65 results on '"Petrof EO"'
Search Results
2. Recours à des bactéries en tant que médicaments : traitements de l’écosystème microbien
- Author
-
Allen-Vercoe, E, primary and Petrof, EO, additional
- Published
- 2015
- Full Text
- View/download PDF
3. Using bugs as drugs: Microbial ecosystem therapeutics
- Author
-
Allen-Vercoe, E, primary and Petrof, EO, additional
- Published
- 2015
- Full Text
- View/download PDF
4. Probiotics in the critically ill: a systematic review of the randomized trial evidence.
- Author
-
Petrof EO, Dhaliwal R, Manzanares W, Johnstone J, Cook D, and Heyland DK
- Abstract
OBJECTIVE: Critical illness results in changes to the microbiology of the gastrointestinal tract, leading to a loss of commensal flora and an overgrowth of potentially pathogenic bacteria. Administering certain strains of live bacteria (probiotics) to critically ill patients may restore balance to the microbiota and have positive effects on immune function and gastrointestinal structure and function. The purpose of this systematic review was to evaluate the effect of probiotics in critically ill patients on clinical outcomes. DESIGN: Systematic review. INTERVENTIONS: None. MEASUREMENTS AND MAIN RESULTS: We searched computerized databases, reference lists of pertinent articles, and personal files from 1980 to 2011. We included randomized controlled trials enrolling critically ill adults, which evaluated probiotics compared to a placebo and reported clinically important outcomes (infections, mortality, and length of stay). A total of 23 randomized controlled trials met inclusion criteria. Probiotics were associated with reduced infectious complications as documented in 11 trials (risk ratio 0.82; 95% confidence interval 0.69-0.99; p = .03; test for heterogeneity p = .05; I 44%). When data from the seven trials reporting ventilator-associated pneumonia were pooled, ventilator-associated pneumonia rates were also significantly reduced with probiotics (risk ratio 0.75; 95% confidence interval 0.59-0.97; p = .03; test for heterogeneity p = .16; I 35%). Probiotics were associated with a trend toward reduced intensive care unit mortality (risk ratio 0.80; 95% confidence interval 0.59-1.09; p = .16; test for heterogeneity p = .89; I 0%) but did not influence hospital mortality. Probiotics had no effect on intensive care unit or hospital length of stay. Compared to trials of higher methodological quality, greater treatment effects were observed in trials of a lower methodological quality. CONCLUSIONS: Probiotics appear to reduce infectious complications including ventilator-associated pneumonia and may influence intensive care unit mortality. However, clinical and statistical heterogeneity and imprecise estimates preclude strong clinical recommendations. Further research on probiotics in the critically ill is warranted. [ABSTRACT FROM AUTHOR]
- Published
- 2012
- Full Text
- View/download PDF
5. Urinary tract infections and a multidrug-resistant Escherichia coli clonal group.
- Author
-
Sandel DC, Wang C, Kessler S, Petrof EO, Schwartz DN, Quinn JP, Barlam T, Moellering R, Johnson JR, Manges AR, and Riley LW
- Published
- 2002
6. Defined microbial communities and their soluble products protect mice from Clostridioides difficile infection.
- Author
-
Douchant K, He SM, Noordhof C, Greenlaw J, Vancuren S, Schroeter K, Allen-Vercoe E, Sjaarda C, Vanner SJ, Petrof EO, Sheth PM, and Guzman M
- Subjects
- Animals, Mice, Anti-Bacterial Agents pharmacology, Gastrointestinal Tract microbiology, Diarrhea prevention & control, Diarrhea microbiology, Bacteria, Microbiota, Clostridium Infections prevention & control, Clostridium Infections microbiology
- Abstract
Clostridioides difficile is the leading cause of antibiotic-associated infectious diarrhea. The development of C.difficile infection is tied to perturbations of the bacterial community in the gastrointestinal tract, called the gastrointestinal microbiota. Repairing the gastrointestinal microbiota by introducing lab-designed bacterial communities, or defined microbial communities, has recently shown promise as therapeutics against C.difficile infection, however, the mechanisms of action of defined microbial communities remain unclear. Using an antibiotic- C.difficile mouse model, we report the ability of an 18-member community and a refined 4-member community to protect mice from two ribotypes of C.difficile (CD027, CD078; p < 0.05). Furthermore, bacteria-free supernatant delivered orally to mice from the 4-member community proteolyzed C.difficile toxins in vitro and protected mice from C.difficile infection in vivo (p < 0.05). This study demonstrates that bacteria-free supernatant is sufficient to protect mice from C.difficile; and could be further explored as a therapeutic strategy against C.difficile infection., (© 2024. The Author(s).)
- Published
- 2024
- Full Text
- View/download PDF
7. The effect of a microbial ecosystem therapeutic (MET-2) on recurrent Clostridioides difficile infection: a phase 1, open-label, single-group trial.
- Author
-
Kao D, Wong K, Franz R, Cochrane K, Sherriff K, Chui L, Lloyd C, Roach B, Bai AD, Petrof EO, and Allen-Vercoe E
- Subjects
- Aged, Disease-Free Survival, Female, Humans, Male, Middle Aged, Recurrence, Treatment Outcome, Clostridium Infections therapy, Fecal Microbiota Transplantation
- Abstract
Background: Faecal microbiota transplantation (FMT) is highly effective for recurrent Clostridioides difficile infection but has inherent risks. Microbial Ecosystem Therapeutic 2 (MET-2) is an oral encapsulated formulation of 40 lyophilised bacterial species initially isolated from stool of a healthy donor, but subsequently manufactured independently of donors, eliminating potential risks introduced by changes in donor health. The aim of this study was to determine MET-2 activity, safety, and tolerability., Methods: This phase 1, open-label, single-group feasibility study was done in Alberta, Canada. The main inclusion criteria were mild to moderate C difficile infection and at least one episode of C difficile infection recurrence (ie, two episodes of C difficile infection) within 12 months. Initial daily treatment was ten oral capsules for 2 days, then three capsules for 8 days. If C difficile infection recurred, a higher dose was offered: 20 capsules for 2 days, then three capsules for 8 days. Patients were followed for adverse events and C difficile infection recurrence up to day 130. The primary outcome was absence of C difficile infection recurrence (fewer than three unformed bowel movements in 24 h persisting for at least 2 days) at day 40 by intention-to-treat analysis. Secondary outcomes were mortality or hospitalisation due to C difficile infection, infections attributed to treatment, nausea, abdominal pain, vomiting, or diarrhoea during treatment, quality of life (C difficile Health Related Quality of Life Questionnaire) before and after treatment, and engrafted MET-2 bacteria in patient stool. Absence of C difficile infection recurrence at day 130 was an exploratory outcome. This study is registered with ClinicalTrials.gov, NCT02865616 FINDINGS: Between Sept 19, 2018, and Feb 28, 2020, we enrolled 19 adult patients with at least two episodes of mild to moderate C difficile infection (median age 65 years [IQR 56-67]; 12 women [63%], seven men [37%]). Recurrent C difficile infection was absent at day 40 in 15 (79%) of 19 patients after initial treatment, increasing to 18 (95%) 40 days after retreatment. No mortality associated with C difficile infection, infections associated with MET-2 treatment, or other serious adverse events were observed. The most common self-limited, mild to moderate symptoms reported during treatment were diarrhoea in 12 (63%) of 19 patients and abdominal cramps in 12 (63%). After MET-2 treatment, quality of life improved significantly, as did alpha diversity in stool microbial composition (p=1·93×10
-6 ). MET-2 associated taxa were found in greater abundance in most patients after treatment compared with baseline. 16 (84%) of 19 patients did not have recurrence of C difficile infection by day 130., Interpretation: MET-2 appears to be safe, efficacious, and well tolerated among patients with recurrent C difficile infection. Results must be validated in controlled studies., Funding: NuBiyota., (Copyright © 2021 Elsevier Ltd. All rights reserved.)- Published
- 2021
- Full Text
- View/download PDF
8. Protease-dependent excitation of nodose ganglion neurons by commensal gut bacteria.
- Author
-
Pradhananga S, Tashtush AA, Allen-Vercoe E, Petrof EO, and Lomax AE
- Subjects
- Animals, Bacteria, Ecosystem, Mice, Neurons, Neurons, Afferent, Peptide Hydrolases, Vagus Nerve, Gastrointestinal Microbiome, Nodose Ganglion
- Abstract
Key Points: The vagus nerve has been implicated in mediating behavioural effects of the gut microbiota on the central nervous system. This study examined whether the secretory products of commensal gut bacteria can modulate the excitability of vagal afferent neurons with cell bodies in nodose ganglia. Cysteine proteases from commensal bacteria increased the excitability of vagal afferent neurons via activation of protease-activated receptor 2 and modulation of the voltage dependence of Na
+ conductance activation. Lipopolysaccharide, a component of the cell wall of gram-negative bacteria, increased the excitability of nodose ganglia neurons via TLR4-dependent activation of nuclear factor kappa B. Our study identified potential mechanisms by which gut microbiota influences the activity of vagal afferent pathways, which may in turn impact on autonomic reflexes and behaviour., Abstract: Behavioural studies have implicated vagal afferent neurons as an important component of the microbiota-gut-brain axis. However, the mechanisms underlying the ability of the gut microbiota to affect vagal afferent pathways are unclear. We examined the effect of supernatant from a community of 33 commensal gastrointestinal bacterial derived from a healthy human donor (microbial ecosystem therapeutics; MET-1) on the excitability of mouse vagal afferent neurons. Perforated patch clamp electrophysiology was used to measure the excitability of dissociated nodose ganglion (NG) neurons. NG neuronal excitability was assayed by measuring the amount of current required to elicit an action potential, the rheobase. MET-1 supernatant increased the excitability of NG neurons by hyperpolarizing the voltage dependence of activation of Na+ conductance. The increase in excitability elicited by MET-1 supernatant was blocked by the cysteine protease inhibitor E-64 (30 nm). The protease activated receptor-2 (PAR2 ) antagonist (GB 83, 10 μm) also blocked the effect of MET-1 supernatant on NG neurons. Supernatant from Lactobacillus paracasei 6MRS, a component of MET-1, recapitulated the effect of MET-1 supernatant on NG neurons. Lastly, we compared the effects of MET-1 supernatant and lipopolysaccharide (LPS) from Escherichia coli 05:B5 on NG neuron excitability. LPS increased the excitability of NG neurons in a toll-like receptor 4 (TLR4 )-dependent and PAR2 -independent manner, whereas the excitatory effects of MET-1 supernatant were independent of TLR4 activation. Together, our findings suggest that cysteine proteases from commensal bacteria increase the excitability of vagal afferent neurons by the activation of PAR2 ., (© 2020 The Authors. The Journal of Physiology © 2020 The Physiological Society.)- Published
- 2020
- Full Text
- View/download PDF
9. The infant microbiome and implications for central nervous system development.
- Author
-
Parsons E, Claud K, and Petrof EO
- Subjects
- Humans, Infant, Infant, Extremely Premature, Developmental Disabilities microbiology, Dysbiosis physiopathology, Gastrointestinal Microbiome, Infant, Premature, Diseases microbiology, Nervous System Diseases microbiology
- Abstract
Neurodevelopmental impairment remains a significant morbidity in former very low birth weight premature infants. There is increasing evidence the microbiome affects neurodevelopment but mechanistic causes are largely unknown. There are many factors which affect the developing microbiome in infants including mode of delivery, feeding, medications, and environmental exposures. The overall impact of these factors may differ between premature and term infants. The microbiome and brain have well recognized bidirectional communication pathways via neural, hormonal, and immunologic mechanisms. Understanding the interplay between these different pathways has been possible with the use of animal models, particularly germ-free mice. The intricate relationship between the microbiome and the brain remains a research priority not only to improve the care, but to also improve the long-term neurodevelopmental outcomes in this vulnerable population., (© 2020 Elsevier Inc. All rights reserved.)
- Published
- 2020
- Full Text
- View/download PDF
10. In search of stool donors: a multicenter study of prior knowledge, perceptions, motivators, and deterrents among potential donors for fecal microbiota transplantation.
- Author
-
McSweeney B, Allegretti JR, Fischer M, Xu H, Goodman KJ, Monaghan T, McLeod C, Mullish BH, Petrof EO, Phelps EL, Chis R, Edmison A, Juby A, Ennis-Davis R, Roach B, Wong K, and Kao D
- Subjects
- Canada, Feces microbiology, Female, Humans, Male, Middle Aged, Surveys and Questionnaires, Young Adult, Clostridium Infections therapy, Fecal Microbiota Transplantation statistics & numerical data, Tissue Donors statistics & numerical data
- Abstract
Fecal microbiota transplantation (FMT) is a highly effective therapy for recurrent Clostridioides difficile infection. Stool donors are essential, but difficult to recruit and retain. We aimed to identify factors influencing willingness to donate stool. This multi-center study with a 32-item questionnaire targeted young adults and health care workers via social media and university email lists in Edmonton and Kingston, Canada; London and Nottingham, England; and Indianapolis and Boston, USA. Items included baseline demographics and FMT knowledge and perception. Investigated motivators and deterrents included economic compensation, screening process, time commitment, and stool donation logistics. Logistic regression and linear regression models estimated associations of study variables with self-assessed willingness to donate stool. 802 respondents completed our questionnaire: 387 (48.3%) age 21-30 years, 573 (71.4%) female, 323 (40%) health care workers. Country of residence, age and occupation were not associated with willingness to donate stool. Factors increasing willingness to donate were: already a blood donor (OR 1.64), male, altruism, economic benefit, knowledge of how FMT can help patients (OR 1.32), and positive attitudes towards FMT (OR 1.39). Factors decreasing willingness to donate were: stool collection unpleasant (OR 0.92), screening process invasive (OR 0.92), higher stool donation frequency, negative social perception of stool, and logistics of collection/transporting feces. We conclude that 1) blood donors and males are more willing to consider stool donation; 2) altruism, economic compensation, and positive feedback are motivators; and 3) screening process, high donation frequency, logistics of collection/transporting feces, lack of public awareness, and negative social perception are deterrents. Considering these variables could maximize donor recruitment and retention.
- Published
- 2020
- Full Text
- View/download PDF
11. Microbial bile salt hydrolases mediate the efficacy of faecal microbiota transplant in the treatment of recurrent Clostridioides difficile infection.
- Author
-
Mullish BH, McDonald JAK, Pechlivanis A, Allegretti JR, Kao D, Barker GF, Kapila D, Petrof EO, Joyce SA, Gahan CGM, Glegola-Madejska I, Williams HRT, Holmes E, Clarke TB, Thursz MR, and Marchesi JR
- Subjects
- Animals, Clostridium Infections microbiology, Disease Models, Animal, Female, Glycocholic Acid, Humans, Mice, Mice, Inbred C57BL, Recurrence, Tandem Mass Spectrometry, Amidohydrolases pharmacology, Clostridioides difficile genetics, Clostridium Infections therapy, DNA, Bacterial genetics, Fecal Microbiota Transplantation methods, Gastrointestinal Microbiome physiology
- Abstract
Objective: Faecal microbiota transplant (FMT) effectively treats recurrent Clostridioides difficile infection (rCDI), but its mechanisms of action remain poorly defined. Certain bile acids affect C. difficile germination or vegetative growth. We hypothesised that loss of gut microbiota-derived bile salt hydrolases (BSHs) predisposes to CDI by perturbing gut bile metabolism, and that BSH restitution is a key mediator of FMT's efficacy in treating the condition., Design: Using stool collected from patients and donors pre-FMT/post-FMT for rCDI, we performed 16S rRNA gene sequencing, ultra performance liquid chromatography mass spectrometry (UPLC-MS) bile acid profiling, BSH activity measurement, and qPCR of bsh / bai CD genes involved in bile metabolism. Human data were validated in C. difficile batch cultures and a C57BL/6 mouse model of rCDI., Results: From metataxonomics, pre-FMT stool demonstrated a reduced proportion of BSH-producing bacterial species compared with donors/post-FMT. Pre-FMT stool was enriched in taurocholic acid (TCA, a potent C. difficile germinant); TCA levels negatively correlated with key bacterial genera containing BSH-producing organisms. Post-FMT samples demonstrated recovered BSH activity and bsh / bai CD gene copy number compared with pretreatment (p<0.05). In batch cultures, supernatant from engineered bsh -expressing E. coli and naturally BSH-producing organisms ( Bacteroides ovatus, Collinsella aerofaciens, Bacteroides vulgatus and Blautia obeum ) reduced TCA-mediated C. difficile germination relative to culture supernatant of wild-type (BSH-negative) E. coli. C. difficile total viable counts were ~70% reduced in an rCDI mouse model after administration of E. coli expressing highly active BSH relative to mice administered BSH-negative E. coli (p<0.05)., Conclusion: Restoration of gut BSH functionality contributes to the efficacy of FMT in treating rCDI., Competing Interests: Competing interests: JRA consults for Finch Therapeutics. DK has received research funding from Rebiotix. EOP is a co-founder of Nubiyota and serves on its scientific advisory board. All other authors declared no conflict of interests., (© Author(s) (or their employer(s)) 2019. Re-use permitted under CC BY. Published by BMJ.)
- Published
- 2019
- Full Text
- View/download PDF
12. Effects of defined gut microbial ecosystem components on virulence determinants of Clostridioides difficile.
- Author
-
Carlucci C, Jones CS, Oliphant K, Yen S, Daigneault M, Carriero C, Robinson A, Petrof EO, Weese JS, and Allen-Vercoe E
- Subjects
- Anti-Bacterial Agents pharmacology, Bacterial Proteins genetics, Bacterial Toxins pharmacology, Ciprofloxacin pharmacology, Clostridiales genetics, Clostridiales metabolism, Clostridioides difficile metabolism, Clostridium Infections microbiology, Diarrhea microbiology, Dysbiosis microbiology, Enterotoxins pharmacology, Fecal Microbiota Transplantation methods, Humans, Microbiota genetics, Virulence genetics, Virulence Factors pharmacology, Clostridioides difficile genetics, Feces microbiology, Gastrointestinal Microbiome genetics
- Abstract
Many cases of Clostridioides difficile infection (CDI) are poorly responsive to standard antibiotic treatment strategies, and often patients suffer from recurrent infections characterized by severe diarrhea. Our group previously reported the successful cure of two patients with recurrent CDI using a standardized stool-derived microbial ecosystem therapeutic (MET-1). Using an in vitro model of the distal gut to support bacterial communities, we characterized the metabolite profiles of two defined microbial ecosystems derived from healthy donor stool (DEC58, and a subset community, MET-1), as well as an ecosystem representative of a dysbiotic state (ciprofloxacin-treated DEC58). The growth and virulence determinants of two C. difficile strains were then assessed in response to components derived from the ecosystems. CD186 (ribotype 027) and CD973 (ribotype 078) growth was decreased upon treatment with DEC58 metabolites compared to ciprofloxacin-treated DEC58 metabolites. Furthermore, CD186 TcdA and TcdB secretion was increased following treatment with ciprofloxacin-treated DEC58 spent medium compared to DEC58 spent medium alone. The net metabolic output of C. difficile was also modulated in response to spent media from defined microbial ecosystems, although several metabolite levels were divergent across the two strains examined. Further investigation of these antagonistic properties will guide the development of microbiota-based therapeutics for CDI.
- Published
- 2019
- Full Text
- View/download PDF
13. Using bioreactors to study the effects of drugs on the human microbiota.
- Author
-
Guzman-Rodriguez M, McDonald JAK, Hyde R, Allen-Vercoe E, Claud EC, Sheth PM, and Petrof EO
- Subjects
- Anti-Bacterial Agents pharmacology, Gastrointestinal Diseases metabolism, Gastrointestinal Diseases microbiology, Gastrointestinal Tract drug effects, Gastrointestinal Tract metabolism, Gastrointestinal Tract microbiology, Humans, Bioreactors microbiology, Gastrointestinal Microbiome drug effects, Gastrointestinal Microbiome physiology, Pharmaceutical Preparations administration & dosage
- Abstract
The study of complex microbial communities has become a major research focus as mounting evidence suggests the pivotal role microbial communities play in host health and disease. Microbial communities of the gastrointestinal tract, known as the gut microbiota, have been implicated in aiding the host with vitamin biosynthesis, regulation of host energy metabolism, immune system development, and resistance to pathogen invasion. Conversely, disruptions of the gut microbiota have been linked to host morbidity, including the development of inflammatory diseases, metabolic disorders, increased cardiovascular risk, and increased risk of infectious diseases. However, studying the gut microbiota in humans and animals is challenging, as many microorganisms are fastidious with unique nutritional or environmental requirements that are often not met using conventional culture techniques. Bioreactors provide a unique solution to overcome some of the limitations of conventional culture techniques. Bioreactors have been used to propagate and establish complex microbial communities in vitro by recapitulating the physiological conditions found in the GI tract. These systems further our understanding of microbial physiology and facilitate our understanding of the impact of medications and xenobiotics on microbial communities. Here, we review the versatility and breadth of bioreactor systems that are currently available and how they are being used to study faecal and defined microbial communities. Bioreactors provide a unique opportunity to study complex microbial interactions and perturbations in vitro in a controlled environment without confounding biotic and abiotic variables., (Copyright © 2018. Published by Elsevier Inc.)
- Published
- 2018
- Full Text
- View/download PDF
14. Protease-Mediated Suppression of DRG Neuron Excitability by Commensal Bacteria.
- Author
-
Sessenwein JL, Baker CC, Pradhananga S, Maitland ME, Petrof EO, Allen-Vercoe E, Noordhof C, Reed DE, Vanner SJ, and Lomax AE
- Subjects
- Animals, Ganglia, Spinal drug effects, Ganglia, Spinal microbiology, Gastrointestinal Microbiome drug effects, Humans, Male, Mice, Mice, Inbred C57BL, Neurons drug effects, Neurons microbiology, Peptide Hydrolases administration & dosage, Symbiosis drug effects, Ganglia, Spinal enzymology, Gastrointestinal Microbiome physiology, Granzymes administration & dosage, Neurons enzymology, Symbiosis physiology
- Abstract
Peripheral pain signaling reflects a balance of pronociceptive and antinociceptive influences; the contribution by the gastrointestinal microbiota to this balance has received little attention. Disorders, such as inflammatory bowel disease and irritable bowel syndrome, are associated with exaggerated visceral nociceptive actions that may involve altered microbial signaling, particularly given the evidence for bacterial dysbiosis. Thus, we tested whether a community of commensal gastrointestinal bacteria derived from a healthy human donor (microbial ecosystem therapeutics; MET-1) can affect the excitability of male mouse DRG neurons. MET-1 reduced the excitability of DRG neurons by significantly increasing rheobase, decreasing responses to capsaicin (2 μm) and reducing action potential discharge from colonic afferent nerves. The increase in rheobase was accompanied by an increase in the amplitude of voltage-gated K
+ currents. A mixture of bacterial protease inhibitors abrogated the effect of MET-1 effects on DRG neuron rheobase. A serine protease inhibitor but not inhibitors of cysteine proteases, acid proteases, metalloproteases, or aminopeptidases abolished the effects of MET-1. The serine protease cathepsin G recapitulated the effects of MET-1 on DRG neurons. Inhibition of protease-activated receptor-4 (PAR-4), but not PAR-2, blocked the effects of MET-1. Furthermore, Faecalibacterium prausnitzii recapitulated the effects of MET-1 on excitability of DRG neurons. We conclude that serine proteases derived from commensal bacteria can directly impact the excitability of DRG neurons, through PAR-4 activation. The ability of microbiota-neuronal interactions to modulate afferent signaling suggests that therapies that induce or correct microbial dysbiosis may impact visceral pain. SIGNIFICANCE STATEMENT Commercially available probiotics have the potential to modify visceral pain. Here we show that secretory products from gastrointestinal microbiota derived from a human donor signal to DRG neurons. Their secretory products contain serine proteases that suppress excitability via activation of protease-activated receptor-4. Moreover, from this community of commensal microbes, Faecalibacterium prausnitzii strain 16-6-I 40 fastidious anaerobe agar had the greatest effect. Our study suggests that therapies that induce or correct microbial dysbiosis may affect the excitability of primary afferent neurons, many of which are nociceptive. Furthermore, identification of the bacterial strains capable of suppressing sensory neuron excitability, and their mechanisms of action, may allow therapeutic relief for patients with gastrointestinal diseases associated with pain., (Copyright © 2017 the authors 0270-6474/17/3711758-11$15.00/0.)- Published
- 2017
- Full Text
- View/download PDF
15. A human gut ecosystem protects against C. difficile disease by targeting TcdA.
- Author
-
Martz SL, Guzman-Rodriguez M, He SM, Noordhof C, Hurlbut DJ, Gloor GB, Carlucci C, Weese S, Allen-Vercoe E, Sun J, Claud EC, and Petrof EO
- Subjects
- Animals, Bacterial Toxins metabolism, Caco-2 Cells, Clostridioides difficile isolation & purification, Cytoskeleton pathology, Disease Models, Animal, Enterocolitis, Pseudomembranous microbiology, Enterocolitis, Pseudomembranous pathology, Enterotoxins metabolism, Feces chemistry, Feces cytology, Feces microbiology, Fibroblasts pathology, Humans, Mice, Inbred C57BL, Bacterial Toxins antagonists & inhibitors, Biological Therapy methods, Clostridioides difficile growth & development, Enterocolitis, Pseudomembranous prevention & control, Enterotoxins antagonists & inhibitors, Gastrointestinal Microbiome
- Abstract
Background: A defined Microbial Ecosystem Therapeutic (MET-1, or "RePOOPulate") derived from the feces of a healthy volunteer can cure recurrent C. difficile infection (rCDI) in humans. The mechanisms of action whereby healthy microbiota protect against rCDI remain unclear. Since C. difficile toxins are largely responsible for the disease pathology of CDI, we hypothesized that MET-1 exerts its protective effects by inhibiting the effects of these toxins on the host., Methods: A combination of in vivo (antibiotic-associated mouse model of C. difficile colitis, mouse ileal loop model) and in vitro models (FITC-phalloidin staining, F actin Western blots and apoptosis assay in Caco2 cells, transepithelial electrical resistance measurements in T84 cells) were employed., Results: MET-1 decreased both local and systemic inflammation in infection and decreased both the cytotoxicity and the amount of TcdA detected in stool, without an effect on C. difficile viability. MET-1 protected against TcdA-mediated damage in a murine ileal loop model. MET-1 protected the integrity of the cytoskeleton in cells treated with purified TcdA, as indicated by FITC-phalloidin staining, F:G actin assays and preservation of transepithelial electrical resistance. Finally, co-incubation of MET-1 with purified TcdA resulted in decreased detectable TcdA by Western blot analysis., Conclusions: MET-1 intestinal microbiota confers protection against C. difficile and decreases C. difficile-mediated inflammation through its protective effects against C. difficile toxins, including enhancement of host barrier function and degradation of TcdA. The effect of MET-1 on C. difficile viability seems to offer little, if any, contribution to its protective effects on the host.
- Published
- 2017
- Full Text
- View/download PDF
16. Microbial therapeutic interventions.
- Author
-
Grady NG, Petrof EO, and Claud EC
- Subjects
- Humans, Dietary Supplements, Fecal Microbiota Transplantation, Microbiota, Prebiotics, Probiotics
- Abstract
The microbiome comprises all the microbes living in and on the human body. Human cells are greatly outnumbered by bacterial cells; thus human health depends on the health of the microbial ecosystem. For the immature preterm infant, the microbiome also influences intestinal and immune system development. This has implications for short term morbidities such as neonatal necrotizing enterocolitis and sepsis, but also long term health outcomes. Optimization of the preterm infant microbiome is a growing topic of interest. The microbial world is not one of good versus evil, but rather one of community; thus optimization includes not only minimizing pathogens, but also enhancing beneficial organisms. Options for optimization include judicious antibiotic use, administration of supplements such as prebiotics or probiotics, and transfaunation procedures such as fecal microbial transplant or microbial ecosystem therapeutics. Potential for benefit as well as risk for each of these options will be discussed., (Copyright © 2016 Elsevier Ltd. All rights reserved.)
- Published
- 2016
- Full Text
- View/download PDF
17. Fecal Microbiota-based Therapeutics for Recurrent Clostridium difficile Infection, Ulcerative Colitis and Obesity.
- Author
-
Carlucci C, Petrof EO, and Allen-Vercoe E
- Subjects
- Animals, Clostridium Infections microbiology, Colitis, Ulcerative microbiology, Dysbiosis, Humans, Obesity microbiology, Recurrence, Treatment Outcome, Clostridioides difficile, Clostridium Infections therapy, Colitis, Ulcerative therapy, Fecal Microbiota Transplantation, Gastrointestinal Microbiome, Obesity therapy
- Abstract
The human gut microbiome is a complex ecosystem of fundamental importance to human health. Our increased understanding of gut microbial composition and functional interactions in health and disease states has spurred research efforts examining the gut microbiome as a valuable target for therapeutic intervention. This review provides updated insight into the state of the gut microbiome in recurrent Clostridium difficile infection (CDI), ulcerative colitis (UC), and obesity while addressing the rationale for the modulation of the gut microbiome using fecal microbiota transplant (FMT)-based therapies. Current microbiome-based therapeutics in pre-clinical or clinical development are discussed. We end by putting this within the context of the current regulatory framework surrounding FMT and related therapies., (Copyright © 2016 The Authors. Published by Elsevier B.V. All rights reserved.)
- Published
- 2016
- Full Text
- View/download PDF
18. Preterm infant gut microbiota affects intestinal epithelial development in a humanized microbiome gnotobiotic mouse model.
- Author
-
Yu Y, Lu L, Sun J, Petrof EO, and Claud EC
- Subjects
- Animals, Germ-Free Life, Humans, Mice, Mice, Inbred C57BL, Intestinal Mucosa microbiology, Microbiota genetics
- Abstract
Development of the infant small intestine is influenced by bacterial colonization. To promote establishment of optimal microbial communities in preterm infants, knowledge of the beneficial functions of the early gut microbiota on intestinal development is needed. The purpose of this study was to investigate the impact of early preterm infant microbiota on host gut development using a gnotobiotic mouse model. Histological assessment of intestinal development was performed. The differentiation of four epithelial cell lineages (enterocytes, goblet cells, Paneth cells, enteroendocrine cells) and tight junction (TJ) formation was examined. Using weight gain as a surrogate marker for health, we found that early microbiota from a preterm infant with normal weight gain (MPI-H) induced increased villus height and crypt depth, increased cell proliferation, increased numbers of goblet cells and Paneth cells, and enhanced TJs compared with the changes induced by early microbiota from a poor weight gain preterm infant (MPI-L). Laser capture microdissection (LCM) plus qRT-PCR further revealed, in MPI-H mice, a higher expression of stem cell marker Lgr5 and Paneth cell markers Lyz1 and Cryptdin5 in crypt populations, along with higher expression of the goblet cell and mature enterocyte marker Muc3 in villus populations. In contrast, MPI-L microbiota failed to induce the aforementioned changes and presented intestinal characteristics comparable to a germ-free host. Our data demonstrate that microbial communities have differential effects on intestinal development. Future studies to identify pioneer settlers in neonatal microbial communities necessary to induce maturation may provide new insights for preterm infant microbial ecosystem therapeutics., (Copyright © 2016 the American Physiological Society.)
- Published
- 2016
- Full Text
- View/download PDF
19. Rebooting the microbiome.
- Author
-
Munoz S, Guzman-Rodriguez M, Sun J, Zhang YG, Noordhof C, He SM, Allen-Vercoe E, Claud EC, and Petrof EO
- Subjects
- Animals, Clostridioides difficile physiology, Clostridium Infections therapy, Colitis therapy, Humans, Mice, Mice, Inbred C57BL, Clostridium Infections microbiology, Colitis microbiology, Gastrointestinal Microbiome, Intestinal Mucosa microbiology
- Abstract
Using a murine Salmonella model of colitis, we recently reported that mice receiving a community of defined gut microbiota (MET-1) lost less weight, had reduced systemic inflammation and splenic S. typhimurium infection, and decreased neutrophil infiltration in the cecum, compared to vehicle controls. In addition, animals receiving MET-1 exhibited preserved tight junction protein expression (Zonula occludens-1, claudin-1), suggesting important effects on barrier function. In this addendum, we describe additional in vitro experiments examining effects of MET-1, as well as in vivo experiments demonstrating that MET-1 is protective in a DSS model of colitis after administration of antibiotics. Placed in the context of our findings and those of others, we discuss differences in our findings between the Salmonella colitis and DSS colitis models, provide speculation as to which bacteria may be important in the protective effects of MET-1, and discuss potential implications for other GI diseases such as IBD.
- Published
- 2016
- Full Text
- View/download PDF
20. Fecal microbiota transplantation: in perspective.
- Author
-
Gupta S, Allen-Vercoe E, and Petrof EO
- Abstract
There has been increasing interest in understanding the role of the human gut microbiome to elucidate the therapeutic potential of its manipulation. Fecal microbiota transplantation (FMT) is the administration of a solution of fecal matter from a donor into the intestinal tract of a recipient in order to directly change the recipient's gut microbial composition and confer a health benefit. FMT has been used to successfully treat recurrent Clostridium difficile infection. There are preliminary indications to suggest that it may also carry therapeutic potential for other conditions such as inflammatory bowel disease, obesity, metabolic syndrome, and functional gastrointestinal disorders.
- Published
- 2016
- Full Text
- View/download PDF
21. Frozen vs Fresh Fecal Microbiota Transplantation and Clinical Resolution of Diarrhea in Patients With Recurrent Clostridium difficile Infection: A Randomized Clinical Trial.
- Author
-
Lee CH, Steiner T, Petrof EO, Smieja M, Roscoe D, Nematallah A, Weese JS, Collins S, Moayyedi P, Crowther M, Ropeleski MJ, Jayaratne P, Higgins D, Li Y, Rau NV, and Kim PT
- Subjects
- Aged, Aged, 80 and over, Diarrhea etiology, Double-Blind Method, Enterocolitis, Pseudomembranous complications, Female, Humans, Male, Middle Aged, Recurrence, Treatment Outcome, Clostridioides difficile, Cryopreservation, Diarrhea therapy, Enterocolitis, Pseudomembranous therapy, Fecal Microbiota Transplantation
- Abstract
Importance: Clostridium difficile infection (CDI) is a major burden in health care and community settings. CDI recurrence is of particular concern because of limited treatment options and associated clinical and infection control issues. Fecal microbiota transplantation (FMT) is a promising, but not readily available, intervention., Objective: To determine whether frozen-and-thawed (frozen, experimental) FMT is noninferior to fresh (standard) FMT in terms of clinical efficacy among patients with recurrent or refractory CDI and to assess the safety of both types of FMT., Design, Setting, and Participants: Randomized, double-blind, noninferiority trial enrolling 232 adults with recurrent or refractory CDI, conducted between July 2012 and September 2014 at 6 academic medical centers in Canada., Interventions: Patients were randomly allocated to receive frozen (n = 114) or fresh (n = 118) FMT via enema., Main Outcomes and Measures: The primary outcome measures were clinical resolution of diarrhea without relapse at 13 weeks and adverse events. Noninferiority margin was set at 15%., Results: A total of 219 patients (n = 108 in the frozen FMT group and n = 111 in the fresh FMT group) were included in the modified intention-to-treat (mITT) population and 178 (frozen FMT: n = 91, fresh FMT: n = 87) in the per-protocol population. In the per-protocol population, the proportion of patients with clinical resolution was 83.5% for the frozen FMT group and 85.1% for the fresh FMT group (difference, -1.6% [95% CI, -10.5% to ∞]; P = .01 for noninferiority). In the mITT population the clinical resolution was 75.0% for the frozen FMT group and 70.3% for the fresh FMT group (difference, 4.7% [95% CI, -5.2% to ∞]; P < .001 for noninferiority). There were no differences in the proportion of adverse or serious adverse events between the treatment groups., Conclusions and Relevance: Among adults with recurrent or refractory CDI, the use of frozen compared with fresh FMT did not result in worse proportion of clinical resolution of diarrhea. Given the potential advantages of providing frozen FMT, its use is a reasonable option in this setting., Trial Registration: clinicaltrials.gov Identifier:NCT01398969.
- Published
- 2016
- Full Text
- View/download PDF
22. Recurrent Clostridium difficile infection and the microbiome.
- Author
-
Almeida R, Gerbaba T, and Petrof EO
- Subjects
- Dysbiosis complications, Enterocolitis, Pseudomembranous therapy, Fecal Microbiota Transplantation adverse effects, Fecal Microbiota Transplantation methods, Humans, Recurrence, Risk Factors, Clostridioides difficile, Enterocolitis, Pseudomembranous microbiology, Gastrointestinal Microbiome
- Abstract
The diverse and densely populated gastrointestinal microbiota is essential for the regulation of host physiology and immune function. As our knowledge of the composition and function of the intestinal microbiota continues to expand, there is new interest in using these developments to tailor fecal microbiota transplantation (FMT) and microbial ecosystem therapeutics (MET) for a variety of diseases. The potential role of FMT and MET in the treatment of Clostridium difficile infection (CDI)-currently the leading nosocomial gastrointestinal infection-has proven highly effective for recurrent CDI, and has emerged as a paradigm shift in the treatment of this disease. The current review will serve as a summary of the key aspects of CDI, and will introduce the essential framework and challenges of FMT, as is currently practiced. MET represents the progression of conventional bacteriotherapy that fundamentally capitalizes on the restorative properties of intestinal bacterial communities and may be viewed as the culmination of a rationally designed therapeutic modality. As our understanding of the composition and function of the intestinal microbiota evolves, it will likely drive next-generation microbiota therapies for a range of medical conditions, such as inflammatory bowel disease, obesity, and metabolic syndrome.
- Published
- 2016
- Full Text
- View/download PDF
23. A Practical Method for Preparation of Fecal Microbiota Transplantation.
- Author
-
Perez E, Lee CH, and Petrof EO
- Subjects
- Clostridioides difficile physiology, Colon microbiology, Colon pathology, Enterocolitis, Pseudomembranous microbiology, Enterocolitis, Pseudomembranous pathology, Fecal Microbiota Transplantation instrumentation, Feces microbiology, Gastrointestinal Microbiome physiology, Humans, Patient Selection ethics, Tissue Donors ethics, Clostridioides difficile pathogenicity, Enterocolitis, Pseudomembranous therapy, Fecal Microbiota Transplantation methods
- Abstract
Clostridium difficile is a challenging infection that can be difficult to treat with antibiotic therapy. This chapter outlines the processing material for fecal microbiota transplantation (FMT), also known as stool transplant. Fecal transplantations are effective in treating recurrent C. difficile infection (CDI). FMT uses a stool sample collected from a healthy, screened donor to restore healthy microbiota in the colon of a patient with CDI for symptom resolution. Here, we describe a rapid method for FMT preparation that uses inexpensive and disposable materials.
- Published
- 2016
- Full Text
- View/download PDF
24. Is Promoting Gut Microbial Diversity in Neonatal Enterocolitis the NECst Step?
- Author
-
Claud EC and Petrof EO
- Subjects
- Female, Humans, Male, Bacteria classification, Enterocolitis, Necrotizing microbiology, Enterocolitis, Necrotizing pathology, Inflammation metabolism, Opportunistic Infections microbiology
- Published
- 2015
- Full Text
- View/download PDF
25. Administration of defined microbiota is protective in a murine Salmonella infection model.
- Author
-
Martz SL, McDonald JA, Sun J, Zhang YG, Gloor GB, Noordhof C, He SM, Gerbaba TK, Blennerhassett M, Hurlbut DJ, Allen-Vercoe E, Claud EC, and Petrof EO
- Subjects
- Animals, Bacteria genetics, Bacteria isolation & purification, Body Weight, Cecum metabolism, Claudin-1 metabolism, Colitis microbiology, Colitis pathology, Cytokines blood, Disease Models, Animal, Feces microbiology, Humans, Intestinal Mucosa microbiology, Mice, Mice, Inbred C57BL, Mucins metabolism, NF-kappa B metabolism, Neutrophils immunology, Phylogeny, RNA, Ribosomal, 16S chemistry, RNA, Ribosomal, 16S genetics, RNA, Ribosomal, 16S metabolism, Salmonella Infections, Animal microbiology, Salmonella Infections, Animal pathology, Salmonella typhimurium growth & development, Salmonella typhimurium pathogenicity, Sequence Analysis, DNA, Spleen microbiology, Tight Junctions metabolism, Tumor Necrosis Factor-alpha genetics, Tumor Necrosis Factor-alpha metabolism, Zonula Occludens-1 Protein metabolism, Bacteria growth & development, Colitis therapy, Intestines microbiology, Microbiota, Salmonella Infections, Animal therapy
- Abstract
Salmonella typhimurium is a major cause of diarrhea and causes significant morbidity and mortality worldwide, and perturbations of the gut microbiota are known to increase susceptibility to enteric infections. The purpose of this study was to investigate whether a Microbial Ecosystem Therapeutic (MET-1) consisting of 33 bacterial strains, isolated from human stool and previously used to cure patients with recurrent Clostridium difficile infection, could also protect against S. typhimurium disease. C57BL/6 mice were pretreated with streptomycin prior to receiving MET-1 or control, then gavaged with S. typhimurium. Weight loss, serum cytokine levels, and S. typhimurium splenic translocation were measured. NF-κB nuclear staining, neutrophil accumulation, and localization of tight junction proteins (claudin-1, ZO-1) were visualized by immunofluorescence. Infected mice receiving MET-1 lost less weight, had reduced serum cytokines, reduced NF-κB nuclear staining, and decreased neutrophil infiltration in the cecum. MET-1 also preserved cecum tight junction protein expression, and reduced S. typhimurium translocation to the spleen. Notably, MET-1 did not decrease CFUs of Salmonella in the intestine. MET-1 may attenuate systemic infection by preserving tight junctions, thereby inhibiting S. typhimurium from gaining access to the systemic circulation. We conclude that MET-1 may be protective against enteric infections besides C. difficile infection.
- Published
- 2015
- Full Text
- View/download PDF
26. Vitamin D receptor pathway is required for probiotic protection in colitis.
- Author
-
Wu S, Yoon S, Zhang YG, Lu R, Xia Y, Wan J, Petrof EO, Claud EC, Chen D, and Sun J
- Subjects
- Animals, Colitis, Ulcerative microbiology, Colitis, Ulcerative prevention & control, Female, HCT116 Cells, Humans, Lactobacillus plantarum, Lacticaseibacillus rhamnosus, Mice, Mice, Inbred C57BL, Paneth Cells drug effects, Paneth Cells metabolism, Probiotics therapeutic use, RNA, Messenger genetics, RNA, Messenger metabolism, Receptors, Calcitriol genetics, Colitis, Ulcerative metabolism, Microbiota, Probiotics pharmacology, Receptors, Calcitriol metabolism
- Abstract
Low expression of vitamin D receptor (VDR) and dysfunction of vitamin D/VDR signaling are reported in patients with inflammatory bowel disease (IBD); therefore, restoration of VDR function to control inflammation in IBD is desirable. Probiotics have been used in the treatment of IBD. However, the role of probiotics in the modulation of VDR signaling to effectively reduce inflammation is unknown. We identified a novel role of probiotics in activating VDR activity, thus inhibiting inflammation, using cell models and VDR knockout mice. We found that the probiotics Lactobacillus rhamnosus strain GG (LGG) and Lactobacillus plantarum (LP) increased VDR protein expression in both mouse and human intestinal epithelial cells. Using the VDR luciferase reporter vector, we detected increased transcriptional activity of VDR after probiotic treatment. Probiotics increased the expression of the VDR target genes, such as antimicrobial peptide cathelicidin, at the transcriptional level. Furthermore, the role of probiotics in regulating VDR signaling was tested in vivo using a Salmonella-colitis model in VDR knockout mice. Probiotic treatment conferred physiological and histologic protection from Salmonella-induced colitis in VDR(+/+) mice, whereas probiotics had no effects in the VDR(-/-) mice. Probiotic treatment also enhanced numbers of Paneth cells, which secrete AMPs for host defense. These data indicate that the VDR pathway is required for probiotic protection in colitis. Understanding how probiotics enhance VDR signaling and inhibit inflammation will allow probiotics to be used effectively, resulting in innovative approaches to the prevention and treatment of chronic inflammation., (Copyright © 2015 the American Physiological Society.)
- Published
- 2015
- Full Text
- View/download PDF
27. Intestinal epithelial vitamin D receptor deletion leads to defective autophagy in colitis.
- Author
-
Wu S, Zhang YG, Lu R, Xia Y, Zhou D, Petrof EO, Claud EC, Chen D, Chang EB, Carmeliet G, and Sun J
- Subjects
- Aged, Aged, 80 and over, Animals, Colitis immunology, Down-Regulation physiology, Female, Humans, Immunohistochemistry, Immunoprecipitation, Intestinal Mucosa immunology, Mice, Inbred Strains, Mice, Knockout, Middle Aged, Paneth Cells metabolism, Receptors, Calcitriol immunology, Autophagy physiology, Colitis physiopathology, Intestinal Mucosa metabolism, Receptors, Calcitriol physiology
- Abstract
Objective: Vitamin D and the vitamin D receptor (VDR) appear to be important immunological regulators of inflammatory bowel diseases (IBD). Defective autophagy has also been implicated in IBD, where interestingly, polymorphisms of genes such as ATG16L1 have been associated with increased risk. Although vitamin D, the microbiome and autophagy are all involved in pathogenesis of IBD, it remains unclear whether these processes are related or function independently., Design: We investigated the effects and mechanisms of intestinal epithelial VDR in healthy and inflamed states using cell culture models, a conditional VDR knockout mouse model (VDR(ΔIEC)), colitis models and human samples., Results: Absence of intestinal epithelial VDR affects microbial assemblage and increases susceptibility to dextran sulfate sodium-induced colitis. Intestinal epithelial VDR downregulates expressions of ATG16L1 and lysozyme, and impairs antimicrobial function of Paneth cells. Gain and loss-of-function assays showed that VDR levels regulate ATG16L1 and lysozyme at the transcriptional and translational levels. Moreover, low levels of intestinal epithelial VDR correlated with reduced ATG16L1 and representation by intestinal Bacteroides in patients with IBD. Administration of the butyrate (a fermentation product of gut microbes) increases intestinal VDR expression and suppresses inflammation in a colitis model., Conclusions: Our study demonstrates fundamental relationship between VDR, autophagy and gut microbial assemblage that is essential for maintaining intestinal homeostasis, but also in contributing to the pathophysiology of IBD. These insights can be leveraged to define therapeutic targets for restoring VDR expression and function., (Published by the BMJ Publishing Group Limited. For permission to use (where not already granted under a licence) please go to http://group.bmj.com/group/rights-licensing/permissions.)
- Published
- 2015
- Full Text
- View/download PDF
28. Microbial composition analysis of Clostridium difficile infections in an ulcerative colitis patient treated with multiple fecal microbiota transplantations.
- Author
-
Brace C, Gloor GB, Ropeleski M, Allen-Vercoe E, and Petrof EO
- Subjects
- Aged, 80 and over, Colitis, Ulcerative complications, Colitis, Ulcerative diagnosis, Enterocolitis, Pseudomembranous complications, Enterocolitis, Pseudomembranous diagnosis, Feces microbiology, Humans, Male, Sigmoidoscopy, Clostridioides difficile isolation & purification, Colitis, Ulcerative microbiology, Enterocolitis, Pseudomembranous microbiology, Microbiota
- Abstract
Fecal microbiota transplantation (FMT) is a promising therapy for Clostridium difficile infection (CDI). However, questions remain regarding efficacy and safety in inflammatory bowel disease (IBD) patients, as well as longitudinal stability of donor stool composition. This report describes an IBD patient with two CDIs 18 months apart, each successfully treated with FMT with no IBD flares or complications. Microbiome composition analysis of patient samples during each infection revealed low-diversity microbiota patterns similar to those previously described in non-IBD patients with CDI and active IBD alone. Samples taken after each transplant demonstrated quick remodeling towards the donor's sample composition coinciding with symptom resolution. Of note, samples taken from the same donor 18 months apart reflected marked differences in microbiota abundances, suggesting that the use of single donors in FMT programs offers little benefit in ensuring predictability of donor stool composition over time. This report describes similar microbial composition patterns during CDI in IBD patients to those described previously in non-IBD patients, and supports FMT as safe and effective treatment for recurring CDI in this patient population., (Copyright © 2014 European Crohn's and Colitis Organisation. Published by Elsevier B.V. All rights reserved.)
- Published
- 2014
- Full Text
- View/download PDF
29. Differential expression of 26S proteasome subunits and functional activity during neonatal development.
- Author
-
Claud EC, McDonald JA, He SM, Yu Y, Duong L, Sun J, and Petrof EO
- Subjects
- Adaptive Immunity, Adenosine Triphosphatases genetics, Adenosine Triphosphatases metabolism, Animals, Animals, Newborn, Chymotrypsin metabolism, Female, Hydrolysis, Organ Specificity, Pregnancy, Proteasome Endopeptidase Complex chemistry, Protein Subunits chemistry, Protein Subunits genetics, Protein Subunits metabolism, Rats, Time Factors, Trypsin metabolism, Weaning, Gene Expression Regulation, Developmental, Proteasome Endopeptidase Complex genetics, Proteasome Endopeptidase Complex metabolism
- Abstract
Proteasomes regulate many essential cellular processes by degrading intracellular proteins. While aging is known to be associated with dysfunction of the proteasome, there are few reports detailing activity and function of proteasomes in the early stages of life. To elucidate the function and development of mammalian proteasomes, 26S proteasomes were affinity-purified from rat intestine, spleen and liver. The developmental expression of core, regulatory and immunoproteasome subunits was analyzed by immunoblotting and reverse-transcriptase PCR of mRNA subunits, and proteasome catalytic function was determined by fluorogenic enzymatic assays. The expression of core (β2, β5, α7 and β1) and regulatory (Rpt5) subunits was found to be present at low levels at birth and increased over time particularly at weaning. In contrast, while gradual developmental progression of proteasome structure was also seen with the immunoproteasome subunits (β1i, β5i, and β2i), these were not present at birth. Our studies demonstrate a developmental pattern to 26S proteasome activity and subunit expression, with low levels of core proteasome components and absence of immunoproteasomes at birth followed by increases at later developmental stages. This correlates with findings from other studies of a developmental hyporesponsiveness of the adaptive immune system to allow establishment of microbial colonization immediately after birth.
- Published
- 2014
- Full Text
- View/download PDF
30. From stool transplants to next-generation microbiota therapeutics.
- Author
-
Petrof EO and Khoruts A
- Subjects
- Animals, Clostridioides difficile pathogenicity, Dysbiosis, Enterocolitis, Pseudomembranous epidemiology, Enterocolitis, Pseudomembranous microbiology, Epidemics, Host-Pathogen Interactions, Humans, Recurrence, Risk Factors, Treatment Outcome, Biological Therapy methods, Clostridioides difficile growth & development, Enterocolitis, Pseudomembranous therapy, Feces microbiology, Intestines microbiology, Microbiota
- Abstract
The epidemic of Clostridium difficile infection fueled by new virulent strains of the organism has led to increased use of fecal microbiota transplantation (FMT). The procedure is effective for even the most desperate cases after failure of multiple courses of antibiotics. The approach recognizes microbiota to be integral to normal human physiology, and microbiota being used in FMT represents a new class of therapeutics. Imbalance in the composition and altered activity of the microbiota are associated with many diseases. Consequently, there is growing interest in applying FMT to non-C difficile indications. However, this may succeed only if microbiota therapeutics are developed systematically, based on mechanistic understanding, and applying up-to-date principles of microbial ecology. We discuss 2 pathways in the development of this new therapeutic class: whole microbial communities separated from donor stool and an assembly of specific fecal microorganisms grown in vitro., (Copyright © 2014 AGA Institute. Published by Elsevier Inc. All rights reserved.)
- Published
- 2014
- Full Text
- View/download PDF
31. The microbiome: what it means for medicine.
- Author
-
Allen-Vercoe E and Petrof EO
- Subjects
- Anti-Bacterial Agents administration & dosage, Anti-Bacterial Agents adverse effects, Bacterial Typing Techniques, Host-Pathogen Interactions, Humans, Precision Medicine, Biomedical Research trends, Microbiota
- Published
- 2014
- Full Text
- View/download PDF
32. Small molecule immunomodulins from cultures of the human microbiome member Lactobacillus plantarum.
- Author
-
Zvanych R, Lukenda N, Kim JJ, Li X, Petrof EO, Khan WI, and Magarvey NA
- Subjects
- Animals, Dipeptides chemistry, Dipeptides isolation & purification, Dipeptides pharmacology, Humans, Immunologic Factors chemistry, Immunologic Factors isolation & purification, Inflammation Mediators metabolism, Male, Mice, Mice, Inbred C57BL, Microbiota, Phenylalanine chemistry, Phenylalanine isolation & purification, Phenylalanine pharmacology, Principal Component Analysis, Pyrrolidonecarboxylic Acid chemistry, Tryptophan chemistry, Tryptophan isolation & purification, Tryptophan pharmacology, Immunologic Factors pharmacology, Interferon-gamma metabolism, Lactobacillus plantarum metabolism, Metabolomics
- Abstract
Lactobacillus plantarum strains are noted for their presence in the human gastrointestinal tract and are distinguished for their immunomodulatory actions and therapeutic applications. Despite the uncertainty in the underlining molecular mechanisms, recent evidence suggests that L. plantarum secretes immunomodulatory agents that alter immunological signaling cascades. Elaboration of these metabolic products from L. plantarum strain WCFS1 was demonstrated previously to correlate with the mid-log-stationary transition, perhaps consistent with secondary metabolite expression. Here, we present the metabolomic shifts revealed by principal component analysis that correspond to the mid-log-stationary transition of L. plantarum, and identify pyroglutamic (pyro) dipeptides within this transition as correlative with the immunomodulatory actions. Four of these (pyro-phenylalanine, pyro-leucine, pyro-isoleucine, pyro-tryptophan) were characterized and the two dominant members, pyro-phenylalanine and pyro-tryptophan, were directly interrogated for immunomodulatory activity through in vivo administration using C57BL/6 mice. Administration of these compounds resulted in decreased production of pro-inflammatory cytokine interferon (IFN)-gamma, which is of noted importance in gastrointestinal immune homeostasis.
- Published
- 2014
- Full Text
- View/download PDF
33. Oral administration of transforming growth factor-β1 (TGF-β1) protects the immature gut from injury via Smad protein-dependent suppression of epithelial nuclear factor κB (NF-κB) signaling and proinflammatory cytokine production.
- Author
-
Shiou SR, Yu Y, Guo Y, Westerhoff M, Lu L, Petrof EO, Sun J, and Claud EC
- Subjects
- Administration, Oral, Animals, Cell Line, Gastrointestinal Diseases metabolism, Gastrointestinal Diseases pathology, Humans, Immunity, Innate genetics, Inflammation drug therapy, Inflammation genetics, Interferon-gamma metabolism, Interleukin-6 metabolism, Interleukin-8, Intestinal Mucosa injuries, Intestinal Mucosa metabolism, Promoter Regions, Genetic, Rats, Signal Transduction, Smad2 Protein metabolism, Transforming Growth Factor beta1 metabolism, Gastrointestinal Diseases drug therapy, Inflammation metabolism, NF-kappa B metabolism, Smad Proteins metabolism, Transforming Growth Factor beta1 administration & dosage
- Abstract
Inflammatory immune responses play an important role in mucosal homeostasis and gut diseases. Nuclear factor κB (NF-κB), central to the proinflammatory cascade, is activated in necrotizing enterocolitis (NEC), a devastating condition of intestinal injury with extensive inflammation in premature infants. TGF-β is a strong immune suppressor and a factor in breast milk, which has been shown to be protective against NEC. In an NEC animal model, oral administration of the isoform TGF-β1 activated the downstream effector Smad2 in intestine and significantly reduced NEC incidence. In addition, TGF-β1 suppressed NF-κB activation, maintained levels of the NF-κB inhibitor IκBα in the intestinal epithelium, and systemically decreased serum levels of IL-6 and IFN-γ. The immature human fetal intestinal epithelial cell line H4 was used as a reductionistic model of the immature enterocyte to investigate mechanism. TGF-β1 pretreatment inhibited the TNF-α-induced IκBα phosphorylation that targets the IκBα protein for degradation and inhibited NF-κB activation. Chromatin immunoprecipitation (ChIP) assays demonstrated decreased NF-κB binding to the promoters of IL-6, IL-8, and IκBα in response to TNF-α with TGF-β1 pretreatment. These TGF-β1 effects appear to be mediated through the canonical Smad pathway as silencing of the TGF-β central mediator Smad4 resulted in loss of the TGF-β1 effects. Thus, TGF-β1 is capable of eliciting anti-inflammatory effects by inhibiting NF-κB specifically in the intestinal epithelium as well as by decreasing systemic IL-6 and IFN-γ levels. Oral administration of TGF-β1 therefore can potentially be used to protect against gastrointestinal diseases.
- Published
- 2013
- Full Text
- View/download PDF
34. Erythropoietin protects epithelial cells from excessive autophagy and apoptosis in experimental neonatal necrotizing enterocolitis.
- Author
-
Yu Y, Shiou SR, Guo Y, Lu L, Westerhoff M, Sun J, Petrof EO, and Claud EC
- Subjects
- Animals, Animals, Newborn, Apoptosis Regulatory Proteins genetics, Apoptosis Regulatory Proteins metabolism, Beclin-1, Caspase 3 genetics, Caspase 3 metabolism, Disease Models, Animal, Enterocolitis, Necrotizing genetics, Enterocolitis, Necrotizing metabolism, Enterocolitis, Necrotizing pathology, Epithelial Cells metabolism, Epithelial Cells pathology, Erythropoietin genetics, Erythropoietin metabolism, Extracellular Signal-Regulated MAP Kinases genetics, Extracellular Signal-Regulated MAP Kinases metabolism, Gene Expression, Humans, Infant, Newborn, Intestinal Mucosa metabolism, Intestinal Mucosa pathology, Intestines pathology, Membrane Proteins genetics, Membrane Proteins metabolism, Microtubule-Associated Proteins genetics, Microtubule-Associated Proteins metabolism, Milk, Human chemistry, Proto-Oncogene Proteins c-akt genetics, Proto-Oncogene Proteins c-akt metabolism, Rats, Signal Transduction, TOR Serine-Threonine Kinases genetics, TOR Serine-Threonine Kinases metabolism, Apoptosis, Autophagy, Enterocolitis, Necrotizing prevention & control, Epithelial Cells drug effects, Erythropoietin pharmacology, Intestinal Mucosa drug effects, Intestines drug effects
- Abstract
Neonatal necrotizing enterocolitis (NEC) is a devastating gastrointestinal disease of preterm infants. Increased intestinal epithelium permeability is an early event in NEC pathogenesis. Autophagy and apoptosis are induced by multiple stress pathways which may impact the intestinal barrier, and they have been associated with pathogenesis of diverse gastrointestinal diseases including inflammatory bowel disease. Using both in vitro and in vivo models, this study investigates autophagy and apoptosis under experimental NEC stresses. Furthermore this study evaluates the effect of erythropoietin (Epo), a component of breast milk previously shown to decrease the incidence of NEC and to preserve intestinal barrier function, on intestinal autophagy and apoptosis. It was found that autophagy and apoptosis are both rapidly up regulated in NEC in vivo as indicated by increased expression of the autophagy markers Beclin 1 and LC3II, and by evidence of apoptosis by TUNEL and cleaved caspase-3 staining. In the rat NEC experimental model, autophagy preceded the onset of apoptosis in intestine. In vitro studies suggested that Epo supplementation significantly decreased both autophagy and apoptosis via the Akt/mTOR signaling pathway and the MAPK/ERK pathway respectively. These results suggest that Epo protects intestinal epithelium from excessive autophagy and apoptosis in experimental NEC.
- Published
- 2013
- Full Text
- View/download PDF
35. Synergistic protection of combined probiotic conditioned media against neonatal necrotizing enterocolitis-like intestinal injury.
- Author
-
Shiou SR, Yu Y, Guo Y, He SM, Mziray-Andrew CH, Hoenig J, Sun J, Petrof EO, and Claud EC
- Subjects
- Animals, Animals, Newborn, Enterocolitis, Necrotizing metabolism, Female, Intestinal Mucosa metabolism, Intestines drug effects, Intestines microbiology, Intestines pathology, NF-kappa B metabolism, Pregnancy, Proteasome Endopeptidase Complex metabolism, Rats, Tight Junctions drug effects, Tight Junctions metabolism, Tumor Necrosis Factor-alpha biosynthesis, Zonula Occludens-1 Protein metabolism, Culture Media, Conditioned pharmacology, Enterocolitis, Necrotizing prevention & control, Probiotics administration & dosage
- Abstract
Balance among the complex interactions of the gut microbial community is important for intestinal health. Probiotic bacteria can improve bacterial balance and have been used to treat gastrointestinal diseases. Neonatal necrotizing enterocolitis (NEC) is a life-threatening inflammatory bowel disorder primarily affecting premature infants. NEC is associated with extensive inflammatory NF-κB signaling activation as well as intestinal barrier disruption. Clinical studies have shown that probiotic administration may protect against NEC, however there are safety concerns associated with the ingestion of large bacterial loads in preterm infants. Bacteria-free conditioned media (CM) from certain probiotic organisms have been shown to retain bioactivity including anti-inflammatory and cytoprotective properties without the risks of live organisms. We hypothesized that the CM from Lactobacillus acidophilus (La), Bifidobacterium infantis (Bi), and Lactobacillus plantarum (Lp), used separately or together would protect against NEC. A rodent model with intestinal injury similar to NEC was used to study the effect of CM from Lp, La/Bi, and La/Bi/Lp on the pathophysiology of NEC. All the CM suppressed NF-κB activation via preserved IκBα expression and this protected IκBα was associated with decreased liver activity of the proteasome, which is the degrading machinery for IκBα. These CM effects also caused decreases in intestinal production of the pro-inflammatory cytokine TNF-α, a downstream target of the NF-κB pathway. Combined La/Bi and La/Bi/Lp CM in addition protected intestinal barrier function by maintaining tight junction protein ZO-1 levels and localization at the tight junction. Double combined La/Bi CM significantly reduced intestinal injury incidence from 43% to 28% and triple combined La/Bi/Lp CM further reduced intestinal injury incidence to 20%. Thus, this study demonstrates different protective mechanisms and synergistic bioactivity of the CM from different organisms in ameliorating NEC-like intestinal injury in an animal model.
- Published
- 2013
- Full Text
- View/download PDF
36. Artificial stool transplantation: progress towards a safer, more effective and acceptable alternative.
- Author
-
Allen-Vercoe E and Petrof EO
- Subjects
- Anti-Bacterial Agents therapeutic use, Diarrhea diagnosis, Diarrhea microbiology, Enterocolitis, Pseudomembranous diagnosis, Enterocolitis, Pseudomembranous microbiology, Humans, Recurrence, Severity of Illness Index, Treatment Outcome, Biological Therapy methods, Clostridioides difficile pathogenicity, Colonoscopy, Diarrhea therapy, Enterocolitis, Pseudomembranous therapy, Feces microbiology
- Published
- 2013
- Full Text
- View/download PDF
37. Microbial ecosystems therapeutics: a new paradigm in medicine?
- Author
-
Petrof EO, Claud EC, Gloor GB, and Allen-Vercoe E
- Subjects
- Clostridioides difficile pathogenicity, Clostridium Infections microbiology, Clostridium Infections therapy, Humans, Biological Products therapeutic use, Biota, Gastrointestinal Tract microbiology, Gastrointestinal Tract physiology, Metagenome
- Abstract
Increasing evidence indicates that the complex microbial ecosystem of the human intestine plays a critical role in protecting the host against disease. This review discusses gut dysbiosis (here defined as a state of imbalance in the gut microbial ecosystem, including overgrowth of some organisms and loss of others) as the foundation for several diseases, and the applicability of refined microbial ecosystem replacement therapies as a future treatment modality. Consistent with the concept of a 'core' microbiome encompassing key functions required for normal intestinal homeostasis, 'Microbial Ecosystem Therapeutics' (MET) would entail replacing a dysfunctional, damaged ecosystem with a fully developed and healthy ecosystem of 'native' intestinal bacteria. Its application in treating Clostridium difficile infection is discussed and possible applications to other diseases such as ulcerative colitis, obesity, necrotising enterocolitis, and regressive-type autism are reviewed. Unlike conventional probiotic therapies that are generally limited to a single strain or at most a few strains of bacteria 'Microbial Ecosystem Therapeutics' would utilise whole bacterial communities derived directly from the human gastrointestinal tract. By taking into account the intrinsic needs of the entire microbial ecosystem, MET would emphasise the rational design of healthy, resilient and robust microbial communities that could be used to maintain or restore human health. More than simply a new probiotic treatment, this emerging paradigm in medicine may lead to novel strategies in treating and managing a wide variety of human diseases.
- Published
- 2013
- Full Text
- View/download PDF
38. Stool substitute transplant therapy for the eradication of Clostridium difficile infection: 'RePOOPulating' the gut.
- Author
-
Petrof EO, Gloor GB, Vanner SJ, Weese SJ, Carter D, Daigneault MC, Brown EM, Schroeter K, and Allen-Vercoe E
- Abstract
Background: Fecal bacteriotherapy ('stool transplant') can be effective in treating recurrent Clostridium difficile infection, but concerns of donor infection transmission and patient acceptance limit its use. Here we describe the use of a stool substitute preparation, made from purified intestinal bacterial cultures derived from a single healthy donor, to treat recurrent C. difficile infection that had failed repeated standard antibiotics. Thirty-three isolates were recovered from a healthy donor stool sample. Two patients who had failed at least three courses of metronidazole or vancomycin underwent colonoscopy and the mixture was infused throughout the right and mid colon. Pre-treatment and post-treatment stool samples were analyzed by 16 S rRNA gene sequencing using the Ion Torrent platform., Results: Both patients were infected with the hyper virulent C. difficile strain, ribotype 078. Following stool substitute treatment, each patient reverted to their normal bowel pattern within 2 to 3 days and remained symptom-free at 6 months. The analysis demonstrated that rRNA sequences found in the stool substitute were rare in the pre-treatment stool samples but constituted over 25% of the sequences up to 6 months after treatment., Conclusion: This proof-of-principle study demonstrates that a stool substitute mixture comprising a multi-species community of bacteria is capable of curing antibiotic-resistant C. difficile colitis. This benefit correlates with major changes in stool microbial profile and these changes reflect isolates from the synthetic mixture., Clinical Trial Registration Number: CinicalTrials.gov NCT01372943.
- Published
- 2013
- Full Text
- View/download PDF
39. Consistent activation of the β-catenin pathway by Salmonella type-three secretion effector protein AvrA in chronically infected intestine.
- Author
-
Lu R, Liu X, Wu S, Xia Y, Zhang YG, Petrof EO, Claud EC, and Sun J
- Subjects
- Animals, Bacterial Proteins pharmacology, Cecum microbiology, Cell Proliferation drug effects, Epithelial Cells physiology, Mice, Bacterial Proteins physiology, Salmonella Infections, Animal physiopathology, beta Catenin metabolism
- Abstract
Salmonella infection is a common public health problem that can become chronic and increase the risk of cancer. Live, mutated Salmonella is used to target cancer cells. However, few studies have addressed chronic Salmonella infection in vivo. AvrA is a Salmonella type-three secretion effector that is multifunctional, inhibiting intestinal inflammation and enhancing proliferation. β-catenin is a key player in intestinal renewal, inflammation, and tumorigenesis. We hypothesize that in Salmonella-infected intestine, AvrA chronically activates the β-catenin pathway and increases cell proliferation, thus deregulating the intestinal responses to bacterial infection. We followed mice with Salmonella infection for 27 wk and investigated the physiological effects and role of AvrA on β-catenin in chronically infected intestine. We found that AvrA persistently regulated β-catenin posttranslational modifications, including phosphorylation and acetylation. Moreover, the upstream regulator Akt, transcription factors, T cell factors, nuclear β-catenin, and β-catenin target genes were enhanced in mice infected with Salmonella-expressing AvrA. AvrA has a chronic functional role in promoting intestinal renewal. In summary, we have uncovered an essential role of Salmonella AvrA in chronically activating β-catenin and impacting intestinal renewal in small intestine and colon. Our study emphasizes the importance of AvrA in chronic bacterial infection.
- Published
- 2012
- Full Text
- View/download PDF
40. A Canadian Working Group report on fecal microbial therapy: microbial ecosystems therapeutics.
- Author
-
Allen-Vercoe E, Reid G, Viner N, Gloor GB, Hota S, Kim P, Lee C, O'Doherty K, Vanner SJ, Weese JS, and Petrof EO
- Subjects
- Canada, Computational Biology, Congresses as Topic, Ecosystem, Gastroenterology ethics, Gastroenterology trends, Humans, Metagenome, Enterocolitis, Pseudomembranous therapy, Feces microbiology
- Abstract
A working group from across Canada comprised of clinician and basic scientists, epidemiologists, ethicists, Health Canada regulatory authorities and representatives of major funding agencies (Canadian Institutes of Health Research and the Crohn's and Colitis Foundation of Canada) met to review the current experience with fecal microbial therapy and to identify the key areas of study required to move this field forward. The report highlights the promise of fecal microbial therapy and related synthetic stool therapy (together called 'microbial ecosystems therapeutics') for the treatment of Clostridium difficile colitis and, possibly, other disorders. It identifies pressing clinical issues that need to be addressed as well as social, ethical and regulatory barriers to the use of these important therapies.
- Published
- 2012
- Full Text
- View/download PDF
41. Axin1 prevents Salmonella invasiveness and inflammatory response in intestinal epithelial cells.
- Author
-
Zhang YG, Wu S, Xia Y, Chen D, Petrof EO, Claud EC, Hsu W, and Sun J
- Subjects
- Animals, Caco-2 Cells, Disease Models, Animal, Epithelial Cells immunology, Epithelial Cells metabolism, Gene Expression, HCT116 Cells, HT29 Cells, Host-Pathogen Interactions immunology, Humans, Inflammation genetics, Mice, Mice, Inbred C57BL, Salmonella Infections genetics, Ubiquitination, Wnt Signaling Pathway, beta Catenin genetics, beta Catenin metabolism, Axin Protein genetics, Axin Protein immunology, Host-Pathogen Interactions genetics, Immunity, Innate genetics, Salmonella pathogenicity, Salmonella Infections immunology
- Abstract
Background: Axin1 and its homolog Axin2 are scaffold proteins essential for regulating Wnt signaling. Axin-dependent regulation of Wnt is important for various developmental processes and human diseases. However, the involvement of Axin1 and Axin2 in host defense and inflammation remains to be determined., Methods/principal Findings: Here, we report that Axin1, but not Axin2, plays an essential role in host-pathogen interaction mediated by the Wnt pathway. Pathogenic Salmonella colonization greatly reduces the level of Axin1 in intestinal epithelial cells. This reduction is regulated at the posttranslational level in early onset of the bacterial infection. Further analysis reveals that the DIX domain and Ser614 of Axin1 are necessary for the Salmonella-mediated modulation through ubiquitination and SUMOylation., Conclusion/significance: Axin1 apparently has a preventive effect on bacterial invasiveness and inflammatory response during the early stages of infection. The results suggest a distinct biological function of Axin1 and Axin2 in infectious disease and intestinal inflammation while they are functionally equivalent in developmental settings.
- Published
- 2012
- Full Text
- View/download PDF
42. Influence of 1,25-dihydroxy vitamin D3 on TLR4-induced activation of antigen presenting cells is dependent on the order of receptor engagement.
- Author
-
Gambhir V, Kim J, Siddiqui S, Taylor M, Byford V, Petrof EO, Jones G, and Basta S
- Subjects
- Animals, Blotting, Western, Dendritic Cells immunology, Dendritic Cells metabolism, Enzyme Activation drug effects, Flow Cytometry, Immunity physiology, Immunomodulation, Interleukin-6 immunology, Interleukin-6 metabolism, Interleukin-6 pharmacology, Lipopolysaccharides pharmacology, Macrophages drug effects, Macrophages immunology, Macrophages metabolism, Mice, Mice, Inbred C57BL, Microscopy, Fluorescence, Monocytes drug effects, Monocytes immunology, Monocytes metabolism, Receptors, Calcitriol genetics, Receptors, Calcitriol immunology, Signal Transduction immunology, Steroid Hydroxylases genetics, Toll-Like Receptor 4 genetics, Toll-Like Receptor 4 immunology, Toll-Like Receptor 4 metabolism, Tumor Necrosis Factor-alpha immunology, Tumor Necrosis Factor-alpha metabolism, Tumor Necrosis Factor-alpha pharmacology, Vitamin D pharmacology, Vitamin D3 24-Hydroxylase, Dendritic Cells drug effects, Immunity drug effects, Receptors, Calcitriol metabolism, Signal Transduction drug effects, Steroid Hydroxylases metabolism, Vitamin D analogs & derivatives
- Abstract
The vitamin D metabolite, 1,25-(OH)₂D₃, binds the vitamin D receptor (VDR) to exert its regulatory effects at the transcription level. VDR is expressed in professional antigen-presenting cells (pAPCs), such as macrophages (Mø) and dendritic cells (DCs). We show for the first time that the 24-hydroxylase enzyme is activated in bone marrow-derived dendritic cell (BMDC), due to 1,25(OH)₂D₃ stimulation which resulted in the induction of its gene, CYP24A1. Furthermore, we provide evidence that the influence of 1,25-(OH)₂D₃ on TLR-4-L-induced activation of pAPC is dependent on the order of VDR and TLR-4 engagement. Thus, pre-treatment of pAPC with 1,25-(OH)₂D₃ partially inhibited LPS-induced nitric oxide (NO) production. However, these inhibitory effects were not observed when LPS and 1,25-(OH)₂D₃ were added simultaneously or when LPS preceded 1,25-(OH)₂D₃. Moreover, we found that 1,25-(OH)₂D₃ pre-treatment of pAPCs did not cause general suppression since it interfered with NO levels but not with the cytokines IL-6 or TNF-α. Consequently, engagement of VDR by 1,25-(OH)₂D₃ can partially interfere with TLR-4-L-induced activation of pAPCs only when it occurs before TLR-4 stimulation., (Copyright © 2011 Elsevier GmbH. All rights reserved.)
- Published
- 2011
- Full Text
- View/download PDF
43. Mother's milk-induced Hsp70 expression preserves intestinal epithelial barrier function in an immature rat pup model.
- Author
-
Liedel JL, Guo Y, Yu Y, Shiou SR, Chen S, Petrof EO, Hu S, Musch MW, and Claud EC
- Subjects
- Animals, Animals, Newborn, Bacterial Infections metabolism, Bacterial Infections microbiology, Cytoprotection, Disease Models, Animal, HSP70 Heat-Shock Proteins genetics, Hypoxia metabolism, Ileum microbiology, Ileum pathology, Intestinal Mucosa microbiology, Intestinal Mucosa pathology, Membrane Proteins metabolism, Oxidative Stress, Permeability, Phosphoproteins metabolism, RNA, Messenger metabolism, Rats, Tight Junctions metabolism, Up-Regulation, Zonula Occludens-1 Protein, HSP70 Heat-Shock Proteins metabolism, Ileum metabolism, Intestinal Mucosa metabolism, Milk metabolism
- Abstract
Preterm infants face many challenges in transitioning from the in utero to extrauterine environment while still immature. Failure of the preterm gut to successfully mature to accommodate bacteria and food substrate leads to significant morbidity such as neonatal necrotizing enterocolitis. The intestinal epithelial barrier plays a critical role in gut protection. Heat shock protein 70 (Hsp70) is an inducible cytoprotective molecule shown to protect the intestinal epithelium in adult models. To investigate the hypothesis that Hsp70 may be important for early protection of the immature intestine, Hsp70 expression was evaluated in intestine of immature rat pups. Data demonstrate that Hsp70 is induced by exposure to mother's milk. Hsp70 is found in mother's milk, and increased Hsp70 transcription is induced by mother's milk. This Hsp70 colocalizes with the tight junction protein ZO-1. Mother's milk-induced Hsp70 may contribute to maintenance of barrier function in the face of oxidant stress. Further understanding of the means by which mother's milk increases Hsp70 in the ileum will allow potential means of strengthening the intestinal barrier in at-risk preterm infants.
- Published
- 2011
- Full Text
- View/download PDF
44. Erythropoietin protects intestinal epithelial barrier function and lowers the incidence of experimental neonatal necrotizing enterocolitis.
- Author
-
Shiou SR, Yu Y, Chen S, Ciancio MJ, Petrof EO, Sun J, and Claud EC
- Subjects
- Animals, Animals, Newborn, Cell Line, Tumor, Disease Models, Animal, Electric Impedance, Enterocytes cytology, Enterocytes drug effects, Enterocytes metabolism, Erythropoietin therapeutic use, Fluorescent Antibody Technique, Humans, Immunoblotting, Intestinal Mucosa cytology, Membrane Proteins genetics, Membrane Proteins metabolism, Phosphatidylinositol 3-Kinases metabolism, Phosphoinositide-3 Kinase Inhibitors, Phosphoproteins genetics, Phosphoproteins metabolism, Proto-Oncogene Proteins c-akt antagonists & inhibitors, Proto-Oncogene Proteins c-akt metabolism, RNA Interference, Rats, Tight Junctions drug effects, Tight Junctions metabolism, Zonula Occludens-1 Protein, Enterocolitis, Necrotizing drug therapy, Erythropoietin pharmacology, Intestinal Mucosa metabolism, Intestines cytology, Intestines drug effects
- Abstract
The impermeant nature of the intestinal barrier is maintained by tight junctions (TJs) formed between adjacent intestinal epithelial cells. Disruption of TJs and loss of barrier function are associated with a number of gastrointestinal diseases, including neonatal necrotizing enterocolitis (NEC), the leading cause of death from gastrointestinal diseases in preterm infants. Human milk is protective against NEC, and the human milk factor erythropoietin (Epo) has been shown to protect endothelial cell-cell and blood-brain barriers. We hypothesized that Epo may also protect intestinal epithelial barriers, thereby lowering the incidence of NEC. Our data demonstrate that Epo protects enterocyte barrier function by supporting expression of the TJ protein ZO-1. As immaturity is a key factor in NEC, Epo regulation of ZO-1 in the human fetal immature H4 intestinal epithelial cell line was examined and demonstrated Epo-stimulated ZO-1 expression in a dose-dependent manner through the PI3K/Akt pathway. In a rat NEC model, oral administration of Epo lowered the incidence of NEC from 45 to 23% with statistical significance. In addition, Epo treatment protected intestinal barrier function and prevented loss of ZO-1 at the TJs in vivo. These effects were associated with elevated Akt phosphorylation in the intestine. This study reveals a novel role of Epo in the regulation of intestinal epithelial TJs and barrier function and suggests the possible use of enteral Epo as a therapeutic agent for gut diseases.
- Published
- 2011
- Full Text
- View/download PDF
45. Salmonella typhimurium infection increases p53 acetylation in intestinal epithelial cells.
- Author
-
Wu S, Ye Z, Liu X, Zhao Y, Xia Y, Steiner A, Petrof EO, Claud EC, and Sun J
- Subjects
- Acetylation, Animals, Cell Cycle drug effects, Cells, Cultured, Female, Gene Expression drug effects, HeLa Cells, Humans, Intestinal Mucosa cytology, Mice, Tumor Necrosis Factor-alpha pharmacology, Bacterial Proteins metabolism, Intestinal Mucosa metabolism, Salmonella Infections physiopathology, Salmonella typhimurium, Tumor Suppressor Protein p53 metabolism
- Abstract
The ability of Salmonella typhimurium to enter intestinal epithelial cells constitutes a crucial step in pathogenesis. Salmonella invasion of the intestinal epithelium requires bacterial type three secretion system. Type three secretion system is a transport device that injects virulence proteins, called effectors, to paralyze or reprogram the eukaryotic cells. Avirulence factor for Salmonella (AvrA) is a Salmonella effector that inhibits the host's inflammatory responses. The mechanism by which AvrA modulates host cell signaling is not entirely clear. p53 is situated at the crossroads of a network of signaling pathways that are essential for genotoxic and nongenotoxic stress responses. We hypothesized that Salmonella infection activates the p53 pathway. We demonstrated that Salmonella infection increased p53 acetylation. Cells infected with AvrA-sufficient Salmonella have increased p53 acetylation, whereas cells infected with AvrA-deficient Salmonella have less p53 acetylation. In a cell-free system, AvrA possessed acetyltransferase activity and used p53 as a substrate. AvrA expression increased p53 transcriptional activity and induced cell cycle arrest. HCT116 p53-/- cells had less inflammatory responses. In a mouse model of Salmonella infection, intestinal epithelial p53 acetylation was increased by AvrA expression. Our studies provide novel mechanistic evidence that Salmonella modulates the p53 pathway during intestinal inflammation and infection.
- Published
- 2010
- Full Text
- View/download PDF
46. Cross, but not direct, presentation of cell-associated virus antigens by spleen macrophages is influenced by their differentiation state.
- Author
-
Alatery A, Siddiqui S, Chan M, Kus A, Petrof EO, and Basta S
- Subjects
- Animals, Bone Marrow immunology, Cells, Cultured, Epitopes, T-Lymphocyte immunology, Humans, Macrophages cytology, Mice, Mice, Inbred C57BL, Phagocytosis, Spleen cytology, T-Lymphocytes immunology, Antigen Presentation, Antigens, Viral immunology, Cell Differentiation, Cross-Priming, Lymphocytic choriomeningitis virus immunology, Macrophages immunology, Spleen immunology
- Abstract
The initiation of T-cell immune responses requires professional antigen-presenting cells. Emerging data point towards an important role for macrophages (Mphi) in the priming of naïve T cells. In this study we analyzed the efficiency and the mechanisms by which Mphi derived from spleen (Sp-Mphi) or bone marrow (BM-Mphi) present Lymphocytic choriomeningitis virus (LCMV) antigens to epitope-specific T cells. We demonstrate that because of phagosomal maturation, Sp-Mphi downregulate their ability to cross-present cell-associated, but not soluble, antigens, as they are further differentiated in culture without altering their capacity to directly present virus antigens after infection. We propose that Sp-Mphi are extremely efficient at direct and cross-presentation. However, if these cells undergo further M-CSF-dependent maturation, they will adapt to be more scavenger and phagocytic and concurrently reduce their cross-presenting capacity. Accordingly, Sp-Mphi can have an important role in regulating T-cell responses through cross-presentation depending on their differentiation state.
- Published
- 2010
- Full Text
- View/download PDF
47. Bacteria-free solution derived from Lactobacillus plantarum inhibits multiple NF-kappaB pathways and inhibits proteasome function.
- Author
-
Petrof EO, Claud EC, Sun J, Abramova T, Guo Y, Waypa TS, He SM, Nakagawa Y, and Chang EB
- Subjects
- Animals, Blotting, Western, Cells, Cultured, Chemokine CCL2 metabolism, Dendritic Cells metabolism, Dendritic Cells microbiology, Electrophoretic Mobility Shift Assay, Enzyme-Linked Immunosorbent Assay, Female, Intestinal Mucosa immunology, Intestinal Mucosa microbiology, Intestines immunology, Intestines microbiology, Macrophages metabolism, Macrophages microbiology, Mice, Mice, Inbred C57BL, Myeloid Differentiation Factor 88 metabolism, NF-kappa B metabolism, Proteasome Endopeptidase Complex metabolism, Rats, Receptors, Tumor Necrosis Factor, Type I metabolism, Receptors, Tumor Necrosis Factor, Type II metabolism, Ubiquitin metabolism, Culture Media, Conditioned pharmacology, Intestinal Mucosa metabolism, Lactobacillus plantarum, NF-kappa B antagonists & inhibitors, Probiotics pharmacology, Proteasome Inhibitors
- Abstract
Background: Bacteria play a role in inflammatory bowel disease and other forms of intestinal inflammation. Although much attention has focused on the search for a pathogen or inciting inflammatory bacteria, another possibility is a lack of beneficial bacteria that normally confer anti-inflammatory properties in the gut. The purpose of this study was to determine whether normal commensal bacteria could inhibit inflammatory pathways important in intestinal inflammation., Methods: Conditioned media from Lactobacillus plantarum (Lp-CM) and other gut bacteria was used to treat intestinal epithelial cell (YAMC) and macrophage (RAW 264.7) or primary culture murine dendritic cells. NF-kappaB was activated through TNF-Receptor, MyD88-dependent and -independent pathways and effects of Lp-CM on the NF-kappaB pathway were assessed. NF-kappaB binding activity was measured using ELISA and EMSA. 1kappaB expression was assessed by Western blot analysis, and proteasome activity determined using fluorescence-based proteasome assays. MCP-1 release was determined by ELISA., Results: Lp-CM inhibited NF-kappaB binding activity, degradation of IkappaBalpha and the chymotrypsin-like activity of the proteasome. Moreover, Lp-CM directly inhibited the activity of purified mouse proteasomes. This effect was specific, since conditioned media from other bacteria had no inhibitory effect. Unlike other proteasome inhibitors, Lp-CM was not toxic in cell death assays. Lp-CM inhibited MCP-1 release in all cell types tested., Conclusions: These studies confirm, and provide a mechanism for, the anti-inflammatory effects of the probiotic and commensal bacterium Lactobacillus plantarum. The use of bacteria-free Lp-CM provides a novel strategy for treatment of intestinal inflammation which would eliminate the risk of bacteremia reported with conventional probiotics.
- Published
- 2009
- Full Text
- View/download PDF
48. Probiotics and Gastrointestinal Disease: Clinical Evidence and Basic Science.
- Author
-
Petrof EO
- Abstract
Our intestinal microbiota serve many roles vital to the normal daily function of the human gastrointestinal tract. Many probiotics are derived from our intestinal bacteria, and have been shown to provide clinical benefit in a variety of gastrointestinal conditions. Current evidence indicates that probiotic effects are strain-specific, they do not act through the same mechanisms, and nor are all probiotics indicated for the same health conditions. However, they do share several common features in that they exert anti-inflammatory effects, they employ different strategies to antagonize competing microorganisms, and they induce cytoprotective changes in the host either through enhancement of barrier function, or through the upregulation of cytoprotective host proteins. In this review we focus on a few selected probiotics - a bacterial mixture (VSL#3), a Gram-negative probiotic (E. coli Nissle 1917), two Gram-positive probiotic bacteria (LGG, L. reuteri), and a yeast probiotic (S. boulardii) - for which sound clinical and mechanistic data is available. Safety of probiotic formulations is also discussed.
- Published
- 2009
- Full Text
- View/download PDF
49. 16S rRNA gene-based analysis of fecal microbiota from preterm infants with and without necrotizing enterocolitis.
- Author
-
Wang Y, Hoenig JD, Malin KJ, Qamar S, Petrof EO, Sun J, Antonopoulos DA, Chang EB, and Claud EC
- Subjects
- Bacteria genetics, Case-Control Studies, Cluster Analysis, DNA, Bacterial chemistry, DNA, Bacterial genetics, DNA, Ribosomal chemistry, DNA, Ribosomal genetics, Female, Humans, Infant, Infant, Newborn, Infant, Premature, Male, Molecular Sequence Data, Phylogeny, RNA, Ribosomal, 16S genetics, Sequence Analysis, DNA, Bacteria classification, Bacteria isolation & purification, Biodiversity, Enterocolitis, Necrotizing microbiology, Feces microbiology
- Abstract
Neonatal necrotizing enterocolitis (NEC) is an inflammatory intestinal disorder affecting preterm infants. Intestinal bacteria have an important function; however no causative pathogen has been identified. The purpose of this study was to determine if there are differences in microbial patterns that may be critical to the development of this disease. Fecal samples from 20 preterm infants, 10 with NEC and 10 matched controls (including 4 twin pairs) were obtained from patients in a single site level III neonatal intensive care unit. Bacterial DNA from individual fecal samples was PCR-amplified and subjected to terminal restriction fragment length polymorphism analysis and library sequencing of the 16S rRNA gene to characterize diversity and structure of the enteric microbiota. The distribution of samples from NEC patients distinctly clustered separately from controls. Intestinal bacterial colonization in all preterm infants was notable for low diversity. Patients with NEC had even less diversity, an increase in abundance of Gammaproteobacteria, a decrease in other bacteria species, and had received a higher mean number of previous days of antibiotics. Our results suggest that NEC is associated with severe lack of microbiota diversity that may accentuate the impact of single dominant microorganisms favored by empiric and widespread use of antibiotics.
- Published
- 2009
- Full Text
- View/download PDF
50. Salmonella type III effector AvrA stabilizes cell tight junctions to inhibit inflammation in intestinal epithelial cells.
- Author
-
Liao AP, Petrof EO, Kuppireddi S, Zhao Y, Xia Y, Claud EC, and Sun J
- Subjects
- Animals, Cell Line, Female, Fluorescent Antibody Technique, Humans, Mice, Mice, Inbred C57BL, Bacterial Proteins physiology, Intestinal Mucosa microbiology, Salmonella physiology, Tight Junctions physiology
- Abstract
Salmonella Typhimurium is a major cause of human gastroenteritis. The Salmonella type III secretory system secretes virulence proteins, called effectors. Effectors are responsible for the alteration of tight junction (TJ) structure and function in intestinal epithelial cells. AvrA is a newly described bacterial effector found in Salmonella. We report here that AvrA expression stabilizes cell permeability and tight junctions in intestinal epithelial cells. Cells colonized with an AvrA-deficient bacterial strain (AvrA-) displayed decreased cell permeability, disruption of TJs, and an increased inflammatory response. Western blot data showed that TJ proteins, such as ZO-1, claudin-1, decreased after AvrA- colonization for only 1 hour. In contrast, cells colonized with AvrA-sufficient bacteria maintained cell permeability with stabilized TJ structure. This difference was confirmed in vivo. Fluorescent tracer studies showed increased fluorescence in the blood of mice infected with AvrA- compared to those infected with the AvrA-sufficient strains. AvrA- disrupted TJ structure and function and increased inflammation in vivo, compared to the AvrA- sufficient strain. Additionally, AvrA overexpression increased TJ protein expression when transfected into colonic epithelial cells. An intriguing aspect of this study is that AvrA stabilized TJs, even though the other TTSS proteins, SopB, SopE, and SopE2, are known to disrupt TJs. AvrA may play a role in stabilizing TJs and balancing the opposing action of other bacterial effectors. Our findings indicate an important role for the bacterial effector AvrA in regulation of intestinal epithelial cell TJs during inflammation. The role of AvrA represents a highly refined bacterial strategy that helps the bacteria survive in the host and dampen the inflammatory response.
- Published
- 2008
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.