |
Collapse
|
27
|
Azimirad M, Alebouyeh M, Naji T. Inhibition of Lipopolysaccharide-Induced Interleukin 8 in Human Adenocarcinoma Cell Line HT-29 by Spore Probiotics: B. coagulans and B. subtilis (natto). Probiotics Antimicrob Proteins 2016; 9:56-63. [DOI: 10.1007/s12602-016-9234-x] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
|
28
|
Ahmed AMS, Soares Magalhaes RJ, Long KZ, Ahmed T, Alam MA, Hossain MI, Islam MM, Mahfuz M, Mondal D, Haque R, Mamun AA. Association of vitamin D status with incidence of enterotoxigenic, enteropathogenic and enteroaggregative Escherichia coli diarrhoea in children of urban Bangladesh. Trop Med Int Health 2016; 21:973-984. [PMID: 27253178 DOI: 10.1111/tmi.12731] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
OBJECTIVE To evaluate the association between vitamin D status and diarrhoeal episodes by enterotoxigenic (ETEC), enteropathogenic (EPEC) and enteroaggregative (EAEC) E. coli in underweight and normal-weight children aged 6-24 months in urban Bangladesh. METHODS Cohorts of 446 normal-weight and 466 underweight children were tested separately for ETEC, EPEC and EAEC from diarrhoeal stool samples collected during 5 months of follow-up while considering vitamin D status at enrolment as the exposure. Cox proportional hazards models with unordered failure events of the same type were used to determine diarrhoeal risk factors after adjusting for sociodemographic and concurrent micronutrient status. RESULTS Vitamin D status was not independently associated with the risk of incidence of ETEC, EPEC and EAEC diarrhoea in underweight children, but moderate-to-severe retinol deficiency was associated with reduced risk for EPEC diarrhoea upon adjustment. Among normal-weight children, insufficient vitamin D status and moderate-to-severe retinol deficiency were independently associated with 44% and 38% reduced risk of incidence of EAEC diarrhoea, respectively. These children were at higher risk of ETEC diarrhoea with vitamin D deficiency status when adjusted for micronutrient status only. CONCLUSION This study demonstrates for the first time that normal-weight children with insufficient vitamin D status have a reduced risk of EAEC diarrhoea than children with sufficient status. Moderate-to-severe deficiency of serum retinol is associated with reduced risk of EPEC and EAEC diarrhoea in underweight and normal-weight children.
Collapse
Affiliation(s)
- A M S Ahmed
- School of Public Health, The University of Queensland, Brisbane, Qld, Australia.,Nutrition and Clinical Services Division, International Centre for Diarrhoeal Disease Research, Bangladesh, Dhaka, Bangladesh
| | - R J Soares Magalhaes
- School of Veterinary Science, The University of Queensland, Gatton, Qld, Australia.,Child Health Research Centre, The University of Queensland, Brisbane, Qld, Australia
| | - K Z Long
- Child Health Research Centre, The University of Queensland, Brisbane, Qld, Australia.,Department of Epidemiology and Public Health, Swiss Tropical and Public Health Institute, Basel, Switzerland
| | - T Ahmed
- Nutrition and Clinical Services Division, International Centre for Diarrhoeal Disease Research, Bangladesh, Dhaka, Bangladesh
| | - Md A Alam
- Nutrition and Clinical Services Division, International Centre for Diarrhoeal Disease Research, Bangladesh, Dhaka, Bangladesh
| | - Md I Hossain
- Nutrition and Clinical Services Division, International Centre for Diarrhoeal Disease Research, Bangladesh, Dhaka, Bangladesh
| | - Md M Islam
- Nutrition and Clinical Services Division, International Centre for Diarrhoeal Disease Research, Bangladesh, Dhaka, Bangladesh
| | - M Mahfuz
- Nutrition and Clinical Services Division, International Centre for Diarrhoeal Disease Research, Bangladesh, Dhaka, Bangladesh
| | - D Mondal
- Nutrition and Clinical Services Division, International Centre for Diarrhoeal Disease Research, Bangladesh, Dhaka, Bangladesh
| | - R Haque
- Nutrition and Clinical Services Division, International Centre for Diarrhoeal Disease Research, Bangladesh, Dhaka, Bangladesh
| | - A A Mamun
- School of Public Health, The University of Queensland, Brisbane, Qld, Australia.,Nutrition and Clinical Services Division, International Centre for Diarrhoeal Disease Research, Bangladesh, Dhaka, Bangladesh
| |
Collapse
|
29
|
Srinivasan P, Lawa HR, Rosado JL, Al Mamun A, Khatun M, Santos JI, Utzinger J, Long KZ. Household and personal factors are sources of heterogenity in intestinal parasite clearance among Mexican children 6-15 months of age supplemented with vitamin A and zinc. Acta Trop 2016; 156:48-56. [PMID: 26772449 DOI: 10.1016/j.actatropica.2015.12.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2015] [Revised: 12/08/2015] [Accepted: 12/11/2015] [Indexed: 11/30/2022]
Abstract
A randomised, double-blind, placebo-controlled trial was carried out among Mexico children aged 6-15 months to determine how household characteristics modify vitamin A and zinc supplementation efficacy on Ascaris lumbricoides, Giardia intestinalis and Entamoeba histolytica/E. dispar infection durations. Children assigned to receive vitamin A every 2 months, a daily zinc supplement, a combined vitamin A-zinc supplement or a placebo were followed for 1 year. Parametric hazard models were fit to infection durations stratified by personal and household factors. Children supplemented with vitamin A and zinc combined from households lacking piped water and children in all three treatment arms from households with dirt floors had longer G. intestinalis and A. lumbricoides infection durations than their counterparts, respectively. Shorter E. histolytica/E.dispar durations were found among zinc-supplemented children of mothers who had <6 years of education and no indoor bathrooms. Heterogeneity in supplementation efficacy among children may reflect differences in exposure risk and baseline immune responses.
Collapse
Affiliation(s)
| | | | - Jorge L Rosado
- School of Natural Sciences, Universidad Autónoma de Querétaro, Querétaro, Mexico
| | - Abdullah Al Mamun
- University of Queensland School of Population Health, Herston, Qld, Australia
| | - Mohsina Khatun
- QIMR Berghofer Medical Research Institute, Herston, Australia
| | - José I Santos
- School of Medicine, National Autonomous University of Mexico, Mexico City, Mexico
| | - Jürg Utzinger
- Department of Epidemiology and Public Health, Swiss Tropical and Public Health Institute, Basel, Switzerland; University of Basel, Basel, Switzerland
| | - Kurt Z Long
- Department of Epidemiology and Public Health, Swiss Tropical and Public Health Institute, Basel, Switzerland; University of Basel, Basel, Switzerland.
| |
Collapse
|
30
|
Lactobacillus acidophilus modulates inflammatory activity by regulating the TLR4 and NF-κB expression in porcine peripheral blood mononuclear cells after lipopolysaccharide challenge. Br J Nutr 2016; 115:567-75. [DOI: 10.1017/s0007114515004857] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
AbstractA total of forty weaned pigs ((Landrace×Yorkshire)×Duroc) were used to evaluate the effects of Lactobacillus acidophilus on inflammatory activity after lipopolysaccharide (LPS) challenge. Experimental treatments were as follows: (T1) control diet+saline challenge; (T2) control diet with 0·1 % L. acidophilus+saline challenge; (T3) control diet+LPS challenge; and (T4) control diet with 0·1 % L. acidophilus+LPS challenge. On d-14, piglets were challenged with saline (T1 and T2) or LPS (T3 and T4). Blood samples were obtained at 0, 2, 4, 6 and 12 h after being challenged and analysed for immune cell cytokine production and gene expression pattern. The L. acidophilus treatment increased the average daily weight gain (ADWG) and average daily feed intake (ADFI) compared with the control diet. With the control diet, the LPS challenge (T3) increased the number of immune cells and expression of TNF-α and IL-6 compared with the saline challenge (T1). Whereas with the saline challenge L. acidophilus treatment (T2) increased the number of leucocytes and CD4 compared with the control diet (T1), with the LPS challenge L. acidophilus treatment (T4) decreased the number of leucocytes, lymphocytes, CD4+ and CD8+ and expression of TNF-α and IL-6 compared with the control diet (T3). L. acidophilus treatment decreased the expression of TRL4 and NF-κB in peripheral blood mononuclear cells (PBMC) after LPS challenge, which leads to inhibition of TNF-α, IFN-γ, IL-6, IL-8 and IL1B1 and to induction of IL-4 and IL-10. We suggested that L. acidophilus improved ADWG and ADFI and protected against LPS-induced inflammatory responses by regulating TLR4 and NF-κB expression in porcine PBMC.
Collapse
|
31
|
Biomarkers of Gastrointestinal Host Responses to Microbial Infections. Mol Microbiol 2016. [DOI: 10.1128/9781555819071.ch46] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
32
|
|
33
|
Šlapeta J, Dowd SE, Alanazi AD, Westman ME, Brown GK. Differences in the faecal microbiome of non-diarrhoeic clinically healthy dogs and cats associated with Giardia duodenalis infection: impact of hookworms and coccidia. Int J Parasitol 2015; 45:585-94. [DOI: 10.1016/j.ijpara.2015.04.001] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2015] [Revised: 04/07/2015] [Accepted: 04/09/2015] [Indexed: 02/07/2023]
|
34
|
Lopez-Romero G, Quintero J, Astiazarán-García H, Velazquez C. Host defences againstGiardia lamblia. Parasite Immunol 2015; 37:394-406. [DOI: 10.1111/pim.12210] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2014] [Accepted: 06/08/2015] [Indexed: 02/06/2023]
Affiliation(s)
- G. Lopez-Romero
- Coordinación de Nutrición; Centro de Investigación en Alimentación y Desarrollo A.C.; Hermosillo Sonora México
| | - J. Quintero
- Department of Chemistry-Biology; University of Sonora; Hermosillo Sonora México
| | - H. Astiazarán-García
- Coordinación de Nutrición; Centro de Investigación en Alimentación y Desarrollo A.C.; Hermosillo Sonora México
| | - C. Velazquez
- Department of Chemistry-Biology; University of Sonora; Hermosillo Sonora México
| |
Collapse
|
35
|
Bartelt LA, Sartor RB. Advances in understanding Giardia: determinants and mechanisms of chronic sequelae. F1000PRIME REPORTS 2015; 7:62. [PMID: 26097735 PMCID: PMC4447054 DOI: 10.12703/p7-62] [Citation(s) in RCA: 78] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Giardia lamblia is a flagellated protozoan that is the most common cause of intestinal parasitic infection in children living in resource-limited settings. The pathogenicity of Giardia has been debated since the parasite was first identified, and clinical outcomes vary across studies. Among recent perplexing findings are diametrically opposed associations between Giardia and acute versus persistent diarrhea and a poorly understood potential for long-term sequelae, including impaired child growth and cognitive development. The mechanisms driving these protean clinical outcomes remain elusive, but recent advances suggest that variability in Giardia strains, host nutritional status, the composition of microbiota, co-infecting enteropathogens, host genetically determined mucosal immune responses, and immune modulation by Giardia are all relevant factors influencing disease manifestations after Giardia infection.
Collapse
Affiliation(s)
- Luther A. Bartelt
- Division of Infectious Diseases and International Health, University of VirginiaBox 801340, Charlottesville, VA 22908USA
| | - R. Balfour Sartor
- Division of Gastroenterology, Center for Gastrointestinal Biology and Disease, University of North Carolina at Chapel HillCampus Box 7032, Chapel Hill, NC 27599-7032USA
| |
Collapse
|
36
|
Colostrum whey down-regulates the expression of early and late inflammatory response genes induced byEscherichia coliandSalmonella entericaTyphimurium components in intestinal epithelial cells. Br J Nutr 2014; 113:200-11. [DOI: 10.1017/s0007114514003481] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Pathogenic invasion byEscherichia coliandSalmonellaeremains a constant threat to the integrity of the intestinal epithelium and can rapidly induce inflammatory responses. At birth, colostrum consumption exerts numerous beneficial effects on the properties of intestinal epithelial cells and protects the gastrointestinal tract of newborns from pathogenic invasion. The present study aimed to investigate the effect of colostrum on the early and late inflammatory responses induced by pathogens. The short-term (2 h) and long-term (24 h) effects of exposure to heat-killed (HK)E. coliandSalmonella entericaTyphimurium on gene expression in the porcine intestinal epithelial cell (IPEC-J2) model were first evaluated by microarray and quantitative PCR analyses. Luciferase assays were performed using a NF-κB-luc reporter construct to investigate the effect of colostrum whey treatment on the activation of NF-κB induced by HK bacteria. Luciferase assays were also performed using NF-κB-luc, IL-8-luc and IL-6-luc reporter constructs in human colon adenocarcinoma Caco-2/15 cells exposed to dose–response stimulations with HK bacteria and colostrum whey. Bovine colostrum whey treatment decreased the expression of early and late inflammatory genes induced by HK bacteria in IPEC-J2, as well as the transcriptional activation of NF-κB-luc induced by HK bacteria. Unlike that with colostrum whey, treatment with other milk fractions failed to decrease the activation of NF-κB-luc induced by HK bacteria. Lastly, the reduction of the HK bacteria-induced activation of NF-κB-luc, IL-8-luc and IL-6-luc by colostrum whey was dose dependent. The results of the present study indicate that bovine colostrum may protect and preserve the integrity of the intestinal mucosal barrier in the host by controlling the expression levels of early and late inflammatory genes following invasion by enteric pathogens.
Collapse
|
37
|
Tomova A, Husarova V, Lakatosova S, Bakos J, Vlkova B, Babinska K, Ostatnikova D. Gastrointestinal microbiota in children with autism in Slovakia. Physiol Behav 2014; 138:179-87. [PMID: 25446201 DOI: 10.1016/j.physbeh.2014.10.033] [Citation(s) in RCA: 381] [Impact Index Per Article: 38.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2014] [Revised: 10/28/2014] [Accepted: 10/30/2014] [Indexed: 02/07/2023]
Abstract
Development of Autism Spectrum Disorders (ASD), including autism, is based on a combination of genetic predisposition and environmental factors. Recent data propose the etiopathogenetic role of intestinal microflora in autism. The aim of this study was to elucidate changes in fecal microbiota in children with autism and determine its role in the development of often present gastrointestinal (GI) disorders and possibly other manifestations of autism in Slovakia. The fecal microflora of 10 children with autism, 9 siblings and 10 healthy children was investigated by real-time PCR. The fecal microbiota of autistic children showed a significant decrease of the Bacteroidetes/Firmicutes ratio and elevation of the amount of Lactobacillus spp. Our results also showed a trend in the incidence of elevated Desulfovibrio spp. in children with autism reaffirmed by a very strong association of the amount of Desulfovibrio spp. with the severity of autism in the Autism Diagnostic Interview (ADI) restricted/repetitive behavior subscale score. The participants in our study demonstrated strong positive correlation of autism severity with the severity of GI dysfunction. Probiotic diet supplementation normalized the Bacteroidetes/Firmicutes ratio, Desulfovibrio spp. and the amount of Bifidobacterium spp. in feces of autistic children. We did not find any correlation between plasma levels of oxytocin, testosterone, DHEA-S and fecal microbiota, which would suggest their combined influence on autism development. This pilot study suggests the role of gut microbiota in autism as a part of the "gut-brain" axis and it is a basis for further investigation of the combined effect of microbial, genetic, and hormonal changes for development and clinical manifestation of autism.
Collapse
Affiliation(s)
| | | | | | - Jan Bakos
- Institute of Physiology, Comenius University, Bratislava, Slovakia
| | - Barbora Vlkova
- Institute of Molecular Biomedicine, Comenius University, Bratislava, Slovakia
| | | | | |
Collapse
|
38
|
Abstract
ABSTRACT
The inflammatory response is an integral part of host defense against enterohemorrhagic
Escherichia coli
(EHEC) infection and also contributes to disease pathology. In this article we explore the factors leading to inflammation during EHEC infection and the mechanisms EHEC and other attaching and effacing (A/E) pathogens have evolved to suppress inflammatory signaling. EHEC stimulates an inflammatory response in the intestine through host recognition of bacterial components such as flagellin and lipopolysaccharide. In addition, the activity of Shiga toxin and some type III secretion system effectors leads to increased tissue inflammation. Various infection models of EHEC and other A/E pathogens have revealed many of the immune factors that mediate this response. In particular, the outcome of infection is greatly influenced by the ability of an infected epithelial cell to mount an effective host inflammatory response. The inflammatory response of infected enterocytes is counterbalanced by the activity of type III secretion system effectors such as NleE and NleC that modify and inhibit components of the signaling pathways that lead to proinflammatory cytokine production. Overall, A/E pathogens have taught us that innate mucosal immune responses in the gastrointestinal tract during infection with A/E pathogens are highly complex and ultimate clearance of the pathogen depends on multiple factors, including inflammatory mediators, bacterial burden, and the function and integrity of resident intestinal epithelial cells.
Collapse
|
39
|
Modulatory effects of vasoactive intestinal peptide on intestinal mucosal immunity and microbial community of weaned piglets challenged by an enterotoxigenic Escherichia coli (K88). PLoS One 2014; 9:e104183. [PMID: 25101851 PMCID: PMC4125177 DOI: 10.1371/journal.pone.0104183] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2014] [Accepted: 07/11/2014] [Indexed: 01/08/2023] Open
Abstract
Toll-like receptors (TLRs) recognize microbial pathogens and trigger immune response, but their regulation by neuropeptide-vasoactive intestinal peptide (VIP) in weaned piglets infected by enterotoxigenic Escherichia coli (ETEC) K88 remains unexplored. Therefore, the study was conducted to investigate its role using a model of early weaned piglets infected by ETEC K88. Male Duroc×Landrace×Yorkshire piglets (n = 24) were randomly divided into control, ETEC K88, VIP, and ETEC K88+VIP groups. On the first three days, ETEC K88 and ETEC K88+VIP groups were orally administrated with ETEC K88, other two groups were given sterile medium. Then each piglet from VIP and ETEC K88+VIP group received 10 nmol VIP intraperitoneally (i.p.) once daily, on day four and six. On the seventh day, the piglets were sacrificed. The results indicated that administration of VIP improved the growth performance, reduced diarrhea incidence of ETEC K88 challenged pigs, and mitigated the histopathological changes of intestine. Serum levels of IL-2, IL-6, IL-12p40, IFN-γ and TNF-α in the ETEC K88+ VIP group were significantly reduced compared with those in the ETEC group. VIP significantly increased IL-4, IL-10, TGF-β and S-IgA production compared with the ETEC K88 group. Besides, VIP could inhibit the expression of TLR2, TLR4, MyD88, NF-κB p65 and the phosphorylation of IκB-α, p-ERK, p-JNK, and p-38 induced by ETEC K88. Moreover, VIP could upregulate the expression of occludin in the ileum mucosa compared with the ETEC K88 group. Colon and caecum content bacterial richness and diversity were lower for pigs in the ETEC group than the unchallenged groups. These results demonstrate that VIP is beneficial for the maturation of the intestinal mucosal immune system and elicited local immunomodulatory activities. The TLR2/4-MyD88 mediated NF-κB and MAPK signaling pathway may be critical to the mechanism underlying the modulatory effect of VIP on intestinal mucosal immune function and bacterial community.
Collapse
|
40
|
Cytokine patterns in paediatric patients presenting serious gastrointestinal and respiratory bacterial infections. Cent Eur J Immunol 2014; 39:223-7. [PMID: 26155128 PMCID: PMC4440032 DOI: 10.5114/ceji.2014.43727] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2014] [Accepted: 03/25/2014] [Indexed: 01/29/2023] Open
Abstract
In the adaptive immune response, the types of cytokines produced define whether there is a cellular (T1) or a humoral (T2) response. Specifically, in the T1 response, interleukin 2 (IL-2), interferon γ (IFN-γ) and tumor necrosis factor β (TNF-β) are produced, whereas in the T2 response, IL-4, IL-5, IL- 6, IL-10 and IL-13 are primarily produced. Cytokines are primarily involved in the regulation of immune system cells. The aim of the present study was to evaluate the cytokine patterns (Type 1/Type 2) and TNF-α expression levels in children with severe gastrointestinal and respiratory bacterial infections. The enzyme-linked immunosorbent assay (ELISA) technique was used to identify the cytokines and the infectious agents. The results obtained demonstrated that, in general, children with bacterial infections experienced an increase in IL-2, IFN-γ and IL-4 concentrations and a decrease in TNF-α, IL-5 and IL-6 concentrations when compared to healthy children. Specifically, type 1 cytokines and an increased TNF-α concentration were found in children with gastrointestinal infections. However, patients with respiratory infections showed increased concentrations of both T2 (IL-4, IL-6 and IL-10) and T1 (IL-2 and IFN-γ) components. Thus, it was concluded that children with gastrointestinal infections exclusively developed a T1 response, whereas children with respiratory infections developed a T1/T2 response to fight the infection.
Collapse
|
41
|
Li E, Liu M, Singer SM. Resistance to reinfection in mice as a vaccine model for giardiasis. Hum Vaccin Immunother 2014; 10:1536-43. [PMID: 24805818 DOI: 10.4161/hv.29116] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Infection with Giardia is the most commonly diagnosed parasitic cause of diarrhea in the developed world, yet no vaccine exists for human use and a commercially available veterinary vaccine is of limited utility. We have used the adult C57BL/6 mouse model of infection with Giardia duodenalis to better understand immunity to secondary infections with this parasite. Mice were primed by infection with either the GS or WB strains of Giardia and treated with metronidazole on day 7-12 to eliminate the primary infections. Challenge infections on day 21 or day 60 after the primary infections resulted in ~50-fold fewer parasites at day 5 than were found in unprimed mice that only received the challenge infection. Resistance to challenge infections was also observed in B cell deficient µMT mice and when primed mice were challenged with parasites of a different strain. While primed mice developed IgA, mast cell, and T cell responses against the parasite, no specific responses correlated with protection against challenge infections. Together these data suggest that development of an effective vaccine for giardiasis should be feasible since strong immunity can be developed against reinfection in the adult mouse model. Moreover we show that antibody responses are not essential for a protective vaccine and that protection is not parasite strain-specific.
Collapse
Affiliation(s)
- Erqiu Li
- Department of Biology and Center for Infectious Disease; Georgetown University; Washington, DC USA
| | - Mingqiu Liu
- Department of Biology and Center for Infectious Disease; Georgetown University; Washington, DC USA; State Key Laboratory of Genetic Engineering; School of Life Sciences; Fudan University; Shanghai, PR China
| | - Steven M Singer
- Department of Biology and Center for Infectious Disease; Georgetown University; Washington, DC USA
| |
Collapse
|
42
|
Finamore A, Roselli M, Imbinto A, Seeboth J, Oswald IP, Mengheri E. Lactobacillus amylovorus inhibits the TLR4 inflammatory signaling triggered by enterotoxigenic Escherichia coli via modulation of the negative regulators and involvement of TLR2 in intestinal Caco-2 cells and pig explants. PLoS One 2014; 9:e94891. [PMID: 24733511 PMCID: PMC3986366 DOI: 10.1371/journal.pone.0094891] [Citation(s) in RCA: 106] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2014] [Accepted: 03/20/2014] [Indexed: 12/30/2022] Open
Abstract
Inflammation derived from pathogen infection involves the activation of toll-like receptor (TLR) signaling. Despite the established immunomodulatory activities of probiotics, studies relating the ability of such bacteria to inhibit the TLR signaling pathways are limited or controversial. In a previous study we showed that Lactobacillus amylovorus DSM 16698T, a novel lactobacillus isolated from unweaned pigs, protects the intestinal cells from enterotoxigenic Escherichia coli (ETEC) K88 infection through cytokine regulation. In the present study we investigated whether the ability of L. amylovorus to counteract the inflammatory status triggered by ETEC in intestine is elicited through inhibition of the TLR4 signaling pathway. We used the human intestinal Caco-2/TC7 cells and intestinal explants isolated from 5 week-old crossbreed Pietrain/Duroc/Large-White piglets, treated with ETEC, L. amylovorus or L. amylovorus cell free supernatant, either alone or simultaneously with ETEC. Western blot analysis showed that L. amylovorus and its cell free supernatant suppress the activation of the different steps of TLR4 signaling in Caco-2/TC7 cells and pig explants, by inhibiting the ETEC induced increase in the level of TLR4 and MyD88, the phosphorylation of the IKKα, IKKβ, IκBα and NF-κB subunit p65, as well as the over-production of inflammatory cytokines IL-8 and IL-1β. The immunofluorescence analysis confirms the lack of phospho-p65 translocation into the nucleus. These anti-inflammatory effects are achieved through modulation of the negative regulators Tollip and IRAK-M. We also found that L. amylovorus blocks the up-regulation of the extracellular heat shock protein (Hsp)72 and Hsp90, that are critical for TLR4 function. By using anti-TLR2 antibody, we demonstrate that TLR2 is required for the suppression of TLR4 signaling activation. These results may contribute to develop therapeutic interventions using L. amylovorus in intestinal disorders of piglets and humans.
Collapse
Affiliation(s)
- Alberto Finamore
- Consiglio per la Ricerca e la Sperimentazione in Agricoltura (CRA), Centro di Ricerca per gli Alimenti e la Nutrizione (Research Center on Food and Nutrition, CRA-NUT), Rome, Italy
| | - Marianna Roselli
- Consiglio per la Ricerca e la Sperimentazione in Agricoltura (CRA), Centro di Ricerca per gli Alimenti e la Nutrizione (Research Center on Food and Nutrition, CRA-NUT), Rome, Italy
| | - Ambra Imbinto
- Consiglio per la Ricerca e la Sperimentazione in Agricoltura (CRA), Centro di Ricerca per gli Alimenti e la Nutrizione (Research Center on Food and Nutrition, CRA-NUT), Rome, Italy
| | - Julie Seeboth
- INRA, UMR 1331 Toxalim, Research Center in Food Toxicology, Toulouse, France
- University of Toulouse, National Polytechnic Institute of Toulouse (INP), UMR 1331 Toxalim, Toulouse, France
| | - Isabelle P. Oswald
- INRA, UMR 1331 Toxalim, Research Center in Food Toxicology, Toulouse, France
- University of Toulouse, National Polytechnic Institute of Toulouse (INP), UMR 1331 Toxalim, Toulouse, France
| | - Elena Mengheri
- Consiglio per la Ricerca e la Sperimentazione in Agricoltura (CRA), Centro di Ricerca per gli Alimenti e la Nutrizione (Research Center on Food and Nutrition, CRA-NUT), Rome, Italy
| |
Collapse
|
43
|
Islam D, Ruamsap N, Aksomboon A, Khantapura P, Srijan A, Mason CJ. Immune responses to Campylobacter (C. jejuni or C. coli) infections: a two-year study of US forces deployed to Thailand. APMIS 2014; 122:1102-13. [PMID: 24730456 DOI: 10.1111/apm.12266] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2013] [Accepted: 01/21/2014] [Indexed: 11/28/2022]
Abstract
Campylobacter spp. is a leading cause of diarrheal disease among US troops deployed to Thailand for exercise. We investigated the importance of immunological analysis and immune responses against Campylobacter infection in US troops deployed to Thailand. Blood and fecal samples were collected from volunteered soldiers with diarrhea and from healthy controls. Stool culture was performed to identify the pathogens. Campylobacter-specific antibodies, antibody secreting cells and cytokines were measured. Several bacterial protein fragments in the outer membrane extract of Campylobacter spp., were identified by an immunoblot analysis with plasma and fecal antibodies. Among all of the diarrheal cases, 35% were Campylobacter-positive. Based on antibody titers in plasma and in fecal extract and antibody secreting cells: 6% of healthy controls, 32% of the Campylobacter culture-negative diarrheal cases, and 85% of the Campylobacter culture-positive diarrheal cases were positive for Campylobacter. Our results indicate that the measurement of Campylobacter-specific antibodies in plasma and fecal extract samples is a good marker of exposure to Campylobacter, and this test may be a useful diagnostic tool for seroepidemiological studies. Elicited antibodies against several bacterial outer membrane protein fragments suggest that these protein fragments are vital in providing protective immunity against Campylobacter.
Collapse
Affiliation(s)
- Dilara Islam
- Department of Enteric Diseases, Armed Forces Research Institute of Medical Sciences (AFRIMS), Bangkok, Thailand
| | | | | | | | | | | |
Collapse
|
44
|
Supplementation transgenic cow's milk containing recombinant human lactoferrin enhances systematic and intestinal immune responses in piglets. Mol Biol Rep 2014; 41:2119-28. [PMID: 24420858 DOI: 10.1007/s11033-014-3061-5] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2013] [Accepted: 01/04/2014] [Indexed: 10/25/2022]
Abstract
Lactoferrin (LF) plays an important role in the body's immune system. However, the immunomodulatory effects of supplementation transgenic cow's milk containing recombinant human LF (rhLF) on the systemic and intestinal immune systems in infants remain unclear. Our laboratory has used genetic engineer to produce transgenic cow secreted rhLF. To assess the immune responses we took piglets as an animal model for infants. Eighteen piglets at 7 days of age were fed ordinary milk, 1:1 mix of ordinary and rhLF milk, or rhLF milk (LFM) for 30 days. The incidence of diarrhea in piglets in natural condition was observed. The protein abundances of immunoglobulin (Ig)G, IgA, IgM, IgE, histamine, interleukin (IL)-1β, IL-2, IL-4, IL-5, IL-8, IL-10, IL-12 interferon, tumor necrosis factor in the plasma, spleen or intestine were measured by enzyme-linked immunosorbent assay. Intestinal structure was assessed by hematoxylin and eosin. The mRNA levels of immune and allergy-related genes were measured by quantitative reverse transcription-polymerase chain reaction. The results showed that LFM-fed significantly reduced incidence of diarrhea, enhanced humoral immunity, T helper (Th) 1, and Th2 cell responses, improved the structure of the intestinal mucosal and did not induce food allergy. LFM increased mRNA levels of toll-like receptor 2 and nuclear factor-κB p65 and decreased that of FCεRI β. In conclusion, rhLF-enriched formula could improve systematic and intestinal immune responses and did not elicit food allergies in neonatal piglets.
Collapse
|
45
|
DuPont HL. Giardia: both a harmless commensal and a devastating pathogen. J Clin Invest 2013; 123:2352-4. [PMID: 23728170 DOI: 10.1172/jci69932] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023] Open
Abstract
The highly prevalent protozoan Giardia lamblia is an enteropathogen that can be asymptomatic in some individuals, while leading to persistent diarrhea and substantial morbidity in others. In this issue of the JCI, Bartelt et al. describe a mouse model of the disease and investigate the contribution of coincident malnutrition with the development of symptomatic infection. This work in part explains how Giardia infection can lead to growth retardation, and may offer insights that guide future therapeutic strategies.
Collapse
Affiliation(s)
- Herbert L DuPont
- University of Texas School of Public Health and Medical School, St. Luke's Episcopal Hospital, Baylor College of Medicine, and Kelsey Research Foundation, Houston, Texas, USA.
| |
Collapse
|
46
|
Bartelt LA, Roche J, Kolling G, Bolick D, Noronha F, Naylor C, Hoffman P, Warren C, Singer S, Guerrant R. Persistent G. lamblia impairs growth in a murine malnutrition model. J Clin Invest 2013; 123:2672-84. [PMID: 23728173 PMCID: PMC3668820 DOI: 10.1172/jci67294] [Citation(s) in RCA: 77] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2012] [Accepted: 03/15/2013] [Indexed: 12/14/2022] Open
Abstract
Giardia lamblia infections are nearly universal among children in low-income countries and are syndemic with the triumvirate of malnutrition, diarrhea, and developmental growth delays. Amidst the morass of early childhood enteropathogen exposures in these populations, G. lamblia–specific associations with persistent diarrhea, cognitive deficits, stunting, and nutrient deficiencies have demonstrated conflicting results, placing endemic pediatric giardiasis in a state of equipoise. Many infections in endemic settings appear to be asymptomatic/ subclinical, further contributing to uncertainty regarding a causal link between G. lamblia infection and developmental delay. We used G. lamblia H3 cyst infection in a weaned mouse model of malnutrition to demonstrate that persistent giardiasis leads to epithelial cell apoptosis and crypt hyperplasia. Infection was associated with a Th2-biased inflammatory response and impaired growth. Malnutrition accentuated the severity of these growth decrements. Faltering malnourished mice exhibited impaired compensatory responses following infection and demonstrated an absence of crypt hyperplasia and subsequently blunted villus architecture. Concomitantly, severe malnutrition prevented increases in B220+ cells in the lamina propria as well as mucosal Il4 and Il5 mRNA in response to infection. These findings add insight into the potential role of G. lamblia as a "stunting" pathogen and suggest that, similarly, malnourished children may be at increased risk of G. lamblia– potentiated growth decrements.
Collapse
Affiliation(s)
- Luther A Bartelt
- Division of Infectious Diseases and Center for Global Health, University of Virginia, Charlottesville, Virginia 22903, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Generation of recombinant bacillus Calmette–Guérin and Mycobacterium smegmatis expressing BfpA and intimin as vaccine vectors against enteropathogenic Escherichia coli. Vaccine 2012; 30:5999-6005. [DOI: 10.1016/j.vaccine.2012.05.083] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2012] [Revised: 05/11/2012] [Accepted: 05/30/2012] [Indexed: 11/22/2022]
|
48
|
Role of heat-stable enterotoxins in the induction of early immune responses in piglets after infection with enterotoxigenic Escherichia coli. PLoS One 2012; 7:e41041. [PMID: 22815904 PMCID: PMC3398878 DOI: 10.1371/journal.pone.0041041] [Citation(s) in RCA: 55] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2011] [Accepted: 06/21/2012] [Indexed: 01/01/2023] Open
Abstract
Enterotoxigenic Escherichia coli (ETEC) strains that produce heat-stable (ST) and/or heat - labile (LT) enterotoxins are cause of post – weaning diarrhea in piglets. However, the relative importance of the different enterotoxins in host immune responses against ETEC infection has been poorly defined. In the present study, several isogenic mutant strains of an O149:F4ac+, LT+ STa+ STb+ ETEC strain were constructed that lack the expression of LT in combination with one or both types of ST enterotoxins (STa and/or STb). The small intestinal segment perfusion (SISP) technique and microarray analysis were used to study host early immune responses induced by these mutant strains 4 h after infection in comparison to the wild type strain and a PBS control. Simultaneously, net fluid absorption of pig small intestinal mucosa was measured 4 h after infection, allowing us to correlate enterotoxin secretion with gene regulation. Microarray analysis showed on the one hand a non-toxin related general antibacterial response comprising genes such as PAP, MMP1 and IL8. On the other hand, results suggest a dominant role for STb in small intestinal secretion early after post-weaning infection, as well as in the induced innate immune response through differential regulation of immune mediators like interleukin 1 and interleukin 17.
Collapse
|
49
|
Enterohemorrhagic Escherichia coli O157:H7 Shiga toxins inhibit gamma interferon-mediated cellular activation. Infect Immun 2012; 80:2307-15. [PMID: 22526675 DOI: 10.1128/iai.00255-12] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Enterohemorrhagic Escherichia coli (EHEC) serotype O157:H7 is a food-borne pathogen that causes significant morbidity and mortality in developing and industrialized nations. EHEC infection of host epithelial cells is capable of inhibiting the gamma interferon (IFN-γ) proinflammatory pathway through the inhibition of Stat-1 phosphorylation, which is important for host defense against microbial pathogens. The aim of this study was to determine the bacterial factors involved in the inhibition of Stat-1 tyrosine phosphorylation. Human HEp-2 and Caco-2 epithelial cells were challenged directly with either EHEC or bacterial culture supernatants and stimulated with IFN-γ, and then the protein extracts were analyzed by immunoblotting. The data showed that IFN-γ-mediated Stat-1 tyrosine phosphorylation was inhibited by EHEC secreted proteins. Using two-dimensional difference gel electrophoresis, EHEC Shiga toxins were identified as candidate inhibitory factors. EHEC Shiga toxin mutants were then generated and complemented in trans, and mutant culture supernatant was supplemented with purified Stx to confirm their ability to subvert IFN-γ-mediated cell activation. We conclude that while other factors are likely involved in the suppression of IFN-γ-mediated Stat-1 tyrosine phosphorylation, E. coli-derived Shiga toxins represent a novel mechanism by which EHEC evades the host immune system.
Collapse
|
50
|
Duell BL, Tan CK, Carey AJ, Wu F, Cripps AW, Ulett GC. Recent insights into microbial triggers of interleukin-10 production in the host and the impact on infectious disease pathogenesis. ACTA ACUST UNITED AC 2012; 64:295-313. [PMID: 22268692 DOI: 10.1111/j.1574-695x.2012.00931.x] [Citation(s) in RCA: 69] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2011] [Revised: 01/17/2012] [Accepted: 01/17/2012] [Indexed: 02/06/2023]
Abstract
Since its initial description as a Th2-cytokine antagonistic to interferon-alpha and granulocyte-macrophage colony-stimulating factor, many studies have shown various anti-inflammatory actions of interleukin-10 (IL-10), and its role in infection as a key regulator of innate immunity. Studies have shown that IL-10 induced in response to microorganisms and their products plays a central role in shaping pathogenesis. IL-10 appears to function as both sword and shield in the response to varied groups of microorganisms in its capacity to mediate protective immunity against some organisms but increase susceptibility to other infections. The nature of IL-10 as a pleiotropic modulator of host responses to microorganisms is explained, in part, by its potent and varied effects on different immune effector cells which influence antimicrobial activity. A new understanding of how microorganisms trigger IL-10 responses is emerging, along with recent discoveries of how IL-10 produced during disease might be harnessed for better protective or therapeutic strategies. In this review, we summarize studies from the past 5 years that have reported the induction of IL-10 by different classes of pathogenic microorganisms, including protozoa, nematodes, fungi, viruses and bacteria and discuss the impact of this induction on the persistence and/or clearance of microorganisms in the host.
Collapse
Affiliation(s)
- Benjamin L Duell
- School of Medical Sciences, Centre for Medicine and Oral Health, Griffith University, Gold Coast Campus, Gold Coast, Qld, Australia
| | | | | | | | | | | |
Collapse
|