101
|
Ekmekciu I, von Klitzing E, Neumann C, Bacher P, Scheffold A, Bereswill S, Heimesaat MM. Fecal Microbiota Transplantation, Commensal Escherichia coli and Lactobacillus johnsonii Strains Differentially Restore Intestinal and Systemic Adaptive Immune Cell Populations Following Broad-spectrum Antibiotic Treatment. Front Microbiol 2017; 8:2430. [PMID: 29321764 PMCID: PMC5732213 DOI: 10.3389/fmicb.2017.02430] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2017] [Accepted: 11/23/2017] [Indexed: 12/28/2022] Open
Abstract
The essential role of the intestinal microbiota in the well-functioning of host immunity necessitates the investigation of species-specific impacts on this interplay. Aim of this study was to examine the ability of defined Gram-positive and Gram-negative intestinal commensal bacterial species, namely Escherichia coli and Lactobacillus johnsonii, respectively, to restore immune functions in mice that were immunosuppressed by antibiotics-induced microbiota depletion. Conventional mice were subjected to broad-spectrum antibiotic treatment for 8 weeks and perorally reassociated with E. coli, L. johnsonii or with a complex murine microbiota by fecal microbiota transplantation (FMT). Analyses at days (d) 7 and 28 revealed that immune cell populations in the small and large intestines, mesenteric lymph nodes and spleens of mice were decreased after antibiotic treatment but were completely or at least partially restored upon FMT or by recolonization with the respective bacterial species. Remarkably, L. johnsonii recolonization resulted in the highest CD4+ and CD8+ cell numbers in the small intestine and spleen, whereas neither of the commensal species could stably restore those cell populations in the colon until d28. Meanwhile less efficient than FMT, both species increased the frequencies of regulatory T cells and activated dendritic cells and completely restored intestinal memory/effector T cell populations at d28. Furthermore, recolonization with either single species maintained pro- and anti-inflammatory immune functions in parallel. However, FMT could most effectively recover the decreased frequencies of cytokine producing CD4+ lymphocytes in mucosal and systemic compartments. E. coli recolonization increased the production of cytokines such as TNF, IFN-γ, IL-17, and IL-22, particularly in the small intestine. Conversely, only L. johnsonii recolonization maintained colonic IL-10 production. In summary, FMT appears to be most efficient in the restoration of antibiotics-induced collateral damages to the immune system. However, defined intestinal commensals such as E. coli and L. johnsonii have the potential to restore individual functions of intestinal and systemic immunity. In conclusion, our data provide novel insights into the distinct role of individual commensal bacteria in maintaining immune functions during/following dysbiosis induced by antibiotic therapy thereby shaping host immunity and might thus open novel therapeutical avenues in conditions of perturbed microbiota composition.
Collapse
Affiliation(s)
- Ira Ekmekciu
- Intestinal Microbiology Research Group, Institute of Microbiology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Eliane von Klitzing
- Intestinal Microbiology Research Group, Institute of Microbiology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Christian Neumann
- Department of Cellular Immunology, Clinic for Rheumatology and Clinical Immunology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany.,German Rheumatism Research Center, Leibniz Association, Berlin, Germany
| | - Petra Bacher
- Department of Cellular Immunology, Clinic for Rheumatology and Clinical Immunology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Alexander Scheffold
- Department of Cellular Immunology, Clinic for Rheumatology and Clinical Immunology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany.,German Rheumatism Research Center, Leibniz Association, Berlin, Germany
| | - Stefan Bereswill
- Intestinal Microbiology Research Group, Institute of Microbiology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Markus M Heimesaat
- Intestinal Microbiology Research Group, Institute of Microbiology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| |
Collapse
|
102
|
Yang X, Liu L, Chen J, Xiao A. Response of Intestinal Bacterial Flora to the Long-term Feeding of Aflatoxin B1 (AFB1) in Mice. Toxins (Basel) 2017; 9:toxins9100317. [PMID: 29023377 PMCID: PMC5666364 DOI: 10.3390/toxins9100317] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2017] [Revised: 09/28/2017] [Accepted: 09/30/2017] [Indexed: 12/20/2022] Open
Abstract
In order to investigate the influence of aflatoxin B1 (AFB1) on intestinal bacterial flora, 24 Kunming mice (KM mice) were randomly placed into four groups, which were labeled as control, low-dose, medium-dose, and high-dose groups. They were fed intragastrically with 0.4 mL of 0 mg/L, 2.5 mg/L, 4 mg/L, or 10 mg/L of AFB1 solutions, twice a day for 2 months. The hypervariable region V3 + V4 on 16S rDNA of intestinal bacterial flora was sequenced by the use of a high-flux sequencing system on a Miseq Illumina platform; then, the obtained sequences were analyzed. The results showed that, when compared with the control group, both genera and phyla of intestinal bacteria in the three treatment groups decreased. About one third of the total genera and one half of the total phyla remained in the high-dose group. The dominant flora were Lactobacillus and Bacteroides in all groups. There were significant differences in the relative abundance of intestinal bacterial flora among groups. Most bacteria decreased as a whole from the control to the high-dose groups, but several beneficial and pathogenic bacterial species increased significantly with increasing dose of AFB1. Thus, the conclusion was that intragastric feeding with 2.5~10 mg/mL AFB1 for 2 months could decrease the majority of intestinal bacterial flora and induce the proliferation of some intestinal bacteria flora.
Collapse
Affiliation(s)
- Xiai Yang
- Institute of Bast Fiber Crops, Chinese Academy of Agricultural Sciences, Changsha 410205, China.
| | - Liangliang Liu
- Institute of Bast Fiber Crops, Chinese Academy of Agricultural Sciences, Changsha 410205, China.
| | - Jing Chen
- Institute of Bast Fiber Crops, Chinese Academy of Agricultural Sciences, Changsha 410205, China.
| | - Aiping Xiao
- Institute of Bast Fiber Crops, Chinese Academy of Agricultural Sciences, Changsha 410205, China.
| |
Collapse
|
103
|
Lukic J, Jancic I, Mirkovic N, Bufan B, Djokic J, Milenkovic M, Begovic J, Strahinic I, Lozo J. Lactococcus lactis and Lactobacillus salivarius differently modulate early immunological response of Wistar rats co-administered with Listeria monocytogenes. Benef Microbes 2017; 8:809-822. [PMID: 28856909 DOI: 10.3920/bm2017.0007] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
In the light of the increasing resistance of bacterial pathogens to antibiotics, one of the main global strategies in applied science is development of alternative treatments, which would be safe both for the host and from the environmental perspective. Accordingly, the aim of this study was to test whether two lactic acid bacteria (LAB) strains, Lactococcus lactis BGBU1-4 and Lactobacillus salivarius BGHO1, could be applied as safe supplements for Listeria infection. Two major research objectives were set: to compare the effects of BGBU1-4 and BGHO1 on early immune response in gut tissue of Wistar rats co-administered with Listeria monocytogenes ATCC19111 and next, to test how this applies to their usage as therapeutics in acute ATCC19111 infection. Intestinal villi (IV), Peyer's patches (PP) and mesenteric lymph nodes (MLN) were used for the analysis. The results showed that BGHO1 increased the mRNA expression of innate immune markers CD14, interleukin (IL)-1β and tumour necrosis factor (TNF)-α in PP and IV, and, in parallel, caused a decrease of listeriolysin O (LLO) mRNA expression in same tissues. In MLN of BGHO1 treated rats, LLO expression was increased, along with an increase of the expression of OX-62 mRNA and CD69, pointing to the activation of adaptive immunity. On the other hand, in BGBU1-4 treated rats, there was no reduction of LLO mRNA expression and no induction of innate immunity markers in intestinal tissue. Additionally, CD14 and IL-1β, as well as LLO, but not OX-62 mRNA and CD69 expression, were elevated in MLN of BGBU1-4 treated rats. However, when applied therapeutically, both, BGBU1-4 and BGHO1, lowered Listeria count in spleens of infected rats. Our results not only reveal the potential of LAB to ameliorate Listeria infections, but suggest different immunological effects of two different LAB strains, both of which could be effective in Listeria elimination.
Collapse
Affiliation(s)
- J Lukic
- 1 Laboratory for Molecular Microbiology, Institute of Molecular Genetics and Genetic Engineering (IMGGE), University of Belgrade, Vojvode Stepe 444a, 11010 Belgrade, Serbia
| | - I Jancic
- 2 Faculty of Pharmacy, University of Belgrade, Vojvode Stepe 450, 11010 Belgrade, Serbia
| | - N Mirkovic
- 1 Laboratory for Molecular Microbiology, Institute of Molecular Genetics and Genetic Engineering (IMGGE), University of Belgrade, Vojvode Stepe 444a, 11010 Belgrade, Serbia
| | - B Bufan
- 2 Faculty of Pharmacy, University of Belgrade, Vojvode Stepe 450, 11010 Belgrade, Serbia
| | - J Djokic
- 1 Laboratory for Molecular Microbiology, Institute of Molecular Genetics and Genetic Engineering (IMGGE), University of Belgrade, Vojvode Stepe 444a, 11010 Belgrade, Serbia
| | - M Milenkovic
- 2 Faculty of Pharmacy, University of Belgrade, Vojvode Stepe 450, 11010 Belgrade, Serbia
| | - J Begovic
- 1 Laboratory for Molecular Microbiology, Institute of Molecular Genetics and Genetic Engineering (IMGGE), University of Belgrade, Vojvode Stepe 444a, 11010 Belgrade, Serbia
| | - I Strahinic
- 1 Laboratory for Molecular Microbiology, Institute of Molecular Genetics and Genetic Engineering (IMGGE), University of Belgrade, Vojvode Stepe 444a, 11010 Belgrade, Serbia
| | - J Lozo
- 1 Laboratory for Molecular Microbiology, Institute of Molecular Genetics and Genetic Engineering (IMGGE), University of Belgrade, Vojvode Stepe 444a, 11010 Belgrade, Serbia.,3 Faculty of Biology, University of Belgrade, Studentski trg 16, 11000 Belgrade, Serbia
| |
Collapse
|
104
|
Topical Antimicrobial Treatments Can Elicit Shifts to Resident Skin Bacterial Communities and Reduce Colonization by Staphylococcus aureus Competitors. Antimicrob Agents Chemother 2017. [PMID: 28630195 DOI: 10.1128/aac.00774-17] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
The skin microbiome is a complex ecosystem with important implications for cutaneous health and disease. Topical antibiotics and antiseptics are often employed to preserve the balance of this population and inhibit colonization by more pathogenic bacteria. However, despite their widespread use, the impact of these interventions on broader microbial communities remains poorly understood. Here, we report the longitudinal effects of topical antibiotics and antiseptics on skin bacterial communities and their role in Staphylococcus aureus colonization resistance. In response to antibiotics, cutaneous populations exhibited an immediate shift in bacterial residents, an effect that persisted for multiple days posttreatment. By contrast, antiseptics elicited only minor changes to skin bacterial populations, with few changes to the underlying microbiota. While variable in scope, both antibiotics and antiseptics were found to decrease colonization by commensal Staphylococcus spp. by sequencing- and culture-based methods, an effect which was highly dependent on baseline levels of Staphylococcus Because Staphylococcus residents have been shown to compete with the skin pathogen S. aureus, we also tested whether treatment could influence S. aureus levels at the skin surface. We found that treated mice were more susceptible to exogenous association with S. aureus and that precolonization with the same Staphylococcus residents that were previously disrupted by treatment reduced S. aureus levels by over 100-fold. In all, the results of this study indicate that antimicrobial drugs can alter skin bacterial residents and that these alterations can have critical implications for cutaneous host defense.
Collapse
|
105
|
Lourbopoulos A, Mamrak U, Roth S, Balbi M, Shrouder J, Liesz A, Hellal F, Plesnila N. Inadequate food and water intake determine mortality following stroke in mice. J Cereb Blood Flow Metab 2017; 37:2084-2097. [PMID: 27449604 PMCID: PMC5464703 DOI: 10.1177/0271678x16660986] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Experimental stroke models producing clinically relevant functional deficits are often associated with high mortality. Because the mechanisms that underlie post-stroke mortality are largely unknown, results obtained using these models are often difficult to interpret, thereby limiting their translational potential. Given that specific forms of post-stroke care reduce mortality in patients, we hypothesized that inadequate food and water intake may underlie mortality following experimental stroke. C57BL/6 mice were subjected to 1 h of intraluminal filament middle cerebral artery occlusion. Nutritional support beginning on the second day after filament middle cerebral artery occlusion reduced the 14-day mortality rate from 59% to 15%. The surviving mice in the post-stroke support group had the same infarct size as non-surviving control mice, suggesting that post-stroke care was not neuroprotective and that inadequate food and/or water intake are the main reasons for filament middle cerebral artery occlusion-induced mortality. This notion was supported by the presence of significant hypoglycemia, ketonemia, and dehydration in control mice. Taken together, these data suggest that post-filament middle cerebral artery occlusion mortality in mice is not primarily caused by ischemic brain damage, but secondarily by inadequate food and/or water intake. Thus, providing nutritional support following filament middle cerebral artery occlusion greatly minimizes mortality bias and allows the study of long-term morphological and functional sequelae of stroke in mice.
Collapse
Affiliation(s)
- Athanasios Lourbopoulos
- 1 Laboratory of Experimental Stroke Research, Institute for Stroke and Dementia Research (ISD), University of Munich Medical Center, Munich, Germany
| | - Uta Mamrak
- 1 Laboratory of Experimental Stroke Research, Institute for Stroke and Dementia Research (ISD), University of Munich Medical Center, Munich, Germany
| | - Stefan Roth
- 1 Laboratory of Experimental Stroke Research, Institute for Stroke and Dementia Research (ISD), University of Munich Medical Center, Munich, Germany
| | - Matilde Balbi
- 1 Laboratory of Experimental Stroke Research, Institute for Stroke and Dementia Research (ISD), University of Munich Medical Center, Munich, Germany
| | - Joshua Shrouder
- 1 Laboratory of Experimental Stroke Research, Institute for Stroke and Dementia Research (ISD), University of Munich Medical Center, Munich, Germany
| | - Arthur Liesz
- 1 Laboratory of Experimental Stroke Research, Institute for Stroke and Dementia Research (ISD), University of Munich Medical Center, Munich, Germany.,2 Munich Cluster for Systems Neurology (Synergy), LMU Munich, Munich, Germany
| | - Farida Hellal
- 1 Laboratory of Experimental Stroke Research, Institute for Stroke and Dementia Research (ISD), University of Munich Medical Center, Munich, Germany
| | - Nikolaus Plesnila
- 1 Laboratory of Experimental Stroke Research, Institute for Stroke and Dementia Research (ISD), University of Munich Medical Center, Munich, Germany.,2 Munich Cluster for Systems Neurology (Synergy), LMU Munich, Munich, Germany
| |
Collapse
|
106
|
Ekmekciu I, von Klitzing E, Fiebiger U, Escher U, Neumann C, Bacher P, Scheffold A, Kühl AA, Bereswill S, Heimesaat MM. Immune Responses to Broad-Spectrum Antibiotic Treatment and Fecal Microbiota Transplantation in Mice. Front Immunol 2017; 8:397. [PMID: 28469619 PMCID: PMC5395657 DOI: 10.3389/fimmu.2017.00397] [Citation(s) in RCA: 99] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2017] [Accepted: 03/21/2017] [Indexed: 12/12/2022] Open
Abstract
Compelling evidence demonstrates the pivotal role of the commensal intestinal microbiota in host physiology and the detrimental effects of its perturbations following antibiotic treatment. Aim of this study was to investigate the impact of antibiotics induced depletion and subsequent restoration of the intestinal microbiota composition on the murine mucosal and systemic immunity. To address this, conventional C57BL/6j mice were subjected to broad-spectrum antibiotic treatment for 8 weeks. Restoration of the intestinal microbiota by peroral fecal microbiota transplantation (FMT) led to reestablishment of small intestinal CD4+, CD8+, and B220+ as well as of colonic CD4+ cell numbers as early as 7 days post-FMT. However, at d28 following FMT, colonic CD4+ and B220+ cell numbers were comparable to those in secondary abiotic (ABx) mice. Remarkably, CD8+ cell numbers were reduced in the colon upon antibiotic treatment, and FMT was not sufficient to restore this immune cell subset. Furthermore, absence of gut microbial stimuli resulted in decreased percentages of memory/effector T cells, regulatory T cells, and activated dendritic cells in the small intestine, colon, mesenteric lymph nodes (MLN), and spleen. Concurrent antibiotic treatment caused decreased cytokine production (IFN-γ, IL-17, IL-22, and IL-10) of CD4+ cells in respective compartments. These effects were, however, completely restored upon FMT. In summary, broad-spectrum antibiotic treatment resulted in profound local (i.e., small and large intestinal), peripheral (i.e., MLN), and systemic (i.e., splenic) changes in the immune cell repertoire that could, at least in part, be restored upon FMT. Further studies need to unravel the distinct molecular mechanisms underlying microbiota-driven changes in immune homeostasis subsequently providing novel therapeutic or even preventive approaches in human immunopathologies.
Collapse
Affiliation(s)
- Ira Ekmekciu
- Department of Microbiology and Hygiene, Charité - University Medicine Berlin, Berlin, Germany
| | - Eliane von Klitzing
- Department of Microbiology and Hygiene, Charité - University Medicine Berlin, Berlin, Germany
| | - Ulrike Fiebiger
- Department of Microbiology and Hygiene, Charité - University Medicine Berlin, Berlin, Germany
| | - Ulrike Escher
- Department of Microbiology and Hygiene, Charité - University Medicine Berlin, Berlin, Germany
| | - Christian Neumann
- Department of Cellular Immunology, Clinic for Rheumatology and Clinical Immunology, Charité - University Medicine Berlin, Berlin, Germany.,German Rheumatism Research Center (DRFZ), Leibniz Association, Berlin, Germany
| | - Petra Bacher
- Department of Cellular Immunology, Clinic for Rheumatology and Clinical Immunology, Charité - University Medicine Berlin, Berlin, Germany
| | - Alexander Scheffold
- Department of Cellular Immunology, Clinic for Rheumatology and Clinical Immunology, Charité - University Medicine Berlin, Berlin, Germany.,German Rheumatism Research Center (DRFZ), Leibniz Association, Berlin, Germany
| | - Anja A Kühl
- Department of Medicine I for Gastroenterology, Infectious Diseases and Rheumatology, Research Center ImmunoSciences (RCIS), Charité - University Medicine Berlin, Berlin, Germany
| | - Stefan Bereswill
- Department of Microbiology and Hygiene, Charité - University Medicine Berlin, Berlin, Germany
| | - Markus M Heimesaat
- Department of Microbiology and Hygiene, Charité - University Medicine Berlin, Berlin, Germany
| |
Collapse
|
107
|
Metabolic adaptation to the aqueous leaf extract of Moringa oleifera Lam.-supplemented diet is related to the modulation of gut microbiota in mice. Appl Microbiol Biotechnol 2017; 101:5115-5130. [PMID: 28382453 DOI: 10.1007/s00253-017-8233-5] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2017] [Revised: 03/05/2017] [Accepted: 03/09/2017] [Indexed: 12/12/2022]
Abstract
The aqueous leaf extract of Moringa oleifera Lam. (LM-A) is reported to have many health beneficial bioactivities and no obvious toxicity, but have mild adverse effects. Little is known about the mechanism of these reported adverse effects. Notably, there has been no report about the influence of LM-A on intestinal microecology. In this study, animal experiments were performed to explore the relationships between metabolic adaptation to an LM-A-supplemented diet and gut microbiota changes. After 8-week feeding with normal chow diet, the body weight of mice entered a stable period, and one of the group received daily doses of 750-mg/kg body weight LM-A by gavage for 4 weeks (assigned as LM); the other group received the vehicle (assigned as NCD). The liver weight to body weight ratio was enhanced, and the ceca were enlarged in the LM group compared with the NCD group. LM-A-supplemented-diet mice elicited a uniform metabolic adaptation, including slightly influenced fasting glucose and blood lipid profiles, significantly reduced liver triglycerides content, enhanced serum lipopolysaccharide level, activated inflammatory responses in the intestine and liver, compromised gut barrier function, and broken intestinal homeostasis. Many metabolic changes in mice were significantly correlated with altered specific gut bacteria. Changes in Firmicutes, Eubacterium rectale/Clostridium coccoides group, Faecalibacterium prausnitzii, Akkermansia muciniphila, segmented filamentous bacteria, Enterococcus spp., and Sutterella spp. may play an important role in the process of host metabolic adaptation to LM-A administration. Our research provides an explanation of the adverse effects of LM-A administration on normal adult individuals in the perspective of microecology.
Collapse
|
108
|
Vogt SL, Finlay BB. Gut microbiota-mediated protection against diarrheal infections. J Travel Med 2017; 24:S39-S43. [PMID: 28520994 PMCID: PMC5731444 DOI: 10.1093/jtm/taw086] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/19/2016] [Accepted: 11/10/2016] [Indexed: 12/21/2022]
Abstract
BACKGROUND The mammalian gut microbiota is a highly abundant and diverse microbial community that resides in the gastrointestinal tract. One major benefit that the gut microbiota provides to its host is colonization resistance-the ability to prevent colonization by foreign microbes, including diarrheal pathogens such as Clostridium difficile , Salmonella enterica serovar Typhimurium and diarrheagenic Escherichia coli . METHODS We conducted a literature review of the effects of the gut microbiota on infection by diarrheal pathogens. We used PubMed to search for relevant articles published before July 2016, as well as incorporated data from our laboratory. RESULTS The gut microbiota provides protection from diarrheal infections both by direct inhibition of pathogens and by indirect effects on host functions. Direct effects of the microbiota on diarrheal pathogens include competing for nutrients and producing metabolites that inhibit pathogen growth or virulence. Indirect effects of the gut microbiota include promoting maintenance of the gut mucosal barrier and stimulating innate and adaptive immunity. CONCLUSIONS Human epidemiological studies and experimental infections of laboratory animals both demonstrate that antibiotic treatment can alter the gut microbial community and thereby reduce colonization resistance against diarrheal pathogens. Further research might lead to the development of next-generation probiotics that could be used to bolster colonization resistance and thus prevent travellers' diarrheal.
Collapse
Affiliation(s)
- Stefanie L Vogt
- Michael Smith Laboratories, University of British Columbia, Vancouver, BC, Canada
| | - B Brett Finlay
- Michael Smith Laboratories, University of British Columbia, Vancouver, BC, Canada.,Department of Microbiology and Immunology, University of British Columbia, Vancouver, BC, Canada.,Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
109
|
Ericsson AC, Personett AR, Turner G, Dorfmeyer RA, Franklin CL. Variable Colonization after Reciprocal Fecal Microbiota Transfer between Mice with Low and High Richness Microbiota. Front Microbiol 2017; 8:196. [PMID: 28280484 PMCID: PMC5322181 DOI: 10.3389/fmicb.2017.00196] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2016] [Accepted: 01/26/2017] [Indexed: 12/29/2022] Open
Abstract
Several associations have been made between characteristics of the resident gut microbiota and human health and disease susceptibility. Animal models provide the means to test these correlations prospectively and evaluate causality. Experimental fecal microbiota transfer (FMT), or the intentional transplantation of gut microbes into recipient mice depleted of their autochthonous microbes with antibiotics, is a commonly used method of testing these relationships. The true completeness of microbial transfer through such procedures is poorly documented in the literature, particularly in the context of reciprocal transfer of microbes between recipient and donor mice harboring microbial populations of differing richness and diversity. Moreover, it is unclear whether the use of frozen fecal contents or cecal contents would confer any difference in the outcomes of transfer. Herein, groups of mice colonized with distinct gut microbiota of differing richness and composition were used in a reciprocal FMT study, with different groups receiving transfer of material prepared from fresh cecal contents, fresh feces, or frozen feces. Targeted 16S rRNA gene amplicon sequencing was used at intervals throughout the study to characterize the microbiota. Notably, despite comparable depletion of the microbiota in recipient mice prior to transfer, donor-specific taxa reliably colonized recipients only when relatively rich donor material was transferred to mice originally colonized with a simpler microbiota. It is unclear whether these differences were due to differences in the endogenous recipient microbiota or host factors induced in early life by microbial factors. These findings are of practical import for researchers using FMT to prospectively assess the influence of the gut microbiota in mouse models, and to those studying host-microbial interactions and their influence on gut barrier function.
Collapse
Affiliation(s)
- Aaron C Ericsson
- University of Missouri Metagenomics Center, University of MissouriColumbia, MO, USA; University of Missouri Mutant Mouse Resource and Research Center, University of MissouriColumbia, MO, USA; Comparative Metagenomics Laboratory, Department of Veterinary Pathobiology, University of MissouriColumbia, MO, USA
| | - Alexa R Personett
- Comparative Metagenomics Laboratory, Department of Veterinary Pathobiology, University of Missouri Columbia, MO, USA
| | - Giedre Turner
- University of Missouri Mutant Mouse Resource and Research Center, University of MissouriColumbia, MO, USA; Comparative Metagenomics Laboratory, Department of Veterinary Pathobiology, University of MissouriColumbia, MO, USA
| | - Rebecca A Dorfmeyer
- University of Missouri Mutant Mouse Resource and Research Center, University of MissouriColumbia, MO, USA; Comparative Metagenomics Laboratory, Department of Veterinary Pathobiology, University of MissouriColumbia, MO, USA
| | - Craig L Franklin
- University of Missouri Metagenomics Center, University of MissouriColumbia, MO, USA; University of Missouri Mutant Mouse Resource and Research Center, University of MissouriColumbia, MO, USA; Comparative Metagenomics Laboratory, Department of Veterinary Pathobiology, University of MissouriColumbia, MO, USA
| |
Collapse
|
110
|
The effect of gut microbiome on tolerance to morphine mediated antinociception in mice. Sci Rep 2017; 7:42658. [PMID: 28211545 PMCID: PMC5314392 DOI: 10.1038/srep42658] [Citation(s) in RCA: 116] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2016] [Accepted: 01/12/2017] [Indexed: 02/06/2023] Open
Abstract
There is growing appreciation for the importance of gastrointestinal microbiota in many physiological and pathophysiological processes. While morphine and other narcotics are the most widely prescribed therapy for moderate to severe pain clinically, they have been noted to alter microbial composition and promote bacterial translocation to other tissues. Here we examined the pharmacodynamic properties of chronic morphine in mice following bacterial depletion with oral gavage of an antibiotic cocktail (ABX). ABX significantly reduced gut bacteria and prevented chronic morphine induced increases in gut permeability, colonic mucosal destruction, and colonic IL-1β expression. In addition, ABX prevented the development of antinociceptive tolerance to chronic morphine in both the tail-immersion and acetic acid stretch assays. Morphine tolerance was also reduced by oral vancomycin that has 0% bioavailability. These findings were recapitulated in primary afferent neurons isolated from dorsal root ganglia (DRG) innervating the lower gastrointestinal tract, wherein in-vivo administration of ABX prevented tolerance to morphine-induced hypoexcitability. Finally, though ABX repeatedly demonstrated an ability to prevent tolerance, we show that it did not alter susceptibility to precipitation of withdrawal by naloxone. Collectively, these finding indicate that the gastrointestinal microbiome is an important modulator of physiological responses induced by chronic morphine administration.
Collapse
|
111
|
Gomes-Neto JC, Mantz S, Held K, Sinha R, Segura Munoz RR, Schmaltz R, Benson AK, Walter J, Ramer-Tait AE. A real-time PCR assay for accurate quantification of the individual members of the Altered Schaedler Flora microbiota in gnotobiotic mice. J Microbiol Methods 2017; 135:52-62. [PMID: 28189782 DOI: 10.1016/j.mimet.2017.02.003] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2016] [Revised: 01/31/2017] [Accepted: 02/07/2017] [Indexed: 01/22/2023]
Abstract
Changes in the gastrointestinal microbial community are frequently associated with chronic diseases such as Inflammatory Bowel Diseases. However, understanding the relationship of any individual taxon within the community to host physiology is made complex due to the diversity and individuality of the gut microbiota. Defined microbial communities such as the Altered Schaedler Flora (ASF) help alleviate the challenges of a diverse microbiota by allowing one to interrogate the relationship between individual bacterial species and host responses. An important aspect of studying these relationships with defined microbial communities is the ability to measure the population abundance and dynamics of each member. Herein, we describe the development of an improved ASF species-specific and sensitive real-time quantitative polymerase chain reaction (qPCR) for use with SYBR Green chemistry to accurately assess individual ASF member abundance. This approach targets hypervariable regions V1 through V3 of the 16S rRNA gene of each ASF taxon to enhance assay specificity. We demonstrate the reproducibility, sensitivity and application of this new method by quantifying each ASF bacterium in two inbred mouse lines. We also used it to assess changes in ASF member abundance before and after acute antibiotic perturbation of the community as well as in mice fed two different diets. Additionally, we describe a nested PCR assay for the detection of lowly abundant ASF members. Altogether, this improved qPCR method will facilitate gnotobiotic research involving the ASF community by allowing for reproducible quantification of its members under various physiological conditions.
Collapse
Affiliation(s)
- João Carlos Gomes-Neto
- Department of Food Science and Technology, University of Nebraska-Lincoln, Lincoln, NE, USA
| | - Sara Mantz
- Department of Food Science and Technology, University of Nebraska-Lincoln, Lincoln, NE, USA
| | - Kyler Held
- Department of Food Science and Technology, University of Nebraska-Lincoln, Lincoln, NE, USA
| | - Rohita Sinha
- Department of Food Science and Technology, University of Nebraska-Lincoln, Lincoln, NE, USA
| | - Rafael R Segura Munoz
- Department of Food Science and Technology, University of Nebraska-Lincoln, Lincoln, NE, USA
| | - Robert Schmaltz
- Department of Food Science and Technology, University of Nebraska-Lincoln, Lincoln, NE, USA
| | - Andrew K Benson
- Department of Food Science and Technology, University of Nebraska-Lincoln, Lincoln, NE, USA
| | - Jens Walter
- Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, Alberta, Canada; Department of Biological Sciences, University of Alberta, Edmonton, Alberta, Canada
| | - Amanda E Ramer-Tait
- Department of Food Science and Technology, University of Nebraska-Lincoln, Lincoln, NE, USA.
| |
Collapse
|
112
|
Rice TC, Armocida SM, Kuethe JW, Midura EF, Jain A, Hildeman DA, Healy DP, Gulbins E, Caldwell CC. Burn injury influences the T cell homeostasis in a butyrate-acid sphingomyelinase dependent manner. Cell Immunol 2016; 313:25-31. [PMID: 28063598 DOI: 10.1016/j.cellimm.2016.12.004] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2016] [Revised: 12/23/2016] [Accepted: 12/24/2016] [Indexed: 01/12/2023]
Abstract
Following burn injury, a key factor for patients susceptible to opportunistic infections is immune suppression. Butyrate levels are important in maintaining a functional immune system and these levels can be altered after injury. The acid sphingomyelinase (Asm) lipid signaling system has been implicated in a T cell actions with some evidence of being influenced by butyrate. Here, we hypothesized that burn-injury changes in butyrate levels would mediate Asm activity and, consequently, T cell homeostasis. We demonstrate that burn injury temporally decreases butyrate levels. We further determined that T cell Asm activity is increased by butyrate and decreased after burn injury. We additionally observed decreased T cell numbers in Asm-deficient, burn-injured, and microbiota-depleted mice. Finally, we demonstrate that butyrate reduced T cell death in an Asm-dependent manner. These data suggest that restoration of butyrate after burn injury may ameliorate the T cell lost observed in burn-injured patients by Asm regulation.
Collapse
Affiliation(s)
- Teresa C Rice
- Division of Research, Department of Surgery, College of Medicine, University of Cincinnati, Cincinnati, OH, USA
| | - Stephanie M Armocida
- Division of Research, Department of Surgery, College of Medicine, University of Cincinnati, Cincinnati, OH, USA
| | - Joshua W Kuethe
- Division of Research, Department of Surgery, College of Medicine, University of Cincinnati, Cincinnati, OH, USA
| | - Emily F Midura
- Division of Research, Department of Surgery, College of Medicine, University of Cincinnati, Cincinnati, OH, USA
| | - Ayushi Jain
- Division of Research, Department of Surgery, College of Medicine, University of Cincinnati, Cincinnati, OH, USA
| | - David A Hildeman
- Division of Immunobiology, Cincinnati Children's Hospital Research Foundation, Cincinnati, OH, USA
| | - Daniel P Healy
- James L. Winkle College of Pharmacy, Division of Pharmacy Practice and Administrative Sciences, University of Cincinnati Academic Health Center, Cincinnati, OH, USA
| | - Erich Gulbins
- Division of Research, Department of Surgery, College of Medicine, University of Cincinnati, Cincinnati, OH, USA; Department of Molecular Biology, University of Duisburg-Essen, Hufelandstrasse 55, 45122 Essen, Germany
| | - Charles C Caldwell
- Division of Research, Department of Surgery, College of Medicine, University of Cincinnati, Cincinnati, OH, USA.
| |
Collapse
|
113
|
Gart EV, Suchodolski JS, Welsh TH, Alaniz RC, Randel RD, Lawhon SD. Salmonella Typhimurium and Multidirectional Communication in the Gut. Front Microbiol 2016; 7:1827. [PMID: 27920756 PMCID: PMC5118420 DOI: 10.3389/fmicb.2016.01827] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2016] [Accepted: 10/31/2016] [Indexed: 12/20/2022] Open
Abstract
The mammalian digestive tract is home to trillions of microbes, including bacteria, archaea, protozoa, fungi, and viruses. In monogastric mammals the stomach and small intestine harbor diverse bacterial populations but are typically less populated than the colon. The gut bacterial community (microbiota hereafter) varies widely among different host species and individuals within a species. It is influenced by season of the year, age of the host, stress and disease. Ideally, the host and microbiota benefit each other. The host provides nutrients to the microbiota and the microbiota assists the host with digestion and nutrient metabolism. The resident microbiota competes with pathogens for space and nutrients and, through this competition, protects the host in a phenomenon called colonization resistance. The microbiota participates in development of the host immune system, particularly regulation of autoimmunity and mucosal immune response. The microbiota also shapes gut–brain communication and host responses to stress; and, indeed, the microbiota is a newly recognized endocrine organ within mammalian hosts. Salmonella enterica serovar Typhimurium (S. Typhimurium hereafter) is a food-borne pathogen which adapts to and alters the gastrointestinal (GI) environment. In the GI tract, S. Typhimurium competes with the microbiota for nutrients and overcomes colonization resistance to establish infection. To do this, S. Typhimurium uses multiple defense mechanisms to resist environmental stressors, like the acidic pH of the stomach, and virulence mechanisms which allow it to invade the intestinal epithelium and disseminate throughout the host. To coordinate gene expression and disrupt signaling within the microbiota and between host and microbiota, S. Typhimurium employs its own chemical signaling and may regulate host hormone metabolism. This review will discuss the multidirectional interaction between S. Typhimurium, host and microbiota as well as mechanisms that allow S. Typhimurium to succeed in the gut.
Collapse
Affiliation(s)
- Elena V Gart
- Department of Veterinary Pathobiology, College of Veterinary Medicine, Texas A&M University, College Station TX, USA
| | - Jan S Suchodolski
- Department of Small Animal Clinical Sciences, College of Veterinary Medicine, Texas A&M University, College Station TX, USA
| | - Thomas H Welsh
- Department of Animal Science, College of Agriculture and Life Sciences, Texas A&M University, College Station TX, USA
| | - Robert C Alaniz
- Department of Microbial Pathogenesis and Immunology, College of Medicine, Texas A&M Health Science Center, Texas A&M University, College Station TX, USA
| | | | - Sara D Lawhon
- Department of Veterinary Pathobiology, College of Veterinary Medicine, Texas A&M University, College Station TX, USA
| |
Collapse
|
114
|
Chen L, Ishigami T, Nakashima-Sasaki R, Kino T, Doi H, Minegishi S, Umemura S. Commensal Microbe-specific Activation of B2 Cell Subsets Contributes to Atherosclerosis Development Independently of Lipid Metabolism. EBioMedicine 2016; 13:237-247. [PMID: 27810309 PMCID: PMC5264349 DOI: 10.1016/j.ebiom.2016.10.030] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2016] [Revised: 10/06/2016] [Accepted: 10/19/2016] [Indexed: 01/11/2023] Open
Abstract
The relation between B2 cells and commensal microbes during atherosclerosis remains largely unexplored. Here we show that under hyperlipidemic conditions intestinal microbiota resulted in recruitment and ectopic activation of B2 cells in perivascular adipose tissue, followed by an increase in circulating IgG, promoting disease development. In contrast, disruption of the intestinal microbiota by a broad-spectrum antibiotic cocktail (AVNM) led to the attenuation of atherosclerosis by suppressing B2 cells, despite the persistence of serum lipid abnormalities. Furthermore, pharmacological depletion of B2 cells with an anti-B2-cell surface CD23 antibody also attenuated commensal microbe-induced atherosclerosis. Moreover, expression analysis of TLR-signaling-related genes in the activated B2 cell subsets, assessed using the Toll-Like Receptor Signaling Pathway RT2 Profiler PCR Array, confirmed activation of the B2-cell autoantibody-production axis, which was associated with an increased capacity of B2 cells to bind to intestinal microbiota. Together, our findings reveal the critical role of commensal microbe-specific activation of B2 cells in the development of atherogenesis through lipid metabolism-independent mechanisms.
Collapse
Affiliation(s)
- Lin Chen
- Department of Medical Science and Cardio-Renal Medicine, Yokohama City University, Graduate School of Medicine, 3-9 Fukuura, Kanazawa-ku, Yokohama, Kanagawa, Japan
| | - Tomoaki Ishigami
- Department of Medical Science and Cardio-Renal Medicine, Yokohama City University, Graduate School of Medicine, 3-9 Fukuura, Kanazawa-ku, Yokohama, Kanagawa, Japan.
| | - Rie Nakashima-Sasaki
- Department of Medical Science and Cardio-Renal Medicine, Yokohama City University, Graduate School of Medicine, 3-9 Fukuura, Kanazawa-ku, Yokohama, Kanagawa, Japan
| | - Tabito Kino
- Department of Medical Science and Cardio-Renal Medicine, Yokohama City University, Graduate School of Medicine, 3-9 Fukuura, Kanazawa-ku, Yokohama, Kanagawa, Japan
| | - Hiroshi Doi
- Department of Medical Science and Cardio-Renal Medicine, Yokohama City University, Graduate School of Medicine, 3-9 Fukuura, Kanazawa-ku, Yokohama, Kanagawa, Japan
| | - Shintaro Minegishi
- Department of Medical Science and Cardio-Renal Medicine, Yokohama City University, Graduate School of Medicine, 3-9 Fukuura, Kanazawa-ku, Yokohama, Kanagawa, Japan
| | - Satoshi Umemura
- Department of Medical Science and Cardio-Renal Medicine, Yokohama City University, Graduate School of Medicine, 3-9 Fukuura, Kanazawa-ku, Yokohama, Kanagawa, Japan
| |
Collapse
|
115
|
Kubelkova K, Benuchova M, Kozakova H, Sinkora M, Krocova Z, Pejchal J, Macela A. Gnotobiotic mouse model's contribution to understanding host-pathogen interactions. Cell Mol Life Sci 2016; 73:3961-9. [PMID: 27544211 PMCID: PMC11108488 DOI: 10.1007/s00018-016-2341-8] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2016] [Revised: 07/25/2016] [Accepted: 08/11/2016] [Indexed: 02/07/2023]
Abstract
This brief review is dedicated to the legacy of Prof. Jaroslav Šterzl and his colleagues, who laid the foundation for gnotobiology in the former Czechoslovakia 55 years. Prof. Sterzl became one of the founders of modern Czechoslovak immunology, which was characterized by work on a wide range of problems needing to be solved. While examining the mechanisms of innate immunity, he focused his studies on the induction of antibody production by immunocompetent cells involved in adaptive immune transmission while using the model of pig fetuses and germ-free piglets and characterizing immunoglobulins in the sera of these piglets. Although not fully appreciated to this day, his experimental proof of the hypothesis focused on the common precursor of cell-forming antibodies of different isotypes was later confirmed in experiments at the gene level. Prof. Sterzl's work represented a true milestone in the development of not solely Czechoslovak but also European and global immunology. He collaborated closely with the World Health Organization for many years, serving there as leader of the Reference Laboratory for Factors of Innate Immunity.
Collapse
Affiliation(s)
- Klara Kubelkova
- Department of Molecular Pathology and Biology, Faculty of Military Health Sciences, University of Defence, 1575 Trebesska, 500 01, Hradec Kralove, Czech Republic.
| | - Milota Benuchova
- Department of Molecular Pathology and Biology, Faculty of Military Health Sciences, University of Defence, 1575 Trebesska, 500 01, Hradec Kralove, Czech Republic
| | - Hana Kozakova
- Laboratory of Gnotobiology, Czech Academy of Sciences, Institute of Microbiology, 549 22, Novy Hradek, Czech Republic
| | - Marek Sinkora
- Laboratory of Gnotobiology, Czech Academy of Sciences, Institute of Microbiology, 549 22, Novy Hradek, Czech Republic
| | - Zuzana Krocova
- Department of Molecular Pathology and Biology, Faculty of Military Health Sciences, University of Defence, 1575 Trebesska, 500 01, Hradec Kralove, Czech Republic
| | - Jaroslav Pejchal
- Department of Toxicology and Military Pharmacy, Faculty of Military Health Sciences, University of Defence, 1575 Trebesska, 500 01, Hradec Kralove, Czech Republic
| | - Ales Macela
- Department of Molecular Pathology and Biology, Faculty of Military Health Sciences, University of Defence, 1575 Trebesska, 500 01, Hradec Kralove, Czech Republic
| |
Collapse
|
116
|
Abstract
BACKGROUND The interplay between host genetics, immunity, and microbiota is central to the pathogenesis of inflammatory bowel disease. Previous population-based studies suggested a link between antibiotic use and increased inflammatory bowel disease risk, but the mechanisms are unknown. The purpose of this study was to determine the long-term effects of antibiotic administration on microbiota composition, innate immunity, and susceptibility to colitis, as well as the mechanism by which antibiotics alter host colitogenicity. METHODS Wild-type mice were given broad-spectrum antibiotics or no antibiotics for 2 weeks, and subsequent immunophenotyping and 16S rRNA gene sequencing-based analysis of the fecal microbiome were performed 6 weeks later. In a separate experiment, control and antibiotic-treated mice were given 7 days of dextran sulfate sodium, 6 weeks after completing antibiotic treatment, and the severity of colitis scored histologically. Fecal transfer was performed from control or antibiotic-treated mice to recipient mice whose endogenous microbiota had been cleared with antibiotics, and the susceptibility of the recipients to dextran sulfate sodium-induced colitis was analyzed. Naive CD4 T cells were transferred from control and antibiotic-treated mice to immunodeficient Rag-1 recipients and the severity of colitis compared. RESULTS Antibiotics led to sustained dysbiosis and changes in T-cell subpopulations, including reductions in colonic lamina propria total T cells and CD4 T cells. Antibiotics conferred protection against dextran sulfate sodium colitis, and this effect was transferable by fecal transplant but not by naive T cells. CONCLUSIONS Antibiotic exposure protects against colitis, and this effect is transferable with fecal microbiota from antibiotic-treated mice, supporting a protective effect of the microbial community.
Collapse
|
117
|
Rajagopala SV, Yooseph S, Harkins DM, Moncera KJ, Zabokrtsky KB, Torralba MG, Tovchigrechko A, Highlander SK, Pieper R, Sender L, Nelson KE. Gastrointestinal microbial populations can distinguish pediatric and adolescent Acute Lymphoblastic Leukemia (ALL) at the time of disease diagnosis. BMC Genomics 2016; 17:635. [PMID: 27527070 PMCID: PMC4986186 DOI: 10.1186/s12864-016-2965-y] [Citation(s) in RCA: 97] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2016] [Accepted: 07/22/2016] [Indexed: 12/12/2022] Open
Abstract
Background An estimated 15,000 children and adolescents under the age of 19 years are diagnosed with leukemia, lymphoma and other tumors in the USA every year. All children and adolescent acute leukemia patients will undergo chemotherapy as part of their treatment regimen. Fortunately, survival rates for most pediatric cancers have improved at a remarkable pace over the past three decades, and the overall survival rate is greater than 90 % today. However, significant differences in survival rate have been found in different age groups (94 % in 1–9.99 years, 82 % in ≥10 years and 76 % in ≥15 years). ALL accounts for about three out of four cases of childhood leukemia. Intensive chemotherapy treatment coupled with prophylactic or therapeutic antibiotic use could potentially have a long-term effect on the resident gastrointestinal (GI) microbiome. The composition of GI microbiome and its changes upon chemotherapy in pediatric and adolescent leukemia patients is poorly understood. In this study, using 16S rRNA marker gene sequences we profile the GI microbial communities of pediatric and adolescent acute leukemia patients before and after chemotherapy treatment and compare with the microbiota of their healthy siblings. Results Our study cohort consisted of 51 participants, made up of matched pediatric and adolescent patients with ALL and a healthy sibling. We elucidated and compared the GI microbiota profiles of patients and their healthy sibling controls via analysis of 16S rRNA gene sequencing data. We assessed the GI microbiota composition in pediatric and adolescent patients with ALL during the course of chemotherapy by comparing stool samples taken before chemotherapy with stool samples collected at varying time points during the chemotherapeutic treatment. The microbiota profiles of both patients and control sibling groups are dominated by members of Bacteroides, Prevotella, and Faecalibacterium. At the genus level, both groups share many taxa in common, but the microbiota diversity of the patient group is significantly lower than that of the control group. It was possible to distinguish between the patient and control groups based on their microbiota profiles. The top taxa include Anaerostipes, Coprococcus, Roseburia, and Ruminococcus2 with relatively higher abundance in the control group. The observed microbiota changes are likely the result of several factors including a direct influence of therapeutic compounds on the gut flora and an indirect effect of chemotherapy on the immune system, which, in turn, affects the microbiota. Conclusions This study provides significant information on GI microbiota populations in immunocompromised children and opens up the potential for developing novel diagnostics based on stool tests and therapies to improve the dysbiotic condition of the microbiota at the time of diagnosis and in the earliest stages of chemotherapy. Electronic supplementary material The online version of this article (doi:10.1186/s12864-016-2965-y) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
| | | | - Derek M Harkins
- J. Craig Venter Institute (JCVI), 9714 Medical Center Drive, Rockville, MD, 20850, USA
| | | | - Keri B Zabokrtsky
- Hyundai Cancer Genomics Center, Children's Hospital Orange County (CHOC Children's), Orange, CA, USA.,Division of Hematology-Oncology, Department of Medicine, School of Medicine, University of California-Irvine, Orange, CA, USA
| | | | - Andrey Tovchigrechko
- J. Craig Venter Institute (JCVI), 9714 Medical Center Drive, Rockville, MD, 20850, USA
| | | | - Rembert Pieper
- J. Craig Venter Institute (JCVI), 9714 Medical Center Drive, Rockville, MD, 20850, USA
| | - Leonard Sender
- Hyundai Cancer Genomics Center, Children's Hospital Orange County (CHOC Children's), Orange, CA, USA.,Division of Hematology-Oncology, Department of Medicine, School of Medicine, University of California-Irvine, Orange, CA, USA.,Division of Oncology, Hyundai Cancer Institute, CHOC Children's, Orange, CA, USA.,Department of Pediatrics, School of Medicine, University of California-Irvine, Orange, CA, USA
| | - Karen E Nelson
- J. Craig Venter Institute (JCVI), 9714 Medical Center Drive, Rockville, MD, 20850, USA. .,JCVI, La Jolla, 92037, CA, USA.
| |
Collapse
|
118
|
Lam V, Su J, Hsu A, Gross GJ, Salzman NH, Baker JE. Intestinal Microbial Metabolites Are Linked to Severity of Myocardial Infarction in Rats. PLoS One 2016; 11:e0160840. [PMID: 27505423 PMCID: PMC4978455 DOI: 10.1371/journal.pone.0160840] [Citation(s) in RCA: 107] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2016] [Accepted: 07/26/2016] [Indexed: 12/13/2022] Open
Abstract
Intestinal microbiota determine severity of myocardial infarction in rats. We determined whether low molecular weight metabolites derived from intestinal microbiota and transported to the systemic circulation are linked to severity of myocardial infarction. Plasma from rats treated for seven days with the non-absorbed antibiotic vancomycin or a mixture of streptomycin, neomycin, polymyxin B and bacitracin was analyzed using mass spectrometry-based metabolite profiling platforms. Antibiotic-induced changes in the abundance of individual groups of intestinal microbiota dramatically altered the host’s metabolism. Hierarchical clustering of dissimilarities separated the levels of 284 identified metabolites from treated vs. untreated rats; 193 were altered by the antibiotic treatments with a tendency towards decreased metabolite levels. Catabolism of the aromatic amino acids phenylalanine, tryptophan and tyrosine was the most affected pathway comprising 33 affected metabolites. Both antibiotic treatments decreased the severity of an induced myocardial infarction in vivo by 27% and 29%, respectively. We then determined whether microbial metabolites of the amino acids phenylalanine, tryptophan and tyrosine were linked to decreased severity of myocardial infarction. Vancomycin-treated rats were administered amino acid metabolites prior to ischemia/reperfusion studies. Oral or intravenous pretreatment of rats with these amino acid metabolites abolished the decrease in infarct size conferred by vancomycin. Inhibition of JAK-2 (AG-490, 10 μM), Src kinase (PP1, 20 μM), Akt/PI3 kinase (Wortmannin, 100 nM), p44/42 MAPK (PD98059, 10 μM), p38 MAPK (SB203580, 10 μM), or KATP channels (glibenclamide, 3 μM) abolished cardioprotection by vancomycin, indicating microbial metabolites are interacting with cell surface receptors to transduce their signals through Src kinase, cell survival pathways and KATP channels. These inhibitors have no effect on myocardial infarct size in untreated rats. This study links gut microbiota metabolites to severity of myocardial infarction and may provide future opportunities for novel diagnostic tests and interventions for the prevention of cardiovascular disease.
Collapse
Affiliation(s)
- Vy Lam
- Division of Cardiothoracic Surgery, Medical College of Wisconsin, Milwaukee, WI, United States of America
| | - Jidong Su
- Division of Cardiothoracic Surgery, Medical College of Wisconsin, Milwaukee, WI, United States of America
| | - Anna Hsu
- Department of Pharmacology and Toxicology, Medical College of Wisconsin, Milwaukee, WI, United States of America
| | - Garrett J Gross
- Department of Pharmacology and Toxicology, Medical College of Wisconsin, Milwaukee, WI, United States of America
| | - Nita H Salzman
- Division of Pediatric Gastroenterology, Medical College of Wisconsin, Milwaukee, WI, United States of America
| | - John E Baker
- Division of Cardiothoracic Surgery, Medical College of Wisconsin, Milwaukee, WI, United States of America.,Department of Pharmacology and Toxicology, Medical College of Wisconsin, Milwaukee, WI, United States of America.,Department of Biochemistry, Medical College of Wisconsin, Milwaukee, WI, United States of America.,The Cardiovascular Center, Medical College of Wisconsin, Milwaukee, WI, United States of America
| |
Collapse
|
119
|
Knoop KA, McDonald KG, Kulkarni DH, Newberry RD. Antibiotics promote inflammation through the translocation of native commensal colonic bacteria. Gut 2016; 65:1100-9. [PMID: 26045138 PMCID: PMC4670297 DOI: 10.1136/gutjnl-2014-309059] [Citation(s) in RCA: 182] [Impact Index Per Article: 20.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2014] [Accepted: 05/17/2015] [Indexed: 12/11/2022]
Abstract
OBJECTIVE Antibiotic use is associated with an increased risk of developing multiple inflammatory disorders, which in turn are linked to alterations in the intestinal microbiota. How these alterations in the intestinal microbiota translate into an increased risk for inflammatory responses is largely unknown. Here we investigated whether and how antibiotics promote inflammation via the translocation of live native gut commensal bacteria. DESIGN Oral antibiotics were given to wildtype and induced mutant mouse strains, and the effects on bacterial translocation, inflammatory responses and the susceptibility to colitis were evaluated. The sources of the bacteria and the pathways required for bacterial translocation were evaluated using induced mutant mouse strains, 16s rRNA sequencing to characterise the microbial communities, and in vivo and ex vivo imaging techniques. RESULTS Oral antibiotics induced the translocation of live native commensal bacteria across the colonic epithelium, promoting inflammatory responses, and predisposing to increased disease in response to coincident injury. Bacterial translocation resulted from decreased microbial signals delivered to colonic goblet cells (GCs), was associated with the formation of colonic GC-associated antigen passages, was abolished when GCs were depleted and required CX3CR1(+) dendritic cells. Bacterial translocation occurred following a single dose of most antibiotics tested, and the predisposition for increased inflammation was only associated with antibiotics inducing bacterial translocation. CONCLUSIONS These findings reveal an unexpected outcome of antibiotic therapy and suggest that bacterial translocation as a result of alterations in the intestinal microflora may provide a link between increasing antibiotic use and the increased incidence of inflammatory disorders.
Collapse
Affiliation(s)
- Kathryn A Knoop
- Department of Internal Medicine, Washington University School of Medicine, St. Louis, Missouri, USA, 63110
| | - Keely G McDonald
- Department of Internal Medicine, Washington University School of Medicine, St. Louis, Missouri, USA, 63110
| | - Devesha H Kulkarni
- Department of Internal Medicine, Washington University School of Medicine, St. Louis, Missouri, USA, 63110
| | - Rodney D Newberry
- Department of Internal Medicine, Washington University School of Medicine, St. Louis, Missouri, USA, 63110,Corresponding author: Rodney Newberry MD, Washington University School of Medicine, 660 S. Euclid Ave. Campus Box 8124, St. Louis, MO 63110, Ph: 314 362-2671 Fax: 314 362-2609,
| |
Collapse
|
120
|
Fecal Microbiota and Diet of Children with Chronic Constipation. Int J Pediatr 2016; 2016:6787269. [PMID: 27418934 PMCID: PMC4935906 DOI: 10.1155/2016/6787269] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/29/2015] [Accepted: 05/18/2016] [Indexed: 12/12/2022] Open
Abstract
Many factors explain dysbiosis in chronic constipation (CC), such as a low-fiber diet. The objective of this study was to compare the fecal microbiota of constipated and nonconstipated children and their intake frequencies of food. Methods. This observational study included 79 children (M/F 43/36) aged six to 36 months divided into two groups: cases (39 constipated children) and controls (40 nonconstipated children). We used a structured form to collect demographic variables, conducted anthropometric assessment, and collected food intake frequency data. The fecal microbiota of the stool samples was analyzed by real-time polymerase chain reaction (PCR) using the fluorophore SYBR® Green. Results. Constipated children had a smaller concentration of Lactobacillus per milligram of stool (p = 0.015) than nonconstipated children, but the concentration of Bifidobacterium per milligram of stool (p = 0.323) and the intake of fruits, vegetables (p = 0.563), and junk food (p = 0.093) of the two groups did not differ. Constipated children consumed more dairy products (0.45 ± 0.8; p > 0.001), were more frequently delivered via caesarean section (69.2%), were weaned earlier (median: 120; 60Q1–240Q3), and had a family history of constipation (71.8%). Conclusions. Children with CC have a smaller concentration of Lactobacillus in their stools and consume more dairy products.
Collapse
|
121
|
A Pathogen-Selective Antibiotic Minimizes Disturbance to the Microbiome. Antimicrob Agents Chemother 2016; 60:4264-73. [PMID: 27161626 DOI: 10.1128/aac.00535-16] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2016] [Accepted: 04/18/2016] [Indexed: 01/18/2023] Open
Abstract
Broad-spectrum antibiotic therapy decimates the gut microbiome, resulting in a variety of negative health consequences. Debio 1452 is a staphylococcus-selective enoyl-acyl carrier protein reductase (FabI) inhibitor under clinical development and was used to determine whether treatment with pathogen-selective antibiotics would minimize disturbance to the microbiome. The effect of oral Debio 1452 on the microbiota of mice was compared to the effects of four commonly used broad-spectrum oral antibiotics. During the 10 days of oral Debio 1452 treatment, there was minimal disturbance to the gut bacterial abundance and composition, with only the unclassified S24-7 taxon reduced at days 6 and 10. In comparison, broad-spectrum oral antibiotics caused ∼100- to 4,000-fold decreases in gut bacterial abundance and severely altered the microbial composition. The gut bacterial abundance and composition of Debio 1452-treated mice were indistinguishable from those of untreated mice 2 days after the antibiotic treatment was stopped. In contrast, the bacterial abundance in broad-spectrum-antibiotic-treated mice took up to 7 days to recover, and the gut composition of the broad-spectrum-antibiotic-treated mice remained different from that of the control group 20 days after the cessation of antibiotic treatment. These results illustrate that a pathogen-selective approach to antibiotic development will minimize disturbance to the gut microbiome.
Collapse
|
122
|
A single gene of a commensal microbe affects host susceptibility to enteric infection. Nat Commun 2016; 7:11606. [PMID: 27173141 PMCID: PMC5482719 DOI: 10.1038/ncomms11606] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2015] [Accepted: 04/11/2016] [Indexed: 02/07/2023] Open
Abstract
Indigenous microbes inside the host intestine maintain a complex self-regulating
community. The mechanisms by which gut microbes interact with intestinal pathogens
remain largely unknown. Here we identify a commensal Escherichia coli strain
whose expansion predisposes mice to infection by Vibrio cholerae, a human
pathogen. We refer to this strain as ‘atypical’ E. coli
(atEc) because of its inability to ferment lactose. The atEc
strain is resistant to reactive oxygen species (ROS) and proliferates extensively in
antibiotic-treated adult mice. V. cholerae infection is more severe in
neonatal mice transplanted with atEc compared with those transplanted with a
typical E. coli strain. Intestinal ROS levels are decreased in
atEc-transplanted mice, favouring proliferation of ROS-sensitive V.
cholerae. An atEc mutant defective in ROS degradation fails to
facilitate V. cholerae infection when transplanted, suggesting that host
infection susceptibility can be regulated by a single gene product of one particular
commensal species. The interactions between gut bacteria and enteric pathogens are poorly
understood. Here, Yoon et al. show that subinhibitory antibiotic treatment in a
mouse model leads to overgrowth of an E. coli strain carrying a catalase-encoding
gene that enhances infection with the human pathogen Vibrio cholerae.
Collapse
|
123
|
Galtier M, De Sordi L, Maura D, Arachchi H, Volant S, Dillies MA, Debarbieux L. Bacteriophages to reduce gut carriage of antibiotic resistant uropathogens with low impact on microbiota composition. Environ Microbiol 2016; 18:2237-45. [PMID: 26971586 DOI: 10.1111/1462-2920.13284] [Citation(s) in RCA: 104] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2016] [Accepted: 02/26/2016] [Indexed: 12/30/2022]
Abstract
Uropathogenic Escherichia coli (UPEC) is the leading cause of urinary tract infections (UTIs) worldwide, causing over 150 million clinical cases annually. There is currently no specific treatment addressing the asymptomatic carriage in the gut of UPEC before they initiate UTIs. This study investigates the efficacy of virulent bacteriophages to decrease carriage of gut pathogens. Three virulent bacteriophages infecting an antibiotic-resistant UPEC strain were isolated and characterized both in vitro and in vivo. A new experimental murine model of gut carriage of E. coli was elaborated and the impact of virulent bacteriophages on colonization levels and microbiota diversity was assessed. A single dose of a cocktail of the three bacteriophages led to a sharp decrease in E. coli levels throughout the gut. We also observed that microbiota diversity was much less affected by bacteriophages than by antibiotics. Therefore, virulent bacteriophages can efficiently target UPEC strains residing in the gut, with potentially profound public health and economic impacts. These results open a new area with the possibility to manipulate specifically the microbiota using virulent bacteriophages, which could have broad applications in many gut-related disorders/diseases and beyond.
Collapse
Affiliation(s)
- Matthieu Galtier
- Department of Microbiology, Institut Pasteur, Paris, France.,Université Paris Diderot, Sorbonne Paris Cité, Cellule Pasteur, Paris, France
| | - Luisa De Sordi
- Department of Microbiology, Institut Pasteur, Paris, France
| | - Damien Maura
- Department of Microbiology, Institut Pasteur, Paris, France.,Université Paris Diderot, Sorbonne Paris Cité, Cellule Pasteur, Paris, France
| | - Harindra Arachchi
- Institut Pasteur Hub Bioinformatique et Biostatistique - C3BI, USR 3756 IP CNRS - Paris, France
| | - Stevenn Volant
- Institut Pasteur Hub Bioinformatique et Biostatistique - C3BI, USR 3756 IP CNRS - Paris, France
| | - Marie-Agnès Dillies
- Institut Pasteur Hub Bioinformatique et Biostatistique - C3BI, USR 3756 IP CNRS - Paris, France
| | - Laurent Debarbieux
- Institut Pasteur Hub Bioinformatique et Biostatistique - C3BI, USR 3756 IP CNRS - Paris, France
| |
Collapse
|
124
|
Gonzalez-Perez G, Hicks AL, Tekieli TM, Radens CM, Williams BL, Lamousé-Smith ESN. Maternal Antibiotic Treatment Impacts Development of the Neonatal Intestinal Microbiome and Antiviral Immunity. THE JOURNAL OF IMMUNOLOGY 2016; 196:3768-79. [PMID: 27036912 DOI: 10.4049/jimmunol.1502322] [Citation(s) in RCA: 104] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/30/2015] [Accepted: 02/29/2016] [Indexed: 12/20/2022]
Abstract
Microbial colonization of the infant gastrointestinal tract (GIT) begins at birth, is shaped by the maternal microbiota, and is profoundly altered by antibiotic treatment. Antibiotic treatment of mothers during pregnancy influences colonization of the GIT microbiota of their infants. The role of the GIT microbiota in regulating adaptive immune function against systemic viral infections during infancy remains undefined. We used a mouse model of perinatal antibiotic exposure to examine the effect of GIT microbial dysbiosis on infant CD8(+) T cell-mediated antiviral immunity. Maternal antibiotic treatment/treated (MAT) during pregnancy and lactation resulted in profound alterations in the composition of the GIT microbiota in mothers and infants. Streptococcus spp. dominated the GIT microbiota of MAT mothers, whereas Enterococcus faecalis predominated within the MAT infant GIT. MAT infant mice subsequently exhibited increased and accelerated mortality following vaccinia virus infection. Ag-specific IFN-γ-producing CD8(+) T cells were reduced in sublethally infected MAT infant mice. MAT CD8(+) T cells from uninfected infant mice also demonstrated a reduced capacity to sustain IFN-γ production following in vitro activation. We additionally determined that control infant mice became more susceptible to infection if they were born in an animal facility using stricter standards of hygiene. These data indicate that undisturbed colonization and progression of the GIT microbiota during infancy are necessary to promote robust adaptive antiviral immune responses.
Collapse
Affiliation(s)
- Gabriela Gonzalez-Perez
- Division of Pediatric Gastroenterology, Hepatology, and Nutrition, Department of Pediatrics, Columbia University, New York, NY 10032; and
| | - Allison L Hicks
- Center for Infection and Immunity, Columbia University, New York, NY 10032
| | - Tessa M Tekieli
- Division of Pediatric Gastroenterology, Hepatology, and Nutrition, Department of Pediatrics, Columbia University, New York, NY 10032; and
| | - Caleb M Radens
- Division of Pediatric Gastroenterology, Hepatology, and Nutrition, Department of Pediatrics, Columbia University, New York, NY 10032; and
| | - Brent L Williams
- Center for Infection and Immunity, Columbia University, New York, NY 10032
| | - Esi S N Lamousé-Smith
- Division of Pediatric Gastroenterology, Hepatology, and Nutrition, Department of Pediatrics, Columbia University, New York, NY 10032; and
| |
Collapse
|
125
|
Yao J, Rock CO. Resistance Mechanisms and the Future of Bacterial Enoyl-Acyl Carrier Protein Reductase (FabI) Antibiotics. Cold Spring Harb Perspect Med 2016; 6:a027045. [PMID: 26931811 DOI: 10.1101/cshperspect.a027045] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
Missense mutations leading to clinical antibiotic resistance are a liability of single-target inhibitors. The enoyl-acyl carrier protein reductase (FabI) inhibitors have one intracellular protein target and drug resistance is increased by the acquisition of single-base-pair mutations that alter drug binding. The spectrum of resistance mechanisms to FabI inhibitors suggests criteria that should be considered during the development of single-target antibiotics that would minimize the impact of missense mutations on their clinical usefulness. These criteria include high-affinity, fast on/off kinetics, few drug contacts with residue side chains, and no toxicity. These stringent criteria are achievable by structure-guided design, but this approach will only yield pathogen-specific drugs. Single-step acquisition of resistance may limit the clinical application of broad-spectrum, single-target antibiotics, but appropriately designed pathogen-specific antibiotics have the potential to overcome this liability.
Collapse
Affiliation(s)
- Jiangwei Yao
- Department of Infectious Diseases, St. Jude Children's Research Hospital, Memphis, Tennessee 38105
| | - Charles O Rock
- Department of Infectious Diseases, St. Jude Children's Research Hospital, Memphis, Tennessee 38105
| |
Collapse
|
126
|
Troche C, Aydemir TB, Cousins RJ. Zinc transporter Slc39a14 regulates inflammatory signaling associated with hypertrophic adiposity. Am J Physiol Endocrinol Metab 2016; 310:E258-68. [PMID: 26646099 PMCID: PMC4971811 DOI: 10.1152/ajpendo.00421.2015] [Citation(s) in RCA: 49] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/23/2015] [Accepted: 12/02/2015] [Indexed: 12/20/2022]
Abstract
Zinc is a signaling molecule in numerous metabolic pathways, the coordination of which occurs through activity of zinc transporters. The expression of zinc transporter Zip14 (Slc39a14), a zinc importer of the solute carrier 39 family, is stimulated under proinflammatory conditions. Adipose tissue upregulates Zip14 during lipopolysaccharide-induced endotoxemia. A null mutation of Zip14 (KO) revealed that phenotypic changes in adipose include increased cytokine production, increased plasma leptin, hypertrophied adipocytes, and dampened insulin signaling. Adipose tissue from KO mice had increased levels of preadipocyte markers, lower expression of the differentiation marker (PPARγ), and activation of NF-κB and STAT3 pathways. Our overall hypothesis was that ZIP14 would play a role in adipocyte differentiation and inflammatory obesity. Global Zip14 KO causes systemic endotoxemia. The observed metabolic changes in adipose metabolism were reversed when oral antibiotics were administrated, indicating that circulating levels of endotoxin were in part responsible for the adipose phenotype. To evaluate a mechanism, 3T3-L1 cells were differentiated into adipocytes and treated with siRNA to knock down Zip14. These cells had an impaired ability to mobilize zinc, which caused dysregulation of inflammatory pathways (JAK2/STAT3 and NF-κB). The Zip14 deletion may limit the availability of intracellular zinc, yielding the unique phenotype of inflammation coupled with hypertrophy. Taken together, these results suggest that aberrant zinc distribution observed with Zip14 ablation impacts adipose cytokine production and metabolism, ultimately increasing fat deposition when exposed to endotoxin. To our knowledge, this is the first investigation into the mechanistic role of ZIP14 in adipose tissue regulation and metabolism.
Collapse
Affiliation(s)
- Catalina Troche
- Food Science and Human Nutrition Department and Center for Nutritional Sciences, College of Agricultural and Life Sciences, University of Florida, Gainesville, Florida
| | - Tolunay Beker Aydemir
- Food Science and Human Nutrition Department and Center for Nutritional Sciences, College of Agricultural and Life Sciences, University of Florida, Gainesville, Florida
| | - Robert J Cousins
- Food Science and Human Nutrition Department and Center for Nutritional Sciences, College of Agricultural and Life Sciences, University of Florida, Gainesville, Florida
| |
Collapse
|
127
|
Abstract
Antibiotics are by far the most common medications prescribed for children. Recent epidemiological data suggests an association between early antibiotic use and disease phenotypes in adulthood. Antibiotic use during infancy induces imbalances in gut microbiota, called dysbiosis. The gut microbiome's responses to antibiotics and its potential link to disease development are especially complex to study in the changing infant gut. Here, we synthesize current knowledge linking antibiotics, dysbiosis, and disease and propose a framework for studying antibiotic-related dysbiosis in children. We recommend future studies into the microbiome-mediated effects of antibiotics focused on four types of dysbiosis: loss of keystone taxa, loss of diversity, shifts in metabolic capacity, and blooms of pathogens. Establishment of a large and diverse baseline cohort to define healthy infant microbiome development is essential to advancing diagnosis, interpretation, and eventual treatment of pediatric dysbiosis. This approach will also help provide evidence-based recommendations for antibiotic usage in infancy.
Collapse
Affiliation(s)
- Pajau Vangay
- Biomedical Informatics and Computational Biology, University of Minnesota, Minneapolis, MN 55455, USA
| | - Tonya Ward
- Biotechnology Institute, University of Minnesota, Saint Paul, MN 55108, USA
| | - Jeffrey S Gerber
- Division of Infectious Diseases, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Dan Knights
- Biotechnology Institute, University of Minnesota, Saint Paul, MN 55108, USA; Department of Computer Science and Engineering, University of Minnesota, Minneapolis, MN 55455, USA.
| |
Collapse
|
128
|
Wellehan • JF, Lierz • M, Phalen • D, Raidal • S, Styles • DK, Crosta • L, Melillo • A, Schnitzer • P, Lennox • A, Lumeij JT. Infectious disease. CURRENT THERAPY IN AVIAN MEDICINE AND SURGERY 2016. [PMCID: PMC7158187 DOI: 10.1016/b978-1-4557-4671-2.00011-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/04/2022]
|
129
|
DDX5 and its associated lncRNA Rmrp modulate TH17 cell effector functions. Nature 2015; 528:517-22. [PMID: 26675721 PMCID: PMC4762670 DOI: 10.1038/nature16193] [Citation(s) in RCA: 134] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2015] [Accepted: 11/02/2015] [Indexed: 12/12/2022]
Abstract
T helper 17 (TH17) lymphocytes protect mucosal barriers from infections, but also contribute to multiple chronic inflammatory diseases. Their differentiation is controlled by RORγt, a ligand-regulated nuclear receptor. Here we identify the RNA helicase DEAD-box protein 5 (DDX5) as a RORγt partner that coordinates transcription of selective TH17 genes, and is required for TH17-mediated inflammatory pathologies. Surprisingly, the ability of DDX5 to interact with RORγt and coactivate its targets depends on intrinsic RNA helicase activity and binding of a conserved nuclear long noncoding RNA (lncRNA), Rmrp, which is mutated in patients with cartilage-hair hypoplasia. A targeted Rmrp gene mutation in mice, corresponding to a gene mutation in cartilage-hair hypoplasia patients, altered lncRNA chromatin occupancy, and reduced the DDX5-RORγt interaction and RORγt target gene transcription. Elucidation of the link between Rmrp and the DDX5-RORγt complex reveals a role for RNA helicases and lncRNAs in tissue-specific transcriptional regulation, and provides new opportunities for therapeutic intervention in TH17-dependent diseases.
Collapse
|
130
|
Bacteriocin production augments niche competition by enterococci in the mammalian gastrointestinal tract. Nature 2015; 526:719-22. [PMID: 26479034 PMCID: PMC4978352 DOI: 10.1038/nature15524] [Citation(s) in RCA: 315] [Impact Index Per Article: 31.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2015] [Accepted: 08/25/2015] [Indexed: 12/21/2022]
Abstract
Enterococcus faecalis (EF) is both a common commensal of the human gastrointestinal tract (GI) and a leading cause of hospital acquired infections1. Systemic infections with multi-drug resistant enterococci occur subsequent to GI colonization2. Preventing colonization by multi-drug resistant EF could therefore be a valuable approach to limiting infection. However, little is known about mechanisms EF uses to colonize and compete for stable gastrointestinal niches. Pheromone-responsive, conjugative plasmids encoding bacteriocins are common among enterococcal strains3, and could modulate niche competition among enterococci or between enterococci and the intestinal microbiota. We developed a model of mouse gut colonization with EF without disrupting the microbiota, to evaluate the role of the conjugative plasmid pPD1 expressing bacteriocin 214 on enterococcal colonization. Here we show that EF harboring pPD1 replaces indigenous enterococci and outcompetes EF lacking pPD1. Furthermore, in the intestine, pPD1 is transferred to other EF strains by conjugation, enhancing their survival. Moreover, colonization with an EF strain carrying a conjugation-defective pPD1 mutant resulted in clearance of vancomycin-resistant enterococci, without plasmid transfer. Therefore bacteriocin expression by commensal bacteria can influence niche-competition in the GI tract, and bacteriocins, delivered by commensals that occupy a precise intestinal bacterial niche, may be an effective therapeutic approach to specifically eliminate intestinal colonization by multi-drug resistant bacteria, without profound disruption of the indigenous microbiota.
Collapse
|
131
|
Ray A, Basu S, Gharaibeh RZ, Cook LC, Kumar R, Lefkowitz EJ, Walker CR, Morrow CD, Franklin CL, Geiger TL, Salzman NH, Fodor A, Dittel BN. Gut Microbial Dysbiosis Due to Helicobacter Drives an Increase in Marginal Zone B Cells in the Absence of IL-10 Signaling in Macrophages. THE JOURNAL OF IMMUNOLOGY 2015; 195:3071-85. [PMID: 26324769 DOI: 10.4049/jimmunol.1500153] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/22/2015] [Accepted: 07/24/2015] [Indexed: 12/22/2022]
Abstract
It is clear that IL-10 plays an essential role in maintaining homeostasis in the gut in response to the microbiome. However, it is unknown whether IL-10 also facilitates immune homeostasis at distal sites. To address this question, we asked whether splenic immune populations were altered in IL-10-deficient (Il10(-/-)) mice in which differences in animal husbandry history were associated with susceptibility to spontaneous enterocolitis that is microbiome dependent. The susceptible mice exhibited a significant increase in splenic macrophages, neutrophils, and marginal zone (MZ) B cells that was inhibited by IL-10 signaling in myeloid, but not B cells. The increase in macrophages was due to increased proliferation that correlated with a subsequent enhancement in MZ B cell differentiation. Cohousing and antibiotic treatment studies suggested that the alteration in immune homeostasis in the spleen was microbiome dependent. The 16S rRNA sequencing revealed that susceptible mice harbored a different microbiome with a significant increase in the abundance of the bacterial genus Helicobacter. The introduction of Helicobacter hepaticus to the gut of nonsusceptible mice was sufficient to drive macrophage expansion and MZ B cell development. Given that myeloid cells and MZ B cells are part of the first line of defense against blood-borne pathogens, their increase following a breach in the gut epithelial barrier would be protective. Thus, IL-10 is an essential gatekeeper that maintains immune homeostasis at distal sites that can become functionally imbalanced upon the introduction of specific pathogenic bacteria to the intestinal track.
Collapse
Affiliation(s)
- Avijit Ray
- Blood Research Institute, BloodCenter of Wisconsin, Milwaukee, WI 53201
| | - Sreemanti Basu
- Blood Research Institute, BloodCenter of Wisconsin, Milwaukee, WI 53201
| | - Raad Z Gharaibeh
- Bioinformatics Services Division, Department of Bioinformatics and Genomics, University of North Carolina at Charlotte, Charlotte, NC 28223; Department of Bioinformatics and Genomics, University of North Carolina at Charlotte, Charlotte, NC 28223
| | - Lydia C Cook
- Department of Veterinary Pathobiology, University of Missouri, Columbia, MO 65211
| | - Ranjit Kumar
- Center for Clinical and Translational Sciences, University of Alabama at Birmingham, Birmingham, AL 35233
| | - Elliot J Lefkowitz
- Department of Cell, Developmental, and Integrative Biology, University of Alabama at Birmingham, Birmingham AL 35233
| | - Catherine R Walker
- Faculty of Life Sciences, University of Manchester, Manchester M13 9PL, United Kingdom
| | - Casey D Morrow
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, AL 35233
| | - Craig L Franklin
- Department of Veterinary Pathobiology, University of Missouri, Columbia, MO 65211
| | - Terrence L Geiger
- Department of Pathology, St. Jude Children's Research Hospital, Memphis, TN 38105; and
| | - Nita H Salzman
- Division of Gastroenterology, Department of Pediatrics, Medical College of Wisconsin, Milwaukee, WI 53226
| | - Anthony Fodor
- Department of Bioinformatics and Genomics, University of North Carolina at Charlotte, Charlotte, NC 28223
| | - Bonnie N Dittel
- Blood Research Institute, BloodCenter of Wisconsin, Milwaukee, WI 53201;
| |
Collapse
|
132
|
Hammami R, Ben Abdallah N, Barbeau J, Fliss I. Symbiotic maple saps minimize disruption of the mice intestinal microbiota after oral antibiotic administration. Int J Food Sci Nutr 2015. [PMID: 26218660 DOI: 10.3109/09637486.2015.1071340] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
This study was undertaken to evaluate the in vivo impact of new symbiotic products based on liquid maple sap or its concentrate. Sap and concentrate, with or without inulin (2%), were inoculated with Bifidobacterium lactis Bb12 and Lactobacillus rhamnosus GG valio at initial counts of 2-4 × 10(8) cfu mL(-1). The experiments started with intra-gastric administration of antibiotic (kanamycin 40 mg in 0.1 cc) (to induce microbiota disturbance and/or diarrhea) to 3-to-5-week-old C57BL/6 female mice followed by a combination of prebiotic and probiotics included in the maple sap or its concentrate for a week. The combination inulin and probiotics in maple sap and concentrate appeared to minimize the antibiotic-induced breakdown of mice microbiota with a marked effect on bifidobacterium and bacteroides levels, thus permitting a more rapid re-establishment of the baseline microbiota levels. Results suggest that maple sap and its concentrate represent good candidates for the production of non-dairy functional foods.
Collapse
Affiliation(s)
- Riadh Hammami
- a STELA Dairy Research Centre, Institute of Nutrition and Functional Foods, Université Laval , Québec , Canada and
| | - Nour Ben Abdallah
- a STELA Dairy Research Centre, Institute of Nutrition and Functional Foods, Université Laval , Québec , Canada and
| | - Julie Barbeau
- b Fédération des Producteurs Acéricoles du Québec , Longueuil, Québec , Canada
| | - Ismail Fliss
- a STELA Dairy Research Centre, Institute of Nutrition and Functional Foods, Université Laval , Québec , Canada and
| |
Collapse
|
133
|
Smith VH, Rubinstein RJ, Park S, Kelly L, Klepac-Ceraj V. Microbiology and ecology are vitally important to premedical curricula. EVOLUTION MEDICINE AND PUBLIC HEALTH 2015. [PMID: 26198190 PMCID: PMC4536855 DOI: 10.1093/emph/eov014] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Despite the impact of the human microbiome on health, an appreciation of microbial ecology is yet to be translated into mainstream medical training and practice. The human microbiota plays a role in the development of the immune system, in the development and function of the brain, in digestion, and in host defense, and we anticipate that many more functions are yet to be discovered. We argue here that without formal exposure to microbiology and ecology—fields that explore the networks, interactions and dynamics between members of populations of microbes—vitally important links between the human microbiome and health will be overlooked. This educational shortfall has significant downstream effects on patient care and biomedical research, and we provide examples from current research highlighting the influence of the microbiome on human health. We conclude that formally incorporating microbiology and ecology into the premedical curricula is invaluable to the training of future health professionals and critical to the development of novel therapeutics and treatment practices.
Collapse
Affiliation(s)
- Val H Smith
- Department of Ecology and Evolutionary Biology, University of Kansas, Lawrence, KS, USA
| | | | - Serry Park
- Department of Biological Sciences, Wellesley College, Wellesley, MA, USA
| | - Libusha Kelly
- Department of Systems and Computational Biology and Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Vanja Klepac-Ceraj
- Department of Biological Sciences, Wellesley College, Wellesley, MA, USA,
| |
Collapse
|
134
|
Sassone-Corsi M, Raffatellu M. No vacancy: how beneficial microbes cooperate with immunity to provide colonization resistance to pathogens. THE JOURNAL OF IMMUNOLOGY 2015; 194:4081-7. [PMID: 25888704 DOI: 10.4049/jimmunol.1403169] [Citation(s) in RCA: 207] [Impact Index Per Article: 20.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The mammalian intestine harbors a community of trillions of microbes, collectively known as the gut microbiota, which coevolved with the host in a mutually beneficial relationship. Among the numerous gut microbial species, certain commensal bacteria are known to provide health benefits to the host when administered in adequate amounts and, as such, are labeled "probiotics." We review some of the mechanisms by which probiotics and other beneficial commensals provide colonization resistance to pathogens. The battle for similar nutrients and the bacterial secretion of antimicrobials provide a direct means of competition between beneficial and harmful microbes. Beneficial microbes can also indirectly diminish pathogen colonization by stimulating the development of innate and adaptive immunity, as well as the function of the mucosal barrier. Altogether, we gather and present evidence that beneficial microbes cooperate with host immunity in an effort to shut out pathogens.
Collapse
Affiliation(s)
- Martina Sassone-Corsi
- Department of Microbiology and Molecular Genetics and Institute for Immunology, University of California Irvine School of Medicine, Irvine, CA 92697
| | - Manuela Raffatellu
- Department of Microbiology and Molecular Genetics and Institute for Immunology, University of California Irvine School of Medicine, Irvine, CA 92697
| |
Collapse
|
135
|
Wu X, Sun R, Chen Y, Zheng X, Bai L, Lian Z, Wei H, Tian Z. Oral ampicillin inhibits liver regeneration by breaking hepatic innate immune tolerance normally maintained by gut commensal bacteria. Hepatology 2015; 62:253-64. [PMID: 25783863 DOI: 10.1002/hep.27791] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/21/2014] [Accepted: 03/15/2015] [Indexed: 12/13/2022]
Abstract
UNLABELLED Commensal bacteria have been proposed to play a role in liver repair after partial (67%) hepatectomy. However, the underlying immune mechanisms remain elusive. Here, we show that liver regeneration was impaired in antibiotic (Atb) water-treated mice and this impairment strongly correlated with commensal bacterial load. Among the various Atbs used in our cocktail, ampicillin-sensitive commensal bacterial was associated with normal liver regeneration. The number of CD1d-dependent natural killer T (NKT) cells in Atb-treated hepatectomized mice was markedly increased, and these NKT cells were functionally overactivated to produce higher interferon-γ. Deficiency of NKT cells or antibody blockade of the CD1d-NKT interaction increased hepatocyte proliferation, which improved liver regeneration. Importantly, an increased number of Kupffer cells were observed in Atb-treated mice, and these Kupffer cells produced higher interleukin-12, which then functioned to activate hepatic NKT cells. Interleukin-12p40 deficiency or treatment with an anti-interleukin-12 antibody significantly inhibited NKT cell overactivation and recovered liver regeneration in Atb-treated mice. CONCLUSION Commensal bacteria play a critical role in maintaining Kupffer cells in a tolerant state, preventing subsequent NKT cell overactivation during liver regeneration. Moreover, our data suggest that long-term Atb use, which can impair the gut microbiota, may influence liver function by retarding liver regeneration.
Collapse
Affiliation(s)
- Xunyao Wu
- Institute of Immunology and The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences and Medical Center, University of Science & Technology of China, Hefei, Anhui, China
| | - Rui Sun
- Institute of Immunology and The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences and Medical Center, University of Science & Technology of China, Hefei, Anhui, China.,Hefei National Laboratory for Physical Sciences at Microscale, Hefei, Anhui, China
| | - Yongyan Chen
- Institute of Immunology and The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences and Medical Center, University of Science & Technology of China, Hefei, Anhui, China
| | - Xiaodong Zheng
- Institute of Immunology and The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences and Medical Center, University of Science & Technology of China, Hefei, Anhui, China
| | - Li Bai
- Institute of Immunology and The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences and Medical Center, University of Science & Technology of China, Hefei, Anhui, China.,Hefei National Laboratory for Physical Sciences at Microscale, Hefei, Anhui, China
| | - Zhexiong Lian
- Institute of Immunology and The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences and Medical Center, University of Science & Technology of China, Hefei, Anhui, China.,Hefei National Laboratory for Physical Sciences at Microscale, Hefei, Anhui, China
| | - Haiming Wei
- Institute of Immunology and The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences and Medical Center, University of Science & Technology of China, Hefei, Anhui, China.,Hefei National Laboratory for Physical Sciences at Microscale, Hefei, Anhui, China
| | - Zhigang Tian
- Institute of Immunology and The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences and Medical Center, University of Science & Technology of China, Hefei, Anhui, China.,Hefei National Laboratory for Physical Sciences at Microscale, Hefei, Anhui, China.,Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| |
Collapse
|
136
|
Engevik MA, Engevik KA, Yacyshyn MB, Wang J, Hassett DJ, Darien B, Yacyshyn BR, Worrell RT. Human Clostridium difficile infection: inhibition of NHE3 and microbiota profile. Am J Physiol Gastrointest Liver Physiol 2015; 308:G497-509. [PMID: 25552580 PMCID: PMC4422371 DOI: 10.1152/ajpgi.00090.2014] [Citation(s) in RCA: 79] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Clostridium difficile infection (CDI) is principally responsible for hospital acquired, antibiotic-induced diarrhea and colitis and represents a significant financial burden on our healthcare system. Little is known about C. difficile proliferation requirements, and a better understanding of these parameters is critical for development of new therapeutic targets. In cell lines, C. difficile toxin B has been shown to inhibit Na(+)/H(+) exchanger 3 (NHE3) and loss of NHE3 in mice results in an altered intestinal environment coupled with a transformed gut microbiota composition. However, this has yet to be established in vivo in humans. We hypothesize that C. difficile toxin inhibits NHE3, resulting in alteration of the intestinal environment and gut microbiota. Our results demonstrate that CDI patient biopsy specimens have decreased NHE3 expression and CDI stool has elevated Na(+) and is more alkaline compared with stool from healthy individuals. CDI stool microbiota have increased Bacteroidetes and Proteobacteria and decreased Firmicutes phyla compared with healthy subjects. In vitro, C. difficile grows optimally in the presence of elevated Na(+) and alkaline pH, conditions that correlate to changes observed in CDI patients. To confirm that inhibition of NHE3 was specific to C. difficile, human intestinal organoids (HIOs) were injected with C. difficile or healthy and CDI stool supernatant. Injection of C. difficile and CDI stool decreased NHE3 mRNA and protein expression compared with healthy stool and control HIOs. Together these data demonstrate that C. difficile inhibits NHE3 in vivo, which creates an altered environment favored by C. difficile.
Collapse
Affiliation(s)
- Melinda A. Engevik
- 1Department of Molecular and Cellular Physiology, University of Cincinnati College of Medicine, Cincinnati, Ohio;
| | - Kristen A. Engevik
- 1Department of Molecular and Cellular Physiology, University of Cincinnati College of Medicine, Cincinnati, Ohio;
| | - Mary Beth Yacyshyn
- 3Department of Medicine Division of Digestive Diseases, University of Cincinnati College of Medicine, Cincinnati, Ohio;
| | - Jiang Wang
- 4Department of Pathology and Laboratory Medicine, University of Cincinnati College of Medicine, Cincinnati, Ohio;
| | - Daniel J. Hassett
- 2Department of Molecular Genetics, Biochemistry and Microbiology, University of Cincinnati College of Medicine, Cincinnati, Ohio;
| | - Benjamin Darien
- 5Department of Animal Health and Biomedical Sciences, University Wisconsin, Madison, Wisconsin; and
| | - Bruce R. Yacyshyn
- 3Department of Medicine Division of Digestive Diseases, University of Cincinnati College of Medicine, Cincinnati, Ohio; ,6Digestive Health Center of Cincinnati Children's Hospital, Cincinnati, Ohio
| | - Roger T. Worrell
- 1Department of Molecular and Cellular Physiology, University of Cincinnati College of Medicine, Cincinnati, Ohio; ,6Digestive Health Center of Cincinnati Children's Hospital, Cincinnati, Ohio
| |
Collapse
|
137
|
Engevik MA, Engevik KA, Yacyshyn MB, Wang J, Hassett DJ, Darien B, Yacyshyn BR, Worrell RT. Human Clostridium difficile infection: inhibition of NHE3 and microbiota profile. Am J Physiol Gastrointest Liver Physiol 2015. [PMID: 25552580] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/13/2023]
Abstract
Clostridium difficile infection (CDI) is principally responsible for hospital acquired, antibiotic-induced diarrhea and colitis and represents a significant financial burden on our healthcare system. Little is known about C. difficile proliferation requirements, and a better understanding of these parameters is critical for development of new therapeutic targets. In cell lines, C. difficile toxin B has been shown to inhibit Na(+)/H(+) exchanger 3 (NHE3) and loss of NHE3 in mice results in an altered intestinal environment coupled with a transformed gut microbiota composition. However, this has yet to be established in vivo in humans. We hypothesize that C. difficile toxin inhibits NHE3, resulting in alteration of the intestinal environment and gut microbiota. Our results demonstrate that CDI patient biopsy specimens have decreased NHE3 expression and CDI stool has elevated Na(+) and is more alkaline compared with stool from healthy individuals. CDI stool microbiota have increased Bacteroidetes and Proteobacteria and decreased Firmicutes phyla compared with healthy subjects. In vitro, C. difficile grows optimally in the presence of elevated Na(+) and alkaline pH, conditions that correlate to changes observed in CDI patients. To confirm that inhibition of NHE3 was specific to C. difficile, human intestinal organoids (HIOs) were injected with C. difficile or healthy and CDI stool supernatant. Injection of C. difficile and CDI stool decreased NHE3 mRNA and protein expression compared with healthy stool and control HIOs. Together these data demonstrate that C. difficile inhibits NHE3 in vivo, which creates an altered environment favored by C. difficile.
Collapse
Affiliation(s)
- Melinda A Engevik
- Department of Molecular and Cellular Physiology, University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - Kristen A Engevik
- Department of Molecular and Cellular Physiology, University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - Mary Beth Yacyshyn
- Department of Medicine Division of Digestive Diseases, University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - Jiang Wang
- Department of Pathology and Laboratory Medicine, University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - Daniel J Hassett
- Department of Molecular Genetics, Biochemistry and Microbiology, University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - Benjamin Darien
- Department of Animal Health and Biomedical Sciences, University Wisconsin, Madison, Wisconsin; and
| | - Bruce R Yacyshyn
- Department of Medicine Division of Digestive Diseases, University of Cincinnati College of Medicine, Cincinnati, Ohio; Digestive Health Center of Cincinnati Children's Hospital, Cincinnati, Ohio
| | - Roger T Worrell
- Department of Molecular and Cellular Physiology, University of Cincinnati College of Medicine, Cincinnati, Ohio; Digestive Health Center of Cincinnati Children's Hospital, Cincinnati, Ohio
| |
Collapse
|
138
|
Ritchie LE, Sturino JM, Carroll RJ, Rooney LW, Azcarate-Peril MA, Turner ND. Polyphenol-rich sorghum brans alter colon microbiota and impact species diversity and species richness after multiple bouts of dextran sodium sulfate-induced colitis. FEMS Microbiol Ecol 2015; 91:fiv008. [PMID: 25764457 PMCID: PMC4573659 DOI: 10.1093/femsec/fiv008] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Revised: 11/05/2014] [Accepted: 01/08/2015] [Indexed: 02/07/2023] Open
Abstract
The microbiota affects host health, and dysbiosis is involved in colitis. Sorghum bran influences butyrate concentrations during dextran sodium sulfate (DSS) colitis, suggesting microbiota changes. We aimed to characterize the microbiota during colitis, and ascertain if polyphenol-rich sorghum bran diets mitigate these effects. Rats (n = 80) were fed diets containing 6% fiber from cellulose, or Black (3-deoxyanthocyanins), Sumac (condensed tannins), or Hi Tannin black (both) sorghum bran. Inflammation was induced three times using 3% DSS for 48 h (40 rats, 2 week separation), and the microbiota characterized by pyrosequencing. The Firmicutes/Bacteroidetes ratio was higher in Cellulose DSS rats. Colonic injury negatively correlated with Firmicutes, Actinobacteria, Lactobacillales and Lactobacillus, and positively correlated with Unknown/Unclassified. Post DSS#2, richness was significantly lower in Sumac and Hi Tannin black. Post DSS#3 Bacteroidales, Bacteroides, Clostridiales, Lactobacillales and Lactobacillus were reduced, with no Clostridium identified. Diet significantly affected Bacteroidales, Bacteroides, Clostridiales and Lactobacillus post DSS#2 and #3. Post DSS#3 diet significantly affected all genus, including Bacteroides and Lactobacillus, and diversity and richness increased. Sumac and Hi Tannin black DSS had significantly higher richness compared to controls. Thus, these sorghum brans may protect against alterations observed during colitis including reduced microbial diversity and richness, and dysbiosis of Firmicutes/Bacteroidetes.
Collapse
Affiliation(s)
- Lauren E Ritchie
- Intercollegiate Faculty of Genetics, Texas A&M University, College Station, TX 77843-2253, USA
| | - Joseph M Sturino
- Intercollegiate Faculty of Genetics, Texas A&M University, College Station, TX 77843-2253, USA Nutrition and Food Science Department, Texas A&M University, College Station, TX 77843-2253, USA
| | - Raymond J Carroll
- Department of Statistics, Texas A&M University, College Station, TX 77843-3143, USA
| | - Lloyd W Rooney
- Soil and Crop Sciences, Texas A&M University, College Station, TX 77843-2474, USA
| | - M Andrea Azcarate-Peril
- Department of Cell Biology and Physiology, University of North Carolina School of Medicine, Chapel Hill, NC 27599-7545, USA
| | - Nancy D Turner
- Intercollegiate Faculty of Genetics, Texas A&M University, College Station, TX 77843-2253, USA Nutrition and Food Science Department, Texas A&M University, College Station, TX 77843-2253, USA
| |
Collapse
|
139
|
The multifaceted role of commensal microbiota in homeostasis and gastrointestinal diseases. J Immunol Res 2015; 2015:321241. [PMID: 25759839 PMCID: PMC4352444 DOI: 10.1155/2015/321241] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2014] [Accepted: 10/08/2014] [Indexed: 01/09/2023] Open
Abstract
The gastrointestinal tract houses a complex and diverse community of microbes. In recent years, an increased understanding of the importance of intestinal microbiota for human physiology has been gained. In the steady state, commensal microorganisms have a symbiotic relationship with the host and possess critical and distinct functions, including directly influencing immunity. This means that recognition of commensal antigens is necessary for the development of complete immune responses. Therefore, the immune system must face the challenge of maintaining mucosal homeostasis while dealing with undue passage of commensal or pathogenic microbes, as well as the host nutritional status or drug use. Disruption of this fine balance has been associated with the development of several intestinal inflammatory diseases. In this review, we discuss the mechanisms involved in the modulation of host-microbe interactions and how the breakdown of this homeostatic association can lead to intestinal inflammation and pathology.
Collapse
|
140
|
Yao J, Rock CO. How bacterial pathogens eat host lipids: implications for the development of fatty acid synthesis therapeutics. J Biol Chem 2015; 290:5940-6. [PMID: 25648887 DOI: 10.1074/jbc.r114.636241] [Citation(s) in RCA: 84] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Bacterial type II fatty acid synthesis (FASII) is a target for the development of novel therapeutics. Bacteria incorporate extracellular fatty acids into membrane lipids, raising the question of whether pathogens use host fatty acids to bypass FASII and defeat FASII therapeutics. Some pathogens suppress FASII when exogenous fatty acids are present to bypass FASII therapeutics. FASII inhibition cannot be bypassed in many bacteria because essential fatty acids cannot be obtained from the host. FASII antibiotics may not be effective against all bacteria, but a broad spectrum of Gram-negative and -positive pathogens can be effectively treated with FASII inhibitors.
Collapse
Affiliation(s)
- Jiangwei Yao
- From the Department of Infectious Diseases, St. Jude Children's Research Hospital, Memphis, Tennessee 38105
| | - Charles O Rock
- From the Department of Infectious Diseases, St. Jude Children's Research Hospital, Memphis, Tennessee 38105
| |
Collapse
|
141
|
Age-related immune clearance of hepatitis B virus infection requires the establishment of gut microbiota. Proc Natl Acad Sci U S A 2015; 112:2175-80. [PMID: 25646429 DOI: 10.1073/pnas.1424775112] [Citation(s) in RCA: 156] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
A unique feature of hepatitis B virus (HBV) infection in humans is that viral clearance heavily depends on the age of exposure. However, the reason for this remains unclear. Here we show that gut microbiota contribute to the age dependence of HBV immunity in a hydrodynamic transfection mouse model. Although adult (12-wk-old) C3H/HeN mice cleared HBV within 6 wk postinjection (wpi), their young (6-wk-old) counterparts remained HBV-positive at 26 wpi. Sterilization of gut microbiota from 6 to 12 wk of age using antibiotics prevented adult mice from rapidly clearing HBV. Young mice with the Toll-like-receptor (TLR) 4 mutation (C3H/HeJ) exhibited rapid HBV clearance. The results suggest that an immuno-tolerating pathway to HBV prevailed in young mice, before the establishment of gut bacteria, through a TLR4-dependent pathway and that the maturation of gut microbiota in adult mice stimulated liver immunity, resulting in rapid HBV clearance.
Collapse
|
142
|
Aguilera M, Cerdà-Cuéllar M, Martínez V. Antibiotic-induced dysbiosis alters host-bacterial interactions and leads to colonic sensory and motor changes in mice. Gut Microbes 2015; 6:10-23. [PMID: 25531553 PMCID: PMC4615720 DOI: 10.4161/19490976.2014.990790] [Citation(s) in RCA: 106] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
Alterations in the composition of the commensal microbiota (dysbiosis) seem to be a pathogenic component of functional gastrointestinal disorders, mainly irritable bowel syndrome (IBS), and might participate in the secretomotor and sensory alterations observed in these patients.We determined if a state antibiotics-induced intestinal dysbiosis is able to modify colonic pain-related and motor responses and characterized the neuro-immune mechanisms implicated in mice. A 2-week antibiotics treatment induced a colonic dysbiosis (increments in Bacteroides spp, Clostridium coccoides and Lactobacillus spp and reduction in Bifidobacterium spp). Bacterial adherence was not affected. Dysbiosis was associated with increased levels of secretory-IgA, up-regulation of the antimicrobial lectin RegIIIγ, and toll-like receptors (TLR) 4 and 7 and down-regulation of the antimicrobial-peptide Resistin-Like Molecule-β and TLR5. Dysbiotic mice showed less goblet cells, without changes in the thickness of the mucus layer. Neither macroscopical nor microscopical signs of inflammation were observed. In dysbiotic mice, expression of the cannabinoid receptor 2 was up-regulated, while the cannabinoid 1 and the mu-opioid receptors were down-regulated. In antibiotic-treated mice, visceral pain-related responses elicited by intraperitoneal acetic acid or intracolonic capsaicin were significantly attenuated. Colonic contractility was enhanced during dysbiosis. Intestinal dysbiosis induce changes in the innate intestinal immune system and modulate the expression of pain-related sensory systems, an effect associated with a reduction in visceral pain-related responses. Commensal microbiota modulates gut neuro-immune sensory systems, leading to functional changes, at least as it relates to viscerosensitivity. Similar mechanisms might explain the beneficial effects of antibiotics or certain probiotics in the treatment of IBS.
Collapse
Key Words
- AMP, antimicrobial peptide
- CB1/2, cannabinoid receptor type 1 or 2
- FGD, functional gastrointestinal disorder
- FISH, fluorescent in situ hybridization
- GCM, gut commensal microbiota
- GI, gastrointestinal
- IBS, irritable bowel syndrome
- MOR, mu-opioid receptor
- NGF, nerve growth factor
- PPR, pattern recognition receptor
- RELMβ, resistin-like molecule-β
- RT-qPCR, reverse transcription quantitative polymerase chain reaction
- Reg3γ, regenerating islet-derived protein 3 gamma
- SFB, segmented filamentous bacteria
- TLR, toll-like receptor
- TPH 1/2, tryptophan hydroxylase isoforms 1 or 2
- TRPV1/3, transient receptor potential vanilloid types 1 or 3
- cannabinoid receptors
- colonic motility
- gut commensal microbiota
- iNOS, inducible nitric oxide synthase
- innate immune system
- intestinal dysbiosis
- opioid receptors
- sIgA, secretory IgA
- visceral sensitivity
Collapse
Affiliation(s)
- M Aguilera
- Department of Cell Biology; Physiology and Immunology; Universitat Autònoma de Barcelona; Barcelona, Spain,Neuroscience Institute; Universitat Autònoma de Barcelona; Barcelona, Spain
| | - M Cerdà-Cuéllar
- Centre de Recerca en Sanitat Animal (CReSA); UAB-IRTA; Universitat Autònoma de Barcelona, and Institut de Recerca i Tecnologia Agroalimentàries (IRTA); Barcelona, Spain
| | - V Martínez
- Department of Cell Biology; Physiology and Immunology; Universitat Autònoma de Barcelona; Barcelona, Spain,Neuroscience Institute; Universitat Autònoma de Barcelona; Barcelona, Spain,Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd); Instituto de Salud Carlos III; Madrid, Spain,Correspondence to: Vicente Martínez;
| |
Collapse
|
143
|
The pathogen Batrachochytrium dendrobatidis disturbs the frog skin microbiome during a natural epidemic and experimental infection. Proc Natl Acad Sci U S A 2014; 111:E5049-58. [PMID: 25385615 DOI: 10.1073/pnas.1412752111] [Citation(s) in RCA: 182] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Symbiotic microbial communities may interact with infectious pathogens sharing a common host. The microbiome may limit pathogen infection or, conversely, an invading pathogen can disturb the microbiome. Documentation of such relationships during naturally occurring disease outbreaks is rare, and identifying causal links from field observations is difficult. This study documented the effects of an amphibian skin pathogen of global conservation concern [the chytrid fungus Batrachochytrium dendrobatidis (Bd)] on the skin-associated bacterial microbiome of the endangered frog, Rana sierrae, using a combination of population surveys and laboratory experiments. We examined covariation of pathogen infection and bacterial microbiome composition in wild frogs, demonstrating a strong and consistent correlation between Bd infection load and bacterial community composition in multiple R. sierrae populations. Despite the correlation between Bd infection load and bacterial community composition, we observed 100% mortality of postmetamorphic frogs during a Bd epizootic, suggesting that the relationship between Bd and bacterial communities was not linked to variation in resistance to mortal disease and that Bd infection altered bacterial communities. In a controlled experiment, Bd infection significantly altered the R. sierrae microbiome, demonstrating a causal relationship. The response of microbial communities to Bd infection was remarkably consistent: Several bacterial taxa showed the same response to Bd infection across multiple field populations and the laboratory experiment, indicating a somewhat predictable interaction between Bd and the microbiome. The laboratory experiment demonstrates that Bd infection causes changes to amphibian skin bacterial communities, whereas the laboratory and field results together strongly support Bd disturbance as a driver of bacterial community change during natural disease dynamics.
Collapse
|
144
|
Yu LCH, Shih YA, Wu LL, Lin YD, Kuo WT, Peng WH, Lu KS, Wei SC, Turner JR, Ni YH. Enteric dysbiosis promotes antibiotic-resistant bacterial infection: systemic dissemination of resistant and commensal bacteria through epithelial transcytosis. Am J Physiol Gastrointest Liver Physiol 2014; 307:G824-35. [PMID: 25059827 PMCID: PMC4214854 DOI: 10.1152/ajpgi.00070.2014] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Antibiotic usage promotes intestinal colonization of antibiotic-resistant bacteria. However, whether resistant bacteria gain dominance in enteric microflora or disseminate to extraintestinal viscera remains unclear. Our aim was to investigate temporal diversity changes in microbiota and transepithelial routes of bacterial translocation after antibiotic-resistant enterobacterial colonization. Mice drinking water with or without antibiotics were intragastrically gavaged with ampicillin-resistant (Amp-r) nonpathogenic Escherichia coli (E. coli) and given normal water afterward. The composition and spatial distribution of intestinal bacteria were evaluated using 16S rDNA sequencing and fluorescence in situ hybridization. Bacterial endocytosis in epithelial cells was examined using gentamicin resistance assay and transmission electromicroscopy. Paracellular permeability was assessed by tight junctional immunostaining and measured by tissue conductance and luminal-to-serosal dextran fluxes. Our results showed that antibiotic treatment enabled intestinal colonization and transient dominance of orally acquired Amp-r E. coli in mice. The colonized Amp-r E. coli peaked on day 3 postinoculation and was competed out after 1 wk, as evidenced by the recovery of commensals, such as Escherichia, Bacteroides, Lachnospiraceae, Clostridium, and Lactobacillus. Mucosal penetration and extraintestinal dissemination of exogenous and endogenous enterobacteria were correlated with abnormal epithelial transcytosis but uncoupled with paracellular tight junctional damage. In conclusion, antibiotic-induced enteric dysbiosis predisposes to exogenous infection and causes systemic dissemination of both antibiotic-resistant and commensal enterobacteria through transcytotic routes across epithelial layers. These results may help explain the susceptibility to sepsis in antibiotic-resistant enteric bacterial infection.
Collapse
Affiliation(s)
- Linda Chia-Hui Yu
- 1Graduate Institute of Physiology, National Taiwan University College of Medicine and Hospital, Taipei, Taiwan;
| | - Yi-An Shih
- 1Graduate Institute of Physiology, National Taiwan University College of Medicine and Hospital, Taipei, Taiwan;
| | - Li-Ling Wu
- 1Graduate Institute of Physiology, National Taiwan University College of Medicine and Hospital, Taipei, Taiwan;
| | - Yang-Ding Lin
- 2Department of Pediatrics, National Taiwan University College of Medicine, Taipei, Taiwan;
| | - Wei-Ting Kuo
- 1Graduate Institute of Physiology, National Taiwan University College of Medicine and Hospital, Taipei, Taiwan;
| | - Wei-Hao Peng
- 3Graduate Institute of Anatomy and Cell Biology, National Taiwan University College of Medicine, Taipei, Taiwan;
| | - Kuo-Shyan Lu
- 3Graduate Institute of Anatomy and Cell Biology, National Taiwan University College of Medicine, Taipei, Taiwan;
| | - Shu-Chen Wei
- 4Department of Internal Medicine, National Taiwan University College of Medicine and Hospital, Taipei, Taiwan; and
| | | | - Yen-Hsuan Ni
- Department of Pediatrics, National Taiwan University College of Medicine, Taipei, Taiwan;
| |
Collapse
|
145
|
Hurley D, McCusker MP, Fanning S, Martins M. Salmonella-host interactions - modulation of the host innate immune system. Front Immunol 2014; 5:481. [PMID: 25339955 PMCID: PMC4188169 DOI: 10.3389/fimmu.2014.00481] [Citation(s) in RCA: 97] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2014] [Accepted: 09/20/2014] [Indexed: 12/27/2022] Open
Abstract
Salmonella enterica (S. enterica) are Gram-negative bacteria that can invade a broad range of hosts causing both acute and chronic infections. This phenotype is related to its ability to replicate and persist within non-phagocytic host epithelial cells as well as phagocytic dendritic cells and macrophages of the innate immune system. Infection with S. enterica manifests itself through a broad range of clinical symptoms and can result in asymptomatic carriage, gastroenteritis, systemic disease such as typhoid fever and in severe cases, death (1). Exposure to S. enterica serovars Typhi and Paratyphi exhibits clinical symptoms including diarrhea, fatigue, fever, and temperature fluctuations. Other serovars such as the non-typhoidal Salmonella (NTS), of which there are over 2,500, are commonly contracted as, but not limited to, food-borne sources causing gastrointestinal symptoms, which include diarrhea and vomiting. The availability of complete genome sequences for many S. enterica serovars has facilitated research into the genetic determinants of virulence for this pathogen. This work has led to the identification of important bacterial components, including flagella, type III secretion systems, lipopolysaccharides, and Salmonella pathogenicity islands, all of which support the intracellular life cycle of S. enterica. Studies focusing on the host-pathogen interaction have provided insights into receptor activation of the innate immune system. Therefore, characterizing the host-S. enterica interaction is critical to understand the pathogenicity of the bacteria in a clinically relevant context. This review outlines salmonellosis and the clinical manifestations between typhoidal and NTS infections as well as discussing the host immune response to infection and the models that are being used to elucidate the mechanisms involved in Salmonella pathogenicity.
Collapse
Affiliation(s)
- Daniel Hurley
- School of Public Health, Physiotherapy and Population Science, UCD Centre for Food Safety, UCD Centre for Molecular Innovation and Drug Discovery, University College Dublin , Dublin , Ireland
| | - Matthew P McCusker
- School of Public Health, Physiotherapy and Population Science, UCD Centre for Food Safety, UCD Centre for Molecular Innovation and Drug Discovery, University College Dublin , Dublin , Ireland
| | - Séamus Fanning
- School of Public Health, Physiotherapy and Population Science, UCD Centre for Food Safety, UCD Centre for Molecular Innovation and Drug Discovery, University College Dublin , Dublin , Ireland
| | - Marta Martins
- School of Public Health, Physiotherapy and Population Science, UCD Centre for Food Safety, UCD Centre for Molecular Innovation and Drug Discovery, University College Dublin , Dublin , Ireland
| |
Collapse
|
146
|
Kaiko GE, Stappenbeck TS. Host-microbe interactions shaping the gastrointestinal environment. Trends Immunol 2014; 35:538-48. [PMID: 25220948 DOI: 10.1016/j.it.2014.08.002] [Citation(s) in RCA: 107] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2014] [Revised: 08/08/2014] [Accepted: 08/13/2014] [Indexed: 12/18/2022]
Abstract
Tremendous advances have been made in mapping the complexity of the human gut microbiota in both health and disease states. These analyses have revealed that, rather than a constellation of individual species, a healthy microbiota comprises an interdependent network of microbes. The microbial and host interactions that shape both this network and the gastrointestinal environment are areas of intense investigation. Here we review emerging concepts of how microbial metabolic processes control commensal composition, invading pathogens, immune activation, and intestinal barrier function. We posit that all of these factors are critical for the maintenance of homeostasis and avoidance of overt inflammatory disease. A greater understanding of the underlying mechanisms will shed light on the pathogenesis of many diseases and guide new therapeutic interventions.
Collapse
Affiliation(s)
- Gerard E Kaiko
- Department of Pathology and Immunology, Washington University School of Medicine, St Louis, MO 63110, USA
| | - Thaddeus S Stappenbeck
- Department of Pathology and Immunology, Washington University School of Medicine, St Louis, MO 63110, USA.
| |
Collapse
|
147
|
Yurist-Doutsch S, Arrieta MC, Vogt SL, Finlay BB. Gastrointestinal microbiota-mediated control of enteric pathogens. Annu Rev Genet 2014; 48:361-82. [PMID: 25251855 DOI: 10.1146/annurev-genet-120213-092421] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
The gastrointestinal (GI) microbiota is a complex community of microorganisms residing within the mammalian gastrointestinal tract. The GI microbiota is vital to the development of the host immune system and plays a crucial role in human health and disease. The composition of the GI microbiota differs immensely among individuals yet specific shifts in composition and diversity have been linked to inflammatory bowel disease, obesity, atopy, and susceptibility to infection. In this review, we describe the GI microbiota and its role in enteric diseases caused by pathogenic Escherichia coli, Salmonella enterica, and Clostridium difficile. We discuss the central role of the GI microbiota in protective immunity, resistance to enteric pathogens, and resolution of enteric colitis.
Collapse
Affiliation(s)
- Sophie Yurist-Doutsch
- Michael Smith Laboratories, The University of British Columbia, Vancouver, British Columbia, Canada, V6T 1Z4; , , ,
| | | | | | | |
Collapse
|
148
|
Lou Y, Sun H, Morrissey S, Porturas T, Liu S, Hua X, Chen YH. Critical roles of TIPE2 protein in murine experimental colitis. THE JOURNAL OF IMMUNOLOGY 2014; 193:1064-70. [PMID: 24973456 DOI: 10.4049/jimmunol.1400415] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Both commensal bacteria and infiltrating inflammatory cells play essential roles in the pathogenesis of inflammatory bowel disease. The molecular mechanisms whereby these pathogenic factors are regulated during the disease are not fully understood. We report in this article that a member of the TNF-α-induced protein 8 (TNFAIP8) family called TIPE2 (TNFAIP8-like 2) plays a crucial role in regulating commensal bacteria dissemination and inflammatory cell function in experimental colitis induced by dextran sodium sulfate (DSS). Following DSS treatment, TIPE2-deficient mice, or chimeric mice that are deficient in TIPE2 only in their hematopoietic cells, lost less body weight and survived longer than wild-type controls. Consistent with this clinical observation, TIPE2-deficient mice exhibited significantly less severe colitis and colonic damage. This was associated with a marked reduction in the colonic expression of inflammatory cytokines, such as TNF-α, IL-6, and IL-12. Importantly, the ameliorated DSS-induced colitis in TIPE2(-/-) mice also was associated with reduced local dissemination of commensal bacteria and a weaker systemic inflammatory response. Combined with our previous report that TIPE2 is a negative regulator of antibacterial immunity, these results indicate that TIPE2 promotes colitis by inhibiting mucosal immunity to commensal bacteria.
Collapse
Affiliation(s)
- Yunwei Lou
- Department of Pathology and Laboratory Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104; Department of Immunology, Shandong University School of Medicine, Ji'nan 250012, People's Republic of China; and
| | - Honghong Sun
- Department of Pathology and Laboratory Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104
| | - Samantha Morrissey
- Department of Pathology and Laboratory Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104
| | - Thomas Porturas
- Department of Pathology and Laboratory Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104
| | - Suxia Liu
- Department of Immunology, Shandong University School of Medicine, Ji'nan 250012, People's Republic of China; and
| | - Xianxin Hua
- Department of Cancer Biology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104
| | - Youhai H Chen
- Department of Pathology and Laboratory Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104;
| |
Collapse
|
149
|
Martin ME, Solnick JV. The gastric microbial community, Helicobacter pylori colonization, and disease. Gut Microbes 2014; 5:345-50. [PMID: 24642475 PMCID: PMC4153772 DOI: 10.4161/gmic.28573] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
Long thought to be a sterile habitat, the stomach contains a diverse and unique community of bacteria. One particular inhabitant, Helicobacter pylori, colonizes half of the world's human population and establishes a decades-long infection that can be asymptomatic, pathogenic, or even beneficial for the host. Many host and bacterial factors are known to influence an individual's risk of gastric disease, but another potentially important determinant has recently come to light: the host microbiota. Although it is unclear to what extent H. pylori infection perturbs the established gastric microbial community, and H. pylori colonization seems generally resistant to disturbances in the host microbiota, it can modulate H. pylori pathogenicity. Interactions between H. pylori and bacteria at non-gastric sites are likely indirect--via programming of the pro-inflammatory vs. regulatory T lymphocytes--which may have a significant impact on human health.
Collapse
Affiliation(s)
- Miriam E Martin
- Department of Medicine; University of California-Davis; Davis, CA USA,Department of Microbiology & Immunology; University of California-Davis; Davis, CA USA
| | - Jay V Solnick
- Department of Medicine; University of California-Davis; Davis, CA USA,Department of Microbiology & Immunology; University of California-Davis; Davis, CA USA,California National Primate Research Center; University of California-Davis; Davis, CA USA,Correspondence to: Jay V Solnick,
| |
Collapse
|
150
|
Levy R, Borenstein E. Metagenomic systems biology and metabolic modeling of the human microbiome: from species composition to community assembly rules. Gut Microbes 2014; 5:265-70. [PMID: 24637600 PMCID: PMC4063856 DOI: 10.4161/gmic.28261] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
The human microbiome is a key contributor to health and development. Yet little is known about the ecological forces that are at play in defining the composition of such host-associated communities. Metagenomics-based studies have uncovered clear patterns of community structure but are often incapable of distinguishing alternative structuring paradigms. In a recent study, we integrated metagenomic analysis with a systems biology approach, using a reverse ecology framework to model numerous human microbiota species and to infer metabolic interactions between species. Comparing predicted interactions with species composition data revealed that the assembly of the human microbiome is dominated at the community level by habitat filtering. Furthermore, we demonstrated that this habitat filtering cannot be accounted for by known host phenotypes or by the metabolic versatility of the various species. Here we provide a summary of our findings and offer a brief perspective on related studies and on future approaches utilizing this metagenomic systems biology framework.
Collapse
Affiliation(s)
- Roie Levy
- Department of Genome Sciences; University of Washington; Seattle WA USA
| | - Elhanan Borenstein
- Department of Genome Sciences; University of Washington; Seattle WA USA,Department of Computer Science and Engineering; University of Washington; Seattle, WA USA,Santa Fe Institute; Santa Fe, NM USA,Correspondence to: Elhanan Borenstein,
| |
Collapse
|