51
|
Larcombe S, Jiang JH, Hutton ML, Abud HE, Peleg AY, Lyras D. A mouse model of Staphylococcus aureus small intestinal infection. J Med Microbiol 2020; 69:290-297. [PMID: 32004137 PMCID: PMC7431102 DOI: 10.1099/jmm.0.001163] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Introduction Staphylococcus aureus is a recognised cause of foodborne intoxication and antibiotic-associated diarrhoea (AAD), which are both mediated by staphylococcal enterotoxins. However, unlike foodborne intoxication, AAD appears to require infection of the host. While S. aureus intoxication is widely studied, little is known about S. aureus pathogenesis in the context of gastrointestinal infection. Aim To develop a mouse model of S. aureus gastrointestinal infection. Methodology An established AAD mouse model was adapted for S. aureus infection, and damage observed via histopathological analysis and immunostaining of intestinal tissues. Results Various strains colonised the mouse model, and analysis showed that although clinical signs of disease were not seen, S. aureus infection induced damage in the small intestine, disrupting host structures essential for epithelial integrity. Studies using a staphylococcal enterotoxin B mutant showed that this toxin may contribute to damage during gastrointestinal infection. Conclusion This work presents a new mouse model of S. aureus gastrointestinal infection, while also providing insight into the pathogenesis of S. aureus in the gut.
Collapse
Affiliation(s)
- Sarah Larcombe
- Infection and Immunity Program, Monash Biomedicine Discovery Institute and Department of Microbiology, Monash University, Clayton, Victoria, Australia
| | - Jhih-Hang Jiang
- Infection and Immunity Program, Monash Biomedicine Discovery Institute and Department of Microbiology, Monash University, Clayton, Victoria, Australia
| | - Melanie L. Hutton
- Infection and Immunity Program, Monash Biomedicine Discovery Institute and Department of Microbiology, Monash University, Clayton, Victoria, Australia
| | - Helen E. Abud
- Cancer Program, Monash Biomedicine Discovery Institute and Department of Anatomy and Developmental Biology, Monash University, Clayton, Victoria, Australia
| | - Anton Y. Peleg
- Infection and Immunity Program, Monash Biomedicine Discovery Institute and Department of Microbiology, Monash University, Clayton, Victoria, Australia
- Department of Infectious Diseases, The Alfred Hospital and Central Clinical School, Monash University, Melbourne, Victoria, Australia
| | - Dena Lyras
- Infection and Immunity Program, Monash Biomedicine Discovery Institute and Department of Microbiology, Monash University, Clayton, Victoria, Australia
- *Correspondence: Dena Lyras,
| |
Collapse
|
52
|
Mathee K, Cickovski T, Deoraj A, Stollstorff M, Narasimhan G. The gut microbiome and neuropsychiatric disorders: implications for attention deficit hyperactivity disorder (ADHD). J Med Microbiol 2020; 69:14-24. [PMID: 31821133 PMCID: PMC7440676 DOI: 10.1099/jmm.0.001112] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2019] [Accepted: 11/01/2019] [Indexed: 12/11/2022] Open
Abstract
Neuropsychiatric disorders (NPDs) such as depression, anxiety, bipolar disorder, autism spectrum disorder (ASD) and attention deficit hyperactivity disorder (ADHD) all relate to behavioural, cognitive and emotional disturbances that are ultimately rooted in disordered brain function. More specifically, these disorders are linked to various neuromodulators (i.e. serotonin and dopamine), as well as dysfunction in both cognitive and socio-affective brain networks. Increasing evidence suggests that the gut environment, and particularly the microbiome, plays a significant role in individual mental health. Although the presence of a gut-brain communication axis has long been established, recent studies argue that the development and regulation of this axis is dictated by the gut microbiome. Many studies involving both animals and humans have connected the gut microbiome with depression, anxiety and ASD. Microbiome-centred treatments for individuals with these same NPDs have yielded promising results. Despite its recent rise and underlying similarities to other NPDs, both biochemically and symptomatically, connections between the gut microbiome and ADHD currently lag behind those for other NPDs. We demonstrate that all evidence points to the importance of, and dire need for, a comprehensive and in-depth analysis of the role of the gut microbiome in ADHD, to deepen our understanding of a condition that affects millions of individuals worldwide.
Collapse
Affiliation(s)
- Kalai Mathee
- Department of Human and Molecular Genetics, Herbert Wertheim College of Medicine, Florida International University, Florida, USA
| | - Trevor Cickovski
- Bioinformatics Research Group (BioRG), School of Computing and Information Sciences, Florida International University, Florida, USA
| | - Alok Deoraj
- Department of Environmental and Occupational Health, Robert Stempel College of Public Health and Social Work, Florida International University, Florida, USA
| | - Melanie Stollstorff
- Department of Psychology, College of Arts, Science and Education, Florida International University, Florida, USA
| | - Giri Narasimhan
- Bioinformatics Research Group (BioRG), School of Computing and Information Sciences, Florida International University, Florida, USA
| |
Collapse
|
53
|
Abstract
C. perfringens type F strains are a common cause of food poisoning and antibiotic-associated diarrhea. Type F strain virulence requires production of C. perfringens enterotoxin (CPE). In Caco-2 cells, high CPE concentrations cause necrosis while low enterotoxin concentrations induce apoptosis. The current study determined that receptor-interacting serine/threonine-protein kinases 1 and 3 are involved in both CPE-induced apoptosis and necrosis in Caco-2 cells, while mixed-lineage kinase domain-like pseudokinase (MLKL) oligomerization is involved in CPE-induced necrosis, thereby indicating that this form of CPE-induced cell death involves necroptosis. High CPE concentrations also caused necroptosis in T84 and Vero cells. Calpain activation was identified as a key intermediate for CPE-induced necroptosis. These results suggest inhibitors of RIP1, RIP3, MLKL oligomerization, or calpain are useful therapeutics against CPE-mediated diseases. Clostridium perfringens type F strains cause gastrointestinal disease when they produce a pore-forming toxin named C. perfringens enterotoxin (CPE). In human enterocyte-like Caco-2 cells, low CPE concentrations cause caspase-3-dependent apoptosis, while high CPE concentrations cause necrosis. Since necrosis or apoptosis sometimes involves receptor-interacting serine/threonine-protein kinase-1 or 3 (RIP1 or RIP3), this study examined whether those kinases are important for CPE-induced apoptosis or necrosis. Highly specific RIP1 or RIP3 inhibitors reduced both CPE-induced apoptosis and necrosis in Caco-2 cells. Those findings suggested that the form of necrosis induced by treating Caco-2 cells with high CPE concentrations involves necroptosis, which was confirmed when high, but not low, CPE concentrations were shown to induce oligomerization of mixed-lineage kinase domain-like pseudokinase (MLKL), a key late step in necroptosis. Furthermore, an MLKL oligomerization inhibitor reduced cell death caused by high, but not low, CPE concentrations. Supporting RIP1 and RIP3 involvement in CPE-induced necroptosis, inhibitors of those kinases also reduced MLKL oligomerization during treatment with high CPE concentrations. Calpain inhibitors similarly blocked MLKL oligomerization induced by high CPE concentrations, implicating calpain activation as a key intermediate in initiating CPE-induced necroptosis. In two other CPE-sensitive cell lines, i.e., Vero cells and human enterocyte-like T84 cells, low CPE concentrations also caused primarily apoptosis/late apoptosis, while high CPE concentrations mainly induced necroptosis. Collectively, these results establish that high, but not low, CPE concentrations cause necroptosis and suggest that RIP1, RIP3, MLKL, or calpain inhibitors can be explored as potential therapeutics against CPE effects in vivo.
Collapse
|
54
|
Xie G, Tan K, Peng M, Long C, Li D, Tan Z. Bacterial diversity in intestinal mucosa of antibiotic-associated diarrhea mice. 3 Biotech 2019; 9:444. [PMID: 31763122 PMCID: PMC6842370 DOI: 10.1007/s13205-019-1967-2] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2019] [Accepted: 10/23/2019] [Indexed: 12/15/2022] Open
Abstract
To probe into the mechanism of antibiotic-associated diarrhea (AAD), the bacterial diversity and composition in the intestinal mucosa of AAD mice were investigated. Twelve specific pathogen-free Kunming mice were divided into control group and model group. The mouse model of AAD was established by gavaging with antibiotics (mixture of gentamycin sulfate and cefradine) at a total dose of 23.33 ml kg-1 day-1 for 5 days continuously, twice a day. The mice in the control group were given with an equal amount of sterile water. Then, the intestinal mucosa DNA was extracted for 16S rRNA gene sequence analysis by high-throughput sequencing. The results showed that the alpha diversity of the two groups did not differ significantly from each other, while the composition of intestinal mucosa bacteria differed dramatically between the two groups. The model group showed a higher abundance of Proteobacteria and Actinobacteria. More importantly, Lactobacillus was significantly less abundant (p = 0.000), while Enterococcus was significantly more abundant (p = 0.019) in the model group than in the control group. Furthermore, antibiotic treatment increased the abundance of Citrobacter, Stenotrophomonas, and Glutamicibacter,whereas antibiotics decreased the abundance of Mycoplasma and Helicobacter. In addition, 6 and 11 unique genera were found in the control group and model group, respectively. The combination of gentamycin sulfate and cefradine changed the intestinal mucosa bacterial composition, reduced colonization resistance and damaged the intestinal mucosal barrier by reducing the abundance of Lactobacillus.
Collapse
Affiliation(s)
- Guozhen Xie
- Hunan University of Chinese Medicine, Xueshi Road 300, Yuelu District, Changsha, 410208 Hunan Province China
| | - Kai Tan
- College of Fisheries and Life Science, Shanghai Ocean University, Shanghai, 201306 China
| | - Maijiao Peng
- Hunan University of Chinese Medicine, Xueshi Road 300, Yuelu District, Changsha, 410208 Hunan Province China
| | - Chengxing Long
- Hunan University of Chinese Medicine, Xueshi Road 300, Yuelu District, Changsha, 410208 Hunan Province China
| | - Dandan Li
- Hunan University of Chinese Medicine, Xueshi Road 300, Yuelu District, Changsha, 410208 Hunan Province China
| | - Zhoujin Tan
- Hunan University of Chinese Medicine, Xueshi Road 300, Yuelu District, Changsha, 410208 Hunan Province China
| |
Collapse
|
55
|
Xie G, Tan K, Peng M, Long C, Li D, Tan Z. Bacterial diversity in intestinal mucosa of antibiotic-associated diarrhea mice. 3 Biotech 2019. [PMID: 31763122 DOI: 10.1007/s13205-019-1967-2.] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022] Open
Abstract
To probe into the mechanism of antibiotic-associated diarrhea (AAD), the bacterial diversity and composition in the intestinal mucosa of AAD mice were investigated. Twelve specific pathogen-free Kunming mice were divided into control group and model group. The mouse model of AAD was established by gavaging with antibiotics (mixture of gentamycin sulfate and cefradine) at a total dose of 23.33 ml kg-1 day-1 for 5 days continuously, twice a day. The mice in the control group were given with an equal amount of sterile water. Then, the intestinal mucosa DNA was extracted for 16S rRNA gene sequence analysis by high-throughput sequencing. The results showed that the alpha diversity of the two groups did not differ significantly from each other, while the composition of intestinal mucosa bacteria differed dramatically between the two groups. The model group showed a higher abundance of Proteobacteria and Actinobacteria. More importantly, Lactobacillus was significantly less abundant (p = 0.000), while Enterococcus was significantly more abundant (p = 0.019) in the model group than in the control group. Furthermore, antibiotic treatment increased the abundance of Citrobacter, Stenotrophomonas, and Glutamicibacter,whereas antibiotics decreased the abundance of Mycoplasma and Helicobacter. In addition, 6 and 11 unique genera were found in the control group and model group, respectively. The combination of gentamycin sulfate and cefradine changed the intestinal mucosa bacterial composition, reduced colonization resistance and damaged the intestinal mucosal barrier by reducing the abundance of Lactobacillus.
Collapse
Affiliation(s)
- Guozhen Xie
- 1Hunan University of Chinese Medicine, Xueshi Road 300, Yuelu District, Changsha, 410208 Hunan Province China
| | - Kai Tan
- 2College of Fisheries and Life Science, Shanghai Ocean University, Shanghai, 201306 China
| | - Maijiao Peng
- 1Hunan University of Chinese Medicine, Xueshi Road 300, Yuelu District, Changsha, 410208 Hunan Province China
| | - Chengxing Long
- 1Hunan University of Chinese Medicine, Xueshi Road 300, Yuelu District, Changsha, 410208 Hunan Province China
| | - Dandan Li
- 1Hunan University of Chinese Medicine, Xueshi Road 300, Yuelu District, Changsha, 410208 Hunan Province China
| | - Zhoujin Tan
- 1Hunan University of Chinese Medicine, Xueshi Road 300, Yuelu District, Changsha, 410208 Hunan Province China
| |
Collapse
|
56
|
Biofilm Formation and Virulence Determinants of Klebsiella oxytoca Clinical Isolates from Patients with Colorectal Cancer. J Gastrointest Cancer 2019; 51:855-860. [PMID: 31659674 DOI: 10.1007/s12029-019-00317-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
OBJECTIVE Biofilm formation has made the therapy of bacterial infections more difficult. The objective our study was assessment of pan-drug-resistant (PDR) Klebsiella oxytoca pathogenicity and virulence factors causing AAHC in patients with colorectal cancer (CRC). MATERIALS AND METHODS Among a total of 300 healthy and 300 patients with antibiotic-associated hemorrhagic colitis (AAHC) and CRC, 200 K. oxytoca were identified during May 2015-January 2019. The virulence properties and biofilm formation among the isolates were investigated by phenotypic, PCR, and real-time PCR (RT-qPCR) techniques. RESULTS The blaCTX-M1 (20%), blaSHV (11%), blaTEM1 (33%), and AmpC encoding CIT (2%) ESBL genes, carbapenemase-encoding genes blaIM (4%) and blaOXA-48 (2%), and colistin-resistant mcr-1 gene (2.5%) were detected. The virulence-encoding genes including fimA (80%), pilQ (100%), matB (100%), mrkA (80%), and npsB (100%) were amplified. Therefore, PDR K. oxytoca containing adhesins and toxin-encoding genes with ability of biofilm formation causing AAHC and CRC were isolated. There was a significant difference between healthy and patients with CRC regarding the presence of K. oxytoca (p = 00.221). CONCLUSION Bacterial enteric pathogens possibly play a role in CRC. Biofilm formation by K. oxytoca strains prevents the efficient infection elimination; therefore, rapid identification and control measure are chief requirements. Additionally, more investigations are necessary with this regard.
Collapse
|
57
|
Chowdhury AH, Cámara M, Verma C, Eremin O, Kulkarni AD, Lobo DN. Modulation of T Regulatory and Dendritic Cell Phenotypes Following Ingestion of Bifidobacterium longum, AHCC ® and Azithromycin in Healthy Individuals. Nutrients 2019; 11:nu11102470. [PMID: 31618905 PMCID: PMC6835407 DOI: 10.3390/nu11102470] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2019] [Revised: 10/08/2019] [Accepted: 10/09/2019] [Indexed: 12/13/2022] Open
Abstract
The probiotic Bifidus BB536 (BB536), which contains Bifidobacterium longum, has been shown to have enhanced probiotic effects when given together with a standardized extract of cultured Lentinula edodes mycelia (AHCC®, Amino Up Co. Ltd., Sapporo, Japan). BB536 and AHCC® may modulate T cell and dendritic cell (DC) phenotypes, and cytokine profiles to favour anti-inflammatory responses following antibiotic ingestion. We tested the hypothesis that orally administered BB536 and/or AHCC®, results in modulation of immune effector cells with polarisation towards anti-inflammatory responses following antibiotic usage. Forty healthy male volunteers divided into 4 equal groups were randomised to receive either placebo, BB536, AHCC® or a combination for 12 days in a double-blind manner. After 7 days volunteers also received 250 mg azithromycin for 5 days. Cytokine profiles from purified CD3+ T cells stimulated with PDB-ionomycin were assessed. CD4+ CD25+ forkhead box P3 (Foxp3) expression and peripheral blood DC subsets were assessed prior to treatment and subsequently at 7 and 13 days. There was no difference in cytokine secretion from stimulated CD3+ T cells between treatment groups. Compared with baseline, Foxp3 expression (0.45 ± 0.1 vs. 1.3 ± 0.4; p = 0.002) and interferon-gamma/interleukin-4 (IFN-γ/IL-4) ratios were increased post-treatment in volunteers receiving BB536 (p = 0.031), although differences between groups were not significant. For volunteers receiving combination BB536 and AHCC®, there was an increase in myeloid dendritic cells (mDC) compared with plasmacytoid DC (pDC) counts (80% vs. 61%; p = 0.006) at post treatment time points. mDC2 phenotypes were more prevalent, compared with baseline, following combination treatment (0.16% vs. 0.05%; p = 0.002). Oral intake of AHCC® and BB536 may modulate T regulatory and DC phenotypes to favour anti-inflammatory responses following antibiotic usage.
Collapse
Affiliation(s)
- Abeed H Chowdhury
- Gastrointestinal Surgery, Nottingham Digestive Diseases Centre, National Institute for Health Research (NIHR) Nottingham Biomedical Research Centre, Nottingham University Hospitals NHS Trust and University of Nottingham, Queen's Medical Centre, Nottingham NG7 2UH, UK.
| | - Miguel Cámara
- School of Life Sciences, Centre for Biomolecular Sciences, University of Nottingham, Nottingham NG7 2RD, UK.
| | - Chandan Verma
- Gastrointestinal Surgery, Nottingham Digestive Diseases Centre, National Institute for Health Research (NIHR) Nottingham Biomedical Research Centre, Nottingham University Hospitals NHS Trust and University of Nottingham, Queen's Medical Centre, Nottingham NG7 2UH, UK.
| | - Oleg Eremin
- Gastrointestinal Surgery, Nottingham Digestive Diseases Centre, National Institute for Health Research (NIHR) Nottingham Biomedical Research Centre, Nottingham University Hospitals NHS Trust and University of Nottingham, Queen's Medical Centre, Nottingham NG7 2UH, UK.
| | - Anil D Kulkarni
- Department of Surgery, The University of Texas Health Science Center and McGovern Medical School, 6431 Fannin Street, MSB 4022-B, Houston, TX 77030, USA.
| | - Dileep N Lobo
- Gastrointestinal Surgery, Nottingham Digestive Diseases Centre, National Institute for Health Research (NIHR) Nottingham Biomedical Research Centre, Nottingham University Hospitals NHS Trust and University of Nottingham, Queen's Medical Centre, Nottingham NG7 2UH, UK.
- MRC Versus Arthritis Centre for Musculoskeletal Ageing Research, School of Life Sciences, University of Nottingham, Queen's Medical Centre, Nottingham NG7 2UH, UK.
| |
Collapse
|
58
|
Effects of Claudin-1 on the Action of Clostridium perfringens Enterotoxin in Caco-2 Cells. Toxins (Basel) 2019; 11:toxins11100582. [PMID: 31601044 PMCID: PMC6832201 DOI: 10.3390/toxins11100582] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2019] [Revised: 10/01/2019] [Accepted: 10/03/2019] [Indexed: 12/11/2022] Open
Abstract
Clostridium perfringens enterotoxin (CPE) contributes to diarrhea and an often-lethal enterotoxemia. CPE action starts when it binds to claudin receptors, forming a small complex (90 kDa). Six small complexes then oligomerize to create prepores, followed by insertion of beta-hairpins from CPE to form beta-barrel pores named CH-1 or CH-2. Of the ~27 members of the human claudin protein family, only some bind CPE. However, both receptor claudins and the nonreceptor claudin-1 (CLDN-1) are associated with the small and CH-1/CH-2 CPE complexes. Therefore, this study evaluated whether claudin-1 affects CPE action by generating a CLDN-1 null mutant in Caco-2 cells using CRISPR-Cas9. Compared to wild-type Caco-2 cells, paracellular permeability of the CLDN-1 mutant was significantly enhanced, suggesting that claudin-1 may reduce CPE absorption during enterotoxemia. The CLDN-1 mutant was also markedly more sensitive than wild-type Caco-2 cells to apically-applied CPE. The mechanism behind this increased sensitivity involved higher CPE binding by the CLDN-1 mutant vs. wild-type Caco-2 cells, which led to more CH-1/CH-2 complex formation. However, the CH-1/CH-2 complexes formed by the CLDN-1 mutant were less stable or trypsin resistant than those of wild-type cells. These results indicate that, although a nonreceptor, CLDN-1 positively and negatively influences CPE action.
Collapse
|
59
|
Kiu R, Caim S, Painset A, Pickard D, Swift C, Dougan G, Mather AE, Amar C, Hall LJ. Phylogenomic analysis of gastroenteritis-associated Clostridium perfringens in England and Wales over a 7-year period indicates distribution of clonal toxigenic strains in multiple outbreaks and extensive involvement of enterotoxin-encoding (CPE) plasmids. Microb Genom 2019; 5. [PMID: 31553300 PMCID: PMC6861862 DOI: 10.1099/mgen.0.000297] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Clostridium perfringens is a major enteric pathogen known to cause gastroenteritis in human adults. Although major outbreak cases are frequently reported, only limited whole-genome sequencing (WGS) based studies have been performed to understand the genomic epidemiology and virulence gene content of outbreak-associated C. perfringens strains. We performed phylogenomic analysis on 109 C. perfringens isolates (human and food) obtained from disease cases in England and Wales between 2011 and 2017. Initial findings highlighted the enhanced discriminatory power of WGS in profiling outbreak C. perfringens strains, when compared to the current Public Health England referencing laboratory technique of fluorescent amplified fragment length polymorphism analysis. Further analysis identified that isogenic C. perfringens strains were associated with nine distinct care-home-associated outbreaks over the course of a 5-year interval, indicating a potential common source linked to these outbreaks or transmission over time and space. As expected, the enterotoxin cpe gene was encoded in all but 4 isolates (96.3 %; 105/109), with virulence plasmids encoding cpe (particularly pCPF5603 and pCPF4969 plasmids) extensively distributed (82.6 %; 90/109). Genes encoding accessory virulence factors, such as beta-2 toxin, were commonly detected (46.7 %; 51/109), and genes encoding phage proteins were also frequently identified. Overall, this large-scale genomic study of gastroenteritis-associated C. perfringens suggested that three major cpe-encoding (toxinotype F) genotypes underlie these outbreaks: strains carrying (1) pCPF5603 plasmid, (2) pCPF4969 plasmid and (3) chromosomal-cpe strains. Our findings substantially expanded our knowledge on type F C. perfringens involved in human-associated gastroenteritis, with further studies required to fully probe the dissemination and regional reservoirs of this enteric pathogen, which may help devise effective prevention strategies to reduce the food-poisoning disease burden in vulnerable patients, such as the elderly.
Collapse
Affiliation(s)
- Raymond Kiu
- Gut Microbes and Health, Quadram Institute Bioscience, Norwich NR4 7UQ, UK
| | - Shabhonam Caim
- Gut Microbes and Health, Quadram Institute Bioscience, Norwich NR4 7UQ, UK
| | - Anais Painset
- Gastrointestinal Bacteria Reference Unit, Public Health England, London NW9 5EQ, UK
| | - Derek Pickard
- Department of Medicine, University of Cambridge, Cambridge CB2 0QQ, UK
| | - Craig Swift
- Gastrointestinal Bacteria Reference Unit, Public Health England, London NW9 5EQ, UK
| | - Gordon Dougan
- Department of Medicine, University of Cambridge, Cambridge CB2 0QQ, UK
| | - Alison E Mather
- Faculty of Medicine and Health Sciences, University of East Anglia, Norwich NR4 7TJ, UK.,Microbes in the Food Chain, Quadram Institute Bioscience, Norwich NR4 7UQ, UK
| | - Corinne Amar
- Gastrointestinal Bacteria Reference Unit, Public Health England, London NW9 5EQ, UK
| | - Lindsay J Hall
- Gut Microbes and Health, Quadram Institute Bioscience, Norwich NR4 7UQ, UK
| |
Collapse
|
60
|
Yang J, Yang H. Non-antibiotic therapy for Clostridioides difficile infection: a review. Crit Rev Clin Lab Sci 2019; 56:493-509. [PMID: 31411909 DOI: 10.1080/10408363.2019.1648377] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Clostridioides difficile infection (CDI) is a common infectious disease that is mainly caused by antibiotics. Antibiotic therapy is still the dominant treatment for CDI, although it is accompanied by side effects. Probiotics, fecal microbiota transplantation (FMT), engineered microorganisms, bacteriophages, diet, natural active substances, nanoparticles and compounds are examples of emerging non-antibiotic therapies that have received a great amount of attention. In this review, we collected data about different non-antibiotic therapies for CDI and provided a comprehensive analysis and detailed comparison of these therapies. The mechanism of action, therapeutic efficacy, and the strengths and weaknesses of these non-antibiotic therapies have been investigated to provide a basis for the reasonable alternative of non-antibiotic therapies for CDI. In summary, probiotics and FMT are currently the best choice for non-antibiotic therapy for CDI.
Collapse
Affiliation(s)
- Jingpeng Yang
- State Key Laboratory of Microbial Metabolism, and School of Life Science & Biotechnology, Shanghai Jiao Tong University , Shanghai , China
| | - Hong Yang
- State Key Laboratory of Microbial Metabolism, and School of Life Science & Biotechnology, Shanghai Jiao Tong University , Shanghai , China
| |
Collapse
|
61
|
Khan I, Ullah N, Zha L, Bai Y, Khan A, Zhao T, Che T, Zhang C. Alteration of Gut Microbiota in Inflammatory Bowel Disease (IBD): Cause or Consequence? IBD Treatment Targeting the Gut Microbiome. Pathogens 2019; 8:pathogens8030126. [PMID: 31412603 PMCID: PMC6789542 DOI: 10.3390/pathogens8030126] [Citation(s) in RCA: 478] [Impact Index Per Article: 79.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2019] [Revised: 08/03/2019] [Accepted: 08/05/2019] [Indexed: 02/06/2023] Open
Abstract
Inflammatory bowel disease (IBD) is a chronic complex inflammatory gut pathological condition, examples of which include Crohn’s disease (CD) and ulcerative colitis (UC), which is associated with significant morbidity. Although the etiology of IBD is unknown, gut microbiota alteration (dysbiosis) is considered a novel factor involved in the pathogenesis of IBD. The gut microbiota acts as a metabolic organ and contributes to human health by performing various physiological functions; deviation in the gut flora composition is involved in various disease pathologies, including IBD. This review aims to summarize the current knowledge of gut microbiota alteration in IBD and how this contributes to intestinal inflammation, as well as explore the potential role of gut microbiota-based treatment approaches for the prevention and treatment of IBD. The current literature has clearly demonstrated a perturbation of the gut microbiota in IBD patients and mice colitis models, but a clear causal link of cause and effect has not yet been presented. In addition, gut microbiota-based therapeutic approaches have also shown good evidence of their effects in the amelioration of colitis in animal models (mice) and IBD patients, which indicates that gut flora might be a new promising therapeutic target for the treatment of IBD. However, insufficient data and confusing results from previous studies have led to a failure to define a core microbiome associated with IBD and the hidden mechanism of pathogenesis, which suggests that well-designed randomized control trials and mouse models are required for further research. In addition, a better understanding of this ecosystem will also determine the role of prebiotics and probiotics as therapeutic agents in the management of IBD.
Collapse
Affiliation(s)
- Israr Khan
- School of Life Sciences, Lanzhou University, Lanzhou 730000, China
- Key Laboratory of Cell Activities and Stress Adaptations, Ministry of Education, Lanzhou University, Lanzhou 730000, China
- Gansu Key Laboratory of Biomonitoring and Bioremediation for Environmental Pollution, Lanzhou University, Lanzhou 730000, China
| | - Naeem Ullah
- School of Life Sciences, Lanzhou University, Lanzhou 730000, China
- Key Laboratory of Cell Activities and Stress Adaptations, Ministry of Education, Lanzhou University, Lanzhou 730000, China
- Gansu Key Laboratory of Biomonitoring and Bioremediation for Environmental Pollution, Lanzhou University, Lanzhou 730000, China
| | - Lajia Zha
- School of Life Sciences, Lanzhou University, Lanzhou 730000, China
- Key Laboratory of Cell Activities and Stress Adaptations, Ministry of Education, Lanzhou University, Lanzhou 730000, China
- Gansu Key Laboratory of Biomonitoring and Bioremediation for Environmental Pollution, Lanzhou University, Lanzhou 730000, China
| | - Yanrui Bai
- School of Life Sciences, Lanzhou University, Lanzhou 730000, China
- Key Laboratory of Cell Activities and Stress Adaptations, Ministry of Education, Lanzhou University, Lanzhou 730000, China
- Gansu Key Laboratory of Biomonitoring and Bioremediation for Environmental Pollution, Lanzhou University, Lanzhou 730000, China
| | - Ashiq Khan
- School of Life Sciences, Lanzhou University, Lanzhou 730000, China
- Probiotics and Biological Feed Research Center, Lanzhou University, Lanzhou 730000, China
| | - Tang Zhao
- School of Life Sciences, Lanzhou University, Lanzhou 730000, China
- Key Laboratory of Cell Activities and Stress Adaptations, Ministry of Education, Lanzhou University, Lanzhou 730000, China
- Gansu Key Laboratory of Biomonitoring and Bioremediation for Environmental Pollution, Lanzhou University, Lanzhou 730000, China
| | - Tuanjie Che
- Gansu Key Laboratory of Functional Genomics and Molecular Diagnosis, Lanzhou 730000, China
| | - Chunjiang Zhang
- School of Life Sciences, Lanzhou University, Lanzhou 730000, China.
- Key Laboratory of Cell Activities and Stress Adaptations, Ministry of Education, Lanzhou University, Lanzhou 730000, China.
- Gansu Key Laboratory of Biomonitoring and Bioremediation for Environmental Pollution, Lanzhou University, Lanzhou 730000, China.
- Gansu Key Laboratory of Functional Genomics and Molecular Diagnosis, Lanzhou 730000, China.
| |
Collapse
|
62
|
Azimirad M, Gholami F, Yadegar A, Knight DR, Shamloei S, Aghdaei HA, Zali MR. Prevalence and characterization of Clostridium perfringens toxinotypes among patients with antibiotic-associated diarrhea in Iran. Sci Rep 2019; 9:7792. [PMID: 31127185 PMCID: PMC6534674 DOI: 10.1038/s41598-019-44281-5] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2018] [Accepted: 05/14/2019] [Indexed: 12/12/2022] Open
Abstract
Clostridium perfringens has emerged as an important cause of antibiotic-associated diarrhea (AAD), particularly in the hospital environment. Here we investigated the prevalence and molecular epidemiology of C. perfringens isolated from 2280 fecal samples from Iranian diarrheal patients suspected of having AAD. Overall, AAD was diagnosed in 13.3% (303/2280) of patients and associated with advanced age (>50 years, P = 0.001). A total of 106 C. perfringens isolates were cultured from AAD (n = 68) and non-AAD (n = 38) groups, with toxinotypes A and F comprising 84% and 16% of isolates, respectively. Notably, 41.2% of type F strains were also cpb2-positive and enterotoxigenic cpe-positive strains were detected in 13.2% of the isolates from AAD patients. Genes associated with the VirR/VirS signal transduction (virR, virS) and accessory gene regulator (agrB, agrD) systems were detected in 56.6% and 67% of the isolates, respectively, and peptides of the quorum-sensing modulator, AgrD were highly conserved across all strains. The high prevalence of C. perfringens in Iranian AAD patients suggests that diagnostic laboratories in this region should consider screening for C. perfringens in cases of suspected AAD, especially if the specimen is negative for other pathogens and/or the patients are aged >50 years.
Collapse
Affiliation(s)
- Masoumeh Azimirad
- Foodborne and Waterborne Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Fatemeh Gholami
- Foodborne and Waterborne Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Abbas Yadegar
- Foodborne and Waterborne Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Daniel R Knight
- School of Veterinary and Life Sciences, Murdoch University, Murdoch, Western Australia, Australia
| | - Sharareh Shamloei
- Department of Water and Wastewater Quality Control Laboratory, Water and Wastewater Company, Tehran, Iran
| | - Hamid Asadzadeh Aghdaei
- Basic and Molecular Epidemiology of Gastrointestinal Disorders Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammad Reza Zali
- Gastroenterology and Liver Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
63
|
Abstract
In humans and livestock, Clostridium perfringens is an important cause of intestinal infections that manifest as enteritis, enterocolitis, or enterotoxemia. This virulence is largely related to the toxin-producing ability of C. perfringens. This article primarily focuses on the C. perfringens type F strains that cause a very common type of human food poisoning and many cases of nonfoodborne human gastrointestinal diseases. The enteric virulence of type F strains is dependent on their ability to produce C. perfringens enterotoxin (CPE). CPE has a unique amino acid sequence but belongs structurally to the aerolysin pore-forming toxin family. The action of CPE begins with binding of the toxin to claudin receptors, followed by oligomerization of the bound toxin into a prepore on the host membrane surface. Each CPE molecule in the prepore then extends a beta-hairpin to form, collectively, a beta-barrel membrane pore that kills cells by increasing calcium influx. The cpe gene is typically encoded on the chromosome of type F food poisoning strains but is encoded by conjugative plasmids in nonfoodborne human gastrointestinal disease type F strains. During disease, CPE is produced when C. perfringens sporulates in the intestines. Beyond type F strains, C. perfringens type C strains producing beta-toxin and type A strains producing a toxin named CPILE or BEC have been associated with human intestinal infections. C. perfringens is also an important cause of enteritis, enterocolitis, and enterotoxemia in livestock and poultry due to intestinal growth and toxin production.
Collapse
|
64
|
Candel-Pérez C, Ros-Berruezo G, Martínez-Graciá C. A review of Clostridioides [Clostridium] difficile occurrence through the food chain. Food Microbiol 2019; 77:118-129. [DOI: 10.1016/j.fm.2018.08.012] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2018] [Revised: 08/01/2018] [Accepted: 08/21/2018] [Indexed: 12/18/2022]
|
65
|
Identification of an Important Orphan Histidine Kinase for the Initiation of Sporulation and Enterotoxin Production by Clostridium perfringens Type F Strain SM101. mBio 2019; 10:mBio.02674-18. [PMID: 30670619 PMCID: PMC6343041 DOI: 10.1128/mbio.02674-18] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Clostridium perfringens type F enteric diseases, which include a very common form of food poisoning and many cases of antibiotic-associated diarrhea, develop when type F strains sporulate and produce C. perfringens enterotoxin (CPE) in the intestines. Spores are also important for transmission of type F disease. Despite the importance of sporulation for type F disease and the evidence that C. perfringens sporulation begins with phosphorylation of the Spo0A transcriptional regulator, the kinase phosphorylating Spo0A to initiate sporulation and CPE production had not been ascertained. In response, the current report now provides identification of an orphan histidine kinase named CPR0195 that can directly phosphorylate Spo0A. Results using a CPR0195 null mutant indicate that this kinase is very important for initiating C. perfringens sporulation and CPE production. Therefore, the CPR0195 kinase represents a potential target to block type F disease by interfering with intestinal C. perfringens sporulation and CPE production. Clostridium perfringens type F strains cause a common human foodborne illness and many cases of nonfoodborne human gastrointestinal diseases. Sporulation plays two critical roles during type F enteric disease. First, it produces broadly resistant spores that facilitate type F strain survival in the food and nosocomial environments. Second, production of C. perfringens enterotoxin (CPE), the toxin responsible for causing the enteric symptoms of type F diseases, is restricted to cells in the process of sporulation. While later steps in the regulation of C. perfringens sporulation have been discerned, the process leading to phosphorylation of Spo0A, the master early regulator of sporulation and consequent CPE production, has remained unknown. Using an insertional mutagenesis approach, the current study identified the orphan histidine kinase CPR0195 as an important factor regulating C. perfringens sporulation and CPE production. Specifically, a CPR0195 null mutant of type F strain SM101 made 103-fold fewer spores than its wild-type parent and produced no detectable CPE. In contrast, a null mutant of another putative C. perfringens orphan histidine kinase (CPR1055) did not significantly affect sporulation or CPE production. Studies using a spoIIA operon promoter-driven reporter plasmid indicated that CPR0195 functions early during sporulation, i.e., prior to production of sporulation-associated sigma factors. Furthermore, in vitro studies showed that the CPR0195 kinase domain can autophosphorylate and phosphorylate Spo0A. These results support the idea of CPR0195 as an important kinase that initiates C. perfringens sporulation by directly phosphorylating Spo0A. This kinase could represent a novel therapeutic target to block C. perfringens sporulation and CPE production during type F disease.
Collapse
|
66
|
De Filippo C, Di Paola M, Giani T, Tirelli F, Cimaz R. Gut microbiota in children and altered profiles in juvenile idiopathic arthritis. J Autoimmun 2019; 98:1-12. [PMID: 30638708 DOI: 10.1016/j.jaut.2019.01.001] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2018] [Revised: 12/19/2018] [Accepted: 01/02/2019] [Indexed: 12/18/2022]
Abstract
Microbial diversity plays a key role in the maintenance of intestinal homeostasis and in the development of the immune system in the gut mucosa. Maybe one of the most important function of our gut microbiota is the immune system education, in particular the discrimination of friends from foes that occurs during childhood. In addition to bacterial antigens, several metabolites of microbial origin have a crucial role in training of the immune system, such as Short Chain Fatty Acids (SCFAs). There are many evidences on the role of the gut microbiota in rheumatic diseases, in particular modifications of microbiota composition causing dysbiosis that, in turn, can induce gut permeability, and thus immunological imbalance and trigger inflammation. In particular, immune cells can reach extra-intestinal sites, such as joints and trigger local inflammation. Childhood is a crucial period of life for development and evolution of the gut microbiota, especially for the acquisition of fundamental functions such as immunotolerance of commensal microorganisms. For this reason, gut dysbiosis is gaining interest as a potential pathogenetic factor for Juvenile Idiopathic Arthritis (JIA). Here we summarized the studies conducted on JIA patients in which a pro-arthritogenic microbial profiles has been observed; this, together with a depletion of microbial biodiversity, clearly distinguish patients' from healthy subjects' microbiota. Further studies are however needed to better clarify the role of microbiota in JIA pathogenesis.
Collapse
Affiliation(s)
- Carlotta De Filippo
- Institute of Biology and Agrarian Biotechnology (IBBA), National Research Council (CNR), Via Moruzzi 1, 56124 Pisa, Italy
| | - Monica Di Paola
- Department of Biology, University of Florence, Via Madonna del Piano 6, 50019 Sesto Fiorentino, Florence, Italy
| | - Teresa Giani
- Rheumatology Unit, Anna Meyer Children's Hospital, University of Florence, Viale G. Pieraccini 24, 50139, Florence, Italy; Department of Medica Biotechnologies, University of Siena, Viale Mario Bracci, 16 53100, Siena, Italy
| | - Francesca Tirelli
- Rheumatology Unit, Anna Meyer Children's Hospital, University of Florence, Viale G. Pieraccini 24, 50139, Florence, Italy
| | - Rolando Cimaz
- Rheumatology Unit, Anna Meyer Children's Hospital, University of Florence, Viale G. Pieraccini 24, 50139, Florence, Italy; Department of Neuroscience, Psychology, Drug Research and Child Health, Meyer Children's Hospital, University of Florence, Viale G. Pieraccini 6, 50139, Florence, Italy.
| |
Collapse
|
67
|
Usacheva EA, Peterson LR, Mendoza K, Schora DM, Hossain MM, Jin JP. Cytoskeletal Tropomyosin as a Biomarker in Clostridium difficile Infection. J Clin Med Res 2019; 11:98-105. [PMID: 30701001 PMCID: PMC6340674 DOI: 10.14740/jocmr3696] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2018] [Accepted: 12/24/2018] [Indexed: 12/17/2022] Open
Abstract
Background Current diagnostics of Clostridium difficile infection (CDI) heavily relies on detection of the disease-causing organism. The objective of this study was to investigate a cytoskeletal protein, tropomyosin (Tpm), as a CDI biomarker. Methods Fecal Tpm was tested by monoclonal antibodies (mAbs) in a 12-month prospective study. Remnant diarrheal clinical specimens and relevant clinical data were collected. The CDI positive (CDI+, n = 230) and CDI negative (CDI-, n = 228) groups were composed of samples testing positive or negative by polymerase chain reaction (PCR) (Xpert® C. difficile/Epi, Cepheid), respectively. The other enteric pathogen (OEP) group (n = 52) was composed of specimens tested for the presence of other enteric pathogens or parasites by routine testing methods. Extracted fecal Tpm was detected by Western blot and the results were correlated with CDI based on clinical and microbiology laboratory data. Results A total of 510 stool specimens were tested. Tpm is not stable in stool, suggesting the utility of fresh specimens. In the CDI+ group, specificity and sensitivity of Tpm detection in correlation with a CDI were 93.2% and 53.7%, respectively, when only "true CDI" and "not CDI" were analyzed (110 samples). For CDI+ samples, 23% did not satisfy CDI clinical signs. Tpm positives in the CDI- group (8.3%) had inflammatory bowel diseases. Conclusion Tpm has a potential role as a CDI biomarker in combination with C. difficile PCR and an appropriate clinical evaluation. However, non-muscle Tpm, as a biomarker for CDI, suffers from a low sensitivity in our study. Therefore further investigation using larger cohorts is needed.
Collapse
Affiliation(s)
- Elena A Usacheva
- Infectious Disease Research, NorthShore University HealthSystem, 2650 Ridge Ave., Evanston, IL 60201, USA.,University of Chicago Pritzker School of Medicine, Chicago, IL, USA
| | - Lance R Peterson
- Infectious Disease Research, NorthShore University HealthSystem, 2650 Ridge Ave., Evanston, IL 60201, USA.,University of Chicago Pritzker School of Medicine, Chicago, IL, USA
| | - Katherine Mendoza
- Infectious Disease Research, NorthShore University HealthSystem, 2650 Ridge Ave., Evanston, IL 60201, USA
| | - Donna M Schora
- Infectious Disease Research, NorthShore University HealthSystem, 2650 Ridge Ave., Evanston, IL 60201, USA
| | - M Moazzem Hossain
- Department of Physiology, Wayne State University School of Medicine, Detroit, MI 48201, USA
| | - Jian-Ping Jin
- Department of Physiology, Wayne State University School of Medicine, Detroit, MI 48201, USA
| |
Collapse
|
68
|
Lopes Cançado GG, Silveira Silva RO, Rupnik M, Nader AP, Starling de Carvalho J, Miana de Mattos Paixão G, Martins Resende BA, Faria Lobato FC, Vilela EG. Clinical epidemiology of Clostridium difficile infection among hospitalized patients with antibiotic-associated diarrhea in a university hospital of Brazil. Anaerobe 2018; 54:65-71. [DOI: 10.1016/j.anaerobe.2018.08.005] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2018] [Revised: 08/02/2018] [Accepted: 08/12/2018] [Indexed: 02/08/2023]
|
69
|
Kleerebezem M, Binda S, Bron PA, Gross G, Hill C, van Hylckama Vlieg JE, Lebeer S, Satokari R, Ouwehand AC. Understanding mode of action can drive the translational pipeline towards more reliable health benefits for probiotics. Curr Opin Biotechnol 2018; 56:55-60. [PMID: 30296737 DOI: 10.1016/j.copbio.2018.09.007] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2018] [Revised: 08/28/2018] [Accepted: 09/07/2018] [Indexed: 12/18/2022]
Abstract
The different levels of knowledge described in a translational pipeline (the connection of molecular mechanisms with pre-clinical physiological and human health effects) are not complete for many probiotics. At present, we are not in a position to fully understand the mechanistic basis of many well established probiotic health benefits which, in turn, limits our ability to use mechanisms to predict which probiotics are likely to be effective in any given population. Here we suggest that this concept of a translation pipeline connecting mechanistic insights to probiotic efficacy can support the selection and production of improved probiotic products. Such a conceptual pipeline would also provide a framework for the design of clinical trials to convincingly demonstrate the benefit of probiotics to human health in well-defined subpopulations.
Collapse
Affiliation(s)
- Michiel Kleerebezem
- Host Microbe Interactomics Group, Wageningen University, Wageningen, The Netherlands.
| | - Sylvie Binda
- Danone Nutricia Research, Centre Daniel Carasso, Palaiseau, France
| | | | - Gabriele Gross
- Innovative Health Sciences, Reckitt Benckiser, Nijmegen, The Netherlands
| | - Colin Hill
- School of Microbiology and APC Microbiome Ireland, University College Cork, Cork, Ireland
| | | | - Sarah Lebeer
- Department of Bioscience Engineering, University of Antwerp, Antwerp, Belgium
| | - Reetta Satokari
- Immunobiology Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Arthur C Ouwehand
- Global Health and Nutrition Sciences, DuPont Nutrition and Health, Kantvik, Finland
| |
Collapse
|
70
|
Kieser S, Sarker SA, Sakwinska O, Foata F, Sultana S, Khan Z, Islam S, Porta N, Combremont S, Betrisey B, Fournier C, Charpagne A, Descombes P, Mercenier A, Berger B, Brüssow H. Bangladeshi children with acute diarrhoea show faecal microbiomes with increased Streptococcus abundance, irrespective of diarrhoea aetiology. Environ Microbiol 2018; 20:2256-2269. [PMID: 29786169 DOI: 10.1111/1462-2920.14274] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2018] [Accepted: 05/08/2018] [Indexed: 12/22/2022]
Abstract
We report streptococcal dysbiosis in acute diarrhoea irrespective of aetiology. Compared with 20 healthy local controls, 71 Bangladeshi children hospitalized with acute diarrhoea (AD) of viral, mixed viral/bacterial, bacterial and unknown aetiology showed a significantly decreased bacterial diversity with loss of pathways characteristic for the healthy distal colon microbiome (mannan degradation, methylerythritol phosphate and thiamin biosynthesis), an increased proportion of faecal streptococci belonging to the Streptococcus bovis and Streptococcus salivarius species complexes, and an increased level of E. coli-associated virulence genes. No enteropathogens could be attributed to a subgroup of patients. Elevated lytic coliphage DNA was detected in 2 out of 5 investigated enteroaggregative E. coli (EAEC)-infected patients. Streptococcal outgrowth in AD is discussed as a potential nutrient-driven consequence of glucose provided with oral rehydration solution.
Collapse
Affiliation(s)
- Silas Kieser
- Gut Ecosystem Department, Institute of Nutritional Science, Nestlé Research Centre, Vers-chez-les-Blanc, CH-1000, Lausanne 26, Switzerland
| | - Shafiqul A Sarker
- International Centre for Diarrheal Diseases Research, Bangladesh (icddr,b), Nutrition and Clinical Services Division, 68 Shaheed Tajuddin Ahmed Sharani, Mohakhali, Dhaka 1212, Bangladesh
| | - Olga Sakwinska
- Gut Ecosystem Department, Institute of Nutritional Science, Nestlé Research Centre, Vers-chez-les-Blanc, CH-1000, Lausanne 26, Switzerland
| | - Francis Foata
- Gut Ecosystem Department, Institute of Nutritional Science, Nestlé Research Centre, Vers-chez-les-Blanc, CH-1000, Lausanne 26, Switzerland
| | - Shamima Sultana
- International Centre for Diarrheal Diseases Research, Bangladesh (icddr,b), Nutrition and Clinical Services Division, 68 Shaheed Tajuddin Ahmed Sharani, Mohakhali, Dhaka 1212, Bangladesh
| | - Zeenat Khan
- International Centre for Diarrheal Diseases Research, Bangladesh (icddr,b), Nutrition and Clinical Services Division, 68 Shaheed Tajuddin Ahmed Sharani, Mohakhali, Dhaka 1212, Bangladesh
| | - Shoheb Islam
- International Centre for Diarrheal Diseases Research, Bangladesh (icddr,b), Nutrition and Clinical Services Division, 68 Shaheed Tajuddin Ahmed Sharani, Mohakhali, Dhaka 1212, Bangladesh
| | - Nadine Porta
- Gut Ecosystem Department, Institute of Nutritional Science, Nestlé Research Centre, Vers-chez-les-Blanc, CH-1000, Lausanne 26, Switzerland
| | - Séverine Combremont
- Gut Ecosystem Department, Institute of Nutritional Science, Nestlé Research Centre, Vers-chez-les-Blanc, CH-1000, Lausanne 26, Switzerland
| | - Bertrand Betrisey
- Nestlé Institute of Health Sciences, EPFL Innovation Park, CH-1015, Lausanne, Switzerland
| | - Coralie Fournier
- Nestlé Institute of Health Sciences, EPFL Innovation Park, CH-1015, Lausanne, Switzerland
| | - Aline Charpagne
- Nestlé Institute of Health Sciences, EPFL Innovation Park, CH-1015, Lausanne, Switzerland
| | - Patrick Descombes
- Nestlé Institute of Health Sciences, EPFL Innovation Park, CH-1015, Lausanne, Switzerland
| | - Annick Mercenier
- Gut Ecosystem Department, Institute of Nutritional Science, Nestlé Research Centre, Vers-chez-les-Blanc, CH-1000, Lausanne 26, Switzerland
| | - Bernard Berger
- Gut Ecosystem Department, Institute of Nutritional Science, Nestlé Research Centre, Vers-chez-les-Blanc, CH-1000, Lausanne 26, Switzerland
| | - Harald Brüssow
- Gut Ecosystem Department, Institute of Nutritional Science, Nestlé Research Centre, Vers-chez-les-Blanc, CH-1000, Lausanne 26, Switzerland
| |
Collapse
|
71
|
Kiu R, Hall LJ. An update on the human and animal enteric pathogen Clostridium perfringens. Emerg Microbes Infect 2018; 7:141. [PMID: 30082713 PMCID: PMC6079034 DOI: 10.1038/s41426-018-0144-8] [Citation(s) in RCA: 277] [Impact Index Per Article: 39.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2018] [Revised: 06/28/2018] [Accepted: 07/02/2018] [Indexed: 12/18/2022]
Abstract
Clostridium perfringens, a rapid-growing pathogen known to secrete an arsenal of >20 virulent toxins, has been associated with intestinal diseases in both animals and humans throughout the past century. Recent advances in genomic analysis and experimental systems make it timely to re-visit this clinically and veterinary important pathogen. This Review will summarise our understanding of the genomics and virulence-linked factors, including antimicrobial potentials and secreted toxins of this gut pathogen, and then its up-to-date clinical epidemiology and biological role in the pathogenesis of several important human and animal-associated intestinal diseases, including pre-term necrotising enterocolitis. Finally, we highlight some of the important unresolved questions in relation to C. perfringens-mediated infections, and implications for future research directions.
Collapse
Affiliation(s)
- Raymond Kiu
- Gut Microbes and Health Programme, Quadram Institute Bioscience, Norwich Research Park, Norwich, UK.,Norwich Medical School, University of East Anglia, Norwich Research Park, Norwich, UK
| | - Lindsay J Hall
- Gut Microbes and Health Programme, Quadram Institute Bioscience, Norwich Research Park, Norwich, UK.
| |
Collapse
|
72
|
|
73
|
Nasiri MJ, Goudarzi M, Hajikhani B, Ghazi M, Goudarzi H, Pouriran R. Clostridioides (Clostridium) difficile infection in hospitalized patients with antibiotic-associated diarrhea: A systematic review and meta-analysis. Anaerobe 2018; 50:32-37. [DOI: 10.1016/j.anaerobe.2018.01.011] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2017] [Revised: 01/25/2018] [Accepted: 01/28/2018] [Indexed: 02/08/2023]
|
74
|
Roshan N, Hammer KA, Riley TV. Non-conventional antimicrobial and alternative therapies for the treatment of Clostridium difficile infection. Anaerobe 2018; 49:103-111. [DOI: 10.1016/j.anaerobe.2018.01.003] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2017] [Revised: 12/19/2017] [Accepted: 01/05/2018] [Indexed: 02/08/2023]
|
75
|
Kim YJ, Kim SH, Ahn J, Cho S, Kim D, Kim K, Lee H, Son H, Lee HJ, Yong D, Choi JY, Kim HR, Shin JH. Prevalence of Clostridium perfringens toxin in patients suspected of having antibiotic-associated diarrhea. Anaerobe 2017; 48:34-36. [DOI: 10.1016/j.anaerobe.2017.06.015] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2017] [Revised: 06/12/2017] [Accepted: 06/24/2017] [Indexed: 02/08/2023]
|
76
|
Rawat P, Singh PK, Kumar V. Evidence based traditional anti-diarrheal medicinal plants and their phytocompounds. Biomed Pharmacother 2017; 96:1453-1464. [DOI: 10.1016/j.biopha.2017.11.147] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2017] [Revised: 11/21/2017] [Accepted: 11/29/2017] [Indexed: 12/18/2022] Open
|
77
|
Formulation development of SYN-004 (ribaxamase) oral solid dosage form, a β-lactamase to prevent intravenous antibiotic-associated dysbiosis of the colon. Int J Pharm 2017; 534:25-34. [DOI: 10.1016/j.ijpharm.2017.10.001] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2017] [Revised: 09/12/2017] [Accepted: 10/01/2017] [Indexed: 01/05/2023]
|
78
|
Diard M, Hardt WD. Evolution of bacterial virulence. FEMS Microbiol Rev 2017; 41:679-697. [DOI: 10.1093/femsre/fux023] [Citation(s) in RCA: 89] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2016] [Accepted: 04/24/2017] [Indexed: 12/13/2022] Open
|
79
|
Blixt T, Gradel KO, Homann C, Seidelin JB, Schønning K, Lester A, Houlind J, Stangerup M, Gottlieb M, Knudsen JD. Asymptomatic Carriers Contribute to Nosocomial Clostridium difficile Infection: A Cohort Study of 4508 Patients. Gastroenterology 2017; 152:1031-1041.e2. [PMID: 28063955 DOI: 10.1053/j.gastro.2016.12.035] [Citation(s) in RCA: 56] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/27/2016] [Revised: 12/25/2016] [Accepted: 12/27/2016] [Indexed: 01/05/2023]
Abstract
BACKGROUND & AIMS Nosocomial infections with Clostridium difficile present a considerable problem despite numerous attempts by health care workers to reduce risk of transmission. Asymptomatic carriers of C difficile can spread their infection to other patients. We investigated the effects of asymptomatic carriers on nosocomial C difficile infections. METHODS We performed a population-based prospective cohort study at 2 university hospitals in Denmark, screening all patients for toxigenic C difficile in the intestine upon admittance, from October 1, 2012, to January 31, 2013. Screening results were blinded to patients, staff, and researchers. Patients were followed during their hospital stay by daily registration of wards and patient rooms. The primary outcomes were rate of C difficile infection in exposed and unexposed patients and factors associated with transmission. RESULTS C difficile infection was detected in 2.6% of patients not exposed to carriers and in 4.6% of patients exposed to asymptomatic carriers at the ward level (odds ratio for infection if exposed to carrier, 1.79; 95% confidence interval, 1.16-2.76). Amount of exposure correlated with risk of C difficile infection, from 2.2% in the lowest quartile to 4.2% in the highest quartile of exposed patients (P = .026). Combining the load of exposure to carriers and length of stay seemed to have an additive effect on the risk of contracting C difficile. CONCLUSIONS In a population-based prospective cohort study in Denmark, we found that asymptomatic carriers of toxigenic C difficile in hospitals increase risk of infection in other patients.
Collapse
Affiliation(s)
- Thomas Blixt
- Department of Gastroenterology, Frederiksberg Hospital, University of Copenhagen, Frederiksberg, Denmark; Department of Gastroenterology, Bispebjerg Hospital, University of Copenhagen, Copenhagen, Denmark.
| | - Kim Oren Gradel
- Center for Clinical Epidemiology, South, Odense University Hospital, Odense, Denmark; Research Unit of Clinical Epidemiology, Institute of Clinical Research, University of Southern Denmark, Odense, Denmark
| | - Christian Homann
- Department of Gastroenterology, Bispebjerg Hospital, University of Copenhagen, Copenhagen, Denmark
| | - Jakob Benedict Seidelin
- Department of Gastroenterology, Bispebjerg Hospital, University of Copenhagen, Copenhagen, Denmark; Department of Gastroenterology, Herlev Hospital, University of Copenhagen, Herlev, Denmark
| | - Kristian Schønning
- Department of Clinical Microbiology, Hvidovre Hospital, University of Copenhagen, Hvidovre, Denmark; Institute for Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Anne Lester
- Department of Clinical Microbiology, Hvidovre Hospital, University of Copenhagen, Hvidovre, Denmark; Infectious Control, Bispebjerg Hospital, University of Copenhagen, Copenhagen, Denmark; Infection Control, Frederiksberg Hospitals, University of Copenhagen, Frederiksberg, Denmark
| | - Jette Houlind
- Infectious Control, Bispebjerg Hospital, University of Copenhagen, Copenhagen, Denmark; Infection Control, Frederiksberg Hospitals, University of Copenhagen, Frederiksberg, Denmark
| | - Marie Stangerup
- Infectious Control, Bispebjerg Hospital, University of Copenhagen, Copenhagen, Denmark; Infection Control, Frederiksberg Hospitals, University of Copenhagen, Frederiksberg, Denmark
| | - Magnus Gottlieb
- Department of Pulmonary Medicine, Bispebjerg Hospital, University of Copenhagen, Copenhagen, Denmark
| | - Jenny Dahl Knudsen
- Department of Clinical Microbiology, Hvidovre Hospital, University of Copenhagen, Hvidovre, Denmark; Infectious Control, Bispebjerg Hospital, University of Copenhagen, Copenhagen, Denmark; Infection Control, Frederiksberg Hospitals, University of Copenhagen, Frederiksberg, Denmark
| |
Collapse
|
80
|
Guo M, Wu F, Hao G, Qi Q, Li R, Li N, Wei L, Chai T. Bacillus subtilis Improves Immunity and Disease Resistance in Rabbits. Front Immunol 2017; 8:354. [PMID: 28424690 PMCID: PMC5372816 DOI: 10.3389/fimmu.2017.00354] [Citation(s) in RCA: 223] [Impact Index Per Article: 27.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2017] [Accepted: 03/13/2017] [Indexed: 01/06/2023] Open
Abstract
Probiotics such as Lactobacillus and Bifidobacterium have been successfully used to promote growth and prevent diseases. Previous reports have demonstrated that Bacillus subtilis (B. subtilis) was a potential probiotic for animals. In this research, 180 B. subtilis were isolated from the soil, identified, and investigated in vitro. Furthermore, five B. subtilis were selected and mixed to investigate their effect on growth performance, immune response, intestine microbiota, and disease resistance in rabbits. Rabbits with a diet of 106 CFU g-1 mixed B. subtilis exhibited the best growth performance and higher serum IgG and IgA than controls (P < 0.05). Moreover, dairy with B. subtilis can promote the balance of intestinal flora. The major proinflammatory factor and β-defensin were upregulated compared with the controls. After 7 weeks of feeding, the survival rate of the rabbits fed with B. subtilis was significantly higher than those in the control groups postinfected with Escherichia coli. At the same time, this study detected higher expression of β-defensin and reduced bacteria contents of the heart and cecal contents from the diet mixed with B. subtilis compared with the control groups. In conclusion, dietary supplementation with B. subtilis for rabbits could improve growth performance, intestinal homeostasis, and immune organ index and enhance innate immune response as well as disease resistance. These findings showed that the induction of β-defensin by B. subtilis might be an interesting new therapeutic strategy to strengthen innate defense mechanisms.
Collapse
Affiliation(s)
- Mengjiao Guo
- College of Veterinary Medicine, Shandong Agricultural University, Sino-German Cooperative Research Centre for Zoonosis of Animal Origin of Shandong Province, Tai’an City, China
- Collaborative Innovation Center for the Origin and Control of Emerging Infectious Diseases, Taishan Medical University, Tai’an City, China
| | - Fahao Wu
- College of Veterinary Medicine, Shandong Agricultural University, Sino-German Cooperative Research Centre for Zoonosis of Animal Origin of Shandong Province, Tai’an City, China
| | - Guangen Hao
- College of Veterinary Medicine, Shandong Agricultural University, Sino-German Cooperative Research Centre for Zoonosis of Animal Origin of Shandong Province, Tai’an City, China
| | - Qin Qi
- Tai’an City Central Hospital, Tai’an City, China
| | - Rong Li
- College of Veterinary Medicine, Shandong Agricultural University, Sino-German Cooperative Research Centre for Zoonosis of Animal Origin of Shandong Province, Tai’an City, China
| | - Ning Li
- College of Veterinary Medicine, Shandong Agricultural University, Sino-German Cooperative Research Centre for Zoonosis of Animal Origin of Shandong Province, Tai’an City, China
- Collaborative Innovation Center for the Origin and Control of Emerging Infectious Diseases, Taishan Medical University, Tai’an City, China
| | - Liangmeng Wei
- College of Veterinary Medicine, Shandong Agricultural University, Sino-German Cooperative Research Centre for Zoonosis of Animal Origin of Shandong Province, Tai’an City, China
| | - Tongjie Chai
- College of Veterinary Medicine, Shandong Agricultural University, Sino-German Cooperative Research Centre for Zoonosis of Animal Origin of Shandong Province, Tai’an City, China
| |
Collapse
|
81
|
Martínez-Meléndez A, Camacho-Ortiz A, Morfin-Otero R, Maldonado-Garza HJ, Villarreal-Treviño L, Garza-González E. Current knowledge on the laboratory diagnosis of Clostridium difficile infection. World J Gastroenterol 2017; 23:1552-1567. [PMID: 28321156 PMCID: PMC5340807 DOI: 10.3748/wjg.v23.i9.1552] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/15/2016] [Revised: 01/21/2017] [Accepted: 02/17/2017] [Indexed: 02/06/2023] Open
Abstract
Clostridium difficile (C. difficile) is a spore-forming, toxin-producing, gram-positive anaerobic bacterium that is the principal etiologic agent of antibiotic-associated diarrhea. Infection with C. difficile (CDI) is characterized by diarrhea in clinical syndromes that vary from self-limited to mild or severe. Since its initial recognition as the causative agent of pseudomembranous colitis, C. difficile has spread around the world. CDI is one of the most common healthcare-associated infections and a significant cause of morbidity and mortality among older adult hospitalized patients. Due to extensive antibiotic usage, the number of CDIs has increased. Diagnosis of CDI is often difficult and has a substantial impact on the management of patients with the disease, mainly with regards to antibiotic management. The diagnosis of CDI is primarily based on the clinical signs and symptoms and is only confirmed by laboratory testing. Despite the high burden of CDI and the increasing interest in the disease, episodes of CDI are often misdiagnosed. The reasons for misdiagnosis are the lack of clinical suspicion or the use of inappropriate tests. The proper diagnosis of CDI reduces transmission, prevents inadequate or unnecessary treatments, and assures best antibiotic treatment. We review the options for the laboratory diagnosis of CDI within the settings of the most accepted guidelines for CDI diagnosis, treatment, and prevention of CDI.
Collapse
|
82
|
Silverman MA, Konnikova L, Gerber JS. Impact of Antibiotics on Necrotizing Enterocolitis and Antibiotic-Associated Diarrhea. Gastroenterol Clin North Am 2017; 46:61-76. [PMID: 28164853 PMCID: PMC5314436 DOI: 10.1016/j.gtc.2016.09.010] [Citation(s) in RCA: 80] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Antibiotic treatment alters the composition and metabolic function of the intestinal microbiota. These alterations may contribute to the pathogenesis of necrotizing enterocolitis (NEC) and antibiotic-associated diarrhea (AAD). Recent studies are beginning to unravel the contribution of specific groups of microbes and their metabolic pathways to these diseases. Probiotics or other microbiota-targeted therapies may provide effect strategies to prevent and treat NEC and AAD.
Collapse
Affiliation(s)
- Michael A. Silverman
- Division of Infectious Diseases, The Children’s Hospital of Philadelphia, Philadelphia, PA 19104, and Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104
| | - Liza Konnikova
- Department of Pediatric and Newborn Medicine, Brigham and Women’s Hospital, Boston, MA 02115 and Department of Pediatrics, Harvard Medical School, Boston, MA 02115
| | - Jeffrey S. Gerber
- Center for Pediatric Clinical Effectiveness, Division of Infectious Diseases, The Children’s Hospital of Philadelphia, Philadelphia, PA, 19104 and Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104
| |
Collapse
|
83
|
Roberts T, Kokai-Kun JF, Coughlin O, Lopez BV, Whalen H, Bristol JA, Hubert S, Longstreth J, Lasseter K, Sliman J. Tolerability and Pharmacokinetics of SYN-004, an Orally Administered β-Lactamase for the Prevention of Clostridium difficile-Associated Disease and Antibiotic-Associated Diarrhea, in Two Phase 1 Studies. Clin Drug Investig 2017; 36:725-734. [PMID: 27283946 DOI: 10.1007/s40261-016-0420-0] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
BACKGROUND SYN-004 is an orally administered β-lactamase enzyme, designed to be given concurrently with certain intravenous β-lactam antibiotics like cephalosporins. SYN-004 is intended to degrade residual antibiotics excreted into the intestine as a result of hepatobiliary excretion and to prevent the disruption of the gut microbiome by these excess antibiotics. Preserving the gut microbiome is expected to prevent secondary infections by pathogens like Clostridium difficile and protect against other antibiotic-associated diarrheas. METHODS Two, randomized, double blind, placebo-controlled Phase 1 clinical studies were conducted in normal healthy adult volunteers to assess the tolerability and systemic absorption of single and multiple doses of SYN-004. A single-ascending dose study investigated single oral doses of 75-750 mg SYN-004 and was conducted in 40 subjects (five cohorts of six active and two placebo subjects). A multiple-ascending dose study investigated doses of 75-300 mg SYN-004, administered every 6 h for 7 days and was conducted in 24 subjects (three cohorts of six active and two placebo subjects). The safety and tolerability of SYN-004 was assessed and serial plasma and serum samples were collected to assess the pharmacokinetics and potential immunogenicity of SYN-004. RESULTS Minimal and mild adverse events were reported in ~30 % of the subjects who received active drug and placebo and no antidrug antibodies were detected in any subject. Analysis of serial plasma samples demonstrated negligible systemic bioavailability of SYN-004 with most plasma concentrations being below the lower limit of quantitation (0.8 ng/mL) for the assay. SYN-004 was well tolerated in the 48 subjects who received active drug, and adverse events in those subjects were comparable to the 16 subjects who received placebo, up to the maximum doses administered in each study. CONCLUSION SYN-004 was well tolerated up to a single oral dose of 750 mg and multiple doses of 300 mg every 6 h for 7 days. The pharmacokinetic results support that SYN-004 remained localized in the intestine.
Collapse
Affiliation(s)
- Tracey Roberts
- Synthetic Biologics, Inc., 9605 Medical Center Drive, Suite 270, Rockville, MD, 20850, USA
| | - John F Kokai-Kun
- Synthetic Biologics, Inc., 9605 Medical Center Drive, Suite 270, Rockville, MD, 20850, USA.
| | - Olivia Coughlin
- Synthetic Biologics, Inc., 9605 Medical Center Drive, Suite 270, Rockville, MD, 20850, USA
| | | | - Heidi Whalen
- Synthetic Biologics, Inc., 9605 Medical Center Drive, Suite 270, Rockville, MD, 20850, USA
| | - J Andrew Bristol
- Synthetic Biologics, Inc., 9605 Medical Center Drive, Suite 270, Rockville, MD, 20850, USA
| | - Steven Hubert
- Synthetic Biologics, Inc., 9605 Medical Center Drive, Suite 270, Rockville, MD, 20850, USA
| | | | | | - Joseph Sliman
- Synthetic Biologics, Inc., 9605 Medical Center Drive, Suite 270, Rockville, MD, 20850, USA
| |
Collapse
|
84
|
Warriner K, Xu C, Habash M, Sultan S, Weese S. Dissemination ofClostridium difficilein food and the environment: Significant sources ofC. difficilecommunity-acquired infection? J Appl Microbiol 2016; 122:542-553. [DOI: 10.1111/jam.13338] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2016] [Revised: 10/25/2016] [Accepted: 10/27/2016] [Indexed: 12/11/2022]
Affiliation(s)
- K. Warriner
- Department of Food Science; University of Guelph; Guelph ON Canada
| | - C. Xu
- Shanghai Ocean University; Shanghai China
| | - M. Habash
- School of Environmental Biology; University of Guelph; Guelph ON Canada
| | - S. Sultan
- School of Environmental Biology; University of Guelph; Guelph ON Canada
| | - S.J. Weese
- Pathobiology; University of Guelph; Guelph ON Canada
| |
Collapse
|
85
|
The Possible Innovative Use of Bifidobacterium longum W11 in Association With Rifaximin: A New Horizon for Combined Approach? J Clin Gastroenterol 2016; 50 Suppl 2, Proceedings from the 8th Probiotics, Prebiotics & New Foods for Microbiota and Human Health meeting held in Rome, Italy on September 13-15, 2015:S153-S156. [PMID: 27741162 DOI: 10.1097/mcg.0000000000000683] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
GOALS The aim of the study was to unequivocally demonstrate the nontransmissibility of the genes mediating the resistance of the strain Bifidobacterium longum W11 (LMG P-21586) to rifaximin. BACKGROUND Most antibiotic treatments can induce unfavorable side effects such as antibiotic-associated diarrhea, which is largely attributable to the disruption of the intestinal microbiota. The parallel intake of probiotic bacteria might reduce these events, even if with generally very poor results. In this regard, the use of antibiotic-resistant beneficial bacteria could represent a worthy strategy. STUDY Rifaximin was tested in parallel with rifampicin, rifapentine, and rifabutin, all rifamycin derivates, using 5 different concentrations. Susceptibility tests were performed by the disc diffusion method of Kirby-Bauer, and inhibition zones were measured after incubation at 37°C. B. longum BL03 was used as comparison. The B. longum W11 genome was sequenced on Illumina MiSeq with a 250 PE reads module. After mapping the reads with the reference bacterial genome, the alignment data were processed using FreeBayes software. RESULTS B. longum BL03 was inhibited by all antibiotics even at the lowest concentration. In contrast, the W11 strain was inhibited by rifampicin, rifabutin, and rifaximin only at the highest concentration (512 μg/mL). The genomic analysis showed a mutation into the chromosomal DNA. No transposable elements were found, and the genetic locus was not flanked by close mobile genetic elements. CONCLUSIONS B. longum W11 could be used in combined therapy with rifaximin, thus opening new focused frontiers in the probiotic era while preserving the necessary safety of use for consumers.
Collapse
|
86
|
Can Probiotics Reduce Diarrhea and Infant Mortality in Africa?: The Project of a Pilot Study. J Clin Gastroenterol 2016; 50 Suppl 2, Proceedings from the 8th Probiotics, Prebiotics & New Foods for Microbiota and Human Health meeting held in Rome, Italy on September 13-15, 2015:S120-S123. [PMID: 27741153 DOI: 10.1097/mcg.0000000000000677] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
BACKGROUND Diarrhea accounts for 9% of the mortality among children under 5 years of age worldwide, and it is significantly associated with malnutrition. Each year, diarrhea kills around 760,000 children under 5 years of age and most of these are in sub-Saharan Africa.In Uganda, the infant mortality rate of 58 per 1000 is unacceptably high, and the major contributors include malnutrition, diarrhea, pneumonia, malaria, prematurity, sepsis, and newborn illnesses.There is an urgent need for intervention to prevent and control diarrheal diseases. STUDY DESIGN Our open-label, randomized controlled study has the primary endpoint of reducing diarrhea and infectious diseases (number of episodes/severity) and the secondary endpoint of decreasing infant mortality. The trial is currently conducted in Luzira, a suburb of Kampala, the capital of Uganda, and in Gulu and Lira, in the north of Uganda.The study is projected to enroll 4000 babies (control=2000 and treatment=2000) who will be followed till 1 year of life. As controls, 2000 babies of the same community are planned to be considered.The probiotic product selected for the trial is composed of 3 designated microorganisms, namely Bifidobacterium breve BR03 (DSM 16604), B. breve B632 (DSM 24706), and Lactobacillus delbrueckii subsp. delbrueckii LDD01 (DSM 22106). The concentration of the 3 bacteria is 10 viable cells/strain/daily dose (5 drops). PERSPECTIVES For a total sample of 4000 babies, the study has an 80% power at a 5% significance level.
Collapse
|
87
|
Generoso SDV, Lages PC, Correia MITD. Fiber, prebiotics, and diarrhea: what, why, when and how. Curr Opin Clin Nutr Metab Care 2016; 19:388-393. [PMID: 27428350 DOI: 10.1097/mco.0000000000000311] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
PURPOSE OF REVIEW Dietary fiber and prebiotics have been the focus of research and discussion for decades, but there are still pending concepts and definitions, in particular when addressing their use in the prevention and treatment of diarrhea. The purpose of this review is to present the latest advances in the understanding of dietary fiber and prebiotics, to review their proven role in the management of diarrhea, and to postulate the best timings and optimal doses. RECENT FINDINGS The use of prebiotics has encompassed not only prevention but also the treatment of distinct types of diarrhea, at different treatment moments, and with regard to various different markers of outcome. Furthermore, the description of soluble fibers claiming to be prebiotics, and vice versa, has too often been the tone in the literature, which has led to misconceptions in classification and, consequently, confusion over the interpretation of results. It remains difficult to establish a consensus about the real impact of fiber and prebiotics on the prevention and therapy of diarrhea. SUMMARY The review highlights the overlapping concepts of fiber and prebiotics, and supports the need for adequate individualization of their use, according to the goal - either prevention or treatment of diarrhea - as well as the optimal timing and dose to be used. Nonetheless, viscous soluble fibers seem to be the best option in treating diarrhea, whereas prebiotics are more important in preventing and avoiding recurrence.
Collapse
Affiliation(s)
- Simone de Vasconcelos Generoso
- aDepartamento de Nutrição, Escola de Enfermagem bDepartamento de Cirurgia, Faculdade de Medicina, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | | | | |
Collapse
|
88
|
Forbes JD, Van Domselaar G, Bernstein CN. The Gut Microbiota in Immune-Mediated Inflammatory Diseases. Front Microbiol 2016; 7:1081. [PMID: 27462309 PMCID: PMC4939298 DOI: 10.3389/fmicb.2016.01081] [Citation(s) in RCA: 276] [Impact Index Per Article: 30.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2016] [Accepted: 06/28/2016] [Indexed: 12/17/2022] Open
Abstract
The collection of microbes and their genes that exist within and on the human body, collectively known as the microbiome has emerged as a principal factor in human health and disease. Humans and microbes have established a symbiotic association over time, and perturbations in this association have been linked to several immune-mediated inflammatory diseases (IMID) including inflammatory bowel disease, rheumatoid arthritis, and multiple sclerosis. IMID is a term used to describe a group of chronic, highly disabling diseases that affect different organ systems. Though a cornerstone commonality between IMID is the idiopathic nature of disease, a considerable portion of their pathobiology overlaps including epidemiological co-occurrence, genetic susceptibility loci and environmental risk factors. At present, it is clear that persons with an IMID are at an increased risk for developing comorbidities, including additional IMID. Advancements in sequencing technologies and a parallel explosion of 16S rDNA and metagenomics community profiling studies have allowed for the characterization of microbiomes throughout the human body including the gut, in a myriad of human diseases and in health. The main challenge now is to determine if alterations of gut flora are common between IMID or, if particular changes in the gut community are in fact specific to a single disease. Herein, we review and discuss the relationships between the gut microbiota and IMID.
Collapse
Affiliation(s)
- Jessica D. Forbes
- Department of Medical Microbiology and Infectious Diseases, University of Manitoba, WinnipegMB, Canada
- National Microbiology Laboratory, Public Health Agency of Canada, WinnipegMB, Canada
| | - Gary Van Domselaar
- Department of Medical Microbiology and Infectious Diseases, University of Manitoba, WinnipegMB, Canada
- National Microbiology Laboratory, Public Health Agency of Canada, WinnipegMB, Canada
| | - Charles N. Bernstein
- Department of Internal Medicine and the IBD Clinical and Research Centre, University of Manitoba, WinnipegMB, Canada
| |
Collapse
|