Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 231
Filter
1.
Int J Mol Sci ; 25(18)2024 Sep 18.
Article in English | MEDLINE | ID: mdl-39337536

ABSTRACT

Pandemics from viral respiratory tract infections in the 20th and early 21st centuries were associated with high mortality, which was not always caused by a primary viral infection. It has been observed that severe course of infection, complications and mortality were often the result of co-infection with other pathogens, especially Staphylococcus aureus. During the COVID-19 pandemic, it was also noticed that patients infected with S. aureus had a significantly higher mortality rate (61.7%) compared to patients infected with SARS-CoV-2 alone. Our previous studies have shown that S. aureus strains isolated from patients with COVID-19 had a different protein profile than the strains in non-COVID-19 patients. Therefore, this study aims to analyze S. aureus strains isolated from COVID-19 patients in terms of their pathogenicity by analyzing their virulence genes, adhesion, cytotoxicity and penetration to the human pulmonary epithelial cell line A549. We have observed that half of the tested S. aureus strains isolated from patients with COVID-19 had a necrotizing effect on the A549 cells. The strains also showed greater variability in terms of their adhesion to the human cells than their non-COVID-19 counterparts.


Subject(s)
COVID-19 , Coinfection , SARS-CoV-2 , Staphylococcal Infections , Staphylococcus aureus , Humans , COVID-19/complications , COVID-19/microbiology , COVID-19/virology , Staphylococcus aureus/genetics , Staphylococcus aureus/pathogenicity , Coinfection/microbiology , Staphylococcal Infections/microbiology , Staphylococcal Infections/complications , A549 Cells , SARS-CoV-2/genetics , Virulence/genetics , Epithelial Cells/microbiology , Epithelial Cells/virology , Epithelial Cells/metabolism , Bacterial Adhesion , Virulence Factors/genetics
2.
BMC Infect Dis ; 24(1): 1020, 2024 Sep 20.
Article in English | MEDLINE | ID: mdl-39304808

ABSTRACT

BACKGROUND: The association of the oral microbiome with SARS-CoV-2 infections and disease progression has been documented in European, Asian, and American populations but not in Africa. METHODS: We conducted a study in Ghana to evaluate and compare the naso-oropharyngeal microbiome in SARS-CoV-2-infected and uninfected persons before (pre-vaccine) and after vaccine availability (post-vaccine) in the country. 16S rRNA V3-V4 variable region was sequenced and analysed from DNA extracted from naso-oropharyngeal swabs. RESULTS: Considering only the infection status, infected and uninfected groups had no difference in their within-group diversity and was evident in the study population pre- and post-vaccine availability. The introduction of vaccines reduced the diversity of the naso-oropharyngeal microbiome particularly among SARS-CoV-2 positive persons and, vaccinated individuals (both infected and uninfected) had higher microbial diversity compared to their unvaccinated counterparts. SARS-CoV-2-positive and -negative individuals were largely compositionally similar varying by 4-7% but considering vaccination*infection statuses, the genetic distance increased to 12% (P = 0.003) and was mainly influenced by vaccination. Common among the pre- and post-vaccine samples, Atopobium and Finegoldia were abundant in infected and uninfected individuals, respectively. Bacteria belonging to major butyrate-producing phyla, Bacillota (particularly class Clostridia) and Bacteroidota showed increased abundance more strikingly in infected individuals before vaccines were available. They reduced significantly after vaccines were introduced into the country with Fusobacterium and Lachnoanaerobaculum being the only common bacteria between pre-vaccine infected persons and vaccinated individuals, suggesting that natural infection and vaccination correlate with high abundance of short-chain fatty acids. CONCLUSION: Our results show, in an African cohort, the abundance of bacteria taxa known for their protective pathophysiological processes, especially during infection, suggesting that this population is protected against severe COVID-19. The immune-related roles of the members of Bacillota and Bacteroidota that were found associated with infection and vaccination require further studies, and how these may be linked to ethnicity, diet and age. We also recommend expansion of microbiome-disease association studies across Africa to identify possible bacterial-mediated therapeutics for emerging infections.


Subject(s)
Bacteria , COVID-19 , Oropharynx , SARS-CoV-2 , Humans , COVID-19/microbiology , Male , Adult , Female , SARS-CoV-2/genetics , Ghana/epidemiology , Middle Aged , Bacteria/classification , Bacteria/genetics , Bacteria/isolation & purification , Oropharynx/microbiology , Oropharynx/virology , Butyrates/metabolism , Microbiota , RNA, Ribosomal, 16S/genetics , Severity of Illness Index , COVID-19 Vaccines , Nasopharynx/microbiology , Nasopharynx/virology , Aged , Young Adult
3.
Sci Rep ; 14(1): 21997, 2024 09 23.
Article in English | MEDLINE | ID: mdl-39313528

ABSTRACT

This is the primary study at Matrouh Governorate to unveil antibiotic resistance, biofilm formation, silver nanoparticles (Ag-NPs) effect using electron microscopy, and REP-PCR analysis of Staphylococcus aureus strains isolated from COVID-19 patients, contaminated food, and Morel's diseased sheep and goats. A total of 15 S. aureus strains were isolated; five from each of the COVID-19 patients, Morel's diseased sheep and goats, and contaminated food. All strains were considered multidrug-resistant (MDR). All strains showed the presence of biofilm. Morphological changes in the cell surface of the bacterium were evidenced, and penetration with the rupture of some bacterial cells. Based on REP-PCR analysis, 4 clusters (C1-C4) with dissimilarity between clusters C1 and C2 8% and between C3 and C4 15%. Cluster I included 3 strains from contaminated food with a similarity of 97%, and Cluster II included 2 strains from contaminated food and 2 from COVID-19-infected patients with a similarity of 96% (confirming the zoonotic nature of this pathogen). Cluster III contained 4 strains isolated from Morel's diseased sheep & goats with a similarity ratio of 99% in comparison the 4th cluster contained 3 strains isolated from COVID-patients and one from Morel's diseased sheep & goats with a similarity ratio of 92%.


Subject(s)
COVID-19 , Goats , Metal Nanoparticles , Silver , Staphylococcal Infections , Staphylococcus aureus , Staphylococcus aureus/genetics , Staphylococcus aureus/isolation & purification , Staphylococcus aureus/drug effects , Silver/pharmacology , Animals , Humans , Sheep , COVID-19/virology , COVID-19/microbiology , COVID-19/veterinary , Staphylococcal Infections/microbiology , Staphylococcal Infections/veterinary , Biofilms/drug effects , Biofilms/growth & development , Polymerase Chain Reaction/methods , Food Microbiology , SARS-CoV-2/genetics , SARS-CoV-2/isolation & purification , Microbial Sensitivity Tests
4.
Gut Microbes ; 16(1): 2402544, 2024.
Article in English | MEDLINE | ID: mdl-39287023

ABSTRACT

Post-COVID syndrome (PCS) patients have reported a wide range of symptoms, including fatigue, shortness of breath, and diarrhea. Particularly, the presence of gastrointestinal symptoms has led to the hypothesis that the gut microbiome is involved in the development and severity of PCS. The objective of this review is to provide an overview of the role of the gut microbiome in PCS by describing the microbial composition and microbial metabolites in COVID-19 and PCS. Moreover, host-microbe interactions via the microbiota-gut-brain (MGB) and the microbiota-gut-lung (MGL) axes are described. Furthermore, we explore the potential of therapeutically targeting the gut microbiome to support the recovery of PCS by reviewing preclinical model systems and clinical studies. Overall, current studies provide evidence that the gut microbiota is affected in PCS; however, diversity in symptoms and highly individual microbiota compositions suggest the need for personalized medicine. Gut-targeted therapies, including treatments with pre- and probiotics, have the potential to improve the quality of life of affected individuals.


Subject(s)
COVID-19 , Gastrointestinal Microbiome , Host Microbial Interactions , Post-Acute COVID-19 Syndrome , Probiotics , SARS-CoV-2 , Humans , COVID-19/microbiology , COVID-19/virology , Probiotics/therapeutic use , SARS-CoV-2/physiology , Brain-Gut Axis/physiology , Animals , Dysbiosis/microbiology , Lung/microbiology , Lung/virology
5.
Eur J Med Res ; 29(1): 463, 2024 Sep 17.
Article in English | MEDLINE | ID: mdl-39289769

ABSTRACT

BACKGROUND: The epidemiology of fungi identified via next-generation sequencing in bronchoalveolar lavage fluid among patients with COVID-19 is unknown. METHODS: De-identified information, including age, SARS-CoV-2 reads and fungi from bronchoalveolar lavage fluid, were used to analysis. RESULTS: A total of 960 patients with COVID-19 were included. Gender was unknown in 38 patients, and 648 (70.3%) of the rest patients were male. For 876 patients with information on age, their mean ± standard age was 63.4 ± 21.3 years, with the minimum being 0.2 years and the maximum being 101 years. For all the patients, their median [interquartile range] SARS-CoV-2 reads were 26,038 [4421.5, 44,641.5]. The Aspergilli were identified in 159 (16.6%) patients, with Aspergillus fumigatus, Aspergillus flavus and Aspergillus niger in 103 (10.7%), 81 (8.4%) and 17 (1.8%), respectively. The Mucoraceae were identified in 14 (1.5%) patients. Pneumocystis jirovecii was identified in 65 (6.8%) patients, among whom 12 (18.5%) patients also had Aspergilli. The Cryptococcaceae and the Dematiaceae were also identified in some patients, including Cryptococcus in 11 (1.1%) patients. CONCLUSIONS: In bronchoalveolar lavage fluid among patients with COVID-19, the Aspergilli were very commonly identified, as were the Mucoraceae, Pneumocystis jirovecii and Cryptococcus via next-generation sequencing.


Subject(s)
Bronchoalveolar Lavage Fluid , COVID-19 , High-Throughput Nucleotide Sequencing , Humans , Bronchoalveolar Lavage Fluid/microbiology , Bronchoalveolar Lavage Fluid/virology , Male , COVID-19/diagnosis , COVID-19/virology , COVID-19/microbiology , COVID-19/epidemiology , Middle Aged , Female , High-Throughput Nucleotide Sequencing/methods , Aged , Retrospective Studies , Adult , Aged, 80 and over , SARS-CoV-2/genetics , SARS-CoV-2/isolation & purification , Young Adult , Adolescent , Child , Infant , Child, Preschool , Fungi/isolation & purification , Fungi/genetics , Fungi/classification
6.
Curr Microbiol ; 81(10): 338, 2024 Sep 03.
Article in English | MEDLINE | ID: mdl-39223407

ABSTRACT

During the COVID-19 pandemic, fungal infections, especially pulmonary aspergillosis, mucormycosis, and invasive candidiasis, have emerged as a significant health concern. Beyond Candida albicans, the most common cause of invasive candidiasis, other rare ascomycetous yeast species have been described in tertiary care units, potentially posing a broader health threat. We have isolated, from September 2020 to June 2021, nine Diutina catenulata strains from urine samples of six patients. This was intriguing as this fungus had not been previously identified in our institution, nor after June 2021. Therefore, we decided to outline the clinical features of the patients with this rare pathogen, to describe phenotypic characteristics, including antifungal susceptibility profiles, of this yeast species and to identify the genetic makeup through whole-genome sequencing analysis to evaluate if this was a cluster of genetically similar D. catenulata isolates in our institution. The strains were identified through MALDI-TOF MS analyses and Sanger sequencing of two rDNA regions. All patients yielding D. catenulata from urine samples needed ventilator support and used urinary catheters during hospitalization for treatment of COVID-19. None of them had received COVID-19 vaccines. Morphological and biochemical profiles of the nine strains were largely consistent, although fluconazole susceptibility varied, ranging from 4 to 32 µg/mL. Phylogenomic analysis revealed minimal genetic variation among the isolates, with low intrapopulation variation, supported by the identification of only 84 SNPs across all strains. Therefore, we propose that the yeast strains isolated were part of a cluster of D. catenulata funguria in the context of COVID-19.


Subject(s)
Antifungal Agents , COVID-19 , SARS-CoV-2 , Tertiary Care Centers , Humans , COVID-19/microbiology , COVID-19/epidemiology , Tertiary Care Centers/statistics & numerical data , Brazil/epidemiology , Male , Female , Middle Aged , Antifungal Agents/therapeutic use , Antifungal Agents/pharmacology , SARS-CoV-2/genetics , SARS-CoV-2/drug effects , Aged , Adult , Phylogeny , Microbial Sensitivity Tests , Saccharomycetales/genetics , Saccharomycetales/isolation & purification , Saccharomycetales/classification , Whole Genome Sequencing
7.
J Appl Microbiol ; 135(8)2024 Aug 05.
Article in English | MEDLINE | ID: mdl-39143035

ABSTRACT

AIMS: This study aimed to investigate the presence of beta-lactams resistance genes and the clonal relationship of clinical isolates of Enterobacterales obtained from patients with and without COVID-19, in a hospital in northeastern Brazil. METHODS AND RESULTS: The study analyzed 45 carbapenem-resistant clinical isolates using enterobacterial repetitive intergenic consensus (ERIC-PCR), PCR, and amplicon sequencing to detect resistance genes (blaKPC, blaGES, blaNDM, blaVIM, and blaIMP). The main species were Klebsiella pneumoniae, Serratia marcescens, and Proteus mirabilis. Detected genes included blaNDM (46.66%), blaKPC (35.55%), and both (17.79%). ERIC-PCR showed multiclonal dissemination and high genetic variability. The main resistance gene was blaNDM, including blaNDM-5 and blaNDM-7. CONCLUSIONS: The presence of Enterobacterales carrying blaKPC and blaNDM in this study, particularly K. pneumoniae, in infections and colonizations of patients with COVID-19 and non-COVID-19, highlights genetic variability and resistance to carbapenems observed in multiple species of this order.


Subject(s)
COVID-19 , Enterobacteriaceae Infections , SARS-CoV-2 , beta-Lactamases , Humans , COVID-19/microbiology , Brazil , beta-Lactamases/genetics , SARS-CoV-2/genetics , Enterobacteriaceae Infections/microbiology , Genetic Variation , Anti-Bacterial Agents/pharmacology , Microbial Sensitivity Tests , Enterobacteriaceae/genetics , Enterobacteriaceae/drug effects , Enterobacteriaceae/isolation & purification , Carbapenems/pharmacology , Hospitals , Klebsiella pneumoniae/genetics , Klebsiella pneumoniae/isolation & purification , Klebsiella pneumoniae/drug effects
8.
FEBS Open Bio ; 14(10): 1701-1717, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39097990

ABSTRACT

Syrian hamsters (Mesocricetus auratus) have been increasingly used as rodent models in recent years, especially for SARS-CoV-2 since the pandemic. However, the physiology of this animal model is not yet well-understood, even less when considering the digestive tract. Generally, the gastrointestinal microbiome influences the immune system, drug metabolism, and vaccination efficacy. However, a detailed understanding of the gastrointestinal microbiome of hamsters is missing. Therefore, we analyzed 10 healthy 11-week-old RjHan:AURA hamsters fed a pelleted standard diet. Their gastrointestinal content was sampled (i.e., forestomach, glandular stomach, ileum, cecum, and colon) and analyzed using 16S rRNA gene amplicon sequencing. Results displayed a distinct difference in the bacterial community before and after the cecum, possibly due to the available nutrients and digestive functions. Next, we compared hamsters with the literature data of young-adult C57BL/6J mice, another important animal model. We sampled the same gastrointestinal regions and analyzed the differences in the microbiome between both rodents. Surprisingly, we found strong differences in their specific gastrointestinal bacterial communities. For instance, Lactobacillaceae were more abundant in hamsters' forestomach and ileum, while Muribaculaceae dominated in the mouse forestomach and ileum. Similarly, in mouse cecum and colon, Muribaculaceae were dominant, while in hamsters, Lachnospiraceae and Erysipelotrichaceae dominated the bacterial community. Molecular strains of Muribaculaceae in both rodent species displayed some species specificity. This comparison allows a better understanding of the suitability of the Syrian hamster as an animal model, especially regarding its comparability to other rodent models. Thereby, this work contributes to the characterization of the hamster model and allows better experimental planning.


Subject(s)
Gastrointestinal Microbiome , Mesocricetus , Mice, Inbred C57BL , RNA, Ribosomal, 16S , Animals , Mesocricetus/microbiology , Gastrointestinal Microbiome/genetics , Mice , Cricetinae , RNA, Ribosomal, 16S/genetics , Male , Bacteria/classification , Bacteria/genetics , Bacteria/isolation & purification , COVID-19/microbiology , SARS-CoV-2/isolation & purification
9.
Microb Drug Resist ; 30(10): 415-421, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39122252

ABSTRACT

This study aimed to assess the impact of the COVID-19 pandemic on Moraxella catarrhalis infections in pediatric patients hospitalized with community-acquired pneumonia (CAP). The epidemiological features and antimicrobial resistance (AMR) patterns of M. catarrhalis were compared between the pre-pandemic period (2018-2019) and during the pandemic (2020-2022). The results revealed a marked increase in the positivity rate of M. catarrhalis in 2020 and 2021 compared with the pre-pandemic years. The median age of the patients increased significantly in 2021 and 2022, while the proportion of male patients decreased substantially from 2019 to 2021. In addition, there were notable changes in the co-infections of Haemophilus influenzae, parainfluenza virus, and respiratory syncytial virus during the COVID-19 pandemic. The AMR profile of M. catarrhalis also changed significantly, showing increased resistance to ampicillin, but decreased resistance to trimethoprim-sulfamethoxazole and ofloxacin, and a lower proportion of multidrug-resistant isolates. Notably, ampicillin resistance increased among ß-lactamase-producing isolates. Before the pandemic, the number and detection rate of isolates, along with resistance to ampicillin and trimethoprim-sulfamethoxazole, were seasonally distributed, peaking in autumn and winter. However, coinciding with local COVID-19 outbreaks, these indices sharply fell in February 2020, and the number of isolates did not recover during the autumn and winter of 2022. These findings indicate that the COVID-19 pandemic has significantly altered the infection landscape of M. catarrhalis in pediatric CAP patients, as evidenced by shifts in the detection rate, demographic characteristics, respiratory co-infections, AMR profiles, and seasonal patterns.


Subject(s)
Anti-Bacterial Agents , COVID-19 , Community-Acquired Infections , Moraxella catarrhalis , Moraxellaceae Infections , Tertiary Care Centers , Humans , Moraxella catarrhalis/drug effects , Moraxella catarrhalis/isolation & purification , Community-Acquired Infections/epidemiology , Community-Acquired Infections/microbiology , Community-Acquired Infections/drug therapy , COVID-19/epidemiology , COVID-19/microbiology , China/epidemiology , Male , Child, Preschool , Female , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/therapeutic use , Child , Infant , Moraxellaceae Infections/epidemiology , Moraxellaceae Infections/drug therapy , Moraxellaceae Infections/microbiology , Coinfection/epidemiology , Coinfection/microbiology , SARS-CoV-2/drug effects , Drug Resistance, Bacterial , Microbial Sensitivity Tests , Adolescent
10.
Microbiol Spectr ; 12(10): e0059924, 2024 Oct 03.
Article in English | MEDLINE | ID: mdl-39162507

ABSTRACT

The COVID-19 pandemic persists despite the availability of vaccines, and it is, therefore, crucial to develop new therapeutic and preventive approaches. In this study, we investigated the potential role of oral microbiome in SARS-CoV-2 infection. Using an in vitro SARS-CoV-2 pseudovirus infection assay, we found a potent inhibitory effect exerted by Porphyromonas gingivalis on SARS-CoV-2 infection mediated by known P. gingivalis compounds such as phosphoglycerol dihydroceramide (PGDHC) and gingipains as well as by unknown bacterial factors. We found that the gingipain-mediated inhibition of infection is likely due to cytotoxicity, whereas PGDHC inhibited virus infection by an unknown mechanism. Unidentified factors present in P. gingivalis supernatant inhibited SARS-CoV-2 likely via the fusion step of the virus life cycle. We addressed the role of other oral bacteria and found certain periodontal pathogens capable of inhibiting SARS-CoV-2 pseudovirus infection by inducing cytotoxicity on target cells. In the human oral cavity, we observed that the modulatory activity of oral microbial communities varied among individuals, in that some saliva-based cultures were capable of inhibiting while others were enhancing infection. These findings contribute to our understanding of the complex relationship between the oral microbiome and viral infections, offering potential avenues for innovative therapeutic strategies in combating COVID-19. IMPORTANCE: The oral microbiome is important in health and disease, and in this study, we addressed the potential role of the oral microbiome in COVID-19 infection. Our in vitro studies suggest that certain bacteria of the oral microbiome such as P. gingivalis produce compounds that could potentially inhibit SARS-CoV-2 infection. These findings elucidating the interactions between the oral microbiome and SARS-CoV-2 infection will be important in our understanding of COVID-19 pathogenesis and the development of innovative therapeutic and preventive strategies against COVID-19 infection.


Subject(s)
COVID-19 , Microbiota , Mouth , Porphyromonas gingivalis , SARS-CoV-2 , Porphyromonas gingivalis/drug effects , Humans , Microbiota/drug effects , COVID-19/microbiology , SARS-CoV-2/drug effects , SARS-CoV-2/physiology , Mouth/microbiology , Mouth/virology , Saliva/microbiology , Saliva/virology , Animals , Gingipain Cysteine Endopeptidases , Chlorocebus aethiops
11.
BMC Microbiol ; 24(1): 292, 2024 Aug 05.
Article in English | MEDLINE | ID: mdl-39103761

ABSTRACT

Recent observational studies suggest that gut microorganisms are involved in the onset and development of coronavirus disease 2019 (COVID-19), but the potential causal relationship behind them remains unclear. Exposure data were derived from the MiBioGen consortium, encompassing 211 gut microbiota (n = 18,340). The outcome data were sourced from the COVID-19 host genetics initiative (round 7), including COVID-19 severity (n = 1,086,211), hospitalization (n = 2,095,324), and susceptibility (n = 2,597,856). First, a two-sample Mendelian randomization (TSMR) was performed to investigate the causal effect between gut microbiota and COVID-19 outcomes. Second, a two-step MR was used to explore the potential mediators and underlying mechanisms. Third, several sensitivity analyses were performed to verify the robustness of the results. Five gut microbes were found to have a potential causality with COVID-19 severity, namely Betaproteobacteria (beta = 0.096, p = 0.034), Christensenellaceae (beta = -0.092, p = 0.023), Adlercreutzia (beta = 0.072, p = 0.048), Coprococcus 1 (beta = 0.089, p = 0.032), Eisenbergiella (beta = 0.064, p = 0.024). Seven gut microbes were found to have a potential causality with COVID-19 hospitalization, namely Victivallaceae (beta = 0.037, p = 0.028), Actinomyces (beta = 0.047, p = 0.046), Coprococcus 2 (beta = -0.061, p = 0.031), Dorea (beta = 0.067, p = 0.016), Peptococcus (beta = -0.035, p = 0.049), Rikenellaceae RC9 gut group (beta = 0.034, p = 0.018), and Proteobacteria (beta = -0.069, p = 0.035). Two gut microbes were found to have a potential causality with COVID-19 susceptibility, namely Holdemanella (beta = -0.024, p = 0.023) and Lachnospiraceae FCS020 group (beta = 0.026, p = 0.027). Multi-omics mediation analyses indicate that numerous plasma proteins, metabolites, and immune factors are critical mediators linking gut microbiota with COVID-19 outcomes. Sensitivity analysis suggested no significant heterogeneity or pleiotropy. These findings revealed the causal correlation and potential mechanism between gut microbiota and COVID-19 outcomes, which may improve our understanding of the gut-lung axis in the etiology and pathology of COVID-19 in the future.


Subject(s)
COVID-19 , Gastrointestinal Microbiome , SARS-CoV-2 , COVID-19/microbiology , COVID-19/virology , Humans , Gastrointestinal Microbiome/genetics , SARS-CoV-2/genetics , Mendelian Randomization Analysis , Hospitalization , Severity of Illness Index
12.
Diagn Microbiol Infect Dis ; 110(2): 116479, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39116653

ABSTRACT

We report the use of a new multiplex Real-Time PCR platform to simultaneously identify 24 pathogens and 3 antimicrobial-resistance genes directly from respiratory samples of COVID-19 patients. Results were compared to culture-based diagnosis. Secondary infections were detected in 60% of COVID-19 patients by molecular analysis and 73% by microbiological assays, with no significant differences in accuracy, indicating Gram-negative bacteria as the predominant species. Among fungal superinfections, Aspergillus spp. were detected by both methods in more than 7% of COVID-19 patients. Oxacillin-resistant S. aureus and carbapenem-resistant K. pneumoniae were highlighted by both methods. Secondary microbial infections in SARS-CoV-2 patients are associated with poor outcomes and an increased risk of death. Since PCR-based tests significantly reduce the turnaround time to 4 hours and 30 minutes (compared to 48 hours for microbial culture), we strongly support the routine use of molecular techniques, in conjunction with microbiological analysis, to identify co/secondary infections.


Subject(s)
COVID-19 , Coinfection , SARS-CoV-2 , Humans , COVID-19/diagnosis , COVID-19/microbiology , SARS-CoV-2/genetics , Coinfection/diagnosis , Coinfection/microbiology , Coinfection/virology , Male , Middle Aged , Female , Molecular Diagnostic Techniques/methods , Aged , Respiratory Tract Infections/microbiology , Respiratory Tract Infections/diagnosis , Respiratory Tract Infections/virology , Multiplex Polymerase Chain Reaction/methods , Adult , Drug Resistance, Bacterial/genetics , Microbial Sensitivity Tests , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/therapeutic use , Real-Time Polymerase Chain Reaction/methods , Bacteria/genetics , Bacteria/drug effects , Bacteria/isolation & purification , Bacteria/classification , Bacterial Infections/diagnosis , Bacterial Infections/microbiology
13.
Viruses ; 16(8)2024 Jul 23.
Article in English | MEDLINE | ID: mdl-39205154

ABSTRACT

The COVID-19 pandemic has altered respiratory infection patterns in pediatric populations. The emergence of the SARS-CoV-2 Omicron variant and relaxation of public health measures have increased the likelihood of coinfections. Previous studies show conflicting results regarding the impact of viral and bacterial coinfections with SARS-CoV-2 on severity of pediatric disease. This study investigated the prevalence and clinical impact of coinfections among children hospitalized with COVID-19 during the Omicron wave. A retrospective analysis was conducted on 574 hospitalized patients aged under 18 years in Russia, from January 2022 to March 2023. Samples from patients were tested for SARS-CoV-2 and other respiratory pathogens using qRT-PCR, bacterial culture tests and mass spectrometry, and ELISA. Approximately one-third of COVID-19 cases had coinfections, with viral and bacterial coinfections occurring at similar rates. Adenovirus and Staphylococcus aureus were the most common viral and bacterial coinfections, respectively. Viral coinfections were associated with higher fevers and increased bronchitis, while bacterial coinfections correlated with longer duration of illness and higher pneumonia rates. Non-SARS-CoV-2 respiratory viruses were linked to more severe lower respiratory tract complications than SARS-CoV-2 monoinfection. These findings suggest that during the Omicron wave, seasonal respiratory viruses may have posed a greater threat to children's health than SARS-CoV-2.


Subject(s)
Bacterial Infections , COVID-19 , Coinfection , Hospitalization , SARS-CoV-2 , Humans , Coinfection/epidemiology , Coinfection/microbiology , Coinfection/virology , COVID-19/epidemiology , COVID-19/complications , COVID-19/microbiology , Adolescent , Child , Child, Preschool , Female , Male , Infant , Russia/epidemiology , Retrospective Studies , Prevalence , Bacterial Infections/epidemiology , Bacterial Infections/microbiology , Respiratory Tract Infections/epidemiology , Respiratory Tract Infections/microbiology , Respiratory Tract Infections/virology , Infant, Newborn
14.
Gut Microbes ; 16(1): 2394248, 2024.
Article in English | MEDLINE | ID: mdl-39185682

ABSTRACT

Microbiome perturbations can have long-term effects on health. The dynamics of the gut microbiome and virome in women living with HIV (WLHIV) and their newborn infants is poorly understood. Here, we performed metagenomic sequencing analyses on longitudinal stool samples including 23 mothers (13 WLHIV, 10 HIV-negative) and 12 infants that experienced SARS-CoV-2 infection with mild disease, as well as 40 mothers (18 WLHIV, 22 HIV-negative) and 60 infants that remained SARS-CoV-2 seronegative throughout the study follow-up. Regardless of HIV or SARS-CoV-2 status, maternal bacterial and viral profiles were distinct from infants. Using linear mixed effects models, we showed that the microbiome alpha diversity trajectory was not significantly different between SARS-CoV-2 seropositive and seronegative women. However, seropositive women's positive trajectory while uninfected was abruptly reversed after SARS-CoV-2 infection (p = 0.015). Gut virome signatures of women were not associated with SARS-CoV-2. Alterations in infant microbiome and virome diversities were generally not impacted by SARS-CoV-2 but were rather driven by development. We did not find statistically significant interactions between HIV and SARS-CoV-2 on the gut microbiome and virome. Overall, our study provides insights into the complex interplay between maternal and infant bacterial microbiome, virome, and the influence of SARS-CoV-2 and HIV status.


Subject(s)
COVID-19 , Feces , Gastrointestinal Microbiome , HIV Infections , SARS-CoV-2 , Virome , Humans , Female , COVID-19/microbiology , COVID-19/virology , HIV Infections/microbiology , HIV Infections/virology , SARS-CoV-2/genetics , SARS-CoV-2/isolation & purification , Adult , Infant, Newborn , Feces/microbiology , Feces/virology , Infant , Bacteria/classification , Bacteria/isolation & purification , Bacteria/genetics , Longitudinal Studies
15.
PLoS One ; 19(7): e0300408, 2024.
Article in English | MEDLINE | ID: mdl-39052548

ABSTRACT

SARS-CoV-2, a severe respiratory disease primarily targeting the lungs, was the leading cause of death worldwide during the pandemic. Understanding the interplay between the oral microbiome and inflammatory cytokines during acute infection is crucial for elucidating host immune responses. This study aimed to explore the relationship between the oral microbiome and cytokines in COVID-19 patients, particularly those with and without sputum production. Saliva and blood samples from 50 COVID-19 patients were subjected to 16S ribosomal RNA gene sequencing for oral microbiome analysis, and 65 saliva and serum cytokines were assessed using Luminex multiplex analysis. The Mann-Whitney test was used to compare cytokine levels between individuals with and without sputum production. Logistic regression machine learning models were employed to evaluate the predictive capability of oral microbiome, salivary, and blood biomarkers for sputum production. Significant differences were observed in the membership (Jaccard dissimilarity: p = 0.016) and abundance (PhILR dissimilarity: p = 0.048; metagenomeSeq) of salivary microbial communities between patients with and without sputum production. Seven bacterial genera, including Prevotella, Streptococcus, Actinomyces, Atopobium, Filifactor, Leptotrichia, and Selenomonas, were more prevalent in patients with sputum production (p<0.05, Fisher's exact test). Nine genera, including Prevotella, Megasphaera, Stomatobaculum, Selenomonas, Leptotrichia, Veillonella, Actinomyces, Atopobium, and Corynebacteria, were significantly more abundant in the sputum-producing group, while Lachnoanaerobaculum was more prevalent in the non-sputum-producing group (p<0.05, ANCOM-BC). Positive correlations were found between salivary IFN-gamma and Eotaxin2/CCL24 with sputum production, while negative correlations were noted with serum MCP3/CCL7, MIG/CXCL9, IL1 beta, and SCF (p<0.05, Mann-Whitney test). The machine learning model using only oral bacteria input outperformed the model that included all data: blood and saliva biomarkers, as well as clinical and demographic variables, in predicting sputum production in COVID-19 subjects. The performance metrics were as follows, comparing the model with only bacteria input versus the model with all input variables: precision (95% vs. 75%), recall (100% vs. 50%), F1-score (98% vs. 60%), and accuracy (82% vs. 66%).


Subject(s)
COVID-19 , Microbiota , Saliva , Sputum , Humans , COVID-19/microbiology , Sputum/microbiology , Saliva/microbiology , Female , Male , Middle Aged , Adult , Aged , Cytokines/blood , Cytokines/metabolism , Lung/microbiology , Lung/virology , SARS-CoV-2/isolation & purification , RNA, Ribosomal, 16S/genetics , Leptotrichia , Prevotella/isolation & purification , Streptococcus/isolation & purification , Actinomyces/isolation & purification , Mouth/microbiology , Mouth/virology , Machine Learning , Biomarkers/blood
16.
Sci Rep ; 14(1): 15737, 2024 07 08.
Article in English | MEDLINE | ID: mdl-38977804

ABSTRACT

The COVID-19 pandemic has significantly transformed the infection spectrum of various pathogens. This study aimed to evaluate the impact of the COVID-19 pandemic on Staphylococcus aureus (S. aureus) infections among pediatric patients with community acquired pneumonia (CAP). We retrospectively reviewed pediatric CAP admissions before (from 2018 to 2019) and during (from 2020 to 2022) the COVID-19 pandemic. The epidemiology and antimicrobial resistance (AMR) profiles of S. aureus isolates were examined to assess the pandemic's effect. As a result, a total of 399 pediatric CAP patients with S. aureus infections were included. The positivity rate, gender, and age distribution of patients were similar across both periods. There was a marked reduction in respiratory co-infections with Haemophilus influenzae (H. influenzae) during the COVID-19 pandemic, compared to 2019. Additionally, there were significant changes in the resistance profiles of S. aureus isolates to various antibiotics. Resistance to oxacillin and tetracycline increased, whereas resistance to penicillin, gentamicin, and quinolones decreased. Notably, resistance to erythromycin significantly decreased in methicillin-resistant S. aureus (MRSA) strains. The number of S. aureus isolates, the proportion of viral co-infections, and the number of resistant strains typically peaked seasonally, primarily in the first or fourth quarters of 2018, 2019, and 2021. However, shifts in these patterns were noted in the first quarter of 2020 and the fourth quarter of 2022. These findings reveal that the COVID-19 pandemic has significantly altered the infection dynamics of S. aureus among pediatric CAP patients, as evidenced by changes in respiratory co-infections, AMR patterns, and seasonal trends.


Subject(s)
Anti-Bacterial Agents , COVID-19 , Community-Acquired Infections , Staphylococcal Infections , Staphylococcus aureus , Humans , COVID-19/epidemiology , COVID-19/microbiology , COVID-19/complications , Community-Acquired Infections/epidemiology , Community-Acquired Infections/microbiology , Community-Acquired Infections/drug therapy , Female , Male , Child , Child, Preschool , Retrospective Studies , Staphylococcus aureus/drug effects , Staphylococcus aureus/isolation & purification , Infant , Staphylococcal Infections/epidemiology , Staphylococcal Infections/drug therapy , Staphylococcal Infections/microbiology , Anti-Bacterial Agents/therapeutic use , Anti-Bacterial Agents/pharmacology , Adolescent , Coinfection/epidemiology , Coinfection/microbiology , SARS-CoV-2/isolation & purification , SARS-CoV-2/drug effects , Methicillin-Resistant Staphylococcus aureus/drug effects , Methicillin-Resistant Staphylococcus aureus/isolation & purification , Pandemics , Hospitalization , Drug Resistance, Bacterial
17.
J Genet Genomics ; 51(10): 1111-1120, 2024 Oct.
Article in English | MEDLINE | ID: mdl-38960315

ABSTRACT

Cough is one of the most common symptoms observed in patients presenting with COVID-19, persisting for an extended duration following SARS-CoV-2 infection. We aim to describe the distribution of airway microbiota and explore its role in patients with post-COVID-19 chronic cough. A total of 57 patients experiencing persistent cough after infection were recruited during the Omicron wave of SARS-CoV-2 in China. Airway microbiota profiling is assessed in nasopharyngeal swab, nasal lavage, and induced sputum samples at 4 and 8 weeks after SARS-CoV-2 infection. Our findings reveal that bacterial families Staphylococcaceae, Corynebacteriaceae, and Enterobacteriaceae are the most prevalent in the upper airway, while Streptococcaceae, Lachnospiraceae, and Prevotellaceae emerge as the most prevalent bacterial families in the lower airway. An increase in the abundance of Staphylococcus in nasopharyngeal swab samples and of Streptococcus in induced sputum samples is observed after one month. Furthermore, the abundance of Staphylococcus identified in nasopharyngeal swab samples at the baseline period emerges as an insightful predictor for improvement in cough severity. In conclusion, dynamic alterations in the airway microbial composition may contribute to the post-COVID-19 chronic cough progression, while the compositional signatures of nasopharyngeal microbiota could reflect the improvement of this disease.


Subject(s)
COVID-19 , Cough , Microbiota , Sputum , Humans , COVID-19/microbiology , COVID-19/complications , COVID-19/virology , Cough/microbiology , Cough/virology , Male , Female , Microbiota/genetics , Middle Aged , Adult , Sputum/microbiology , Sputum/virology , Prospective Studies , Chronic Disease , SARS-CoV-2/isolation & purification , Disease Progression , Nasopharynx/microbiology , Nasopharynx/virology , China , Chronic Cough
18.
NPJ Biofilms Microbiomes ; 10(1): 66, 2024 Aug 01.
Article in English | MEDLINE | ID: mdl-39085233

ABSTRACT

The clinical course of COVID-19 is variable and often unpredictable. To test the hypothesis that disease progression and inflammatory responses associate with alterations in the microbiome and metabolome, we analyzed metagenome, metabolome, cytokine, and transcriptome profiles of repeated samples from hospitalized COVID-19 patients and uninfected controls, and leveraged clinical information and post-hoc confounder analysis. Severe COVID-19 was associated with a depletion of beneficial intestinal microbes, whereas oropharyngeal microbiota disturbance was mainly linked to antibiotic use. COVID-19 severity was also associated with enhanced plasma concentrations of kynurenine and reduced levels of several other tryptophan metabolites, lysophosphatidylcholines, and secondary bile acids. Moreover, reduced concentrations of various tryptophan metabolites were associated with depletion of Faecalibacterium, and tryptophan decrease and kynurenine increase were linked to enhanced production of inflammatory cytokines. Collectively, our study identifies correlated microbiome and metabolome alterations as a potential contributor to inflammatory dysregulation in severe COVID-19.


Subject(s)
COVID-19 , Cytokines , Dysbiosis , Gastrointestinal Microbiome , SARS-CoV-2 , Tryptophan , Humans , COVID-19/microbiology , COVID-19/immunology , Tryptophan/metabolism , Male , Female , Middle Aged , Cytokines/blood , Cytokines/metabolism , Metabolome , Inflammation , Kynurenine/metabolism , Kynurenine/blood , Aged , Adult
19.
mBio ; 15(8): e0166724, 2024 Aug 14.
Article in English | MEDLINE | ID: mdl-39037272

ABSTRACT

Severe COVID-19 has been associated with coinfections with bacterial and fungal pathogens. Notably, patients with COVID-19 who develop Staphylococcus aureus bacteremia exhibit higher rates of mortality than those infected with either pathogen alone. To understand this clinical scenario, we collected and examined S. aureus blood and respiratory isolates from a hospital in New York City during the early phase of the pandemic from both SARS-CoV-2+ and SARS-CoV-2- patients. Whole genome sequencing of these S. aureus isolates revealed broad phylogenetic diversity in both patient groups, suggesting that SARS-CoV-2 coinfection was not associated with a particular S. aureus lineage. Phenotypic characterization of the contemporary collection of S. aureus isolates from SARS-CoV-2+ and SARS-CoV-2- patients revealed no notable differences in several virulence traits examined. However, we noted a trend toward overrepresentation of S. aureus bloodstream strains with low cytotoxicity in the SARS-CoV-2+ group. We observed that patients coinfected with SARS-CoV-2 and S. aureus were more likely to die during the acute phase of infection when the coinfecting S. aureus strain exhibited high or low cytotoxicity. To further investigate the relationship between SARS-CoV-2 and S. aureus infections, we developed a murine coinfection model. These studies revealed that infection with SARS-CoV-2 renders mice susceptible to subsequent superinfection with low cytotoxicity S. aureus. Thus, SARS-CoV-2 infection sensitizes the host to coinfections, including S. aureus isolates with low intrinsic virulence. IMPORTANCE: The COVID-19 pandemic has had an enormous impact on healthcare across the globe. Patients who were severely infected with SARS-CoV-2, the virus causing COVID-19, sometimes became infected with other pathogens, which is termed coinfection. If the coinfecting pathogen is the bacterium Staphylococcus aureus, there is an increased risk of patient death. We collected S. aureus strains that coinfected patients with SARS-CoV-2 to study the disease outcome caused by the interaction of these two important pathogens. We found that both in patients and in mice, coinfection with an S. aureus strain lacking toxicity resulted in more severe disease during the early phase of infection, compared with infection with either pathogen alone. Thus, SARS-CoV-2 infection can directly increase the severity of S. aureus infection.


Subject(s)
COVID-19 , Coinfection , SARS-CoV-2 , Staphylococcal Infections , Staphylococcus aureus , COVID-19/complications , COVID-19/microbiology , Coinfection/microbiology , Coinfection/virology , Staphylococcus aureus/genetics , Staphylococcus aureus/pathogenicity , Staphylococcal Infections/microbiology , Humans , Animals , Mice , SARS-CoV-2/genetics , Phylogeny , Female , New York City/epidemiology , Male , Virulence , Middle Aged , Whole Genome Sequencing , Bacteremia/microbiology , Disease Models, Animal , Aged
20.
Arch Med Res ; 55(6): 103038, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39018939

ABSTRACT

BACKGROUND: Prolonged hospitalization due to the COVID-19 pandemic gathered risk factors for developing invasive candidiasis. AIM: To describe Candida spp. isolated from patients with clinical suspicion of COVID treated in a public hospital specialized in COVID-19 during the pandemic, considering the susceptibility profiles and the risk factors related to the species detected in a positive yeast culture. METHODS: From different samples of 33 patients with comorbidities, 42 clinical isolates were identified by VITEKⓇ MS Plus. Antifungal susceptibility testing was performed using VITEKⓇ 2 Compact with the AST-YS08 card. RESULTS: The most frequently identified species were C. albicans and C. glabrata, which were also the most common co-infections, Saprochaete capitata, an uncommon yeast was isolated in one patient. 85% of the co-infections were COVID positive and 100% of patients with a co-infection required mechanical ventilation (MV) which has been described as one of the major predisposing factors to candidiasis. Candida species vary in their response to treatment. In this study, 44% of isolates identified as C. glabrata were fluconazole-resistant, which were also immediately susceptible to caspofungin; this profile limits therapeutic options and emphasizes the importance of evaluating the susceptibility profile. CONCLUSIONS: This work highlights the increase in isolation of different Candida species during COVID-19 and the importance of establishing criteria to declare Candida colonization or infection and the correct etiological identification to establish an agent-based antifungal treatment, to reduce the spreading risk of Candida spp. in the hospital environment, mortality, time, and cost of hospitalization.


Subject(s)
Antifungal Agents , COVID-19 , Candida , Humans , COVID-19/microbiology , COVID-19/epidemiology , COVID-19/complications , Male , Female , Prospective Studies , Aged , Candida/isolation & purification , Candida/drug effects , Middle Aged , Antifungal Agents/therapeutic use , Antifungal Agents/pharmacology , SARS-CoV-2 , Aged, 80 and over , Candidiasis/microbiology , Candidiasis/epidemiology , Candidiasis/drug therapy , Microbial Sensitivity Tests , Coinfection/microbiology , Coinfection/epidemiology , Drug Resistance, Fungal , Adult , Risk Factors , Fluconazole/therapeutic use , Fluconazole/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL