47 results on '"Host-Associated Microbial Communities"'
Search Results
2. Vertebrate Hosts as Islands: Dynamics of Selection, Immigration, Loss, Persistence, and Potential Function of Bacteria on Salamander Skin
- Author
-
Loudon, Andrew H, Venkataraman, Arvind, Van Treuren, William, Woodhams, Douglas C, Parfrey, Laura Wegener, McKenzie, Valerie J, Knight, Rob, Schmidt, Thomas M, and Harris, Reid N
- Subjects
Microbiology ,Biological Sciences ,Ecology ,Infectious Diseases ,2.2 Factors relating to the physical environment ,Aetiology ,Infection ,Life Below Water ,neutral model ,host-associated microbial communities ,Island biogeography ,Plethodon cinereus ,Batrachochytrium dendrobatidis ,symbiosis ,antifungal ,Environmental Science and Management ,Soil Sciences ,Medical microbiology - Abstract
Skin bacterial communities can protect amphibians from a fungal pathogen; however, little is known about how these communities are maintained. We used a neutral model of community ecology to identify bacteria that are maintained on salamanders by selection or by dispersal from a bacterial reservoir (soil) and ecological drift. We found that 75% (9/12) of bacteria that were consistent with positive selection,
- Published
- 2016
3. A Co-Association of Streptococcus mutans and Veillonella parvula/dispar in Root Caries Patients and
- Author
-
Amber M, Abram, Michelle M, Szewczyk, Seon G, Park, Sumita S, Sam, Haya B, Eldana, Fadi J, Koria, Joseph M, Ferracciolo, Laura A, Young, Hina, Qadir, Aaron J, Bonham, Fei, Yang, Jonathan S, Zora, Sara A, Abdulelah, Neil A, Patel, Ayah, Koleilat, Malaka A, Saleh, Jamal A, Alhabeil, Shameel, Khan, Ashootosh, Tripathi, John G, Palanci, and Eric S, Krukonis
- Subjects
Streptococcus mutans ,Veillonella ,Sucrose ,Root Caries ,Biofilms ,Humans ,Saliva, Artificial ,Pilot Projects ,Dental Caries ,Host-Associated Microbial Communities ,Aged - Abstract
Root caries in geriatric patients is a growing problem as more people are maintaining their natural teeth into advanced age. We determined the levels of various bacterial species previously implicated in root caries disease or health using quantitative real-time PCR in a pilot study of 7 patients with 1 to 4 root caries lesions per person. Levels of 12 different species on diseased roots compared to healthy (contralateral control) roots were measured. Four species were found at significantly higher levels on diseased roots (Streptococcus mutans, Veillonella parvula/dispar, Actinomyces naeslundii/viscosus, and Capnocytophaga granulosa) compared across all plaque samples. The level of colonization by these species varied dramatically (up to 1,000-fold) between patients, indicating different patients have different bacteria contributing to root caries disease. Neither of the two species previously reported to correlate with healthy roots (C. granulosa and Delftia acidovorans) showed statistically significant protective roles in our population, although D. acidovorans showed a trend toward higher levels on healthy teeth (P = 0.08). There was a significant positive correlation between higher levels of S. mutans and V. parvula/dispar on the same diseased teeth. In vitro mixed biofilm studies demonstrated that co-culturing S. mutans and V. parvula leads to a 50 to 150% increase in sucrose-dependent biofilm mass compared to S. mutans alone, depending on the growth conditions, while V. parvula alone did not form in vitro biofilms. The presence of V. parvula also decreased the acidification of S. mutans biofilms when grown in artificial saliva and enhanced the health of mixed biofilms.
- Published
- 2023
4. Porphyromonas gingivalis Tyrosine Kinase Is a Fitness Determinant in Polymicrobial Infections
- Author
-
John D. Perpich, Lan Yakoumatos, Kendall S. Stocke, Gina R. Lewin, Anayancy Ramos, Deborah R. Yoder-Himes, Marvin Whiteley, and Richard J. Lamont
- Subjects
Mice ,Infectious Diseases ,Fusobacterium nucleatum ,Coinfection ,Immunology ,Animals ,Parasitology ,Protein-Tyrosine Kinases ,Host-Associated Microbial Communities ,Porphyromonas gingivalis ,Microbiology ,Abscess ,Ecosystem - Abstract
Many pathogenic microbial ecosystems are polymicrobial, and community function can be shaped by interbacterial interactions. Little is known, however, regarding the genetic determinants required for fitness in heterotypic community environments. In periodontal diseases, Porphyromonas gingivalis is a primary pathogen, but only within polymicrobial communities. Here, we used a transposon sequencing (Tn-Seq) library of P. gingivalis to screen for genes that influence fitness of the organism in a coinfection murine abscess model with the oral partner species Streptococcus gordonii and Fusobacterium nucleatum. Genes impacting fitness with either organism were involved in diverse processes, including metabolism and energy production, along with cell wall and membrane biogenesis. Despite the overall similarity of function, the majority of identified genes were specific to the partner species, indicating that synergistic mechanisms of P. gingivalis vary to a large extent according to community composition. Only two genes were identified as essential for P. gingivalis fitness in abscess development with both S. gordonii and F. nucleatum: ptk1, encoding a tyrosine kinase, and inlJ, encoding an internalin family surface protein. Ptk1, but not InlJ, is required for community development with S. gordonii, and we found that the action of this kinase is similarly required for P. gingivalis to accumulate in a community with F. nucleatum. A limited number of P. gingivalis genes are therefore required for species-independent synergy, and the Ptk1 tyrosine kinase network may integrate and coordinate input from multiple organisms.
- Published
- 2022
- Full Text
- View/download PDF
5. Vertebrate hosts as islands: dynamics of selection, immigration, loss, persistence and potential function of bacteria on salamander skin
- Author
-
Andrew Howard Loudon, Arvind eVenkataraman, William eVanTreuren, Douglas eWoodhams, Laura Wegener Parfrey, Valerie eMcKenzie, Rob eKnight, Thomas eSchmidt, and Reid eHarris
- Subjects
Symbiosis ,Antifungal ,island biogeography ,Batrachochytrium dendrobatidis ,host-associated microbial communities ,Neutral model ,Microbiology ,QR1-502 - Abstract
Skin bacterial communities can protect amphibians from a fungal pathogen; however, little is known about how these communities are maintained. We used a neutral model of community ecology to identify bacteria that are maintained on salamanders by selection or by dispersal from a bacterial reservoir (soil) and ecological drift. We found that 75% (9/12) of bacteria that were consistent with positive selection, < 1% of bacteria that were consistent with random dispersal and none of the bacteria that were consistent under negative selection had a 97% or greater match to antifungal isolates. Additionally we performed an experiment where salamanders were either provided or denied a bacterial reservoir and estimated immigration and loss (emigration and local extinction) rates of bacteria on salamanders in both treatments. Loss was strongly related to bacterial richness, suggesting competition is important for structuring the community. Bacteria closely related to antifungal isolates were more likely to persist on salamanders with or without a bacterial reservoir, suggesting they had a competitive advantage. Furthermore, over-represented and under-represented OTUs had similar persistence on salamanders when a bacterial reservoir was present. However, under-represented OTUs were less likely to persist in the absence of a bacterial reservoir, suggesting that the over-represented and under-represented bacteria are selected for or against on salamanders through time. Our findings from the neutral model, migration and persistence analyses show that bacteria that exhibit a high similarity to antifungal isolates persist on salamanders, which likely protect hosts against pathogens and improve fitness. This research is one of the first to apply ecological theory to investigate assembly of host associated-bacterial communities, which can provide insights for probiotic bioaugmentation as a conservation strategy against disease.
- Published
- 2016
- Full Text
- View/download PDF
6. Nontypeable Haemophilus influenzae Infection Impedes Pseudomonas aeruginosa Colonization and Persistence in Mouse Respiratory Tract
- Author
-
Lea Novak, Natalie Lindgren, W. Edward Swords, Melissa S. McDaniel, and Benjamin C. Hunt
- Subjects
Haemophilus Infections ,Cystic Fibrosis ,Respiratory System ,Immunology ,Population ,Biology ,medicine.disease_cause ,Microbiology ,Cystic fibrosis ,Proinflammatory cytokine ,Haemophilus influenzae ,Mice ,Immune system ,otorhinolaryngologic diseases ,medicine ,Animals ,Humans ,Colonization ,Respiratory system ,education ,Respiratory Tract Infections ,education.field_of_study ,Lung ,business.industry ,Pseudomonas aeruginosa ,Respiratory infection ,Host-Associated Microbial Communities ,medicine.disease ,Infectious Diseases ,medicine.anatomical_structure ,Biofilms ,Parasitology ,business ,Respiratory tract - Abstract
Patients with cystic fibrosis (CF) experience lifelong respiratory infections which are a significant cause of morbidity and mortality. These infections are polymicrobial in nature, and the predominant bacterial species undergo a predictable series of changes as patients age. Young patients have populations dominated by opportunists that are typically found within the microbiome of the human nasopharynx, such as nontypeable Haemophilus influenzae (NTHi); these are eventually supplanted and the population within the CF lung is later dominated by pathogens such as Pseudomonas aeruginosa (Pa). In this study, we investigated how initial colonization with NTHi impacts colonization and persistence of Pa in the respiratory tract.Analysis of polymicrobial biofilms in vitro by confocal microscopy revealed that NTHi promoted greater levels of Pa biofilm volume and diffusion. However, sequential respiratory infection of mice with NTHi followed by Pa resulted in significantly lower Pa as compared to infection with Pa alone. Coinfected mice also had reduced airway tissue damage and lower levels of inflammatory cytokines as compared with Pa infected mice. Similar results were observed after instillation of heat-inactivated NTHi bacteria or purified NTHi lipooligosaccharide (LOS) endotoxin prior to Pa introduction. Based on these results, we conclude that NTHi significantly reduces susceptibility to subsequent Pa infection, most likely due to priming of host innate immunity rather than a direct competitive interaction between species. These findings have potential significance with regard to therapeutic management of early life infections in patients with CF.
- Published
- 2022
- Full Text
- View/download PDF
7. A Trifecta of New Insights into Ovine Footrot for Infection Drivers, Immune Response, and Host-Pathogen Interactions
- Author
-
Charlotte Back, Catrin S. Rutland, Jule K. Michler, Adam M. Blanchard, Sean Wattegedera, Ceri E. Staley, Christina-Marie Baumbach, Nerissa Newbold, Sabine Tötemeyer, Laurence Shaw, and Gary Entrican
- Subjects
Immunology ,Sheep Diseases ,Virulence ,Dichelobacter nodosus ,Microbiology ,Collagen Type I ,histology ,transcriptomics ,Immune system ,Animals ,Foot Rot ,Pathogen ,veterinary microbiology ,Skin ,Mycoplasma fermentans ,metagenomics ,Sheep ,metatranscriptomics ,Bacteria ,biology ,Host (biology) ,Immunity ,Host-Associated Microbial Communities ,biology.organism_classification ,Immunosurveillance ,Infectious Diseases ,Infectious disease (medical specialty) ,Host-Pathogen Interactions ,Parasitology ,Transcriptome - Abstract
Footrot is a polymicrobial infectious disease in sheep causing severe lameness, leading to one of the industry’s largest welfare problems. The complex etiology of footrot makesin situorin vitroinvestigations difficult. Computational methods offer a solution to understanding the bacteria involved and how they may interact with the host, ultimately providing a way to identify targets for future hypothesis-driven investigative work. Here, we present the first combined global analysis of bacterial community transcripts together with the host immune response in healthy and diseased ovine feet during a natural polymicrobial infection state using metatranscriptomics. The intratissue and surface bacterial populations and the most abundant bacterial transcriptomes were analyzed, demonstrating that footrot-affected skin has reduced diversity and increased abundances of not only the causative bacteriumDichelobacter nodosusbut also other species such asMycoplasma fermentansandPorphyromonas asaccharolytica. Host transcriptomics reveals the suppression of biological processes related to skin barrier function, vascular functions, and immunosurveillance in unhealthy interdigital skin, supported by histological findings that type I collagen (associated with scar tissue formation) is significantly increased in footrot-affected interdigital skin compared to outwardly healthy skin. Finally, we provide some interesting indications of host and pathogen interactions associated with virulence genes and the host spliceosome, which could lead to the identification of future therapeutic targets.
- Published
- 2021
8. The Abundance and Organization of
- Author
-
Mark M, Hahn, Juan F, González, Regan, Hitt, Lauren, Tucker, and John S, Gunn
- Subjects
Salmonella typhimurium ,Mice ,Cholesterol ,Extracellular Polymeric Substance Matrix ,Biofilms ,Animals ,Humans ,Female ,Gallstones ,biochemical phenomena, metabolism, and nutrition ,Salmonella typhi ,Host-Associated Microbial Communities - Abstract
Salmonella enterica serovar Typhi causes chronic infections by establishing biofilms on cholesterol gallstones. The production of extracellular polymeric substances (EPSs) is key to biofilm development, and biofilm architecture depends on which EPSs are made. The presence and spatial distribution of Salmonella EPSs produced in vitro and in vivo were investigated in Salmonella enterica serovar Typhimurium and S. Typhi biofilms by confocal microscopy. Comparisons between serovars and EPS-mutant bacteria were carried out by examining growth on cholesterol-coated surfaces, with human gallstones in ox or human bile, and in mice with gallstones. On cholesterol-coated surfaces, no major differences in EPS biomass were found between serovars. Cocultured biofilms containing wild-type (WT) and EPS-mutant bacteria demonstrated WT compensation for EPS mutations. Analysis of biofilm EPSs from gallbladder-mimicking conditions found that culture in human bile more consistently replicated the relative abundance and spatial organization of each EPS on gallstones from the chronic mouse model than culture in ox bile. S. Typhimurium biofilms cultured in vitro on gallstones in ox bile exhibited colocalized pairings of curli fimbriae/lipopolysaccharide and O-antigen capsule/cellulose, while these associations were not present in S. Typhi biofilms or in mouse gallstone biofilms. In general, the inclusion of human bile with gallstones in vitro replicated biofilm development on gallstones in vivo, demonstrating the strength of this model for studying biofilm parameters or EPS-directed therapeutic treatments.
- Published
- 2021
9. Vibrio cholerae Infection Induces Strain-Specific Modulation of the Zebrafish Intestinal Microbiome
- Author
-
Andrew D. Winters, Kevin R. Theis, Jeffrey H. Withey, and Paul Breen
- Subjects
0301 basic medicine ,030106 microbiology ,Immunology ,ved/biology.organism_classification_rank.species ,medicine.disease_cause ,Microbiology ,03 medical and health sciences ,Cholera ,RNA, Ribosomal, 16S ,Genotype ,medicine ,Animals ,Colonization ,Microbiome ,Model organism ,Vibrio cholerae ,Zebrafish ,Pathogen ,biology ,ved/biology ,fungi ,Host-Associated Microbial Communities ,biology.organism_classification ,Gastrointestinal Microbiome ,Disease Models, Animal ,030104 developmental biology ,Infectious Diseases ,Host-Pathogen Interactions ,Microbial Interactions ,Parasitology ,Disease Susceptibility ,Metagenomics ,Proteobacteria - Abstract
Zebrafish (Danio rerio) is an attractive model organism to use for an array of scientific studies, including host-microbe interactions. Zebrafish contain a core (i.e., consistently detected) intestinal microbiome consisting primarily of Proteobacteria. Furthermore, this core intestinal microbiome is plastic and can be significantly altered due to external factors. Zebrafish are particularly useful for the study of aquatic microbes that can colonize vertebrate hosts, including Vibrio cholerae. As an intestinal pathogen, V. cholerae must colonize the intestine of an exposed host for pathogenicity to occur. Members of the resident intestinal microbial community likely must be reduced or eliminated by V. cholerae for colonization, and subsequent disease, to occur. Many studies have explored a variety of aspects of the pathogenic effects of V. cholerae on zebrafish and other model organisms but few have researched how a V. cholerae infection changes the resident intestinal microbiome. In this study, 16S rRNA gene sequencing was used to examine how five genetically diverse V. cholerae strains alter the intestinal microbiome following an infection. We found that V. cholerae colonization induced significant changes in the zebrafish intestinal microbiome. Notably, changes in the microbial profile were significantly different from each other, based on the particular strain of V. cholerae used to infect zebrafish hosts. We conclude that V. cholerae significantly modulates the zebrafish intestinal microbiota to enable colonization and that specific microbes that are targeted depend on the V. cholerae genotype.
- Published
- 2021
- Full Text
- View/download PDF
10. Anaerobic Microbiota Derived from the Upper Airways Impact Staphylococcus aureus Physiology
- Author
-
Madison M. Ahmad, Sarah K. Lucas, Alex R. Villarreal, Ryan C. Hunter, Holly C. Boyer, Abayo Itabiyi, and Erin Feddema
- Subjects
Staphylococcus aureus ,Immunology ,Virulence ,Physiology ,medicine.disease_cause ,Microbiology ,Pathogenesis ,otorhinolaryngologic diseases ,medicine ,Prevotella ,Humans ,Anaerobiosis ,Respiratory Tract Infections ,biology ,Streptococcus ,Microbiota ,Mucin ,Staphylococcal Infections ,Host-Associated Microbial Communities ,Commensalism ,biology.organism_classification ,Infectious Diseases ,Fusobacterium ,Chronic Disease ,Host-Pathogen Interactions ,Microbial Interactions ,Parasitology ,Disease Susceptibility - Abstract
Staphylococcus aureus is associated with the development of persistent and severe inflammatory diseases of the upper airways. Yet, S. aureus is also carried asymptomatically in the sinonasal cavity of ∼50% of healthy adults. The causes of this duality and host and microbial factors that tip the balance between S. aureus pathogenesis and commensalism are poorly understood. We have shown that by degrading mucins, anaerobic microbiota support the growth of airway pathogens by liberating metabolites that are otherwise unavailable. Given the widely reported culture-based detection of anaerobes from individuals with chronic rhinosinusitis (CRS), here we tested our hypothesis that CRS microbiota is characterized by a mucin-degrading phenotype that alters S. aureus physiology. Using 16S rRNA gene sequencing, we indeed observed an increased prevalence and abundance of anaerobes in CRS relative to non-CRS controls. PICRUSt2-based functional predictions suggested increased mucin degradation potential among CRS microbiota that was confirmed by direct enrichment culture. Prevotella, Fusobacterium, and Streptococcus comprised a core mucin-degrading community across CRS subjects that generated a nutrient pool that augmented S. aureus growth on mucin as a carbon source. Finally, using transcriptome sequencing (RNA-seq), we observed that S. aureus transcription is profoundly altered in the presence of mucin-derived metabolites, though expression of several key metabolism- and virulence-associated pathways varied between CRS-derived bacterial communities. Together, these data support a model in which S. aureus metabolism and virulence in the upper airways are dependent upon the composition of cocolonizing microbiota and the metabolites they exchange.
- Published
- 2021
- Full Text
- View/download PDF
11. The Vibrio cholerae Type Six Secretion System Is Dispensable for Colonization but Affects Pathogenesis and the Structure of Zebrafish Intestinal Microbiome
- Author
-
Paul Breen, Kevin R. Theis, Andrew D. Winters, and Jeffrey H. Withey
- Subjects
Immunology ,ved/biology.organism_classification_rank.species ,medicine.disease_cause ,Microbiology ,Cholera ,RNA, Ribosomal, 16S ,medicine ,Animals ,Secretion ,Microbiome ,Model organism ,Vibrio cholerae ,Pathogen ,Zebrafish ,Host Microbial Interactions ,biology ,ved/biology ,Type VI Secretion Systems ,Host-Associated Microbial Communities ,biology.organism_classification ,Vibrio ,Gastrointestinal Microbiome ,Disease Models, Animal ,Infectious Diseases ,Host-Pathogen Interactions ,Parasitology ,Disease Susceptibility ,Metagenomics ,Bacteria - Abstract
Zebrafish (Danio rerio) are an attractive model organism for a variety of scientific studies, including host-microbe interactions. The organism is particularly useful for the study of aquatic microbes that can colonize vertebrate hosts, including Vibrio cholerae, an intestinal pathogen. V. cholerae must colonize the intestine of an exposed host for pathogenicity to occur. While numerous studies have explored various aspects of the pathogenic effects of V. cholerae on zebrafish and other model organisms, few, if any, have examined how a V. cholerae infection alters the resident intestinal microbiome and the role of the type six secretion system (T6SS) in that process. In this study, 16S rRNA gene sequencing was utilized to investigate how strains of V. cholerae both with and without the T6SS alter the aforementioned microbial profiles following an infection. V. cholerae infection induced significant changes in the zebrafish intestinal microbiome, and while not necessary for colonization, the T6SS was important for inducing mucin secretion, a marker for diarrhea. Additional salient differences to the microbiome were observed based on the presence or absence of the T6SS in the V. cholerae utilized for challenging the zebrafish hosts. We conclude that V. cholerae significantly modulates the zebrafish intestinal microbiome to enable colonization and that the T6SS is important for pathogenesis induced by the examined V. cholerae strains. Furthermore, the presence or absence of T6SS differentially and significantly affected the composition and structure of the intestinal microbiome, with an increased abundance of other Vibrio bacteria observed in the absence of V. cholerae T6SS.
- Published
- 2021
- Full Text
- View/download PDF
12. Pharyngeal Microbial Signatures Are Predictive of the Risk of Fungal Pneumonia in Hematologic Patients
- Author
-
Luca Facchini, Luigina Romani, Daniela Valente, Antonio Spadea, Katerina Coufalikova, Lukas Englmaier, Katia Codeluppi, Monica Borghi, Matteo Puccetti, Teresa Zelante, Angelica Spolzino, Zdenek Spacil, Giulia Dragonetti, Melissa Palmieri, Francesco Merli, Gianpaolo Nadali, Giuseppe Lomurno, Claudio Costantini, Emilia Nunzi, Francesco Marchesi, Roberta Spaccapelo, Franco Aversa, Marina M. Bellet, Vincenzo Nicola Talesa, Enzo Acerbi, Stefano Giovagnoli, Lorella Melillo, Giorgia Renga, Livio Pagano, Gessica Marchesini, and Singapore Centre for Environmental Life Sciences and Engineering
- Subjects
0301 basic medicine ,Indole-3aldehyde ,Antifungal Agents ,Hematological malignancies, invasive fungal infection, airway microbiome, metabolomics, tryptophan, indole-3-aldehyde, antibiotics ,Antibiotics ,invasive fungal infection ,antibiotics ,Hematological malignancies ,Mice ,0302 clinical medicine ,Risk Factors ,Antimicrobial stewardship ,airway microbiome ,biology ,Microbiota ,Biological sciences [Science] ,Host-Associated Microbial Communities ,metabolomics ,3. Good health ,Infectious Diseases ,Hematologic Neoplasms ,030220 oncology & carcinogenesis ,Metabolic profile ,medicine.drug_class ,Hematological Malignancies ,Immunology ,Risk Assessment ,Microbiology ,03 medical and health sciences ,medicine ,Animals ,Humans ,tryptophan ,Clostridiales ,Airway Microbiome ,Bacteroidetes ,indole-3-aldehyde ,Pneumonia ,biology.organism_classification ,Fungal pneumonia ,medicine.disease ,Hematologic Diseases ,Disease Models, Animal ,Settore MED/15 - MALATTIE DEL SANGUE ,030104 developmental biology ,Mycoses ,Murine model ,Metagenome ,Pharynx ,Parasitology ,Metagenomics ,Dysbiosis - Abstract
The ability to predict invasive fungal infections (IFI) in patients with hematological malignancies is fundamental for successful therapy. Although gut dysbiosis is known to occur in hematological patients, whether airway dysbiosis also contributes to the risk of IFI has not been investigated. Nasal and oropharyngeal swabs were collected for functional microbiota characterization in 173 patients with hematological malignancies recruited in a multicenter, prospective, observational study and stratified according to the risk of developing IFI. A lower microbial richness and evenness were found in the pharyngeal microbiota of high-risk patients that were associated with a distinct taxonomic and metabolic profile. A murine model of IFI provided biologic plausibility for the finding that loss of protective anaerobes, such as Clostridiales and Bacteroidetes, along with an apparent restricted availability of tryptophan, is causally linked to the risk of IFI in hematologic patients and indicates avenues for antimicrobial stewardship and metabolic reequilibrium in IFI. This work was supported by FunMeta Project (ERC-2011-AdG 293714), MicroTher(ERC-2018-PoC-813099), and Gilead (IN-IT-131-4525-518872.9) to L.R. and the GrantAgency of the Czech Republic (GACR No 17-24592Y) and the Czech Ministry ofEducation, Youth and Sports (CETOCOEN PLUS CZ.02.1.01/0.0/0.0/15_003/0000469;LM2015051 and CETOCOEN EXCELLENCE Teaming 2 project CZ.02.1.01/0.0/0.0/18_046/0015975; and Horizon2020 project 857560) to Z.S.
- Published
- 2021
- Full Text
- View/download PDF
13. Cage Environment Regulates Gut Microbiota Independent of Toll-Like Receptors
- Author
-
Gary B. Huffnagle, Stephen J. Gurczynski, David N O'Dwyer, Robert P. Dickson, Bethany B. Moore, Jay H Lipinski, John R. Erb-Downward, and Xiaofeng Zhou
- Subjects
0301 basic medicine ,Transgene ,Immunology ,Inflammation ,Gut flora ,Microbiology ,digestive system ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Immunity ,medicine ,Animals ,Homeostasis ,Intestinal Mucosa ,Receptor ,Immunity, Mucosal ,Innate immune system ,biology ,Host Microbial Interactions ,Toll-Like Receptors ,biology.organism_classification ,medicine.disease ,Host-Associated Microbial Communities ,Immunity, Innate ,Gastrointestinal Microbiome ,030104 developmental biology ,Infectious Diseases ,Models, Animal ,Dysbiosis ,Parasitology ,Disease Susceptibility ,medicine.symptom ,030217 neurology & neurosurgery ,Function (biology) - Abstract
The gut microbiome orchestrates epithelial homeostasis and both local and remote immunological responses. Critical to these regulatory interactions are innate immune receptors termed Toll-like receptors (TLRs). Studies to date have implicated innate immunity and Toll-like receptors in shaping key features of the gut microbiome. However, a variety of biological and environmental variables are also implicated in determining gut microbiota composition. In this report, we hypothesized that cohousing and environment dominated the regulation of the gut microbiota in animal models independent of innate immunity. To determine the importance of these variables, innate immunity, or environment in shaping gut microbiota, we used a randomized cohousing strategy and transgenic TLR-deficient mice. We have found that mice cohoused together by genotype exhibited limited changes over time in the composition of the gut microbiota. However, for mice randomized to cage, we report extensive changes in the gut microbiota, independent of TLR function, whereby the fecal microbiota of TLR-deficient mice converges with that of wild-type mice. TLR5-deficient mice in these experiments exhibit greater susceptibility to comparative changes in the microbiota than other TLR-deficient mice and wild-type mice. Our work has broad implications for the study of innate immunity and host-microbiota interactions. Given the profound impact that gut dysbiosis may have on immunity, this report highlights the potential impact of cohousing on the gut microbiota and indices of inflammation as outcomes in biological models of infectious or inflammatory disease.
- Published
- 2021
14. The Arg753Gln Polymorphism of Toll-Like Receptor 2 Has a Lower Occurrence in Patients with Syphilis, Suggesting Its Protective Effect in Czech and Slovak Individuals
- Author
-
Klára Janečková, Petra Pospíšilová, Jana Musilová, David Šmajs, Vladana Woznicová, Linda Grillová, Ivana Kuklová, and Hana Zákoucká
- Subjects
Adult ,Male ,0301 basic medicine ,Nonsynonymous substitution ,Slovakia ,Adolescent ,Genotype ,Immunology ,Polymorphism, Single Nucleotide ,Microbiology ,Proinflammatory cytokine ,Young Adult ,03 medical and health sciences ,0302 clinical medicine ,Gene Frequency ,medicine ,Humans ,Genetic Predisposition to Disease ,Syphilis ,030212 general & internal medicine ,Receptor ,Alleles ,Czech Republic ,Toll-like receptor ,Treponema ,biology ,Case-control study ,Middle Aged ,Host-Associated Microbial Communities ,medicine.disease ,biology.organism_classification ,Toll-Like Receptor 2 ,3. Good health ,TLR2 ,030104 developmental biology ,Infectious Diseases ,Amino Acid Substitution ,Case-Control Studies ,Parasitology - Abstract
Syphilis is a bacterial infection caused by Treponema pallidum subsp. pallidum. Infection with T. pallidum subsp. pallidum and its dissemination lead to the synthesis of proinflammatory cytokines triggered by the interaction of bacterial lipoproteins with Toll-like receptor 2 (TLR2). TLR2 contains several nonsynonymous single-nucleotide polymorphisms that may impact the activation of its signaling cascade and alter the responsiveness to, or the course of, various infectious diseases, including those caused by pathogenic spirochetes. To investigate whether TLR2 polymorphism may influence susceptibility to syphilis, 221 healthy individuals with no history of syphilis (controls) and 137 patients diagnosed with syphilis (cases) were screened for the presence of the Arg753Gln polymorphism in the TLR2 gene (2258G→A; rs5743708). The Arg753Gln variant occurs at a significantly lower frequency in syphilis patients (4 of 137 [3%]) than in controls (24 of 221 [10.9%]). These data suggest that TLR2 Arg753Gln may protect from the development of syphilis due to reduced signaling.
- Published
- 2020
- Full Text
- View/download PDF
15. Interactions between amphibians' symbiotic bacteria cause the production of emergent anti-fungal metabolites.
- Author
-
Loudon, Andrew H., Holland, Jessica A., Umile, Thomas P., Burzynski, Elizabeth A., Minbiole, Kevin P. C., and Harris, Reid N.
- Subjects
BATRACHOCHYTRIUM dendrobatidis ,METABOLITE analysis ,AMPHIBIAN behavior ,ANTIFUNGAL agents ,MICROBIAL biotechnology - Abstract
Amphibians possess beneficial skin bacteria that protect against the disease chytridiomycosis by producing secondary metabolites that inhibit the pathogen Batrachochytrium dendrobatidis (Bd). Metabolite production may be a mechanism of competition between bacterial species that results in host protection as a by-product. We expect that some co-cultures of bacterial species or strains will result in greater Bd inhibition than mono-cultures. To test this, we cultured four bacterial isolates (Bacillus sp., Janthinobacterium sp., Pseudomonas sp. and Chitinophaga arvensicola) from red-backed salamanders (Plethodon cinereus) and cultured isolates both alone and together to collect their cell-free supernatants (CFS). We challenged Bd with CFSs from four bacterial species in varying combinations. This resulted in three experimental treatments: (1) CFSs of single isolates; (2) combined CFSs of two isolates; and (3) CFSs from co-cultures. Pair-wise combinations of four bacterial isolates CFSs were assayed against Bd and revealed additive Bd inhibition in 42.2% of trials, synergistic inhibition in 42.2% and no effect in 16.6% of trials. When bacteria isolates were grown in co-cultures, complete Bd inhibition was generally observed, and synergistic inhibition occurred in four out of six trials. A metabolite profile of the most potent co-culture, Bacillus sp. and Chitinophaga arvensicola, was determined with LC-MS and compared with the profiles of each isolate in mono-culture. Emergent metabolites appearing in the co-culture were inhibitory to Bd, and the most potent inhibitor was identified as tryptophol. Thus mono-cultures of bacteria cultured from red-backed salamanders interacted synergistically and additively to inhibit Bd, and such bacteria produced emergent metabolites when cultured together, with even greater pathogen inhibition. Knowledge of how bacterial species interact to inhibit Bd can be used to select probiotics to provide amphibians with protection against Bd. [ABSTRACT FROM AUTHOR]
- Published
- 2014
- Full Text
- View/download PDF
16. Impact of Diabetes on the Gut and Salivary IgA Microbiomes
- Author
-
Eric L. Brown, David Aguilar, Craig L. Hanis, Joel M. Sederstrom, Sarah M. Gunter, Joseph F. Petrosino, Heather T. Essigmann, Herbert L. DuPont, Goo Jun, Noah W. Palm, Kristi L. Hoffman, and William B. Perkison
- Subjects
0301 basic medicine ,Adult ,Male ,Saliva ,Immunology ,Context (language use) ,Type 2 diabetes ,Disease ,Biology ,Microbiology ,03 medical and health sciences ,Feces ,0302 clinical medicine ,Immune system ,RNA, Ribosomal, 16S ,medicine ,Humans ,Microbiome ,Prediabetes ,Bacteria ,Discriminant Analysis ,030206 dentistry ,Middle Aged ,medicine.disease ,Host-Associated Microbial Communities ,Classification ,Gastrointestinal Microbiome ,030104 developmental biology ,Infectious Diseases ,Diabetes Mellitus, Type 2 ,Immunoglobulin A, Secretory ,Dysbiosis ,Parasitology ,Female - Abstract
Mucosal surfaces like those present in the lung, gut, and mouth interface with distinct external environments. These mucosal gateways are not only portals of entry for potential pathogens but also homes to microbial communities that impact host health. Secretory immunoglobulin A (SIgA) is the single most abundant acquired immune component secreted onto mucosal surfaces and, via the process of immune exclusion, shapes the architecture of these microbiomes. Not all microorganisms at mucosal surfaces are targeted by SIgA; therefore, a better understanding of the SIgA-coated fraction may identify the microbial constituents that stimulate host immune responses in the context of health and disease. Chronic diseases like type 2 diabetes are associated with altered microbial communities (dysbiosis) that in turn affect immune-mediated homeostasis. 16S rRNA gene sequencing of SIgA-coated/uncoated bacteria (IgA-Biome) was conducted on stool and saliva samples of normoglycemic participants and individuals with prediabetes or diabetes (n = 8/group). These analyses demonstrated shifts in relative abundance in the IgA-Biome profiles between normoglycemic, prediabetic, or diabetic samples distinct from that of the overall microbiome. Differences in IgA-Biome alpha diversity were apparent for both stool and saliva, while overarching bacterial community differences (beta diversity) were also observed in saliva. These data suggest that IgA-Biome analyses can be used to identify novel microbial signatures associated with diabetes and support the need for further studies exploring these communities. Ultimately, an understanding of the IgA-Biome may promote the development of novel strategies to restructure the microbiome as a means of preventing or treating diseases associated with dysbiosis at mucosal surfaces.
- Published
- 2020
17. Presence of Gastric Pepsinogen in the Trachea Is Associated with Altered Inflammation and Microbial Composition
- Author
-
Nicodemus Tedla, Nadeem O. Kaakoush, Usha Krishnan, Harveen Singh, and Steven T. Leach
- Subjects
Male ,0301 basic medicine ,Adolescent ,medicine.medical_treatment ,Interleukin-1beta ,030106 microbiology ,Immunology ,Prevotella ,Inflammation ,Microbiology ,Cohort Studies ,03 medical and health sciences ,Pepsin ,Pepsinogen A ,RNA, Ribosomal, 16S ,medicine ,Humans ,Microbiome ,Internal transcribed spacer ,Respiratory system ,Child ,Candida ,biology ,Interleukin-8 ,Respiratory Aspiration ,Infant ,Host-Associated Microbial Communities ,biology.organism_classification ,16S ribosomal RNA ,Gastrointestinal Microbiome ,Chemokine CXCL10 ,Trachea ,030104 developmental biology ,Infectious Diseases ,Cytokine ,Child, Preschool ,Gastroesophageal Reflux ,biology.protein ,Cytokines ,Female ,Parasitology ,medicine.symptom ,Interleukin-1 - Abstract
Gastroesophageal reflux is a common gastrointestinal issue that can lead to aspiration and contribute to respiratory problems. Little is known about how reflux can alter the respiratory microenvironment. We aimed to determine if the presence of gastric pepsinogen in the trachea was associated with changes in the microbial and inflammatory microenvironment. A pediatric cohort at high risk of reflux aspiration was prospectively recruited, and the tracheal microenvironment was examined. Pepsinogen A3 (PGA3) and cytokines were measured. The microbiome (bacterial and fungal) was profiled using 16S rRNA and internal transcribed spacer 2 (ITS2) amplicon sequencing. Increased bacterial richness and an altered composition driven by an enrichment of Prevotella correlated with high PGA3 levels. Fungal richness increased with PGA3, with higher Candida relative abundances observed in a subset of samples with high PGA3 levels. Source tracking of tracheal microbial taxa against taxa from matched oral and gastric samples revealed a significantly greater contribution of oral than of gastric taxa with higher PGA3 levels. Tracheal cytokines were differentially produced when stratified according to PGA3, with higher levels of interleukin-1 (IL-1)-related cytokines and IL-8 being associated with high PGA3 levels. Network analysis across cytokine and microbiome measures identified relationships between IL-1-related proteins and microbial taxa, with the presence of respiratory issues associated with higher levels of IL-1β, IP-10, and Prevotella. In conclusion, PGA3 levels in the trachea are correlated with increases in specific microbial taxa and inflammatory molecules, with an increase in oral microbes with increasing PGA3.
- Published
- 2020
- Full Text
- View/download PDF
18. A Role of Epithelial Cells and Virulence Factors in Biofilm Formation by Streptococcus pyogenes In Vitro
- Author
-
Yashuan Chao, Maria Baumgarten, Anders Håkansson, Feiruz Alamiri, and Kristian Riesbeck
- Subjects
Keratinocytes ,Cell type ,Streptococcus pyogenes ,Virulence Factors ,Immunology ,Virulence ,Biology ,medicine.disease_cause ,Serogroup ,Microbiology ,Extracellular matrix ,Drug Resistance, Bacterial ,medicine ,Biomass ,Bacterial Capsules ,Antigens, Bacterial ,Biofilm ,Epithelial Cells ,biochemical phenomena, metabolism, and nutrition ,Host-Associated Microbial Communities ,Cysteine protease ,In vitro ,Anti-Bacterial Agents ,Extracellular Matrix ,Infectious Diseases ,Biofilms ,Parasitology ,Streptolysin ,Carrier Proteins ,Bacterial Outer Membrane Proteins - Abstract
Biofilm formation by Streptococcus pyogenes (group A streptococcus [GAS]) in model systems mimicking the respiratory tract is poorly documented. Most studies have been conducted on abiotic surfaces, which poorly represent human tissues. We have previously shown that GAS forms mature and antibiotic-resistant biofilms on physiologically relevant epithelial cells. However, the roles of the substratum, extracellular matrix (ECM) components, and GAS virulence factors in biofilm formation and structure are unclear. In this study, biofilm formation was measured on respiratory epithelial cells and keratinocytes by determining biomass and antibiotic resistance, and biofilm morphology was visualized using scanning electron microscopy. All GAS isolates tested formed biofilms that had similar, albeit not identical, biomass and antibiotic resistance for both cell types. Interestingly, functionally mature biofilms formed more rapidly on keratinocytes but were structurally denser and coated with more ECM on respiratory epithelial cells. The ECM was crucial for biofilm integrity, as protein- and DNA-degrading enzymes induced bacterial release from biofilms. Abiotic surfaces supported biofilm formation, but these biofilms were structurally less dense and organized. No major role for M protein, capsule, or streptolysin O was observed in biofilm formation on epithelial cells, although some morphological differences were detected. NAD-glycohydrolase was required for optimal biofilm formation, whereas streptolysin S and cysteine protease SpeB impaired this process. Finally, no correlation was found between cell adherence or autoaggregation and GAS biofilm formation. Combined, these results provide a better understanding of the role of biofilm formation in GAS pathogenesis and can potentially provide novel targets for future treatments against GAS infections.
- Published
- 2020
19. Slipped-Strand Mispairing in the Gene Encoding Sialidase NanH3 in
- Author
-
Shakya P, Kurukulasuriya, Mo H, Patterson, and Janet E, Hill
- Subjects
Gardnerella ,Phenotype ,Genotype ,Genes, Bacterial ,Genetic Code ,Genetic Variation ,Humans ,Neuraminidase ,Female ,Vaginosis, Bacterial ,Host-Associated Microbial Communities ,Sequence Analysis - Abstract
Cell wall proteins with sialidase activity are involved in carbohydrate assimilation, adhesion to mucosal surfaces, and biofilm formation. Gardnerella spp. inhabit the human vaginal microbiome and encode up to three sialidase enzymes, two of which are suspected to be cell wall associated. Here, we demonstrate that the gene encoding extracellular sialidase NanH3 is found almost exclusively in Gardnerella piotii and the closely related species Gardnerella genome sp. 3, and its presence correlates with a sialidase-positive phenotype in a collection of 112 Gardnerella isolates. The nanH3 gene sequence includes a homopolymeric repeat of cytosines that varies in length within cell populations, indicating that this gene is subject to slipped-strand mispairing, a mechanism of phase variation in bacteria. Variation in the length of the homopolymer sequence results in production of either the full-length sialidase protein or truncated peptides lacking the sialidase domain due to introduction of reading-frame shifts and premature stop codons. Phase variation in NanH3 may be involved in immune evasion or modulation of adhesion to host epithelial cells and formation of biofilms characteristic of the vaginal dysbiosis known as bacterial vaginosis.
- Published
- 2020
20. Chlamydia Deficient in Plasmid-Encoded pGP3 Is Prevented from Spreading to Large Intestine
- Author
-
Tianjun Jia, Jie Wang, Zhi Huo, Ying Xu, Conghui He, and Guangming Zhong
- Subjects
Gastrointestinal tract ,Chlamydia ,Inoculation ,Immunology ,Biology ,Host-Associated Microbial Communities ,urologic and male genital diseases ,medicine.disease ,Microbiology ,Phenotype ,female genital diseases and pregnancy complications ,Small intestine ,Infectious Diseases ,medicine.anatomical_structure ,Plasmid ,medicine ,Parasitology ,Large intestine ,Colonization - Abstract
The cryptic plasmid pCM is critical for chlamydial colonization in the gastrointestinal tract. Nevertheless, orally inoculated plasmid-free Chlamydia sp. was still able to colonize the gut. Surprisingly, orally inoculated Chlamydia sp. deficient in only plasmid-encoded pGP3 was no longer able to colonize the gut. A comparison of live organism recoveries from individual gastrointestinal tissues revealed that pGP3-deficient Chlamydia sp. survived significantly better than plasmid-free Chlamydia sp. in small intestinal tissues. However, the small intestinal pGP3-deficient Chlamydia sp. failed to reach the large intestine, explaining the lack of live pGP3-deficient Chlamydia sp. in rectal swabs following an oral inoculation. Interestingly, pGP3-deficient Chlamydia sp. was able to colonize the colon following an intracolon inoculation, suggesting that pGP3-deficient Chlamydia sp. might be prevented from spreading from the small intestine to the large intestine. This hypothesis is supported by the finding that following an intrajejunal inoculation that bypasses the gastric barrier, pGP3-deficient Chlamydia sp. still failed to reach the large intestine, although similarly inoculated plasmid-free Chlamydia sp. was able to do so. Interestingly, when both types of organisms were intrajejunally coinoculated into the same mouse small intestine, plasmid-free Chlamydia sp. was no longer able to spread to the large intestine, suggesting that pGP3-deficient Chlamydia sp. might be able to activate an intestinal resistance for regulating Chlamydia sp. spreading. Thus, the current study has not only provided evidence for reconciling a previously identified conflicting phenotype but also revealed a potential intestinal resistance to chlamydial spreading. Efforts are under way to further define the mechanism of the putative intestinal resistance.
- Published
- 2020
- Full Text
- View/download PDF
21. Cooperativity between Stenotrophomonas maltophilia and Pseudomonas aeruginosa during Polymicrobial Airway Infections
- Author
-
W. Edward Swords, Trenton R. Schoeb, and Melissa S. McDaniel
- Subjects
0301 basic medicine ,congenital, hereditary, and neonatal diseases and abnormalities ,Stenotrophomonas maltophilia ,030106 microbiology ,Immunology ,Population ,Virulence ,Biology ,medicine.disease_cause ,Microbiology ,Cystic fibrosis ,Mice ,03 medical and health sciences ,Pneumonia, Bacterial ,medicine ,Animals ,education ,Lung ,education.field_of_study ,Coinfection ,Pseudomonas aeruginosa ,Body Weight ,Pseudomonas ,biochemical phenomena, metabolism, and nutrition ,Host-Associated Microbial Communities ,bacterial infections and mycoses ,medicine.disease ,biology.organism_classification ,Bacterial Load ,Immunity, Innate ,respiratory tract diseases ,Multiple drug resistance ,Disease Models, Animal ,030104 developmental biology ,Infectious Diseases ,Microbial Interactions ,bacteria ,Parasitology ,Stenotrophomonas ,Gram-Negative Bacterial Infections ,Bronchoalveolar Lavage Fluid - Abstract
Stenotrophomonas maltophilia is a Gram-negative bacterium found ubiquitously in the environment that has historically been regarded as nonpathogenic. S. maltophilia is increasingly observed in patient sputa in cystic fibrosis (CF), and while existing epidemiology indicates that patients with S. maltophilia have poorer diagnoses, its clinical significance remains unclear. Moreover, as multidrug resistance is common among S. maltophilia isolates, treatment options for these infections may be limited. Here, we investigated the pathogenicity of S. maltophilia alone and during polymicrobial infection with Pseudomonas aeruginosa. Colonization, persistence, and virulence of S. maltophilia were assessed in experimental respiratory infections of mice. The results of this study indicate that S. maltophilia transiently colonizes the lung accompanied by significant weight loss and immune cell infiltration and the expression of early inflammatory markers, including interleukin 6 (IL-6), IL-1α, and tumor necrosis factor alpha (TNF-α). Importantly, polymicrobial infection with P. aeruginosa elicited significantly higher S. maltophilia counts in bronchoalveolar lavages and lung tissue homogenates. This increase in bacterial load was directly correlated with the density of the P. aeruginosa population and required viable P. aeruginosa bacteria. Microscopic analysis of biofilms formed in vitro revealed that S. maltophilia formed well-integrated biofilms with P. aeruginosa, and these organisms colocalize in the lung during dual-species infection. Based on these results, we conclude that active cellular processes by P. aeruginosa afford a significant benefit to S. maltophilia during polymicrobial infections. Furthermore, these results indicate that S. maltophilia may have clinical significance in respiratory infections.
- Published
- 2020
- Full Text
- View/download PDF
22. Genetic Variation in Toll-Like Receptor 5 and Colonization with Flagellated Bacterial Vaginosis-Associated Bacteria
- Author
-
Christina M. Kohler, Kristina Garcia, Andrew Wiser, Thomas R. Hawn, Jennifer E. Balkus, Anna Frishman, Jeanne M. Marrazzo, Antoinette Oot, Erin J. dela Cruz, Laura K. Sycuro, Tina L. Fiedler, Matthew M Munch, David N. Fredricks, Congzhou Liu, Sujatha Srinivasan, Jacqueline M Wallis, Jonathan L. Golob, and Junhui Wang
- Subjects
0301 basic medicine ,Adult ,Washington ,Adolescent ,Genotype ,030106 microbiology ,Immunology ,microbiome ,Microbiology ,Cohort Studies ,03 medical and health sciences ,Young Adult ,medicine ,Humans ,Colonization ,Mobiluncus ,biology ,Genetic Variation ,Vaginosis, Bacterial ,Middle Aged ,biology.organism_classification ,Mobiluncus mulieris ,medicine.disease ,Host-Associated Microbial Communities ,Toll-like receptors ,Mobiluncus curtisii ,Toll-Like Receptor 5 ,030104 developmental biology ,Infectious Diseases ,TLR5 ,Genes, Bacterial ,Vagina ,biology.protein ,Parasitology ,Female ,flagella ,Bacterial vaginosis ,Bacteria ,Flagellin ,bacterial vaginosis - Abstract
Bacterial vaginosis (BV) is a vaginal dysbiotic condition linked to negative gynecological and reproductive sequelae. Flagellated bacteria have been identified in women with BV, including Mobiluncus spp. and BV-associated bacterium-1 (BVAB1), an uncultivated, putatively flagellated species., Bacterial vaginosis (BV) is a vaginal dysbiotic condition linked to negative gynecological and reproductive sequelae. Flagellated bacteria have been identified in women with BV, including Mobiluncus spp. and BV-associated bacterium-1 (BVAB1), an uncultivated, putatively flagellated species. The host response to flagellin mediated through Toll-like receptor 5 (TLR5) has not been explored in BV. Using independent discovery and validation cohorts, we examined the hypothesis that TLR5 deficiency—defined by a dominant negative stop codon polymorphism, rs5744168—is associated with an increased risk for BV and increased colonization with flagellated bacteria associated with BV (BVAB1, Mobiluncus curtisii, and Mobiluncus mulieris). TLR5 deficiency was not associated with BV status, and TLR5-deficient women had decreased colonization with BVAB1 in both cohorts. We stimulated HEK-hTLR5-overexpressing NF-κB reporter cells with whole, heat-killed M. mulieris or M. curtisii and with partially purified flagellin from these species; as BVAB1 is uncultivated, we used cervicovaginal lavage (CVL) fluid supernatant from women colonized with BVAB1 for stimulation. While heat-killed M. mulieris and CVL fluid from women colonized with BVAB1 stimulate a TLR5-mediated response, heat-killed M. curtisii did not. In contrast, partially purified flagellin from both Mobiluncus species stimulated a TLR5-mediated response in vitro. We observed no correlation between vaginal interleukin 8 (IL-8) and flagellated BVAB concentrations among TLR5-sufficient women. Interspecies variation in accessibility of flagellin recognition domains may be responsible for these observations, as reflected in the potentially novel flagellin products encoded by Mobiluncus species versus those encoded by BVAB1.
- Published
- 2020
23. The Cryptic Plasmid Improves Chlamydia Fitness in Different Regions of the Gastrointestinal Tract
- Author
-
Jingyue Ma, Conghui He, Ying Xu, Zhi Huo, Quanzhong Liu, Guangming Zhong, and Bernard P. Arulanandam
- Subjects
Chlamydia muridarum ,Immunology ,Colony Count, Microbial ,Microbiology ,Mice ,Plasmid ,medicine ,Animals ,Large intestine ,Colonization ,Gastrointestinal tract ,Chlamydia ,biology ,Inoculation ,Chlamydia Infections ,biology.organism_classification ,medicine.disease ,Host-Associated Microbial Communities ,Small intestine ,Gastrointestinal Tract ,Mice, Inbred C57BL ,Infectious Diseases ,medicine.anatomical_structure ,Organ Specificity ,Host-Pathogen Interactions ,Parasitology ,Female ,Genome, Bacterial ,Plasmids - Abstract
The cryptic plasmid is important for chlamydial colonization in the gastrointestinal tract. We used a combination of intragastric, intrajejunal, and intracolon inoculations to reveal the impact of the plasmid on chlamydial colonization in distinct regions of gastrointestinal tract. Following an intragastric inoculation, the plasmid significantly improved chlamydial colonization. At the tissue level, plasmid-positive Chlamydia produced infectious progenies throughout gastrointestinal tract. However, to our surprise, plasmid-deficient Chlamydia failed to produce infectious progenies in small intestine, although infectious progenies were eventually detected in large intestine, indicating a critical role of the plasmid in chlamydial differentiation into infectious particles in small intestine. The noninfectious status may represent persistent infection, since Chlamydia genomes proliferated in the same tissues. Following an intrajejunal inoculation that bypasses the gastric barrier, plasmid-deficient Chlamydia produced infectious progenies in small intestine but was 530-fold less infectious than plasmid-positive Chlamydia, suggesting that (i) the noninfectious status developed after intragastric inoculation might be induced by a combination of gastric and intestinal effectors and (ii) chlamydial colonization in small intestine was highly dependent on plasmid. Finally, following an intracolon inoculation, the dependence of chlamydial colonization on plasmid increased over time. Thus, we have demonstrated that the plasmid may be able to improve chlamydial fitness in different gut regions via different mechanisms, which has laid a foundation to further reveal the specific mechanisms.
- Published
- 2020
24. Dysregulation of Intestinal Microbiota Elicited by Food Allergy Induces IgA-Mediated Oral Dysbiosis
- Author
-
Mariko Kikuchi, Hideo Kataoka, Hirotaka Kuwata, Nobuo Okahashi, Hirobumi Morisaki, Takashi Takaki, Shoji Hironaka, Kenji Mishima, Shohei Matsui, Hitomi Matsushima, Junichi Tanaka, Yasubumi Maruoka, and Haruka Fukamachi
- Subjects
0301 basic medicine ,Allergy ,030106 microbiology ,Immunology ,Microbiology ,03 medical and health sciences ,Citrobacter ,Immune system ,Food allergy ,medicine ,Animals ,Immunologic Factors ,Mice, Inbred BALB C ,Mouth ,food allergy ,biology ,Microbiota ,digestive, oral, and skin physiology ,Epithelial Cells ,Citrobacter koseri ,Host-Associated Microbial Communities ,biology.organism_classification ,medicine.disease ,Immunoglobulin A ,Gastrointestinal Tract ,Interleukin 33 ,Disease Models, Animal ,Ovalbumin ,030104 developmental biology ,Infectious Diseases ,biology.protein ,Cytokines ,Dysbiosis ,Parasitology ,Food Hypersensitivity - Abstract
Food allergy is a life-threatening response to specific foods, and microbiota imbalance (dysbiosis) in gut is considered a cause of this disease. Meanwhile, the host immune response also plays an important role in the disease. Notably, interleukin 33 (IL-33) released from damaged or necrotic intestinal epithelial cells facilitates IL-2-producing CD4 helper T (Th2) responses. However, causal relationships between the gut and oral dysbiosis and food allergy remain unknown., Food allergy is a life-threatening response to specific foods, and microbiota imbalance (dysbiosis) in gut is considered a cause of this disease. Meanwhile, the host immune response also plays an important role in the disease. Notably, interleukin 33 (IL-33) released from damaged or necrotic intestinal epithelial cells facilitates IL-2-producing CD4 helper T (Th2) responses. However, causal relationships between the gut and oral dysbiosis and food allergy remain unknown. In this study, we analyzed effects of gut and oral dysbiosis on development of food allergy. A murine model of food allergy was established via ovalbumin (OVA) injection in BALB/c mice. Viable fecal bacteria were identified using matrix-assisted laser desorption ionization–time of flight mass spectrometry (MALDI-TOF MS). il33 expression in colon-26 mouse colon cells stimulated by isolated fecal bacteria was quantified by real-time PCR. Intestinal T cells from the mice were analyzed by flow cytometry. Salivary IgA levels were quantified by enzyme-linked immunosorbent assay (ELISA), and IgA-bound oral bacteria were detected by flow cytometry. Among fecal bacteria, the abundance of Citrobacter sp. increased in the feces of allergic mice and induced il33 expression in colon-26 cells. Orally administered Citrobacter koseri JCM1658 exacerbated systemic allergic symptoms and reduced intestinal Th17 cells. Salivary IgA and IgA-bound oral bacteria increased in the allergic mice. Based on the results described above, food allergy induced both gut and oral dysbiosis. Citrobacter sp. aggravated allergy symptoms by inducing IL-33 release from intestinal epithelial cells.
- Published
- 2019
- Full Text
- View/download PDF
25. The Host Immune System Facilitates Disseminated Staphylococcus aureus Disease Due to Phagocytic Attraction to Candida albicans during Coinfection
- Author
-
Hubertine M. E. Willems, Nina Scheres, Carolien S. Bode, Bastiaan P. Krom, Devon L. Allison, Mark E. Shirtliff, Preventive Dentistry, and Preventieve tandheelkunde (OII, ACTA)
- Subjects
Staphylococcus aureus ,Phagocyte ,Immunology ,medicine.disease_cause ,Microbiology ,Cell Line ,Mice ,Immune system ,Phagocytosis ,SDG 3 - Good Health and Well-being ,Candida albicans ,medicine ,Animals ,Innate immune system ,biology ,Coinfection ,Macrophages ,Candidiasis ,Staphylococcal Infections ,Host-Associated Microbial Communities ,biology.organism_classification ,medicine.disease ,Immunity, Innate ,Corpus albicans ,Bacterial adhesin ,Infectious Diseases ,medicine.anatomical_structure ,Parasitology - Abstract
Invasive Staphylococcus aureus infections account for 15 to 50% of fatal bloodstream infections annually. These disseminated infections often arise without a defined portal of entry into the host but cause high rates of mortality. The fungus Candida albicans and the Gram-positive bacterium S. aureus can form polymicrobial biofilms on epithelial tissue, facilitated by the C. albicans adhesin encoded by ALS3. While a bacterium-fungus interaction is required for systemic infection, the mechanism by which bacteria disseminate from the epithelium to internal organs is unclear. In this study, we show that highly immunogenic C. albicans hyphae attract phagocytic cells, which rapidly engulf adherent S. aureus and subsequently migrate to cervical lymph nodes. Following S. aureus-loaded phagocyte translocation from the mucosal surface, S. aureus produces systemic disease with accompanying morbidity and mortality. Our results suggest a novel role for the host in facilitating a bacterium-fungus infectious synergy, leading to disseminated staphylococcal disease.
- Published
- 2019
- Full Text
- View/download PDF
26. Evasion of Innate Lymphoid Cell-Regulated Gamma Interferon Responses by Chlamydia muridarum To Achieve Long-Lasting Colonization in Mouse Colon
- Author
-
John J. Koprivsek, Chenchen Song, Alexei V. Tumanov, Nu Zhang, Guangming Zhong, and Ying He
- Subjects
0301 basic medicine ,Chlamydia muridarum ,Colon ,Immunology ,Mutant ,C-C chemokine receptor type 6 ,Microbiology ,Natural killer cell ,03 medical and health sciences ,Interferon-gamma ,Mice ,0302 clinical medicine ,RAR-related orphan receptor gamma ,medicine ,Animals ,Colonization ,Lymphocytes ,Common gamma chain ,biology ,Innate lymphoid cell ,Chlamydia Infections ,biology.organism_classification ,Host-Associated Microbial Communities ,digestive system diseases ,Immunity, Innate ,Mice, Inbred C57BL ,030104 developmental biology ,Infectious Diseases ,medicine.anatomical_structure ,Parasitology ,030215 immunology - Abstract
Revealing the mechanisms by which bacteria establish long-lasting colonization in the gastrointestinal tract is an area of intensive investigation. The obligate intracellular bacterium Chlamydia is known to colonize mouse colon for long periods. A colonization-deficient mutant strain of this intracellular bacterium is able to regain long-lasting colonization in gamma interferon (IFN-γ) knockout mice following intracolon inoculation. We now report that mice deficient in conventional T lymphocytes or recombination-activating gene (Rag) failed to show rescue of mutant colonization. Nevertheless, antibody depletion of IFN-γ or genetic deletion of interleukin 2 (IL-2) receptor common gamma chain in Rag-deficient mice did rescue mutant colonization. These observations suggest that colonic IFN-γ, responsible for inhibiting the intracellular bacterial mutant, is produced by innate lymphoid cells (ILCs). Consistently, depletion of NK1.1(+) cells in Rag-deficient mice both prevented IFN-γ production and rescued mutant colonization. Furthermore, mice deficient in transcriptional factor RORγt, but not chemokine receptor CCR6, showed full rescue of the long-lasting colonization of the mutant, indicating a role for group 3-like ILCs. However, the inhibitory function of the responsible group 3-like ILCs was not dependent on the natural killer cell receptor (NCR1), since NCR1-deficient mice still inhibited mutant colonization. Consistently, mice deficient in the transcriptional factor T-bet only delayed the clearance of the bacterial mutant without fully rescuing the long-lasting colonization of the mutant. Thus, we have demonstrated that the obligate intracellular bacterium Chlamydia maintains its long-lasting colonization in the colon by evading IFN-γ from group 3-like ILCs.
- Published
- 2019
27. Resolution and Cooccurrence Patterns of Gardnerella leopoldii, G. swidsinskii, G. piotii, and G. vaginalis within the Vaginal Microbiome
- Author
-
Alain Albert, Arianne Albert, and Janet Hill
- Subjects
0301 basic medicine ,Canada ,030106 microbiology ,Immunology ,Biology ,medicine.disease_cause ,Microbiology ,Species description ,03 medical and health sciences ,Gardnerella ,Abundance (ecology) ,RNA, Ribosomal, 16S ,medicine ,Gardnerella vaginalis ,DNA Barcoding, Taxonomic ,Humans ,Microbiome ,Clade ,Relative species abundance ,Genetics ,Microbiota ,High-Throughput Nucleotide Sequencing ,Chaperonin 60 ,Vaginosis, Bacterial ,medicine.disease ,Host-Associated Microbial Communities ,030104 developmental biology ,Infectious Diseases ,Vagina ,Dysbiosis ,Parasitology ,Female - Abstract
Gardnerella vaginalis is a hallmark of vaginal dysbiosis, but it is found in the microbiomes of women with and without vaginal symptoms and those who do not have microbiologically defined dysbiosis. G. vaginalis encompasses diverse taxa differing in attributes that are potentially important for virulence, and there is evidence that clades or subgroups within the species are differentially associated with clinical outcomes. The G. vaginalis species description was recently emended, and three new species within the genus were defined (G. leopoldii, G. swidsinskii, and G. piotii). 16S rRNA sequences for the four Gardnerella species are all >98.5% identical, and no signature sequences differentiate them. We demonstrated that Gardnerella species can be resolved using partial chaperonin 60 (cpn60) sequences, with pairwise percent identities of 87.1 to 97.8% among the type strains. Pairwise cooccurrence patterns of Gardnerella spp. in the vaginal microbiomes of 413 reproductive aged Canadian women were investigated, and several significant cooccurrences of species were identified. Abundance of G. vaginalis and G. swidsinskii was associated with vaginal symptoms of abnormal odor and discharge. cpn60 barcode sequencing can provide a rapid assessment of the relative abundance of Gardnerella spp. in microbiome samples, providing a powerful method of elucidating associations between these diverse organisms and clinical outcomes. Researchers should consider using cpn60 instead of 16S RNA for better resolution of these important organisms.
- Published
- 2019
28. Production of a Functional Factor, p40, by Lactobacillus rhamnosus GG Is Promoted by Intestinal Epithelial Cell-Secreted Extracellular Vesicles
- Author
-
Sari Acra, Luyao Yang, Liping Liu, Gang Zhao, D. Brent Polk, Fang Yan, Hui Li, Richard M. Peek, and James N. Higginbotham
- Subjects
0301 basic medicine ,Lactobacillus GG ,Immunology ,Inflammation ,Microbiology ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Bacterial Proteins ,Lactobacillus rhamnosus ,Heat shock protein ,medicine ,Animals ,Secretion ,HSP90 Heat-Shock Proteins ,Intestinal Mucosa ,biology ,Lacticaseibacillus rhamnosus ,Secretory Vesicles ,Epithelial Cells ,Extracellular vesicle ,Host-Associated Microbial Communities ,biology.organism_classification ,Epithelium ,Cell biology ,Mice, Inbred C57BL ,030104 developmental biology ,Infectious Diseases ,Secretory protein ,medicine.anatomical_structure ,030220 oncology & carcinogenesis ,Parasitology ,medicine.symptom - Abstract
The symbiotic relationship between the gut microbiome and the host provides a nutrient-rich environment for gut microbes and has beneficial effects on host health. Although the composition of the gut microbiome is known to be influenced by both host genetics and environmental factors, host effects on the activities and functions of the gut microbial communities remain poorly understood. Intestinal epithelial cells exert front-line responses to gut microbes and contribute to maintaining a healthy intestinal homeostasis. Here, seeking to elucidate whether intestinal epithelial cells modulate Lactobacillus rhamnosus GG (LGG) functions, we examined the production of p40, an LGG-derived secretory protein that protects intestinal epithelial cells against inflammation. We found that growth medium conditioned with colonic epithelial cell-derived components promotes p40 protein synthesis and secretion by LGG and enhances LGG-stimulated protective responses in intestinal epithelial cells. Furthermore, when LGG was cultured with the colonic luminal contents from healthy mice, p40 production was upregulated but was attenuated with luminal contents from mice with intestinal inflammation. Importantly, the colonic epithelial cell-derived components potentiated LGG-produced p40 levels in a mouse model of colitis and enhanced LGG-mediated amelioration of intestinal inflammation in this model. Notably, we found that colonic epithelial cell-secreted extracellular vesicles participate in communicating with LGG and that heat shock protein 90 (HSP90) in these vesicles might mediate the promotion of p40 production. These results reveal a previously unrecognized mechanism by which the anti-inflammatory effect of LGG is reinforced by intestinal epithelial cells and thereby maintains intestinal health.
- Published
- 2019
- Full Text
- View/download PDF
29. Diversification and Evolution of Vancomycin-Resistant Enterococcus faecium during Intestinal Domination
- Author
-
Peter T. McKenney, Joao B. Xavier, Eric G. Pamer, Deepti Mathur, Krista Dubin, Ying Taur, Eric R. Littmann, Jonathan U. Peled, Marcel R.M. van den Brink, and Bradford P. Taylor
- Subjects
0301 basic medicine ,DNA, Bacterial ,medicine.drug_class ,030106 microbiology ,Immunology ,Population ,Antibiotics ,DNA Mutational Analysis ,Enterococcus faecium ,Biology ,bacterial evolution ,Microbiology ,Vancomycin-Resistant Enterococci ,03 medical and health sciences ,Feces ,Bloodstream infection ,Ampicillin ,RNA, Ribosomal, 16S ,medicine ,Animals ,Humans ,Longitudinal Studies ,Spotlight ,education ,Gram-Positive Bacterial Infections ,Vancomycin resistant Enterococcus faecium ,education.field_of_study ,Marrow transplantation ,Genetic Variation ,Patient data ,biochemical phenomena, metabolism, and nutrition ,biology.organism_classification ,bacterial infections and mycoses ,Host-Associated Microbial Communities ,Editor's Pick ,Biological Evolution ,3. Good health ,030104 developmental biology ,Infectious Diseases ,Parasitology ,medicine.drug - Abstract
Vancomycin-resistant Enterococcus faecium (VRE) is a leading cause of hospital-acquired infections. This is particularly true in immunocompromised patients, where the damage to the microbiota caused by antibiotics can lead to VRE domination of the intestine, increasing a patient’s risk for bloodstream infection., Vancomycin-resistant Enterococcus faecium (VRE) is a leading cause of hospital-acquired infections. This is particularly true in immunocompromised patients, where the damage to the microbiota caused by antibiotics can lead to VRE domination of the intestine, increasing a patient’s risk for bloodstream infection. In previous studies we observed that the intestinal domination by VRE of patients hospitalized to receive allogeneic bone marrow transplantation can persist for weeks, but little is known about subspecies diversification and evolution during prolonged domination. Here we combined a longitudinal analysis of patient data and in vivo experiments to reveal previously unappreciated subspecies dynamics during VRE domination that appeared to be stable from 16S rRNA microbiota analyses. Whole-genome sequencing of isolates obtained from sequential stool samples provided by VRE-dominated patients revealed an unanticipated level of VRE population complexity that evolved over time. In experiments with ampicillin-treated mice colonized with a single CFU, VRE rapidly diversified and expanded into distinct lineages that competed for dominance. Mathematical modeling shows that in vivo evolution follows mostly a parabolic fitness landscape, where each new mutation provides diminishing returns and, in the setting of continuous ampicillin treatment, reveals a fitness advantage for mutations in penicillin-binding protein 5 (pbp5) that increase resistance to ampicillin. Our results reveal the rapid diversification of host-colonizing VRE populations, with implications for epidemiologic tracking of in-hospital VRE transmission and susceptibility to antibiotic treatment.
- Published
- 2019
30. Microbiome Profiles of Ligature-Induced Periodontitis in Nonhuman Primates across the Life Span
- Author
-
Jeffrey L. Ebersole, Octavio A. Gonzalez, Sreenatha Kirakodu, Janis Gonzalez Martinez, and Jin Chen
- Subjects
0301 basic medicine ,Male ,medicine.medical_treatment ,Immunology ,Physiology ,Disease ,Biology ,Microbiology ,03 medical and health sciences ,0302 clinical medicine ,medicine ,Animals ,Microbiome ,Ligature ,Periodontitis ,Phylogeny ,Mouth ,Life span ,Bacteria ,Microbiota ,Age Factors ,Primate Diseases ,Bacteroidetes ,Fusobacteria ,030206 dentistry ,biology.organism_classification ,medicine.disease ,Host-Associated Microbial Communities ,Macaca mulatta ,030104 developmental biology ,Infectious Diseases ,Parasitology ,Female ,Oral Microbiome - Abstract
This investigation compared the microbiomes colonizing teeth during the initiation, progression, and resolution of periodontitis in nonhuman primates (Macaca mulatta) at different ages. Subgingival plaque samples were collected at baseline; 0.5, 1, and 3 months following ligature-induced periodontitis; and following naturally occurring disease resolution at 5 months. Samples were analyzed using 16S amplicon sequencing to identify bacterial profiles across age groups: young (
- Published
- 2019
31. Bacteriophage Resistance Alters Antibiotic-Mediated Intestinal Expansion of Enterococci
- Author
-
Cydney N. Johnson, Kelli L. Palmer, Anushila Chatterjee, Phat Luong, Breck A. Duerkop, Paul E. Carlson, Alyxandria M. Schubert, Karthik Hullahalli, and Sara W. McBride
- Subjects
Male ,Phage therapy ,medicine.drug_class ,medicine.medical_treatment ,viruses ,Immunology ,Antibiotics ,Enterococcus faecium ,Microbial Sensitivity Tests ,Biology ,Microbiology ,Enterococcus faecalis ,Bacteriophage ,Mice ,Antibiotic resistance ,Vancomycin ,medicine ,Animals ,Humans ,Bacteriophages ,Gram-Positive Bacterial Infections ,medicine.disease ,biology.organism_classification ,Host-Associated Microbial Communities ,Anti-Bacterial Agents ,Biological Therapy ,Intestines ,Mice, Inbred C57BL ,Infectious Diseases ,Lytic cycle ,Parasitology ,Female ,Bacterial virus ,Dysbiosis - Abstract
Enterococcus faecalis is a human intestinal pathobiont with intrinsic and acquired resistance to many antibiotics, including vancomycin. Nature provides a diverse and virtually untapped repertoire of bacterial viruses, or bacteriophages (phages), that could be harnessed to combat multidrug-resistant enterococcal infections. Bacterial phage resistance represents a potential barrier to the implementation of phage therapy, emphasizing the importance of investigating the molecular mechanisms underlying the emergence of phage resistance. Using a cohort of 19 environmental lytic phages with tropism against E. faecalis, we found that these phages require the enterococcal polysaccharide antigen (Epa) for productive infection. Epa is a surface-exposed heteroglycan synthesized by enzymes encoded by both conserved and strain-specific genes. We discovered that exposure to phage selective pressure favors mutation in nonconserved epa genes both in culture and in a mouse model of intestinal colonization. Despite gaining phage resistance, epa mutant strains exhibited a loss of resistance to cell wall-targeting antibiotics. Finally, we show that an E. faecalisepa mutant strain is deficient in intestinal colonization, cannot expand its population upon antibiotic-driven intestinal dysbiosis, and fails to be efficiently transmitted to juvenile mice following birth. This study demonstrates that phage therapy could be used in combination with antibiotics to target enterococci within a dysbiotic microbiota. Enterococci that evade phage therapy by developing resistance may be less fit at colonizing the intestine and sensitized to vancomycin, preventing their overgrowth during antibiotic treatment.
- Published
- 2019
32. Multi-omics Analyses Reveal Synergistic Carbohydrate Metabolism in Streptococcus mutans-Candida albicans Mixed-Species Biofilms
- Author
-
Y. Liu, Hyun Koo, T. K. Lim, Yew Mun Lee, Tong Cao, Qingsong Lin, Chaminda Jayampath Seneviratne, Kassapa Ellepola, and Thuyen Truong
- Subjects
0301 basic medicine ,Proteomics ,Immunology ,Quantitative proteomics ,Carbohydrate metabolism ,Dental Caries ,Microbiology ,Streptococcus mutans ,03 medical and health sciences ,0302 clinical medicine ,Bacterial Proteins ,Cell Wall ,Gene Expression Regulation, Fungal ,Candida albicans ,Humans ,Child ,Symbiosis ,Glucans ,biology ,Cell morphogenesis ,Biofilm ,030206 dentistry ,Gene Expression Regulation, Bacterial ,biology.organism_classification ,Host-Associated Microbial Communities ,Corpus albicans ,Coculture Techniques ,omics ,stomatognathic diseases ,030104 developmental biology ,Infectious Diseases ,Glucosyltransferases ,Biofilms ,biology.protein ,Exoenzyme ,Carbohydrate Metabolism ,mixed-species biofilms ,Parasitology ,early-childhood caries ,Transcriptome - Abstract
Candida albicans, a major opportunistic fungal pathogen, is frequently found together with Streptococcus mutans in dental biofilms associated with severe childhood caries (tooth decay), a prevalent pediatric oral disease. However, the impact of this cross-kingdom relationship on C. albicans remains largely uncharacterized. Here, we employed a novel quantitative proteomics approach in conjunction with transcriptomic profiling to unravel molecular pathways of C. albicans when cocultured with S. mutans in mixed biofilms., Candida albicans, a major opportunistic fungal pathogen, is frequently found together with Streptococcus mutans in dental biofilms associated with severe childhood caries (tooth decay), a prevalent pediatric oral disease. However, the impact of this cross-kingdom relationship on C. albicans remains largely uncharacterized. Here, we employed a novel quantitative proteomics approach in conjunction with transcriptomic profiling to unravel molecular pathways of C. albicans when cocultured with S. mutans in mixed biofilms. RNA sequencing and iTRAQ (isobaric tags for relative and absolute quantitation)-based quantitative proteomics revealed that C. albicans genes and proteins associated with carbohydrate metabolism were significantly enhanced, including sugar transport, aerobic respiration, pyruvate breakdown, and the glyoxylate cycle. Other C. albicans genes and proteins directly and indirectly related to cell morphogenesis and cell wall components such as mannan and glucan were also upregulated, indicating enhanced fungal activity in mixed-species biofilm. Further analyses revealed that S. mutans-derived exoenzyme glucosyltransferase B (GtfB), which binds to the fungal cell surface to promote coadhesion, can break down sucrose into glucose and fructose that can be readily metabolized by C. albicans, enhancing growth and acid production. Altogether, we identified key pathways used by C. albicans in the mixed biofilm, indicating an active fungal role in the sugar metabolism and environmental acidification (key virulence traits associated with caries onset) when interacting with S. mutans, and a new cross-feeding mechanism mediated by GtfB that enhances C. albicans carbohydrate utilization. In addition, we demonstrate that comprehensive transcriptomics and quantitative proteomics can be powerful tools to study microbial contributions which remain underexplored in cross-kingdom biofilms.
- Published
- 2019
33. Antibiotic-induced shifts in fecal microbiota density and composition during hematopoietic stem cell transplantation
- Author
-
Ying Taur, Rebecca A. Carter, Bradford P. Taylor, Sejal Morjaria, Jonas Schluter, Emily Fontana, Joao B. Xavier, Marcel R.M. van den Brink, Eric R. Littmann, and Jonathan U. Peled
- Subjects
0301 basic medicine ,Male ,medicine.medical_treatment ,Antibiotics ,microbiome ,Hematopoietic stem cell transplantation ,antibiotics ,Feces ,0302 clinical medicine ,RNA, Ribosomal, 16S ,Spotlight ,skin and connective tissue diseases ,0303 health sciences ,Microbiota ,Hematopoietic Stem Cell Transplantation ,systems biology ,Middle Aged ,Host-Associated Microbial Communities ,Anti-Bacterial Agents ,3. Good health ,Infectious Diseases ,surgical procedures, operative ,Female ,Anaerobic bacteria ,Stem cell ,Adult ,medicine.drug_class ,Immunology ,Biology ,Microbiology ,03 medical and health sciences ,Immune system ,medicine ,Humans ,Microbiome ,hematopoietic cell transplantation ,Aged ,030304 developmental biology ,Bacteria ,commensal anaerobes ,030306 microbiology ,Obligate anaerobe ,biology.organism_classification ,Gastrointestinal Microbiome ,Transplantation ,030104 developmental biology ,Parasitology ,sense organs ,030215 immunology - Abstract
Dramatic microbiota changes and loss of commensal anaerobic bacteria are associated with adverse outcomes in hematopoietic cell transplantation (HCT) recipients. In this study, we demonstrate these dynamic changes at high resolution through daily stool sampling and assess the impact of individual antibiotics on those changes., Dramatic microbiota changes and loss of commensal anaerobic bacteria are associated with adverse outcomes in hematopoietic cell transplantation (HCT) recipients. In this study, we demonstrate these dynamic changes at high resolution through daily stool sampling and assess the impact of individual antibiotics on those changes. We collected 272 longitudinal stool samples (with mostly daily frequency) from 18 patients undergoing HCT and determined their composition by multiparallel 16S rRNA gene sequencing as well as the density of bacteria in stool by quantitative PCR (qPCR). We calculated microbiota volatility to quantify rapid shifts and developed a new dynamic systems inference method to assess the specific impact of antibiotics. The greatest shifts in microbiota composition occurred between stem cell infusion and reconstitution of healthy immune cells. Piperacillin-tazobactam caused the most severe declines among obligate anaerobes. Our approach of daily sampling, bacterial density determination, and dynamic systems modeling allowed us to infer the independent effects of specific antibiotics on the microbiota of HCT patients.
- Published
- 2019
- Full Text
- View/download PDF
34. Interference with Pseudomonas aeruginosa Quorum Sensing and Virulence by the Mycobacterial
- Author
-
Franziska S, Birmes, Ruth, Säring, Miriam C, Hauke, Niklas H, Ritzmann, Steffen L, Drees, Jens, Daniel, Janina, Treffon, Eva, Liebau, Barbara C, Kahl, and Susanne, Fetzner
- Subjects
Mycobacterium abscessus ,Virulence ,Cell Survival ,Virulence Factors ,Quorum Sensing ,Gene Expression Regulation, Bacterial ,Quinolones ,bacterial infections and mycoses ,Host-Associated Microbial Communities ,Coculture Techniques ,Dioxygenases ,A549 Cells ,Antibiosis ,Pseudomonas aeruginosa ,Escherichia coli ,Pyocyanine ,bacteria ,Animals ,Humans ,Caenorhabditis elegans ,Carboxylic Ester Hydrolases ,Oligopeptides - Abstract
The nosocomial pathogen Pseudomonas aeruginosa regulates its virulence via a complex quorum sensing network, which, besides N-acylhomoserine lactones, includes the alkylquinolone signal molecules 2-heptyl-3-hydroxy-4(1H)-quinolone (Pseudomonas quinolone signal [PQS]) and 2-heptyl-4(1H)-quinolone (HHQ). Mycobacteroides abscessus subsp. abscessus, an emerging pathogen, is capable of degrading the PQS and also HHQ. Here, we show that although M. abscessus subsp. abscessus reduced PQS levels in coculture with P. aeruginosa PAO1, this did not suffice for quenching the production of the virulence factors pyocyanin, pyoverdine, and rhamnolipids. However, the levels of these virulence factors were reduced in cocultures of P. aeruginosa PAO1 with recombinant M. abscessus subsp. massiliense overexpressing the PQS dioxygenase gene aqdC of M. abscessus subsp. abscessus, corroborating the potential of AqdC as a quorum quenching enzyme. When added extracellularly to P. aeruginosa cultures, AqdC quenched alkylquinolone and pyocyanin production but induced an increase in elastase levels. When supplementing P. aeruginosa cultures with QsdA, an enzyme from Rhodococcus erythropolis which inactivates N-acylhomoserine lactone signals, rhamnolipid and elastase levels were quenched, but HHQ and pyocyanin synthesis was promoted. Thus, single quorum quenching enzymes, targeting individual circuits within a complex quorum sensing network, may also elicit undesirable regulatory effects. Supernatants of P. aeruginosa cultures grown in the presence of AqdC, QsdA, or both enzymes were less cytotoxic to human epithelial lung cells than supernatants of untreated cultures. Furthermore, the combination of both aqdC and qsdA in P. aeruginosa resulted in a decline of Caenorhabditis elegans mortality under P. aeruginosa exposure.
- Published
- 2019
35. The SOS Response Mediates Sustained Colonization of the Mammalian Gut
- Author
-
Jun Zhu, Mark Goulian, Manuela Roggiani, Rahul M. Kohli, and Amanda N. Samuels
- Subjects
DNA repair ,DNA damage ,Immunology ,Context (language use) ,Genotoxic Stress ,Biology ,medicine.disease_cause ,Microbiology ,Mice ,Escherichia coli ,medicine ,Animals ,Colonization ,Microbiome ,SOS response ,SOS Response, Genetics ,Gene Expression Regulation, Bacterial ,Host-Associated Microbial Communities ,Gastrointestinal Tract ,Mice, Inbred C57BL ,Disease Models, Animal ,Infectious Diseases ,bacteria ,Parasitology ,DNA Damage - Abstract
Bacteria have a remarkable ability to survive, persist, and ultimately adapt to environmental challenges. A ubiquitous environmental hazard is DNA damage, and most bacteria have evolved a network of genes to combat genotoxic stress. This network is known as the SOS response and aids in bacterial survival by regulating genes involved in DNA repair and damage tolerance. Recently, the SOS response has been shown to play an important role in bacterial pathogenesis, and yet the role of the SOS response in nonpathogenic organisms and in physiological settings remains underexplored. Using a commensal Escherichia coli strain, MP1, we showed that the SOS response plays a vital role during colonization of the murine gut. In an unperturbed environment, the SOS-off mutant is impaired for stable colonization relative to a wild-type strain, suggesting the presence of genotoxic stress in the mouse gut. We evaluated the possible origins of genotoxic stress in the mouse gut by examining factors associated with the host versus the competing commensal organisms. In a dextran sulfate sodium (DSS) colitis model, the SOS-off colonization defect persisted but was not exacerbated. In contrast, in a germ-free model, the SOS-off mutant colonized with efficiency equal to that seen with the wild-type strain, suggesting that competing commensal organisms might be a significant source of genotoxic stress. This report extends our understanding of the importance of a functional SOS response for bacterial fitness in the context of a complex physiological environment and highlights the SOS response as a possible mechanism that contributes to ongoing genomic changes, including potential antibiotic resistance, in the microbiome of healthy hosts.
- Published
- 2019
- Full Text
- View/download PDF
36. Sortase-Dependent Proteins Promote Gastrointestinal Colonization by Enterococci
- Author
-
Nita H. Salzman, Leou Ismael Banla, Adam M. Pickrum, Michael Hayward, and Christopher J. Kristich
- Subjects
Male ,Immunology ,Microbiology ,Enterococcus faecalis ,Mice ,Bacterial Proteins ,Sortase ,Cell Wall ,medicine ,Animals ,Humans ,Colonization ,biology ,Mucin ,Human gastrointestinal tract ,biology.organism_classification ,Aminoacyltransferases ,Host-Associated Microbial Communities ,Mucus ,Gastrointestinal Microbiome ,Gastrointestinal Tract ,Mice, Inbred C57BL ,Cysteine Endopeptidases ,Disease Models, Animal ,Infectious Diseases ,medicine.anatomical_structure ,Enterococcus ,Sortase A ,Parasitology - Abstract
The human gastrointestinal tract (GIT) is inhabited by a dense microbial community of symbionts. Enterococci are among the earliest members of this community and remain core members of the GIT microbiota throughout life. Enterococci have also recently emerged as opportunistic pathogens and major causes of nosocomial infections. Although recognized as a prerequisite for infection, colonization of the GIT by enterococci remains poorly understood. One way that bacteria adapt to dynamic ecosystems like the GIT is through the use of their surface proteins to sense and interact with components of their immediate environment. In Gram-positive bacteria, a subset of surface proteins relies on an enzyme called sortase for covalent attachment to the cell wall. Here, we show that the housekeeping sortase A (SrtA) enzyme promotes intestinal colonization by enterococci. Furthermore, we show that the enzymatic activity of SrtA is key to the ability of Enterococcus faecalis to bind mucin (a major component of the GIT mucus). We also report the GIT colonization phenotypes of E. faecalis mutants lacking selected sortase-dependent proteins (SDPs). Further examination of the mucin binding ability of these mutants suggests that adhesion to mucin contributes to intestinal colonization by E. faecalis.
- Published
- 2018
37. Autoinducer 2 (AI-2) Production by Nontypeable Haemophilus influenzae 86-028NP Promotes Expression of a Predicted Glycosyltransferase That Is a Determinant of Biofilm Maturation, Prevention of Dispersal, and Persistence In Vivo
- Author
-
Chelsie E. Armbruster, Brian S. Learman, Gayle C Foster, W. Edward Swords, Bing Pang, and Uma Gandhi
- Subjects
0301 basic medicine ,Haemophilus Infections ,Transcription, Genetic ,Immunology ,Population ,Mutant ,Biology ,medicine.disease_cause ,Real-Time Polymerase Chain Reaction ,Microbiology ,Haemophilus influenzae ,03 medical and health sciences ,chemistry.chemical_compound ,Lactones ,Bacterial Proteins ,Chinchilla ,Glycosyltransferase ,medicine ,otorhinolaryngologic diseases ,Homoserine ,Animals ,education ,education.field_of_study ,Xylose ,Gene Expression Profiling ,Biofilm ,Intracellular Signaling Peptides and Proteins ,Biofilm matrix ,Glycosyltransferases ,biochemical phenomena, metabolism, and nutrition ,biology.organism_classification ,Host-Associated Microbial Communities ,Autoinducer-2 ,Carbon-Sulfur Lyases ,Otitis Media ,030104 developmental biology ,Infectious Diseases ,chemistry ,Biofilms ,Mutation ,biology.protein ,Parasitology ,Carrier Proteins ,Bacteria - Abstract
Nontypeable Haemophilus influenzae (NTHi) is an extremely common human pathobiont that persists on the airway mucosal surface within biofilm communities, and our previous work has shown that NTHi biofilm maturation is coordinated by the production and uptake of autoinducer 2 (AI-2) quorum signals. To directly test roles for AI-2 in maturation and maintenance of NTHi biofilms, we generated an NTHi 86-028NP mutant in which luxS transcription was under the control of the xylA promoter (NTHi 86-028NP luxS xylA::luxS), rendering AI-2 production inducible by xylose. Comparison of biofilms under inducing and noninducing conditions revealed a biofilm defect in the absence of xylose, whereas biofilm maturation increased following xylose induction. The removal of xylose resulted in the interruption of luxS expression and biofilm dispersal. Measurement of luxS transcript levels by real-time reverse transcription-PCR (RT-PCR) showed that luxS expression peaked as biofilms matured and waned before dispersal. Transcript profiling revealed significant changes following the induction of luxS, including increased transcript levels for a predicted family 8 glycosyltransferase (NTHI1750; designated gstA); this result was confirmed by real-time RT-PCR. An isogenic NTHi 86-028NP gstA mutant had a biofilm defect, including decreased levels of sialylated matrix and significantly altered biofilm structure. In experimental chinchilla infections, we observed a significant decrease in the number of bacteria in the biofilm population (but not in effusions) for NTHi 86-028NP gstA compared to the parental strain. Therefore, we conclude that AI-2 promotes NTHi biofilm maturation and the maintenance of biofilm integrity, due at least in part to the expression of a probable glycosyltransferase that is potentially involved in the synthesis of the biofilm matrix.
- Published
- 2018
38. Ceftriaxone Administration Disrupts Intestinal Homeostasis, Mediating Noninflammatory Proliferation and Dissemination of Commensal Enterococci
- Author
-
Michael Hayward, Vy Lam, Sushma Kommineni, Christopher J. Kristich, Rajrupa Chakraborty, Jeremiah Stromich, and Nita H. Salzman
- Subjects
0301 basic medicine ,Male ,medicine.drug_class ,030106 microbiology ,Immunology ,Antibiotics ,Colonisation resistance ,Biology ,Microbiology ,Enterococcus faecalis ,03 medical and health sciences ,Mice ,Antibiotic resistance ,Intestinal mucosa ,Lactobacillus ,medicine ,Animals ,Homeostasis ,Symbiosis ,Gram-Positive Bacterial Infections ,Ceftriaxone ,biochemical phenomena, metabolism, and nutrition ,biology.organism_classification ,Host-Associated Microbial Communities ,Anti-Bacterial Agents ,Gastrointestinal Microbiome ,Intestines ,Mice, Inbred C57BL ,030104 developmental biology ,Infectious Diseases ,Enterococcus ,Bacterial Translocation ,Parasitology ,medicine.drug - Abstract
Enterococci are Gram-positive commensals of the mammalian intestinal tract and harbor intrinsic resistance to broad-spectrum cephalosporins. Disruption of colonization resistance in humans by antibiotics allows enterococci to proliferate in the gut and cause disseminated infections. In this study, we used Enterococcus faecalis (EF)-colonized mice to study the dynamics of enterococci, commensal microbiota, and the host in response to systemic ceftriaxone administration. We found that the mouse model recapitulates intestinal proliferation and dissemination of enterococci seen in humans. Employing a ceftriaxone-sensitive strain of enterococci (E. faecalis JL308), we showed that increased intestinal abundance is critical for the systemic dissemination of enterococci. Investigation of the impact of ceftriaxone on the mucosal barrier defenses and integrity suggested that translocation of enterococci across the intestinal mucosa was not associated with intestinal pathology or increased permeability. Ceftriaxone-induced alteration of intestinal microbial composition was associated with transient increase in the abundance of multiple bacterial operational taxonomic units (OTUs) in addition to enterococci, for example, lactobacilli, which also disseminated to the extraintestinal organs. Collectively, these results emphasize that ceftriaxone-induced disruption of colonization resistance and alteration of mucosal homeostasis facilitate increased intestinal abundance of a limited number of commensals along with enterococci, allowing their translocation and systemic dissemination in a healthy host.
- Published
- 2018
39. Treatment of Dextran Sulfate Sodium-Induced Colitis with Mucosa-Associated Lymphoid Tissue Lymphoma Translocation 1 Inhibitor MI-2 Is Associated with Restoration of Gut Immune Function and the Microbiota
- Author
-
Chang-Hoon Lee, Min-Seok Kim, and Kyung Won Lee
- Subjects
0301 basic medicine ,Immunology ,microbiome ,Inflammation ,Biology ,Microbiology ,Monocytes ,Proinflammatory cytokine ,03 medical and health sciences ,Feces ,Mice ,0302 clinical medicine ,Immune system ,medicine ,Animals ,Humans ,Microbiome ,Colitis ,Cells, Cultured ,ulcerative colitis ,MALT1 ,Macrophages ,Dextran Sulfate ,Fecal Microbiota Transplantation ,Triazoles ,medicine.disease ,Host-Associated Microbial Communities ,Ulcerative colitis ,Gastrointestinal Microbiome ,Intestines ,Mice, Inbred C57BL ,030104 developmental biology ,Infectious Diseases ,Lymphatic system ,Mucosa-Associated Lymphoid Tissue Lymphoma Translocation 1 Protein ,030220 oncology & carcinogenesis ,Cytokines ,Parasitology ,Tumor necrosis factor alpha ,Acetanilides ,Female ,medicine.symptom ,MI-2 - Abstract
Disruption of the healthy intestinal microbiome and homeostasis of the intestinal immune system, which are closely interactive, are two key factors for ulcerative colitis. Here, we show that MI-2, a selective inhibitor of mucosa-associated lymphoid tissue lymphoma translocation-1 (MALT1), alleviated excessive inflammatory responses and was associated with restoration of healthy intestinal microbiome in mice suffering from dextran sulfate sodium (DSS)-induced colitis., Disruption of the healthy intestinal microbiome and homeostasis of the intestinal immune system, which are closely interactive, are two key factors for ulcerative colitis. Here, we show that MI-2, a selective inhibitor of mucosa-associated lymphoid tissue lymphoma translocation-1 (MALT1), alleviated excessive inflammatory responses and was associated with restoration of healthy intestinal microbiome in mice suffering from dextran sulfate sodium (DSS)-induced colitis. We found that the diversity of intestinal microbiome of mice with DSS-induced colitis was significantly lower than that of healthy mice. However, MI-2 treatment in mice with DSS-induced colitis resulted in restored microbially diverse populations. To understand the possibility of the beneficial effect of the restored microbially diverse populations of MI-2-treated mice with DSS-induced colitis, we showed that inserting fecal microbiota from MI-2-treated mice with DSS-induced colitis and healthy control mice into mice with DSS-induced colitis could alleviate symptoms of colitis. The possibility of MI-2 treatment in DSS-induced colitis, associated with restoration of healthy microbially diverse populations in addition to reshaping host immune modulating capacity by reducing inflammatory cytokines (tumor necrosis factor alpha, interleukin-1β [IL-1β], IL-17α, and IL-22), may be considered therapeutic for ulcerative colitis.
- Published
- 2018
40. Host-Derived Metabolites Modulate Transcription of
- Author
-
Caroline C, Gillis, Maria G, Winter, Rachael B, Chanin, Wenhan, Zhu, Luisella, Spiga, and Sebastian E, Winter
- Subjects
Male ,Salmonella typhimurium ,L-Lactate Dehydrogenase ,Gene Expression Regulation, Bacterial ,Host-Associated Microbial Communities ,Mice, Inbred C57BL ,Oxygen ,Mice ,Bacterial Proteins ,Salmonella Infections ,Animals ,Humans ,Female ,Lactic Acid ,Intestinal Mucosa ,Transcription Factors - Abstract
During Salmonella enterica serovar Typhimurium infection, host inflammation alters the metabolic environment of the gut lumen to favor the outgrowth of the pathogen at the expense of the microbiota. Inflammation-driven changes in host cell metabolism lead to the release of l-lactate and molecular oxygen from the tissue into the gut lumen. Salmonella utilizes lactate as an electron donor in conjunction with oxygen as the terminal electron acceptor to support gut colonization. Here, we investigated transcriptional regulation of the respiratory l-lactate dehydrogenase LldD in vitro and in mouse models of Salmonella infection. The two-component system ArcAB repressed transcription of l-lactate utilization genes under anaerobic conditions in vitro. The ArcAB-mediated repression of lldD transcription was relieved under microaerobic conditions. Transcription of lldD was induced by l-lactate but not d-lactate. A mutant lacking the regulatory protein LldR failed to induce lldD transcription in response to l-lactate. Furthermore, the lldR mutant exhibited reduced transcription of l-lactate utilization genes and impaired fitness in murine models of infection. These data provide evidence that the host-derived metabolites oxygen and l-lactate serve as cues for Salmonella to regulate lactate oxidation metabolism on a transcriptional level.
- Published
- 2018
41. Role of Neuraminidase-Producing Bacteria in Exposing Cryptic Carbohydrate Receptors for Streptococcus gordonii Adherence
- Author
-
Shireen A. Woodiga, Samantha J. King, Margaret A. Grau, Anirudh K. Singh, and Alexander Wong
- Subjects
0301 basic medicine ,Glycan ,Glycoconjugate ,030106 microbiology ,Immunology ,Hemagglutinins, Viral ,Neuraminidase ,Microbiology ,Bacterial Adhesion ,03 medical and health sciences ,chemistry.chemical_compound ,stomatognathic system ,Sortase ,Humans ,Adhesins, Bacterial ,chemistry.chemical_classification ,biology ,Streptococcus gordonii ,Mouth Mucosa ,Galactose ,Streptococcus oralis ,biology.organism_classification ,Host-Associated Microbial Communities ,N-Acetylneuraminic Acid ,Sialic acid ,Bacterial adhesin ,stomatognathic diseases ,030104 developmental biology ,Infectious Diseases ,chemistry ,biology.protein ,Parasitology ,Carrier Proteins - Abstract
Streptococcus gordonii is an early colonizer of the oral cavity. Although a variety of S. gordonii adherence mechanisms have been described, current dogma is that the major receptor for S. gordonii is sialic acid. However, as many bacterial species in the oral cavity produce neuraminidase that can cleave terminal sialic acid, it is unclear whether S. gordonii relies on sialic acid for adherence to oral surfaces or if this species has developed alternative binding strategies. Previous studies have examined adherence to immobilized glycoconjugates and identified binding to additional glycans, but no prior studies have defined the contribution of these different glycan structures in adherence to oral epithelial cells. We determined that the majority of S. gordonii strains tested did not rely on sialic acid for efficient adherence. In fact, adherence of some strains was significantly increased following neuraminidase treatment. Further investigation of representative strains that do not rely on sialic acid for adherence revealed binding not only to sialic acid via the serine-rich repeat protein GspB but also to β-1,4-linked galactose. Adherence to this carbohydrate occurs via an unknown adhesin distinct from those utilized by Streptococcus oralis and Streptococcus pneumoniae . Demonstrating the potential biological relevance of binding to this cryptic receptor, we established that S. oralis increases S. gordonii adherence in a neuraminidase-dependent manner. These data suggest that S. gordonii has evolved to simultaneously utilize both terminal and cryptic receptors in response to the production of neuraminidase by other species in the oral environment.
- Published
- 2018
42. Chemotaxis Allows Bacteria To Overcome Host-Generated Reactive Oxygen Species That Constrain Gland Colonization
- Author
-
Karen M. Ottemann, Helmut Grasberger, John Y. Kao, Shuai Hu, Kieran Collins, and Freitag, Nancy E
- Subjects
0301 basic medicine ,Male ,Mutant ,crypts ,chemoreceptors ,Medical and Health Sciences ,chemistry.chemical_compound ,Mice ,2.2 Factors relating to the physical environment ,Colonization ,Aetiology ,Mice, Knockout ,chemistry.chemical_classification ,reactive oxygen species ,0303 health sciences ,Superoxide ,Chemotaxis ,Bacterial ,Biological Sciences ,Host-Associated Microbial Communities ,Infectious Diseases ,Carrier State ,Female ,signal transduction ,stomach ,Knockout ,1.1 Normal biological development and functioning ,030106 microbiology ,Immunology ,Motility ,Biology ,Microbiology ,glands ,03 medical and health sciences ,Immune system ,stomatognathic system ,Bacterial Proteins ,Underpinning research ,Animals ,Humans ,biogeography ,030304 developmental biology ,Reactive oxygen species ,Innate immune system ,Helicobacter pylori ,Agricultural and Veterinary Sciences ,030306 microbiology ,biology.organism_classification ,030104 developmental biology ,Emerging Infectious Diseases ,chemistry ,Gene Expression Regulation ,Genes ,Genes, Bacterial ,Gastric Mucosa ,MCP ,Mutation ,Parasitology ,Reactive Oxygen Species ,Digestive Diseases ,Bacteria - Abstract
The epithelial layer of the gastrointestinal tract contains invaginations, called glands or crypts, which are colonized by symbiotic and pathogenic microorganisms and may function as designated niches for certain species. Factors that control gland colonization are poorly understood, but bacterial chemotaxis aids occupation of these sites. We report here that a Helicobacter pylori cytoplasmic chemoreceptor, TlpD, is required for gland colonization in the stomach. tlpD mutants demonstrate gland colonization defects characterized by a reduction in the percent of glands colonized, but not in number of bacteria per gland. Consistent with TlpD’s reported role in reactive oxygen species (ROS) avoidance, tlpD mutants showed hallmarks of exposure to large amounts of ROS. To assess the role of host-generated ROS in TlpD-dependent gland colonization, we utilized mice that lack either the ability to generate epithelial hydrogen peroxide or immune cell superoxide. tlpD gland colonization defects were rescued to wild-type H. pylori levels in both of these mutants. These results suggest that multiple types of innate immune generated ROS production limit gland colonization and that bacteria have evolved specific mechanisms to migrate through this gauntlet to establish in the glands.Classification: Biological sciences; microbiologySignificance statementMicrobial colonization of the gastrointestinal tract occurs at distinct sites within the tissue including glandular structures found in the stomach and intestine. Multiple lines of evidence suggest that glands supply niches that promote chronic microbial colonization, a process that is critical for symbiotic and pathogenic bacteria to maintain themselves. In this report, we show that host-produced reactive oxygen species (ROS) constrain gland colonization by the gastric pathogen Helicobacter pylori. A bacterial cytoplasmic chemoreceptor, TlpD, allows H. pylori to avoid ROS and enhances H. pylori’s ability to colonize a broad swath of glands. We propose that hosts limit gland access and spread by producing ROS, and bacteria counter with chemotactic responses that allow navigation through this gauntlet.
- Published
- 2018
43. Dysbiosis of Inferior Turbinate Microbiota Is Associated with High Total IgE Levels in Patients with Allergic Rhinitis
- Author
-
Na-Ri Shin, Min-Soo Kim, Joo Heon Yoon, Hyun Jin Min, Jin-Woo Bae, Hyun Sik Kim, Woorim Kang, Tae Woong Whon, June-Young Lee, Dong-Wook Hyun, Augustine M.K. Choi, and Pil Soo Kim
- Subjects
Male ,0301 basic medicine ,Firmicutes ,Immunology ,Mucous membrane of nose ,Turbinates ,Immunoglobulin E ,medicine.disease_cause ,Microbiology ,03 medical and health sciences ,Propionibacterium acnes ,Allergen ,RNA, Ribosomal, 16S ,medicine ,Humans ,House dust mite ,biology ,Microbiota ,Human microbiome ,Biodiversity ,Sequence Analysis, DNA ,Allergens ,Host-Associated Microbial Communities ,medicine.disease ,biology.organism_classification ,Rhinitis, Allergic ,Nasal Mucosa ,030104 developmental biology ,Infectious Diseases ,Case-Control Studies ,biology.protein ,Dysbiosis ,Female ,Immunization ,Parasitology ,Biomarkers - Abstract
Abnormalities in the human microbiota are associated with the etiology of allergic diseases. Although disease site-specific microbiota may be associated with disease pathophysiology, the role of the nasal microbiota is unclear. We sought to characterize the microbiota of the site of allergic rhinitis, the inferior turbinate, in subjects with allergic rhinitis ( n = 20) and healthy controls ( n = 12) and to examine the relationship of mucosal microbiota with disease occurrence, sensitized allergen number, and allergen-specific and total IgE levels. Microbial dysbiosis correlated significantly with total IgE levels representing combined allergic responses but not with disease occurrence, the number of sensitized allergens, or house dust mite allergen-specific IgE levels. Compared to the populations in individuals with low total IgE levels (group IgE low ), low microbial biodiversity with a high relative abundance of Firmicutes phylum ( Staphylococcus aureus ) and a low relative abundance of Actinobacteria phylum ( Propionibacterium acnes ) was observed in individuals with high total serum IgE levels (group IgE high ). Phylogeny-based microbial functional potential predicted by the 16S rRNA gene indicated an increase in signal transduction-related genes and a decrease in energy metabolism-related genes in group IgE high as shown in the microbial features with atopic and/or inflammatory diseases. Thus, dysbiosis of the inferior turbinate mucosa microbiota, particularly an increase in S. aureus and a decrease in P. acnes , is linked to high total IgE levels in allergic rhinitis, suggesting that inferior turbinate microbiota may be affected by accumulated allergic responses against sensitized allergens and that site-specific microbial alterations play a potential role in disease pathophysiology.
- Published
- 2018
- Full Text
- View/download PDF
44. Gut Microbiome Analysis Identifies Potential Etiological Factors in Acute Gastroenteritis
- Author
-
Hazel M. Mitchell, William D. Rawlinson, Nadeem O. Kaakoush, Natalia Castaño-Rodríguez, Juan Merif, Alexander Underwood, and Stephen M. Riordan
- Subjects
0301 basic medicine ,Adult ,Adolescent ,Immunology ,Gut flora ,medicine.disease_cause ,Microbiology ,03 medical and health sciences ,Feces ,Young Adult ,medicine ,Humans ,Microbiome ,Child ,Aged ,Aged, 80 and over ,biology ,Bacteria ,Campylobacter ,Age Factors ,Infant ,Middle Aged ,biology.organism_classification ,Capnocytophaga ,Host-Associated Microbial Communities ,Gastroenteritis ,Gastrointestinal Microbiome ,030104 developmental biology ,Infectious Diseases ,Aeromonas ,Arcobacter ,Child, Preschool ,Acute Disease ,Parasitology ,Enterotype ,Bacteroides - Abstract
The morbidity and mortality resulting from acute gastroenteritis and associated chronic sequelae represent a substantial burden on health care systems worldwide. Few studies have investigated changes in the gut microbiome following an episode of acute gastroenteritis. By using nondirected 16S rRNA gene amplicon sequencing, the fecal microbiota of 475 patients with acute gastroenteritis was examined. Patient age was correlated with the overall microbial composition, with a decrease in the abundance of Faecalibacterium being observed in older patients. We observed the emergence of a potential Escherichia-Shigella -dominated enterotype in a subset of patients, and this enterotype was predicted to be more proinflammatory than the other common enterotypes, with the latter being dominated by Bacteroides or Faecalibacterium . The increased abundance of Escherichia-Shigella did not appear to be associated with infection with an agent of a similar sequence similarity. Stool color and consistency were associated with the diversity and composition of the microbiome, with deviations from the norm (not brown and solid) showing increases in the abundances of bacteria such as Escherichia-Shigella and Veillonella . Analysis of enriched outliers within the data identified a range of genera previously associated with gastrointestinal diseases, including Treponema , Proteus , Capnocytophaga , Arcobacter , Campylobacter , Haemophilus , Aeromonas , and Pseudomonas . Our data represent the first in-depth analysis of gut microbiota in acute gastroenteritis. Phenotypic changes in stool color and consistency were associated with specific changes in the microbiota. Enriched bacterial taxa were detected in cases where no causative agent was identified by using routine diagnostic tests, suggesting that in the future, microbiome analyses may be utilized to improve diagnostics.
- Published
- 2018
45. A Vaginal Tract Signal Detected by the Group B Streptococcus SaeRS System Elicits Transcriptomic Changes and Enhances Murine Colonization
- Author
-
Laura C. Cook, Michael J. Federle, Mark Maienschein-Cline, and Hong Hu
- Subjects
0301 basic medicine ,Immunology ,Virulence ,Biology ,medicine.disease_cause ,Microbiology ,Group B ,Streptococcus agalactiae ,Transcriptome ,03 medical and health sciences ,Mice ,Bacterial Proteins ,In vivo ,Streptococcal Infections ,medicine ,Animals ,Colonization ,Gene ,Streptococcus ,Gene Expression Regulation, Bacterial ,Host-Associated Microbial Communities ,Disease Models, Animal ,030104 developmental biology ,Infectious Diseases ,Vagina ,Parasitology ,Female ,Signal Transduction - Abstract
Streptococcus agalactiae (group B streptococcus [GBS]) can colonize the human vaginal tract, leading to both superficial and serious infections in adults and neonates. To study bacterial colonization of the reproductive tract in a mammalian system, we employed a murine vaginal carriage model. Using transcriptome sequencing (RNA-Seq), the transcriptome of GBS growing in vivo during vaginal carriage was determined. Over one-quarter of the genes in GBS were found to be differentially regulated during in vivo colonization compared to laboratory cultures. A two-component system (TCS) homologous to the staphylococcal virulence regulator SaeRS was identified as being upregulated in vivo . One of the SaeRS targets, pbsP , a proposed GBS vaccine candidate, is shown to be important for colonization of the vaginal tract. A component of vaginal lavage fluid acts as a signal to turn on pbsP expression via SaeRS. These data demonstrate the ability to quantify RNA expression directly from the murine vaginal tract and identify novel genes involved in vaginal colonization by GBS. They also provide more information about the regulation of an important virulence and colonization factor of GBS, pbsP , by the TCS SaeRS.
- Published
- 2017
46. Interactions between amphibians’ symbiotic bacteria cause the production of emergent anti-fungal metabolites
- Author
-
Thomas P. Umile, Elizabeth A. Burzynski, Andrew H. Loudon, Reid N. Harris, Jessica A. Holland, and Kevin P. C. Minbiole
- Subjects
Microbiology (medical) ,Metabolite ,lcsh:QR1-502 ,Bacillus ,anti-fungal metabolites ,Microbiology ,lcsh:Microbiology ,Amphibians ,symbiotic bacteria ,chemistry.chemical_compound ,Tryptophol ,Original Research Article ,Pathogen ,Batrachochytrium dendrobatidis ,biology ,interspecific competition ,Pseudomonas ,emergent metabolites ,biology.organism_classification ,Synergy ,chemistry ,host-associated microbial communities ,Janthinobacterium ,Bacteria ,Symbiotic bacteria - Abstract
Amphibians possess beneficial skin bacteria that protect against the disease chytridiomycosis by producing secondary metabolites that inhibit the pathogen Batrachochytrium dendrobatidis (Bd). Metabolite production may be a mechanism of competition between bacterial species that results in host protection as a by-product. We expect that some co-cultures of bacterial species or strains will result in greater Bd inhibition than mono-cultures. To test this, we cultured four bacterial isolates (Bacillus sp., Janthinobacterium sp., Pseudomonas sp. and Chitinophaga arvensicola) from red-backed salamanders (Plethodon cinereus) and cultured isolates both alone and together to collect their cell-free supernatants (CFS). We challenged Bd with CFSs from four bacterial species in varying combinations. This resulted in three experimental treatments: (1) CFSs of single isolates; (2) combined CFSs of two isolates; and (3) CFSs from co-cultures. Pair-wise combinations of four bacterial isolates CFSs were assayed against Bd and revealed additive Bd inhibition in 42.2% of trials, synergistic inhibition in 42.2% and no effect in 16.6% of trials. When bacteria isolates were grown in co-cultures, complete Bd inhibition was generally observed, and synergistic inhibition occurred in four out of six trials. A metabolite profile of the most potent co-culture, Bacillus sp. and Chitinophaga arvensicola, was determined with LC-MS and compared with the profiles of each isolate in mono-culture. Emergent metabolites appearing in the co-culture were inhibitory to Bd, and the most potent inhibitor was identified as tryptophol. Thus mono-cultures of bacteria cultured from red-backed salamanders interacted synergistically and additively to inhibit Bd, and such bacteria produced emergent metabolites when cultured together, with even greater pathogen inhibition. Knowledge of how bacterial species interact to inhibit Bd can be used to select probiotics to provide amphibians with protection against Bd.
- Published
- 2014
- Full Text
- View/download PDF
47. Habitat filtering at multiple scales : a case study with crab-associated microbial communities
- Author
-
Cuellar-Gempeler, Catalina
- Subjects
- Community assembly, Crab, Host-associated microbial communities, Bacteria, Diversity, Species pools
- Abstract
Local diversity is the result of colonization from a species pool and local habitat filters. The relative contribution of these processes delineates our ability to predict changes in biodiversity, key in a changing world. Although theory suggests that these interactions are critical for the maintenance of biodiversity, empirical work has been restricted by limitations of studies across biogeographical regions and laboratory microcosms. Alternatively, host-associated microbial communities represent discrete, tractable and replicable assemblages. Hosts provide distinct habitat patches with different habitat filters and are surrounded by environmental bacteria that determine the regional species pools. My dissertation focuses on colonization and habitat filtering processes driving species composition of microbial communities associated with coastal crabs. First, I investigated colonization from the water column and filtering associated with the striped shore crab (Pachygrapsus transversus). Using a field experiment I asked whether species pool size interacts with habitat filters to structure microbial communities. Results indicated that the carapace was a stronger filters than the surrounding surfaces and are thus less influenced by changes in the regional species pool. Next, I examined whether the temporal scale of convergence and divergence in community composition was habitat dependent. In a mesocosm experiment, gut communities converged in community composition while carapace communities converged at day 8 and diverged thereafter. These findings indicate that the gut represents a strong filter when compared to carapace habitats, whose communities were driven by species interactions during biofilm formation. Second, I investigated whether multiple colonist pools can influence microbial communities. In a field study, I found that surface and burrow sediment colonize fiddler crab gut and carapace communities To better understand the interaction between multiple colonist pools and habitat filters, I used a mesocosm experiment where I manipulated sediment bacteria. While carapace communities were influenced by burrow sediment, removing bacteria from the surface also impacted carapace microbial composition. In contrast, gut communities responded weakly to colonist pool manipulations suggesting strong filtering. These findings highlight complex interactions between local communities and colonist pools. Importantly, identifying when multiple colonist pools or habitat filters drives community composition should lead to a more predictive community ecology.
- Published
- 2016
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.