Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 3.789
Filter
1.
PLoS One ; 19(9): e0307333, 2024.
Article in English | MEDLINE | ID: mdl-39288108

ABSTRACT

The search for alternative therapies to antimicrobial growth promoters (AGP) in poultry production has gained momentum in the past years because of consumer preference and government restrictions on the use of AGP in animal production. Flavonoids are plant-derived metabolites that have been studied for their health-promoting properties that could potentially be used as an alternative to AGP in poultry. In a previous study, we showed that the inclusion of a flavonoid-rich corn cultivar (PennHFD1) in the diet improved the health of broilers undergoing necrotic enteritis. However, the mechanisms of action by which the PennHFD1-based diet ameliorated necrotic enteritis are unknown. This study describes the microbial diversity and composition of the jejunum and ileum of chickens co-infected with Eimeria maxima and Clostridium perfringens and treated with a high-flavonoid corn-based diet. Luminal content and mucosal samples from the jejunum and ileum were collected for DNA extraction, 16S rRNA amplicon sequencing and data analyses. The infection model and the dietary treatments significantly changed the alfa diversity indices (Mucosal samples: ASVs, P = 0.04; Luminal content samples: ASVs, P = 0.03), and beta diversities (Mucosal samples: P < 0.01, Luminal content: P < 0.01) of the ileal samples but not those of the jejunal samples. The microbial composition revealed that birds fed the high-flavonoid corn diet had a lower relative abundance of C. perfringens compared to birds fed the commercial corn diet. The treatments also changed the relative abundance of other bacteria that are related to gut health, such as Lactobacillus. We concluded that both the infection model and the dietary high-flavonoid corn changed the broilers' gut microbial diversity and composition. In addition, the decrease in the relative abundance of C. perfringens corroborates with a decrease in mortality and intestinal lesions due to necrotic enteritis. Collecting different segments and sample types provided a broader understanding of the changes in the gut microbiota among treatments.


Subject(s)
Chickens , Clostridium perfringens , Enteritis , Flavonoids , Gastrointestinal Microbiome , Poultry Diseases , Zea mays , Animals , Chickens/microbiology , Zea mays/microbiology , Enteritis/veterinary , Enteritis/microbiology , Enteritis/diet therapy , Gastrointestinal Microbiome/drug effects , Clostridium perfringens/pathogenicity , Clostridium perfringens/isolation & purification , Poultry Diseases/microbiology , Animal Feed , Eimeria , RNA, Ribosomal, 16S/genetics , Clostridium Infections/veterinary , Clostridium Infections/microbiology , Necrosis , Jejunum/microbiology , Coccidiosis/veterinary , Ileum/microbiology , Diet/veterinary
2.
BMC Infect Dis ; 24(1): 989, 2024 Sep 17.
Article in English | MEDLINE | ID: mdl-39289598

ABSTRACT

BACKGROUND: The nosocomial transmission of toxin-producing Clostridioides difficile is a significant concern in infection control. C. difficile, which resides in human intestines, poses a risk of transmission, especially when patients are in close contact with medical staff. METHODS: To investigate the nosocomial transmission of C. difficile in a single center, we analyzed the genetic relationships of the bacteria. This was done using draft whole-genome sequencing (WGS) and examining single nucleotide polymorphisms (SNPs) in core-genome, alongside data regarding the patient's hospital wards and room changes. Our retrospective analysis covered 38 strains, each isolated from a different patient, between April 2014 and January 2015. RESULTS: We identified 38 strains that were divided into 11 sequence types (STs). ST81 was the most prevalent (n = 11), followed by ST183 (n = 10) and ST17 (n = 7). A cluster of strains that indicated suspected nosocomial transmission (SNT) was identified through SNP analysis. The draft WGS identified five clusters, with 16 of 38 strains belonging to these clusters. There were two clusters for ST81 (ST81-SNT-1 and ST81-SNT-2), two for ST183 (ST183-SNT-1 and ST183-SNT-2), and one for ST17 (ST17-SNT-1). ST183-SNT-1 was the largest SNT cluster, encompassing five patients who were associated with Wards A, B, and K. The most frequent room changer was a patient labeled Pt08, who changed rooms seven times in Ward B. Patients Pt36 and Pt10, who were also in Ward B, had multiple admissions and discharges during the study period. CONCLUSIONS: Additional culture tests and SNP analysis of C. difficile using draft WGS revealed silent transmission within the wards, particularly in cases involving frequent room changes and repeated admissions and discharges. Monitoring C. difficile transmission using WGS-based analysis could serve as a valuable marker in infection control management.


Subject(s)
Clostridioides difficile , Clostridium Infections , Cross Infection , Molecular Epidemiology , Polymorphism, Single Nucleotide , Whole Genome Sequencing , Humans , Clostridioides difficile/genetics , Clostridioides difficile/classification , Clostridioides difficile/isolation & purification , Clostridium Infections/transmission , Clostridium Infections/epidemiology , Clostridium Infections/microbiology , Cross Infection/transmission , Cross Infection/epidemiology , Cross Infection/microbiology , Retrospective Studies , Female , Male , Genome, Bacterial , Aged , Middle Aged , Hospitals , Aged, 80 and over , Adult
3.
Sci Rep ; 14(1): 21559, 2024 09 16.
Article in English | MEDLINE | ID: mdl-39284883

ABSTRACT

Clostridioides difficile, a cause of healthcare-associated infections, poses a significant global health threat. This multi-institutional retrospective study focuses on epidemic dynamics, emphasizing minor and toxin-negative clinical isolates through high-resolution genotyping. The genotype of the C. difficile clinical isolates during 2005 to 2022 was gathered from 14 hospitals across Japan (N = 982). The total number of unique genotypes was 294. Some genotypes were identified in every hospital (cross-regional genotypes), while others were unique to a specific hospital or those in close geographic proximity (region-specific genotypes). Notably, a hospital located in a sparsely populated prefecture exhibited the highest prevalence of region-specific genotypes. The isolation rate of cross-regional genotypes positively correlated with the human mobility flow. A 6-month interval analysis at a university hospital from 2019 to 2021 revealed a temporal transition of the genotype dominance. The frequent isolation of identical genotypes over a brief timeframe did not always align with the current criteria for defining nosocomial outbreaks. This study highlights the presence of diverse indigenous C. difficile strains in regional environments. The cross-regional strains may have a higher competency to spread in the human community. The longitudinal analysis underscores the need for further investigation into potential nosocomial spread.


Subject(s)
Clostridioides difficile , Clostridium Infections , Cross Infection , Genotype , Clostridioides difficile/genetics , Clostridioides difficile/isolation & purification , Clostridioides difficile/classification , Humans , Japan/epidemiology , Clostridium Infections/epidemiology , Clostridium Infections/microbiology , Cross Infection/microbiology , Cross Infection/epidemiology , Retrospective Studies , Male , Female , Hospitals
4.
Sci Rep ; 14(1): 20607, 2024 09 04.
Article in English | MEDLINE | ID: mdl-39232075

ABSTRACT

Biofilm formation and toxin production are some of the virulence factors of Clostridioides difficile (C. difficile), which causes hospital-acquired C. difficile infection (HA-CDI). This work investigated the prevalence and distribution of different strains recovered from HA-CDI patients hospitalized in 4 medical centres across Israel, and characterized strains' virulence factors and antibiotic susceptibility. One-hundred and eighty-eight faecal samples were collected. C. difficile 's toxins were detected by the CerTest Clostridium difficile GDH + Toxin A + B combo card test kit. Toxin loci PaLoc and PaCdt were detected by whole-genome sequencing (WGS). Multi-locus sequence typing (MLST) was performed to classify strains. Biofilm production was assessed by crystal violet. Antibiotic susceptibility was determined using Etest. Fidaxomicin susceptibility was tested via agar dilution. Sequence type (ST) 42 was the most (13.8%) common strain. All strains harboured the 2 toxins genes; 6.9% had the binary toxin. Most isolates were susceptible to metronidazole (98.9%) and vancomycin (99.5%). Eleven (5.85%) isolates were fidaxomicin-resistant. Biofilm production capacity was associated with ST (p < 0.001). In conclusion, a broad variety of C. difficile strains circulate in Israel's medical centres. Further studies are needed to explore the differences and their contribution to HA-CDI epidemiology.


Subject(s)
Anti-Bacterial Agents , Biofilms , Clostridioides difficile , Clostridium Infections , Cross Infection , Microbial Sensitivity Tests , Virulence Factors , Clostridioides difficile/genetics , Clostridioides difficile/drug effects , Clostridioides difficile/isolation & purification , Clostridioides difficile/pathogenicity , Humans , Israel/epidemiology , Clostridium Infections/microbiology , Clostridium Infections/epidemiology , Anti-Bacterial Agents/pharmacology , Virulence Factors/genetics , Male , Female , Biofilms/drug effects , Biofilms/growth & development , Cross Infection/microbiology , Cross Infection/epidemiology , Aged , Middle Aged , Multilocus Sequence Typing , Adult , Aged, 80 and over , Whole Genome Sequencing , Feces/microbiology
5.
PeerJ ; 12: e17776, 2024.
Article in English | MEDLINE | ID: mdl-39224820

ABSTRACT

Background: The tcdA gene codes for an important toxin produced by Clostridioides difficile (C. difficile), but there is currently no simple and cost-effective method of detecting it. This article establishes and validates a rapid and visual loop-mediated isothermal amplification (LAMP) assay for the detection of the tcdA gene. Methods: Three sets of primers were designed and optimized to amplify the tcdA gene in C. difficile using a LAMP assay. To evaluate the specificity of the LAMP assay, C. difficile VPI10463 was used as a positive control, while 26 pathogenic bacterial strains lacking the tcdA gene and distilled water were utilized as negative controls. For sensitivity analysis, the LAMP assay was compared to PCR using ten-fold serial dilutions of DNA from C. difficile VPI10463, ranging from 207 ng/µl to 0.000207 pg/µl. The tcdA gene of C.difficile was detected in 164 stool specimens using both LAMP and polymerase chain reaction (PCR). Positive and negative results were distinguished using real-time monitoring of turbidity and chromogenic reaction. Results: At a temperature of 66 °C, the target DNA was successfully amplified with a set of primers designated, and visualized within 60 min. Under the same conditions, the target DNA was not amplified with the tcdA12 primers for 26 pathogenic bacterial strains that do not carry the tcdA gene. The detection limit of LAMP was 20.700 pg/µl, which was 10 times more sensitive than that of conventional PCR. The detection rate of tcdA in 164 stool specimens using the LAMP method was 17% (28/164), significantly higher than the 10% (16/164) detection rate of the PCR method (X2 = 47, p < 0.01). Conclusion: LAMP method is an effective technique for the rapid and visual detection of the tcdA gene of C. difficile, and shows potential advantages over PCR in terms of speed, simplicity, and sensitivity. The tcdA-LAMP assay is particularly suitable for medical diagnostic environments with limited resources and is a promising diagnostic strategy for the screening and detection of C. difficile infection in populations at high risk.


Subject(s)
Bacterial Toxins , Clostridioides difficile , Clostridium Infections , Enterotoxins , Feces , Nucleic Acid Amplification Techniques , Sensitivity and Specificity , Clostridioides difficile/genetics , Clostridioides difficile/isolation & purification , Nucleic Acid Amplification Techniques/methods , Humans , Bacterial Toxins/genetics , Clostridium Infections/diagnosis , Clostridium Infections/microbiology , Feces/microbiology , Feces/chemistry , Enterotoxins/genetics , DNA Primers/genetics , Molecular Diagnostic Techniques/methods , Polymerase Chain Reaction/methods , Adult , Middle Aged
6.
Gut Microbes ; 16(1): 2393766, 2024.
Article in English | MEDLINE | ID: mdl-39224076

ABSTRACT

Clostridioides difficile is a major nosocomial pathogen, causing significant morbidity and mortality worldwide. Antibiotic usage, a major risk factor for Clostridioides difficile infection (CDI), disrupts the gut microbiota, allowing C. difficile to proliferate and cause infection, and can often lead to recurrent CDI (rCDI). Fecal microbiota transplantation (FMT) and live biotherapeutic products (LBPs) have emerged as effective treatments for rCDI and aim to restore colonization resistance provided by a healthy gut microbiota. However, much is still unknown about the mechanisms mediating their success. Bile acids, extensively modified by gut microbes, affect C. difficile's germination, growth, and toxin production while also shaping the gut microbiota and influencing host immune responses. Additionally, microbial interactions, such as nutrient competition and cross-feeding, contribute to colonization resistance against C. difficile and may contribute to the success of microbiota-focused therapeutics. Bile acids as well as other microbial mediated interactions could have implications for other diseases being treated with microbiota-focused therapeutics. This review focuses on the intricate interplay between bile acid modifications, microbial ecology, and host responses with a focus on C. difficile, hoping to shed light on how to move forward with the development of new microbiota mediated therapeutic strategies to combat rCDI and other intestinal diseases.


Subject(s)
Bile Acids and Salts , Clostridioides difficile , Clostridium Infections , Fecal Microbiota Transplantation , Gastrointestinal Microbiome , Bile Acids and Salts/metabolism , Humans , Clostridioides difficile/physiology , Clostridium Infections/therapy , Clostridium Infections/microbiology , Animals
7.
Clin Lab ; 70(9)2024 Sep 01.
Article in English | MEDLINE | ID: mdl-39257107

ABSTRACT

BACKGROUND: In February 2024, our hospital confirmed a case of ocular infection with Clostridium tertium caused by a salute gun explosion. The patient sought medical attention at our hospital due to a salute gun explosion injury in the right eye. Two days ago, a patient mistakenly believed that the fuse was not ignited when firing a salute gun. When observing, the salute gun exploded and injured his right eye. The patient immediately went to the local hos-pital for treatment. The CT scan of the local hospital showed rupture of the right eyeball. For additional diagnosis and treatment, the patient came to our hospital. The patient in this case has an acute onset, severe condition, no additional systemic diseases, and no history of drug or food allergies. METHODS: Intraocular exploration, cranial CT, local and systemic anti infection treatment. Pathogen examination items: bacterial smear, bacterial culture and identification. Venous blood test items: blood routine, liver function, kidney function, and coagulation function. RESULTS: Intraocular exploration showed conjunctival congestion and edema in the right eye, corneal haze and ede-ma, shallow anterior chamber, anterior chamber hemorrhage, and unclear intraocular structure. Clinical treatment: debridement and suturing of right eye rupture + repair of eyeball rupture + removal of intraocular foreign body + repair of superior rectus muscle detachment + anterior chamber flushing + anterior chamber shaping + suture of eyelid laceration. Pathogen examination item: Eye secretion bacterial smear (Gram staining): A large number of gram-positive bacilli were found, and the secretion bacterial culture and identification (MALDI-TOF MS): Clostridium tertium. Auxiliary examination: Blood routine (venous blood): White blood cells 10.89 x 109/L, neutrophil count 9.65 x 109/L, whole blood hypersensitive C-reactive protein 20.28 mg/L, renal function: urea 9.15 mmol/L, uric acid 428.5 µmol/L, fasting glucose 6.48 mmol/L, no further abnormalities observed. Clinical drug treatment plan: Tetanus human immunoglobulin 250 IU im, tobramycin eye drops 0.1 g ext qd, vancomycin 0.5 g ih qd, levofloxacin 0.5g ivgtt qd, aluminum magnesium suspension 15 mL po bid, potassium chloride sustained-release tablets 0.5 g po qd. After 7 days of treatment, the patient's body temperature returned to normal, conjunctival congestion and edema decreased, anterior chamber hemorrhage decreased, corneal incision closed properly, and the patient improved and was discharged. CONCLUSIONS: This article reports a case of ocular infection caused by a salute gun explosion with Clostridium tertium. Clostridium tertium was quickly and accurately identified by a mass spectrometer, and reasonable treatment measures were adopted clinically. The patient improved and was discharged. I hope that in the future, this study can provide assistance for the clinical diagnosis and treatment of special site infections caused by Clostridium tertium.


Subject(s)
Explosions , Humans , Male , Anti-Bacterial Agents/therapeutic use , Eye Infections, Bacterial/diagnosis , Eye Infections, Bacterial/microbiology , Eye Infections, Bacterial/etiology , Blast Injuries/microbiology , Blast Injuries/complications , Blast Injuries/diagnosis , Clostridium Infections/diagnosis , Clostridium Infections/microbiology , Clostridium Infections/therapy , Clostridium/isolation & purification , Adult
8.
Cell Host Microbe ; 32(8): 1219-1224, 2024 Aug 14.
Article in English | MEDLINE | ID: mdl-39146793

ABSTRACT

Fecal microbial transplantation (FMT) for inflammatory diseases or refractory immune checkpoint inhibitor therapy is less effective than for preventing recurrent Clostridioides difficile infection. This commentary outlines strategies to use biomarkers of successful FMT to guide newer approaches to restore microbial homeostasis in individuals with dysbiosis-mediated inflammation.


Subject(s)
Dysbiosis , Fecal Microbiota Transplantation , Gastrointestinal Microbiome , Precision Medicine , Humans , Fecal Microbiota Transplantation/methods , Dysbiosis/therapy , Precision Medicine/methods , Clostridium Infections/therapy , Clostridium Infections/microbiology , Inflammation , Clostridioides difficile , Biomarkers
9.
Age Ageing ; 53(8)2024 Aug 06.
Article in English | MEDLINE | ID: mdl-39141079

ABSTRACT

BACKGROUND: Advanced age has been widely identified as a risk factor for recurrent Clostridioides difficile infection (CDI), but most related studies were performed before the introduction of novel therapies. The aim of this study was to compare CDI characteristics and outcomes in patients over and under 80 years old with CDI and their outcomes in the era of new treatments. METHODS: This was a retrospective cohort study of patients diagnosed with CDI from January 2021 to December 2022 in an academic hospital. We compared recurrence and mortality at 12 weeks after the end of treatment. An extension of the Fine and Grey model adjusted for competing events was used to assess the effect of age on recurrence. RESULTS: Four hundred seventy-six patients were considered to have CDI (320 in patients <80 years and 156 in ≥80 years). CDI in older patients was more frequently healthcare-associated and was more severe. Although the Charlson index was almost identical between populations, comorbidities clearly differed. New treatments (bezlotoxumab, fidaxomicin and faecal microbiota transplantation) were more frequently used in older patients without statistical significance (41.3% vs. 33.4%, P = .053). There were 69 (14.5%) recurrences, with no differences by age group after adjusting for competing events. Mortality was greater in the oldest (35.3%) than in the youngest (13.1%); P < .001. CONCLUSIONS: No differences in CDI recurrence rates were found between age groups. However, there was a high mortality rate in patients ≥80 years old, which emphasises the urgent need to improve the prevention and treatment of CDI in this group.


Subject(s)
Clostridium Infections , Recurrence , Humans , Male , Aged, 80 and over , Clostridium Infections/epidemiology , Clostridium Infections/mortality , Clostridium Infections/microbiology , Clostridium Infections/therapy , Clostridium Infections/diagnosis , Clostridium Infections/drug therapy , Female , Retrospective Studies , Risk Factors , Aged , Age Factors , Fecal Microbiota Transplantation , Anti-Bacterial Agents/therapeutic use , Clostridioides difficile , Middle Aged , Fidaxomicin/therapeutic use , Broadly Neutralizing Antibodies/therapeutic use , Antibodies, Monoclonal
10.
Commun Biol ; 7(1): 947, 2024 Aug 06.
Article in English | MEDLINE | ID: mdl-39103440

ABSTRACT

Clostridium septicum infections are highly predictive of certain malignancies in human patients. To initiate infections, C. septicum spores must first germinate and regain vegetative growth. Yet, what triggers the germination of C. septicum spores is still unknown. Here, we observe that C. septicum germinates in response to specific bile salts. Putative bile salt recognition genes are identified in C. septicum based on their similarity in sequence and organization to bile salt-responsive csp genes in Clostridioides difficile. Inactivating two of these csp orthologs (cspC-82 and cspC-1718) results in mutant spores that no longer germinate in the presence of their respective cognate bile salts. Additionally, inactivating the putative cspBA or sleC genes in C. septicum abrogates the germination response to all bile salt germinants, suggesting that both act at a convergent point downstream of cspC-82 and cspC-1718. Molecular dynamics simulations show that both CspC-82 and CspC-1718 bear a strong structural congruence with C. difficile's CspC. The existence of functional bile salt germination sensors in C. septicum may be relevant to the association between infection and malignancy.


Subject(s)
Bacterial Proteins , Bile Acids and Salts , Clostridioides difficile , Clostridium septicum , Spores, Bacterial , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Bile Acids and Salts/metabolism , Spores, Bacterial/genetics , Clostridioides difficile/genetics , Clostridium septicum/genetics , Molecular Dynamics Simulation , Gene Expression Regulation, Bacterial , Clostridium Infections/microbiology , Carrier Proteins
11.
Gut Microbes ; 16(1): 2392872, 2024.
Article in English | MEDLINE | ID: mdl-39189608

ABSTRACT

We sought to better understand how intestinal microbiota confer protection against Clostridioides difficile (C. difficile) infection (CDI). We utilized gnotobiotic altered Schaedler flora (ASF) mice, which lack the abnormalities of germfree (GF) mice as well as the complexity and heterogeneity of antibiotic-treated mice. Like GF mice, ASF mice were highly prone to rapid lethal CDI, without antibiotics, while very low infectious doses resulted in chronic CDI. Administering such chronic CDI mice an undefined preparation of Clostridia lowered C. difficile levels by several logs. Importantly, such resolution of CDI was associated with colonization of Lachnospiraceae. Fractionation of the Clostridia population to enrich for Lachnospiraceae led to the appreciation that its CDI-impeding property strongly associated with a specific Lachnospiraceae strain, namely uncultured bacteria and archaea (UBA) 3401. UBA3401 was recalcitrant to being propagated as a pure culture but could be maintained in ASF mice, wherein it comprised up to about 50% of the intestinal microbiota, which was sufficient to generate a high-quality genomic sequence of this bacterium. Sequence analysis and ex vivo study of UBA3401 indicated that it had the ability to secrete substance(s) that directly impeded C. difficile growth. Moreover, in vivo administration of UBA3401/ASF feces provided strong protection to C. difficile challenge. Thus, UBA3401 may contribute to and/or provide a means to study microbiota-mediated CDI resistance.


Subject(s)
Clostridiales , Clostridioides difficile , Clostridium Infections , Gastrointestinal Microbiome , Germ-Free Life , Animals , Mice , Clostridioides difficile/genetics , Clostridioides difficile/growth & development , Clostridioides difficile/physiology , Clostridioides difficile/pathogenicity , Clostridium Infections/microbiology , Clostridium Infections/prevention & control , Gastrointestinal Microbiome/drug effects , Clostridiales/genetics , Clostridiales/growth & development , Mice, Inbred C57BL , Disease Models, Animal , Feces/microbiology , Female , Anti-Bacterial Agents/pharmacology
12.
PLoS One ; 19(8): e0306622, 2024.
Article in English | MEDLINE | ID: mdl-39116083

ABSTRACT

Clostridioides difficile infection (CDI) is a significant public health threat, associated with antibiotic-induced disruption of the normally protective gastrointestinal microbiota. CDI is thought to occur in two stages: acquisition of asymptomatic colonization from ingesting C. difficile bacteria followed by progression to symptomatic CDI caused by toxins produced during C. difficile overgrowth. The degree to which disruptive antibiotic exposure increases susceptibility at each stage is uncertain, which might contribute to divergent published projections of the impact of hospital antibiotic stewardship interventions on CDI. Here, we model C. difficile transmission and CDI among hospital inpatients, including exposure to high-CDI-risk antibiotics and their effects on each stage of CDI epidemiology. We derive the mathematical relationship, using a deterministic model, between those parameters and observed equilibrium levels of colonization, CDI, and risk ratio of CDI among certain antibiotic-exposed patients relative to patients with no recent antibiotic exposure. We then quantify the sensitivity of projected antibiotic stewardship intervention impacts to alternate assumptions. We find that two key parameters, the antibiotic effects on susceptibility to colonization and to CDI progression, are not identifiable given the data frequently available. Furthermore, the effects of antibiotic stewardship interventions are sensitive to their assumed values. Thus, discrepancies between different projections of antibiotic stewardship interventions may be largely due to model assumptions. Data supporting improved quantification of mechanistic antibiotic effects on CDI epidemiology are needed to understand stewardship effects better.


Subject(s)
Anti-Bacterial Agents , Clostridioides difficile , Clostridium Infections , Humans , Clostridium Infections/epidemiology , Clostridium Infections/microbiology , Clostridium Infections/drug therapy , Anti-Bacterial Agents/adverse effects , Anti-Bacterial Agents/therapeutic use , Anti-Bacterial Agents/pharmacology , Clostridioides difficile/drug effects , Antimicrobial Stewardship , Health Facilities , Cross Infection/epidemiology , Cross Infection/microbiology , Cross Infection/drug therapy , Risk Factors , Models, Theoretical , Gastrointestinal Microbiome/drug effects
14.
Anaerobe ; 89: 102894, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39122138

ABSTRACT

BACKGROUND: Antibiotic exposure is a known risk factor for Clostridioides difficile infection (CDI) and recurrence and can lead to infection with specific C. difficile strains. In this study, we sought to explore the relationship between antecedent antibiotic exposure and C. difficile antimicrobial resistance, and the impact of resistance on clinical outcomes. METHODS: This was a single center retrospective study evaluating patients with CDI between 2011 and 2021. A logistic regression model was used to evaluate the relationship between antecedent antibiotics in the 30 days prior to CDI and resistance among isolates. In addition, an exploratory analysis using a cause-specific Cox proportional hazards model evaluated the association between resistance and a composite outcome of clinical failure, relapse at 30 days or CDI-related death. RESULTS: we analyzed one isolate from 510 patients; resistance was noted in 339 (66.5 %) of the isolates. Exposure to fluoroquinolones and macrolides was associated with 2.4 (95 % CI 1.4-4.4) and 4.7 (95 % CI 1.1-20.5) increased odds of having resistance compared to other antibiotic class exposure, respectively. There were 58 (17.0 %) patients in the resistance group who developed the composite outcome and 24 (14.2 %) patients who lacked resistance who developed the composite outcome (HR 1.32, 95 % CI 0.81-2.14). CONCLUSION: These findings suggest that fluoroquinolone and macrolide exposure were significantly associated with isolating a resistant strain, but we did not find significant differences in clinical outcomes based on the presence of antimicrobial resistance.


Subject(s)
Anti-Bacterial Agents , Clostridioides difficile , Clostridium Infections , Ribotyping , Humans , Anti-Bacterial Agents/adverse effects , Anti-Bacterial Agents/therapeutic use , Anti-Bacterial Agents/pharmacology , Clostridium Infections/microbiology , Clostridium Infections/epidemiology , Clostridium Infections/drug therapy , Retrospective Studies , Male , Female , Aged , Clostridioides difficile/drug effects , Clostridioides difficile/genetics , Clostridioides difficile/isolation & purification , Clostridioides difficile/classification , Middle Aged , Drug Resistance, Bacterial , Treatment Outcome , Aged, 80 and over , Adult , Recurrence
15.
Anaerobe ; 89: 102899, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39142535

ABSTRACT

OBJECTIVE: Flagellin protein, an integral component of flagella, provides motility to several bacterial species and also acts as a candidate antigen in diagnostics and subunit vaccines. The bulk production of flagellin with retention of all conformational epitopes using recombinant protein technology is of paramount importance in the development of pathogen-specific immuno-assays and vaccines. We describe the production of highly soluble and immuno-reactive rFliA(C) protein of Clostridium chauvoei, a causative agent of blackleg or black quarter (BQ) affecting cattle and small ruminants worldwide. The bacterium is known to possess peritrichous flagella that provide motility and also act as a virulence factor with high protective antigenicity. METHODS: Upon sequence and structural analysis, a partial fliA(C) gene from Clostridium chauvoei was cloned and the recombinant mature protein with N- and C- terminal truncation was over-expressed as a His-tagged fusion protein (∼25 kDa) in Escherichia coli. Subsequently, rFliA(C) protein was purified by single-step affinity chromatography and characterized for its immuno-reactivity in laboratory animals, Western blot, and indirect-ELISA format. RESULTS: rFliA(C) was highly soluble and was purified in high quantity and quality. rFliA(C) elicited antigen-specific conformational polyclonal antibodies in rabbit and guinea pig models, as well as anti-Clostridium chauvoei-specific antibodies being specifically detected in BQ-vaccinated and convalescent sera of bovines in Western blot and in indirect-ELISA format. Further, no cross reactivity was noted with antibodies against major bovine diseases (e.g., foot-and-mouth disease, IBR, LSDV, hemorrhagic septicaemia, brucellosis, and leptospirosis). CONCLUSION: The study indicated the production of conformational recombinant flagellin-rFliA(C)-antigen and its potential utility in development of diagnostics for detection of Clostridium chauvoei-specific antibodies in BQ-recovered and/or vaccinated animals.


Subject(s)
Antibodies, Bacterial , Clostridium chauvoei , Flagellin , Recombinant Proteins , Flagellin/immunology , Flagellin/genetics , Animals , Clostridium chauvoei/immunology , Clostridium chauvoei/genetics , Recombinant Proteins/immunology , Recombinant Proteins/genetics , Rabbits , Antibodies, Bacterial/blood , Antibodies, Bacterial/immunology , Guinea Pigs , Bacterial Vaccines/immunology , Bacterial Vaccines/genetics , Clostridium Infections/veterinary , Clostridium Infections/immunology , Clostridium Infections/microbiology , Cattle , Antigens, Bacterial/immunology , Antigens, Bacterial/genetics , Cattle Diseases/immunology , Cattle Diseases/microbiology , Escherichia coli/genetics , Escherichia coli/metabolism , Enzyme-Linked Immunosorbent Assay , Cloning, Molecular
16.
Nat Commun ; 15(1): 7416, 2024 Aug 28.
Article in English | MEDLINE | ID: mdl-39198411

ABSTRACT

The human gut pathogen Clostridioides difficile displays substantial inter-strain genetic variability and confronts a changeable nutrient landscape in the gut. We examined how human gut microbiota inter-species interactions influence the growth and toxin production of various C. difficile strains across different nutrient environments. Negative interactions influencing C. difficile growth are prevalent in an environment containing a single highly accessible resource and sparse in an environment containing C. difficile-preferred carbohydrates. C. difficile toxin production displays significant community-context dependent variation and does not trend with growth-mediated inter-species interactions. C. difficile strains exhibit differences in interactions with Clostridium scindens and the ability to compete for proline. Further, C. difficile shows substantial differences in transcriptional profiles in co-culture with C. scindens or Clostridium hiranonis. C. difficile exhibits massive alterations in metabolism and other cellular processes in co-culture with C. hiranonis, reflecting their similar metabolic niches. C. hiranonis uniquely inhibits the growth and toxin production of diverse C. difficile strains across different nutrient environments and robustly ameliorates disease severity in mice. In sum, understanding the impact of C. difficile strain variability and nutrient environments on inter-species interactions could help improve the effectiveness of anti-C. difficile strategies.


Subject(s)
Clostridioides difficile , Clostridium Infections , Coculture Techniques , Gastrointestinal Microbiome , Clostridioides difficile/genetics , Clostridioides difficile/metabolism , Clostridioides difficile/physiology , Humans , Animals , Mice , Clostridium Infections/microbiology , Nutrients/metabolism , Bacterial Toxins/metabolism , Bacterial Toxins/genetics , Microbial Interactions , Clostridium/metabolism , Clostridium/genetics , Female , Antibiosis , Mice, Inbred C57BL
17.
Int J Mol Sci ; 25(16)2024 Aug 08.
Article in English | MEDLINE | ID: mdl-39201359

ABSTRACT

Clostridioides difficile detection in community settings is time-intensive, resulting in delays in diagnosing and quarantining infected individuals. However, with the advent of semi-automated devices and improved algorithms in recent decades, the ability to discern CDI infection from asymptomatic carriage has significantly improved. This, in turn, has led to efficiently regulated monitoring systems, further reducing endemic risk, with recent concerns regarding a possible surge in hospital-acquired Clostridioides difficile infections post-COVID failing to materialize. This review highlights established and emerging technologies used to detect community-acquired Clostridioides difficile in research and clinical settings.


Subject(s)
Clostridioides difficile , Clostridium Infections , Humans , Clostridium Infections/diagnosis , Clostridium Infections/microbiology , Clostridioides difficile/pathogenicity , COVID-19/diagnosis
18.
Microbiome ; 12(1): 119, 2024 Jul 01.
Article in English | MEDLINE | ID: mdl-38951925

ABSTRACT

BACKGROUND: Fecal microbiota transplantation (FMT) and fecal virome transplantation (FVT, sterile filtrated donor feces) have been effective in treating recurrent Clostridioides difficile infections, possibly through bacteriophage-mediated modulation of the gut microbiome. However, challenges like donor variability, costly screening, coupled with concerns over pathogen transfer (incl. eukaryotic viruses) with FMT or FVT hinder their wider clinical application in treating less acute diseases. METHODS: To overcome these challenges, we developed methods to broaden FVT's clinical application while maintaining efficacy and increasing safety. Specifically, we employed the following approaches: (1) chemostat-fermentation to reproduce the bacteriophage FVT donor component and remove eukaryotic viruses (FVT-ChP), (2) solvent-detergent treatment to inactivate enveloped viruses (FVT-SDT), and (3) pyronin-Y treatment to inhibit RNA virus replication (FVT-PyT). We assessed the efficacy of these processed FVTs in a C. difficile infection mouse model and compared them with untreated FVT (FVT-UnT), FMT, and saline. RESULTS: FVT-SDT, FVT-UnT, and FVT-ChP reduced the incidence of mice reaching the humane endpoint (0/8, 2/7, and 3/8, respectively) compared to FMT, FVT-PyT, and saline (5/8, 7/8, and 5/7, respectively) and significantly reduced the load of colonizing C. difficile cells and associated toxin A/B levels. There was a potential elimination of C. difficile colonization, with seven out of eight mice treated with FVT-SDT testing negative with qPCR. In contrast, all other treatments exhibited the continued presence of C. difficile. Moreover, the results were supported by changes in the gut microbiome profiles, cecal cytokine levels, and histopathological findings. Assessment of viral engraftment following FMT/FVT treatment and host-phage correlations analysis suggested that transfer of phages likely were an important contributing factor associated with treatment efficacy. CONCLUSIONS: This proof-of-concept study shows that specific modifications of FVT hold promise in addressing challenges related to donor variability and infection risks. Two strategies lead to treatments significantly limiting C. difficile colonization in mice, with solvent/detergent treatment and chemostat propagation of donor phages emerging as promising approaches. Video Abstract.


Subject(s)
Bacteriophages , Clostridioides difficile , Clostridium Infections , Fecal Microbiota Transplantation , Feces , Gastrointestinal Microbiome , Fecal Microbiota Transplantation/methods , Animals , Mice , Bacteriophages/physiology , Bacteriophages/isolation & purification , Clostridium Infections/therapy , Clostridium Infections/microbiology , Feces/microbiology , Feces/virology , Disease Models, Animal , Humans , Mice, Inbred C57BL , Female
19.
EBioMedicine ; 106: 105244, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39018757

ABSTRACT

BACKGROUND: Clostridioides difficile infection results in life-threatening short-term outcomes and the potential for subsequent recurrent infection. Predicting these outcomes at diagnosis, when important clinical decisions need to be made, has proven to be a difficult task. METHODS: 52 clinical features from existing models or the literature were collected retrospectively within ±48 h of diagnosis among 1660 inpatient infections. A modified desirability of outcome ranking (DOOR) was designed to encompass clinically-important severe events attributable to the acute infection (intensive care transfer due to sepsis, shock, colectomy/ileostomy, mortality) and/or 60-day recurrence. A deep neural network was constructed and interpreted using SHapley Additive exPlanations (SHAP). High-importance features were used to train a reduced, shallow network and performance was compared to existing conventional models (7 severity, 7 recurrence; after summing DOOR probabilities to align with conventional binary outputs) using area under the ROC curve (AUROC) and DeLong tests. FINDINGS: The full (52-feature) model achieved an out-of-sample AUROC 0.823 for severity and 0.678 for recurrence. SHAP identified 13 unique, highly-important features (age, hypotension, initial treatment, onset, PCR cycle threshold, number of prior episodes, antibiotic exposure, fever, hypotension, pressors, leukocytosis, creatinine, lactate) that were used to train a reduced model, which performed similarly to the full model (severity AUROC difference P = 0.130; recurrence P = 0.426) and significantly better than the top severity model (reduced model predicting severity 0.837, ATLAS 0.749; P = 0.001). The reduced model also outperformed the top recurrence model, but this was not statistically-significant (reduced model recurrence AUROC 0.653, IDSA Recurrence Risk Criteria 0.595; P = 0.196). The final, reduced model was deployed as a web application with real-time SHAP explanations. INTERPRETATION: Our final model outperformed existing severity and recurrence models; however, it requires external validation. A DOOR output allows specific clinical questions to be asked with explainable predictions that can be feasibly implemented with limited computing resources. FUNDING: National Institutes of Health-Institute of Allergy and Infectious Diseases.


Subject(s)
Clostridioides difficile , Clostridium Infections , Machine Learning , Humans , Clostridium Infections/diagnosis , Clostridium Infections/microbiology , Male , Female , ROC Curve , Aged , Middle Aged , Prognosis , Recurrence , Retrospective Studies , Area Under Curve
20.
J Med Microbiol ; 73(7)2024 Jul.
Article in English | MEDLINE | ID: mdl-39028257

ABSTRACT

Clostridioides difficile is the most common cause of nosocomial antibiotic-associated diarrhoea and is responsible for a spectrum of diseases characterized by high levels of recurrence and morbidity. In some cases, complications can lead to death. Currently, several types of animal models have been developed to study various aspects of C. difficile infection (CDI), such as colonization, virulence, transmission and recurrence. These models have also been used to test the role of environmental conditions, such as diet, age and microbiome that modulate infection outcome, and to evaluate several therapeutic strategies. Different rodent models have been used successfully, such as the hamster model and the gnotobiotic and conventional mouse models. These models can be applied to study either the initial CDI infectious process or recurrences. The applications of existing rodent models and their advantages and disadvantages are discussed here.


Subject(s)
Clostridioides difficile , Clostridium Infections , Disease Models, Animal , Animals , Clostridium Infections/microbiology , Clostridioides difficile/pathogenicity , Mice , Cricetinae , Humans , Rodentia/microbiology , Germ-Free Life
SELECTION OF CITATIONS
SEARCH DETAIL