Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 38
Filter
1.
Nat Commun ; 15(1): 2993, 2024 Apr 06.
Article in English | MEDLINE | ID: mdl-38582763

ABSTRACT

Bacteriophage therapy is a promising approach to address antimicrobial infections though questions remain regarding the impact of the immune response on clinical effectiveness. Here, we develop a mouse model to assess phage treatment using a cocktail of five phages from the Myoviridae and Siphoviridae families that target Vancomycin-Resistant Enterococcus gut colonization. Phage treatment significantly reduces fecal bacterial loads of Vancomycin-Resistant Enterococcus. We also characterize immune responses elicited following administration of the phage cocktail. While minimal innate responses are observed after phage administration, two rounds of treatment induces phage-specific neutralizing antibodies and accelerate phage clearance from tissues. Interestingly, the myophages in our cocktail induce a more robust neutralizing antibody response than the siphophages. This anti-phage immunity reduces the effectiveness of the phage cocktail in our murine model. Collectively, this study shows phage-specific immune responses may be an important consideration in the development of phage cocktails for therapeutic use.


Subject(s)
Bacteriophages , Vancomycin-Resistant Enterococci , Humans , Animals , Mice , Bacteriophages/physiology , Vancomycin/pharmacology , Disease Models, Animal , Myoviridae/physiology , Anti-Bacterial Agents/pharmacology
2.
Biologicals ; 86: 101758, 2024 May.
Article in English | MEDLINE | ID: mdl-38518435

ABSTRACT

Fecal microbiota transplantation (FMT) has been demonstrated to be efficacious in preventing recurrent Clostridioides difficile (C. difficile) infections, and is being investigated for treatment of several other diseases including inflammatory bowel disease, cancer, obesity, liver disease, and diabetes. To speed up the translation of FMT into clinical practice as a safe and standardized therapeutic intervention, additional evidence-based technical and regulatory guidance is needed. To this end in May of 2022, the International Alliance for Biological Standardization (IABS) and the BIOASTER Microbiology Technology Institute hosted a second webinar to discuss key issues still impeding the advancement and standardization of FMT. The goal of this two-day webinar was to provide a forum for scientific experts to share and discuss data and key challenges with one another. Discussion included a focus on the evaluation of safety, efficacy, clinical trial design, reproducibility and accuracy in obtained microbiome measurements and data reporting, and the potential for standardization across these areas. It also focused on increasing the application potential and visibility of FMT beyond treating C. difficile infections.


Subject(s)
Clostridium Infections , Fecal Microbiota Transplantation , Humans , Fecal Microbiota Transplantation/standards , Fecal Microbiota Transplantation/methods , Clostridium Infections/therapy , Clostridium Infections/microbiology , Clostridioides difficile , Gastrointestinal Microbiome
3.
AMB Express ; 14(1): 9, 2024 Jan 20.
Article in English | MEDLINE | ID: mdl-38245586

ABSTRACT

Nanobodies are highly specific binding domains derived from naturally occurring single chain camelid antibodies. Live biotherapeutic products (LBPs) are biological products containing preparations of live organisms, such as Lactobacillus, that are intended for use as drugs, i.e. to address a specific disease or condition. Demonstrating potency of multi-strain LBPs can be challenging. The approach investigated here is to use strain-specific nanobody reagents in LBP potency assays. Llamas were immunized with radiation-killed Lactobacillus jensenii or L. crispatus whole cell preparations. A nanobody phage-display library was constructed and panned against bacterial preparations to identify nanobodies specific for each species. Nanobody-encoding DNA sequences were subcloned and the nanobodies were expressed, purified, and characterized. Colony immunoblots and flow cytometry showed that binding by Lj75 and Lj94 nanobodies were limited to a subset of L. jensenii strains while binding by Lc38 and Lc58 nanobodies were limited to L. crispatus strains. Mass spectrometry was used to demonstrate that Lj75 specifically bound a peptidase of L. jensenii, and that Lc58 bound an S-layer protein of L. crispatus. The utility of fluorescent nanobodies in evaluating multi-strain LBP potency assays was assessed by evaluating a L. crispatus and L. jensenii mixture by fluorescence microscopy, flow cytometry, and colony immunoblots. Our results showed that the fluorescent nanobody labelling enabled differentiation and quantitation of the strains in mixture by these methods. Development of these nanobody reagents represents a potential advance in LBP testing, informing the advancement of future LBP potency assays and, thereby, facilitation of clinical investigation of LBPs.

4.
J Bacteriol ; 205(12): e0032423, 2023 12 19.
Article in English | MEDLINE | ID: mdl-37971230

ABSTRACT

IMPORTANCE: This study is the first example of C. difficile growing with siderophores as the sole iron source and describes the characterization of the ferric hydroxamate uptake ABC transporter (FhuDBGC). This transporter shows specificity to the siderophore ferrichrome. While not required for pathogenesis, this transporter highlights the redundancy in iron acquisition mechanisms that C. difficile uses to compete for iron during an infection.


Subject(s)
Clostridioides difficile , Siderophores , Iron/metabolism , Ferrichrome/metabolism , Clostridioides difficile/metabolism , Clostridioides , Membrane Transport Proteins
5.
Appl Microbiol Biotechnol ; 107(12): 4069-4077, 2023 Jun.
Article in English | MEDLINE | ID: mdl-37148337

ABSTRACT

Characterization of live biotherapeutic product (LBP) batches typically includes a measurement of viability, such as colony forming units (CFU). However, strain-specific CFU enumeration assays can be complicated by the presence of multiple organisms in a single product with similar growth requirements. To overcome specific challenges associated with obtaining strain-specific CFU values from multi-strain mixtures, we developed a method combining mass spectrometry-based colony identification with a traditional CFU assay. This method was assessed using defined consortia made from up to eight bacterial strains. Among four replicate batches of an eight-strain mixture, observed values differed from expected values by less than 0.4 log10 CFU among all strains measured (range of differences, -0.318 to + 0.267). The average difference between observed and expected values was + 0.0308 log10 CFU, with 95% limits of agreement from -0.347 to 0.408 (Bland-Altman analysis). To estimate precision, a single batch of eight-strain mixture was assayed in triplicate by three different users, for a total of nine measurements. Pooled standard deviation values ranged from 0.067 to 0.195 log10 CFU for the eight strains measured, and user averages did not differ significantly. Leveraging emerging mass-spectrometry-based colony identification tools, a novel method for simultaneous enumeration and identification of viable bacteria from mixed-strain consortia was developed and tested. This study demonstrates the potential for this approach to generate accurate and consistent measurements of up to eight bacterial strains simultaneously and may provide a flexible platform for future refinements and modifications. KEY POINTS: • Enumeration of live biotherapeutics is essential for product quality and safety. • Conventional CFU counting may not differentiate between strains in microbial products. • This approach was developed for direct enumeration of mixed bacterial strains simultaneously.


Subject(s)
Bacteria , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Colony Count, Microbial
6.
Med ; 3(6): 351-352, 2022 06 10.
Article in English | MEDLINE | ID: mdl-35690052

ABSTRACT

Persistent fecal shedding of SARS-CoV-2 viral RNA has remained a clinical feature of interest throughout the COVID-19 pandemic. In this issue of Med, Natarajan et al. report fecal shedding dynamics of individuals diagnosed with mild-to-moderate COVID-19 disease and sampled longitudinally for up to 10 months1.


Subject(s)
COVID-19 , SARS-CoV-2 , Humans , Pandemics , RNA, Viral/genetics , SARS-CoV-2/genetics , Virus Shedding
7.
Metabolites ; 12(5)2022 Apr 22.
Article in English | MEDLINE | ID: mdl-35629884

ABSTRACT

Mucosal-associated invariant T cells are activated following the recognition of bacterial antigens presented by the major histocompatibility complex class I-related molecule (MR1). Previous metagenomics data showed that MR1-/- knock-out (KO) mice had distinct microbiota and displayed a resistance to Clostridioides difficile (CDI) colonization vs. wild-type (WT) mice. In the present study, LC/MS-based untargeted metabolomics are applied to evaluate the changes in metabolic activities, in accordance with the changes in gut microbiota caused by cefoperazone (Cef) treatment. Adult C57Bl/6J WT and MR1-/- KO mice were given sterile drinking water or spiked with 0.5 mg/mL Cef ad libitum for five days. Fecal pellets were collected daily, and both small intestinal and cecal contents were harvested at sacrifice. The PLS-DA score plots of the metabolomic data indicate that the microbiota is relatively less disturbed by Cef treatment in KO mice, which is consistent with the metagenomics data. The most noticeable differences in the metabolome of KO and WT mice were the increases in carbohydrates in the WT mice, but not in the KO mice. Metabolic functional biomarkers were identified through the correlation analysis of gamma-aminobutyric acid (GABA) and riboflavin. These detected metabolic functional biomarkers could provide information complementary to metagenomics data.

8.
Lancet Microbe ; 2(6): e259-e266, 2021 06.
Article in English | MEDLINE | ID: mdl-33821247

ABSTRACT

BACKGROUND: Faecal shedding of SARS-CoV-2 has raised concerns about transmission through faecal microbiota transplantation procedures. Validation parameters of authorised tests for SARS-CoV-2 RNA detection in respiratory samples are described in product labelling, whereas the published methods for SARS-CoV-2 detection from faecal samples have not permitted a robust description of the assay parameters. We aimed to develop and validate a test specifically for detection of SARS-CoV-2 in human stool. METHODS: In this validation study, we evaluated performance characteristics of a reverse transcriptase real-time PCR (RT-rtPCR) test for detection of SARS-CoV-2 in human stool specimens by spiking stool with inactivated SARS-CoV-2 material. A modified version of the US Centers for Disease Control and Prevention RT-rtPCR SARS-CoV-2 test was used for detection of viral RNA. Analytical sensitivity was evaluated in freshly spiked stool by testing two-fold dilutions in replicates of 20. Masked samples were tested by a second laboratory to evaluate interlaboratory reproducibility. Short-term (7-day) stability of viral RNA in stool samples was assessed with four different stool storage buffers (phosphate-buffered saline, Cary-Blair medium, Stool Transport and Recovery [STAR] buffer, and DNA/RNA Shield) kept at -80°C, 4°C, and ambient temperature (approximately 21°C). We also tested clinical stool and anal swab specimens from patients who were SARS-CoV-2 positive by nasopharyngeal testing. FINDINGS: The lower limit of detection of the assay was found to be 3000 viral RNA copies per g of original stool sample, with 100% detection across 20 replicates assessed at this concentration. Analytical sensitivity was diminished by approximately two times after a single freeze-thaw cycle at -80°C. At 100 times the limit of detection, spiked samples were generally stable in all four stool storage buffers tested for up to 7 days, with maximum changes in mean threshold cycle values observed at -80°C storage in Cary-Blair medium (from 29·4 [SD 0·27] at baseline to 30·8 [0·17] at day 7; p<0·0001), at 4°C storage in DNA/RNA Shield (from 28·5 [0·15] to 29·8 [0·09]; p=0·0019), and at ambient temperature in STAR buffer (from 30·4 [0·24] to 32·4 [0·62]; p=0·0083). 30 contrived SARS-CoV-2 samples were tested by a second laboratory and were correctly identified as positive or negative in at least one of two rounds of testing. Additionally, SARS-CoV-2 RNA was detected using this assay in the stool and anal swab specimens of 11 of 23 individuals known to be positive for SARS-CoV-2. INTERPRETATION: This is a sensitive and reproducible assay for detection of SARS-CoV-2 RNA in human stool, with potential uses in faecal microbiota transplantation donor screening, sewage monitoring, and further research into the effects of faecal shedding on the epidemiology of the COVID-19 pandemic. FUNDING: National Institute of Allergy and Infectious Diseases, US National Institutes of Health; Center for Biologics Evaluation and Research, US Food and Drug Administration.


Subject(s)
COVID-19 , SARS-CoV-2 , COVID-19/diagnosis , Humans , Pandemics , RNA, Viral/genetics , Reproducibility of Results , SARS-CoV-2/genetics
9.
Microbiome ; 9(1): 2, 2021 01 04.
Article in English | MEDLINE | ID: mdl-33397500

ABSTRACT

The inaugural "Microbiome for Mars" virtual workshop took place on July 13, 2020. This event assembled leaders in microbiome research and development to discuss their work and how it may relate to long-duration human space travel. The conference focused on surveying current microbiome research, future endeavors, and how this growing field could broadly impact human health and space exploration. This report summarizes each speaker's presentation in the order presented at the workshop.


Subject(s)
Astronauts , Delivery of Health Care/trends , Mars , Microbiota/physiology , Space Flight , Animals , Gastrointestinal Microbiome/genetics , Gastrointestinal Microbiome/physiology , Humans , Microbiota/genetics
10.
Metabolites ; 10(4)2020 Mar 26.
Article in English | MEDLINE | ID: mdl-32225042

ABSTRACT

Mucosal associated invariant T-cells (MAIT cells) are activated following recognition of bacterial antigens (riboflavin intermediates) presented on major histocompatibility complex class I-related molecule (MR1). Our previous study showed that MR1-/- knock-out (KO) mice (lacking MAIT cells) harbor a unique microbiota that is resistant to antibiotic disruption and Clostridioides difficile colonization. While we have characterized the microbiota of this mouse strain, changes in global metabolic activity in these KO mice have not been assessed. Here, LC/MS-based untargeted metabolomics was applied to investigate the differences in the metabolome, specifically in the bile acid (BA) profile of wild-type (WT) and MR1-/- KO mice, as well as how antibiotics change these profiles. BA changes were evaluated in the intestinal content, cecum content, and stool samples from MR1-/- mice and WT mice treated with cefoperazone (Cef). Fecal pellets were collected daily and both intestinal and cecal contents were harvested at predetermined endpoints on day 0 (D0), day 1 (D1), day 3 (D3), and day 5 (D5). KO mice exhibited no changes in 6-hydroxymethyl-8-D-ribityllumazine (rRL-6-CH2OH; an MR1-restricted riboflavin derivative) in the stool samples at either time point vs. D0, while WT mice showed significant decreases in rRL-6-CH2OH in the stool samples on all treatment days vs. D0. Metabolomics analysis from cecal and stool samples showed that KO mice had more total BA intensity (KO/WT = ~1.7 and ~3.3 fold higher) than that from WT mice prior to Cef treatment, while the fold change difference (KO/WT = ~4.5 and ~4.4 fold) increased after five days of Cef treatment. Both KO and WT mice showed decreases in total BA intensity in response to Cef treatment, however, less dramatic decreases were present in KO vs. WT mice. Increases in taurocholic acid (TCA) intensity and decreases in deoxycholic acid (DCA) intensity in the stool samples from WT mice were associated with the depletion of certain gut bacteria, which was consistent with the previously reported microbiome data. Furthermore, the non-detected TCA and relatively higher DCA intensity in the KO mice might be related to Clostridioides difficile infection resistance, although this needs further investigation.

11.
Cell Host Microbe ; 27(2): 173-175, 2020 02 12.
Article in English | MEDLINE | ID: mdl-32053787

ABSTRACT

Fecal microbiota for transplantation (FMT) is being studied as a potential intervention for numerous conditions. The regulation of FMT by the FDA is discussed along with FMT donor screening and manufacturing considerations. The FDA is committed to ensuring that FMT products can be safely tested in clinical trials.


Subject(s)
Fecal Microbiota Transplantation , Feces/microbiology , Clostridioides difficile , Clostridium Infections/prevention & control , Fecal Microbiota Transplantation/methods , Fecal Microbiota Transplantation/trends , Gastrointestinal Microbiome , Living Donors , Safety , Social Control, Formal , Treatment Outcome
12.
PLoS One ; 14(9): e0223025, 2019.
Article in English | MEDLINE | ID: mdl-31560732

ABSTRACT

Clostridium difficile (Cd) infection (CDI) typically occurs after antibiotic usage perturbs the gut microbiota. Mucosa-associated invariant T cells (MAIT) are found in the gut and their development is dependent on Major histocompatibility complex-related protein 1 (MR1) and the host microbiome. Here we were interested in determining whether the absence of MR1 impacts resistance to CDI. To this end, wild-type (WT) and MR1-/- mice were treated with antibiotics and then infected with Cd spores. Surprisingly, MR1-/- mice exhibited resistance to Cd colonization. 16S rRNA gene sequencing of feces revealed inherent differences in microbial composition. This colonization resistance was transferred from MR1-/- to WT mice via fecal microbiota transplantation, suggesting that MR1-dependent factors influence the microbiota, leading to CDI susceptibility.


Subject(s)
Clostridium Infections/immunology , Disease Resistance/genetics , Gastrointestinal Microbiome/immunology , Histocompatibility Antigens Class I/genetics , Minor Histocompatibility Antigens/genetics , Animals , Anti-Bacterial Agents/administration & dosage , Anti-Bacterial Agents/adverse effects , Cefoperazone/administration & dosage , Cefoperazone/adverse effects , Clostridium Infections/etiology , Clostridium Infections/microbiology , Clostridium Infections/therapy , Disease Models, Animal , Disease Resistance/immunology , Fecal Microbiota Transplantation , Feces/microbiology , Gastrointestinal Microbiome/drug effects , Histocompatibility Antigens Class I/immunology , Humans , Intestinal Mucosa/cytology , Intestinal Mucosa/immunology , Intestinal Mucosa/microbiology , Mice , Mice, Knockout , Minor Histocompatibility Antigens/immunology , Mucosal-Associated Invariant T Cells/immunology , Specific Pathogen-Free Organisms
13.
Infect Immun ; 87(6)2019 06.
Article in English | MEDLINE | ID: mdl-30936157

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.


Subject(s)
Anti-Bacterial Agents/pharmacology , Bacteriophages/physiology , Enterococcus faecalis/drug effects , Enterococcus faecalis/virology , Enterococcus faecium/virology , Gram-Positive Bacterial Infections/therapy , Intestines/microbiology , Animals , Biological Therapy , Enterococcus faecalis/immunology , Enterococcus faecalis/physiology , Enterococcus faecium/drug effects , Enterococcus faecium/immunology , Enterococcus faecium/physiology , Female , Gram-Positive Bacterial Infections/microbiology , Humans , Male , Mice , Mice, Inbred C57BL , Microbial Sensitivity Tests , Vancomycin/pharmacology
14.
mSphere ; 3(5)2018 09 05.
Article in English | MEDLINE | ID: mdl-30185513

ABSTRACT

Clostridium difficile is a Gram-positive obligate anaerobe that forms spores in order to survive for long periods in the unfavorable environment outside a host. C. difficile is the leading cause of nosocomial infectious diarrhea worldwide. C. difficile infection (CDI) arises after a patient treated with broad-spectrum antibiotics ingests infectious spores. The first step in C. difficile pathogenesis is the metabolic reactivation of dormant spores within the gastrointestinal (GI) tract through a process known as germination. In this work, we aim to elucidate the specific conditions and the location within the GI tract that facilitate this process. Our data suggest that C. difficile germination occurs through a two-step biochemical process that is regulated by pH and bile salts, amino acids, and calcium present within the GI tract. Maximal germination occurs at a pH ranging from 6.5 to 8.5 in the terminal small intestine prior to bile salt and calcium reabsorption by the host. Germination can be initiated by lower concentrations of germinants when spores are incubated with a combination of bile salts, calcium, and amino acids, and this synergy is dependent on the availability of calcium. The synergy described here allows germination to proceed in the presence of inhibitory bile salts and at physiological concentrations of germinants, effectively decreasing the concentrations of nutrients required to initiate an essential step of pathogenesis.IMPORTANCEClostridium difficile is an anaerobic spore-forming human pathogen that is the leading cause of nosocomial infectious diarrhea worldwide. Germination of infectious spores is the first step in the development of a C. difficile infection (CDI) after ingestion and passage through the stomach. This study investigates the specific conditions that facilitate C. difficile spore germination, including the following: location within the gastrointestinal (GI) tract, pH, temperature, and germinant concentration. The germinants that have been identified in culture include combinations of bile salts and amino acids or bile salts and calcium, but in vitro, these function at concentrations that far exceed normal physiological ranges normally found in the mammalian GI tract. In this work, we describe and quantify a previously unreported synergy observed when bile salts, calcium, and amino acids are added together. These germinant cocktails improve germination efficiency by decreasing the required concentrations of germinants to physiologically relevant levels. Combinations of multiple germinant types are also able to overcome the effects of inhibitory bile salts. In addition, we propose that the acidic conditions within the GI tract regulate C. difficile spore germination and could provide a biological explanation for why patients taking proton pump inhibitors are associated with increased risk of developing a CDI.


Subject(s)
Bile Acids and Salts/metabolism , Calcium/metabolism , Clostridioides difficile/physiology , Intestine, Small/microbiology , Spores, Bacterial/physiology , Amino Acids/metabolism , Animals , Bacterial Proteins/metabolism , Calcium Signaling , Hydrogen-Ion Concentration , Intestine, Small/metabolism , Mice , Mice, Inbred C57BL
15.
J Bacteriol ; 200(20)2018 10 15.
Article in English | MEDLINE | ID: mdl-30061354

ABSTRACT

In Bordetella pertussis, two serologically distinct fimbriae, FIM2 and FIM3, undergo on/off phase variation independently of each other via variation in the lengths of C stretches in the promoters for their major subunit genes, fim2 and fim3 These two promoters are also part of the BvgAS virulence regulon and therefore, if in an on configuration, are activated by phosporylated BvgA (BvgA~P) under normal growth conditions (Bvg+ mode) but not in the Bvg- mode, inducible by growth in medium containing MgSO4 or other compounds, termed modulators. In the B. pertussis Tohama I strain (FIM2+ FIM3-), the fim3 promoter is in the off state. However, a high level of transcription of the fim3 gene is observed in the Bvg- mode. In this study, we provide an explanation for this anomalous behavior by defining a Bvg-repressed promoter (BRP), located approximately 400 bp upstream of the Pfim3 transcriptional start. Although transcription of the fim3 gene in the Bvg- mode resulted in Fim3 translation, as measured by LacZ translational fusions, no accumulation of Fim3 protein was detectable. We propose that Fim3 protein resulting from translation of mRNA driven by BRP in the Bvg- mode is unstable due to a lack of the fimbrial assembly apparatus encoded by the fimBC genes, located within the fha operon, and therefore is not expressed in the Bvg- mode.IMPORTANCE In Bordetella pertussis, the promoter Pfim3-15C for the major fimbrial subunit gene fim3 is activated by the two-component system BvgAS in the Bvg+ mode but not in the Bvg- mode. However, many transcriptional profiling studies have shown that fim3 is transcribed in the Bvg- mode even when Pfim3 is in a nonpermissive state (Pfim3-13C), suggesting the presence of a reciprocally regulated element upstream of Pfim3 Here, we provide evidence that BRP is the cause of this anomalous behavior of fim3 Although BRP effects vrg-like transcription of fim3 in the Bvg- mode, it does not lead to stable production of FIM3 fimbriae, because expression of the chaperone and usher proteins FimB and FimC occurs only in the Bvg+ mode.


Subject(s)
Antigens, Bacterial/genetics , Bacterial Proteins/genetics , Bordetella pertussis/genetics , Fimbriae Proteins/genetics , Fimbriae, Bacterial/metabolism , Promoter Regions, Genetic , Trans-Activators/genetics , Virulence Factors, Bordetella/genetics , Amino Acid Sequence , Antigens, Bacterial/metabolism , Base Sequence , Fimbriae Proteins/metabolism , Gene Expression Regulation, Bacterial , Lac Operon , Serogroup , Virulence Factors, Bordetella/metabolism
16.
Infect Immun ; 86(8)2018 08.
Article in English | MEDLINE | ID: mdl-29866903

ABSTRACT

Clostridium difficile is an anaerobic, spore-forming bacterium capable of colonizing the gastrointestinal tract of humans following disruption of the normal microbiota, typically from antibiotic therapy for an unrelated infection. With approximately 500,000 confirmed infections leading to 29,000 deaths per year in the United States, C. difficile infection (CDI) is an urgent public health threat. We previously determined that C. difficile survives in up to 3% oxygen. Low levels of oxygen are present in the intestinal tract, with the higher concentrations being associated with the epithelial cell surface. Additionally, antibiotic treatment, the greatest risk factor for CDI, increases the intestinal oxygen concentration. Therefore, we hypothesized that the C. difficile genome encodes mechanisms for survival during oxidative stress. Previous data have shown that cysteine desulfurases involved in iron-sulfur cluster assembly are involved in protecting bacteria from oxidative stress. In this study, deletion of a putative cysteine desulfurase (Cd630_12790/IscS2) involved in the iron-sulfur cluster (Isc) system caused a severe growth defect in the presence of 2% oxygen. Additionally, this mutant delayed colonization in a conventional mouse model of CDI and failed to colonize in a germfree model, which has higher intestinal oxygen levels. These data imply an undefined role for this cysteine desulfurase in protecting C. difficile from low levels of oxygen in the gut.


Subject(s)
Carbon-Sulfur Lyases/metabolism , Clostridioides difficile/enzymology , Clostridioides difficile/physiology , Microbial Viability/drug effects , Oxygen/metabolism , Oxygen/toxicity , Animals , Carbon-Sulfur Lyases/genetics , Clostridioides difficile/growth & development , Clostridium Infections/microbiology , Clostridium Infections/pathology , Disease Models, Animal , Gastrointestinal Tract/microbiology , Gene Deletion , Mice, Inbred C57BL , Oxidative Stress
17.
J Bacteriol ; 200(16)2018 08 15.
Article in English | MEDLINE | ID: mdl-29760211

ABSTRACT

Germination of Clostridium difficile spores is a crucial early requirement for colonization of the gastrointestinal tract. Likewise, C. difficile cannot cause disease pathologies unless its spores germinate into metabolically active, toxin-producing cells. Recent advances in our understanding of C. difficile spore germination mechanisms indicate that this process is both complex and unique. This review defines unique aspects of the germination pathways of C. difficile and compares them to those of two other well-studied organisms, Bacillus anthracis and Clostridium perfringensC. difficile germination is unique, as C. difficile does not contain any orthologs of the traditional GerA-type germinant receptor complexes and is the only known sporeformer to require bile salts in order to germinate. While recent advances describing C. difficile germination mechanisms have been made on several fronts, major gaps in our understanding of C. difficile germination signaling remain. This review provides an updated, in-depth summary of advances in understanding of C. difficile germination and potential avenues for the development of therapeutics, and discusses the major discrepancies between current models of germination and areas of ongoing investigation.


Subject(s)
Clostridioides difficile/physiology , Spores, Bacterial/growth & development , Bacillus anthracis/physiology , Bacterial Proteins/metabolism , Clostridioides difficile/pathogenicity , Clostridium Infections/drug therapy , Clostridium perfringens/physiology , Humans , Membrane Proteins/genetics , Membrane Proteins/metabolism
18.
Pathog Dis ; 76(2)2018 03 01.
Article in English | MEDLINE | ID: mdl-29390060

ABSTRACT

Clostridium difficile (Cd) is an anaerobic, spore-forming bacterium capable of colonizing the gastrointestinal tract of humans. Colonization usually occurs following antibiotic-induced disruption of the host microbiota, which also leads to an increase in oxygen within the gastrointestinal tract. We sought to understand how Cd responds to this microaerophilic condition that is likely experienced within the host. Transcriptome profiling showed differential regulation of genes involved in sugar metabolism, pyruvate metabolism and stress responses. These data provide insight into potential mechanisms of Cd adaptation to the host environment and should lead to the elucidation unknown mechanisms of Cd oxygen resistance and pathogenesis.


Subject(s)
Anaerobiosis , Clostridioides difficile/drug effects , Clostridioides difficile/growth & development , Gene Expression Profiling , Oxygen/toxicity , Adaptation, Physiological , Humans , Stress, Physiological
19.
Pathog Dis ; 76(2)2018 03 01.
Article in English | MEDLINE | ID: mdl-29390127

ABSTRACT

Clostridium difficile (Cd) is the leading cause of antibiotic-associated diarrhea. During an infection, Cd must compete with both the host and other commensal bacteria to acquire iron. Iron is essential for many cell processes, but it can also cause damage if allowed to form reactive hydroxyl radicals. In all organisms, levels of free iron are tightly regulated as are processes utilizing iron molecules. Genome-wide transcriptional analysis of Cd grown in iron-depleted conditions revealed significant changes in expression of genes involved in iron transport, metabolism and virulence. These data will aid future studies examining Cd colonization and the requirements for growth in vivo during an infection.


Subject(s)
Clostridioides difficile/genetics , Clostridioides difficile/metabolism , Gene Expression Regulation, Bacterial , Iron/metabolism , Gene Expression Profiling , Membrane Transport Proteins/biosynthesis , Stress, Physiological , Trace Elements/metabolism , Virulence Factors/biosynthesis
20.
Microbiol Spectr ; 5(5)2017 10.
Article in English | MEDLINE | ID: mdl-28975881

ABSTRACT

Interest in the use of bacteria-containing products for the treatment or prevention of disease has increased in recent years. Bacterial preparations for human consumption are commercially available in the form of dietary supplements and typically contain strains with a history of use in food fermentation. Advances in our understanding of the role of the microbiota in health and disease are likely to lead to development of products containing more novel bacterial species, along with genetic modification of strains to provide specific functions. By law, any substance intended for use in the diagnosis, cure, mitigation, treatment, or prevention of disease in humans meets the definition of a drug, and an Investigational New Drug (IND) application for clinical investigation must be filed with the FDA. This article is meant to provide information about the IND submission process and additional considerations with regard to chemistry, manufacturing, and controls information for live biotherapeutic products.


Subject(s)
Biological Products/standards , Biological Therapy/standards , Investigational New Drug Application , Bacteria/genetics , Bacteria/metabolism , Drug Approval/legislation & jurisprudence , Drug Design , Humans , Investigational New Drug Application/legislation & jurisprudence , Manufacturing and Industrial Facilities/legislation & jurisprudence , Microbiota , Microorganisms, Genetically-Modified , United States , United States Food and Drug Administration/legislation & jurisprudence
SELECTION OF CITATIONS
SEARCH DETAIL
...