Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 208
Filtrar
1.
Front Cell Infect Microbiol ; 14: 1347345, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38828262

RESUMEN

Background: To date, more than 770 million individuals have become coronavirus disease 2019 (COVID-19) convalescents worldwide. Emerging evidence highlights the influence of COVID-19 on the oral microbiome during both acute and convalescent disease phases. Front-line healthcare workers are at an elevated risk of exposure to viral infections, and the effects of COVID-19 on their oral microbiome remain relatively unexplored. Methods: Oropharyngeal swab specimens, collected one month after a negative COVID-19 test from a cohort comprising 55 healthcare workers, underwent 16S rRNA sequencing. We conducted a comparative analysis between this post-COVID-19 cohort and the pre-infection dataset from the same participants. Community composition analysis, indicator species analysis, alpha diversity assessment, beta diversity exploration, and functional prediction were evaluated. Results: The Shannon and Simpson indexes of the oral microbial community declined significantly in the post-COVID-19 group when compared with the pre-infection cohort. Moreover, there was clear intergroup clustering between the two groups. In the post-COVID-19 group, the phylum Firmicutes showed a significant increase. Further, there were clear differences in relative abundance of several bacterial genera in contrast with the pre-infection group, including Streptococcus, Gemella, Granulicatella, Capnocytophaga, Leptotrichia, Fusobacterium, and Prevotella. We identified Gemella enrichment in the post-COVID-19 group, potentially serving as a recovery period performance indicator. Functional prediction revealed lipopolysaccharide biosynthesis downregulation in the post-COVID-19 group, an outcome with host inflammatory response modulation and innate defence mechanism implications. Conclusion: During the recovery phase of COVID-19, the oral microbiome diversity of front-line healthcare workers failed to fully return to its pre-infection state. Despite the negative COVID-19 test result one month later, notable disparities persisted in the composition and functional attributes of the oral microbiota.


Asunto(s)
Bacterias , COVID-19 , Personal de Salud , Microbiota , Orofaringe , ARN Ribosómico 16S , SARS-CoV-2 , Humanos , COVID-19/microbiología , Orofaringe/microbiología , Orofaringe/virología , SARS-CoV-2/aislamiento & purificación , SARS-CoV-2/genética , Adulto , ARN Ribosómico 16S/genética , Masculino , Femenino , Bacterias/clasificación , Bacterias/aislamiento & purificación , Bacterias/genética , Persona de Mediana Edad , Estudios de Cohortes
2.
PLoS One ; 19(6): e0304504, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38870232

RESUMEN

To determine why SARS-CoV-2 appears to thrive specifically well in meat packaging plants, we used SARS-CoV-2 Delta variant and meat packaging plant drain samples to develop mixed-species biofilms on materials commonly found within meat packaging plants (stainless steel (SS), PVC, and ceramic tile). Our data provides evidence that SARS-CoV-2 Delta variant remained viable on all the surfaces tested with and without an environmental biofilm after the virus was inoculated with the biofilm for 5 days at 7°C. We observed that SARS-CoV-2 Delta variant was able to remain infectious with each of the environmental biofilms by conducting plaque assay and qPCR experiments, however, we detected a significant reduction in viability post-exposure to Plant B biofilm on SS, PVC, and on ceramic tile chips, and to Plant C biofilm on SS and PVC chips. The numbers of viable SARS-CoV-2 Delta viral particles was 1.81-4.57-fold high than the viral inoculum incubated with the Plant B and Plant C environmental biofilm on SS, and PVC chips. We did not detect a significant difference in viability when SARS-CoV-2 Delta variant was incubated with the biofilm obtained from Plant A on any of the materials tested and SARS-CoV-2 Delta variant had higher plaque numbers when inoculated with Plant C biofilm on tile chips, with a 2.75-fold difference compared to SARS-CoV-2 Delta variant on tile chips by itself. In addition, we detected an increase in the biofilm biovolume in response to SARS-CoV-2 Delta variant which is also a concern for food safety due to the potential for foodborne pathogens to respond likewise when they come into contact with the virus. These results indicate a complex virus-environmental biofilm interaction which correlates to the different bacteria found in each biofilm. Our results also indicate that there is the potential for biofilms to protect SARS-CoV-2 from disinfecting agents and remaining prevalent in meat packaging plants.


Asunto(s)
Biopelículas , Embalaje de Alimentos , SARS-CoV-2 , Biopelículas/crecimiento & desarrollo , SARS-CoV-2/fisiología , SARS-CoV-2/aislamiento & purificación , SARS-CoV-2/genética , Embalaje de Alimentos/métodos , Humanos , COVID-19/microbiología , COVID-19/virología , COVID-19/transmisión , Acero Inoxidable , Carne/microbiología , Carne/virología
3.
Nat Commun ; 15(1): 4708, 2024 Jun 03.
Artículo en Inglés | MEDLINE | ID: mdl-38830853

RESUMEN

Critical illness can significantly alter the composition and function of the human microbiome, but few studies have examined these changes over time. Here, we conduct a comprehensive analysis of the oral, lung, and gut microbiota in 479 mechanically ventilated patients (223 females, 256 males) with acute respiratory failure. We use advanced DNA sequencing technologies, including Illumina amplicon sequencing (utilizing 16S and ITS rRNA genes for bacteria and fungi, respectively, in all sample types) and Nanopore metagenomics for lung microbiota. Our results reveal a progressive dysbiosis in all three body compartments, characterized by a reduction in microbial diversity, a decrease in beneficial anaerobes, and an increase in pathogens. We find that clinical factors, such as chronic obstructive pulmonary disease, immunosuppression, and antibiotic exposure, are associated with specific patterns of dysbiosis. Interestingly, unsupervised clustering of lung microbiota diversity and composition by 16S independently predicted survival and performed better than traditional clinical and host-response predictors. These observations are validated in two separate cohorts of COVID-19 patients, highlighting the potential of lung microbiota as valuable prognostic biomarkers in critical care. Understanding these microbiome changes during critical illness points to new opportunities for microbiota-targeted precision medicine interventions.


Asunto(s)
COVID-19 , Disbiosis , Microbioma Gastrointestinal , Pulmón , Microbiota , Humanos , Femenino , Masculino , Disbiosis/microbiología , Persona de Mediana Edad , Pulmón/microbiología , COVID-19/microbiología , COVID-19/virología , Anciano , Microbiota/genética , Microbioma Gastrointestinal/genética , Interacciones Microbiota-Huesped/genética , Estudios Longitudinales , ARN Ribosómico 16S/genética , Insuficiencia Respiratoria/microbiología , SARS-CoV-2/genética , SARS-CoV-2/aislamiento & purificación , Adulto , Respiración Artificial , Bacterias/genética , Bacterias/clasificación , Bacterias/aislamiento & purificación , Enfermedad Crítica , Metagenómica/métodos
4.
NPJ Biofilms Microbiomes ; 10(1): 50, 2024 Jun 20.
Artículo en Inglés | MEDLINE | ID: mdl-38902263

RESUMEN

During the COVID-19 pandemic, facemasks played a pivotal role in preventing person-person droplet transmission of viral particles. However, prolonged facemask wearing causes skin irritations colloquially referred to as 'maskne' (mask + acne), which manifests as acne and contact dermatitis and is mostly caused by pathogenic skin microbes. Previous studies revealed that the putative causal microbes were anaerobic bacteria, but the pathogenesis of facemask-associated skin conditions remains poorly defined. We therefore characterized the role of the facemask-associated skin microbiota in the development of maskne using culture-dependent and -independent methodologies. Metagenomic analysis revealed that the majority of the facemask microbiota were anaerobic bacteria that originated from the skin rather than saliva. Previous work demonstrated direct interaction between pathogenic bacteria and antagonistic strains in the microbiome. We expanded this analysis to include indirect interaction between pathogenic bacteria and other indigenous bacteria classified as either 'pathogen helper (PH)' or 'pathogen inhibitor (PIn)' strains. In vitro screening of bacteria isolated from facemasks identified both strains that antagonized and promoted pathogen growth. These data were validated using a mouse skin infection model, where we observed attenuation of symptoms following pathogen infection. Moreover, the inhibitor of pathogen helper (IPH) strain, which did not directly attenuate pathogen growth in vitro and in vivo, functioned to suppress symptom development and pathogen growth indirectly through PH inhibitory antibacterial products such as phenyl lactic acid. Taken together, our study is the first to define a mechanism by which indirect microbiota interactions under facemasks can control symptoms of maskne by suppressing a skin pathogen.


Asunto(s)
COVID-19 , Máscaras , Microbiota , Piel , Animales , Ratones , Humanos , COVID-19/microbiología , COVID-19/virología , Piel/microbiología , Acné Vulgar/microbiología , SARS-CoV-2 , Femenino , Metagenómica/métodos , Modelos Animales de Enfermedad , Bacterias/clasificación , Bacterias/genética , Bacterias/aislamiento & purificación , Interacciones Microbianas , Dermatitis por Contacto/etiología
5.
Viruses ; 16(5)2024 05 02.
Artículo en Inglés | MEDLINE | ID: mdl-38793604

RESUMEN

Recent studies highlight the crucial role of the gut microbiome in post-infectious complications, especially in patients recovering from severe COVID-19. Our research aimed to explore the connection between gut microbiome changes and the cytokine profile of patients with post-COVID syndrome. Using 16S rRNA amplicon sequencing, we analyzed the composition of the gut microbiome in 60 COVID-19 patients over the course of one year. We also measured the levels of serum cytokines and chemokines using the Milliplex system. Our results showed that severe SARS-CoV-2 infection cases, especially those complicated by pneumonia, induce a pro-inflammatory microbial milieu with heightened presence of Bacteroides, Faecalibacterium, and Prevotella_9. Furthermore, we found that post-COVID syndrome is characterized by a cross-correlation of various cytokines and chemokines MDC, IL-1b, Fractalkine, TNFa, FGF-2, EGF, IL-1RA, IFN-a2, IL-10, sCD40L, IL-8, Eotaxin, IL-12p40, and MIP-1b as well as a shift in the gut microbiome towards a pro-inflammatory profile. At the functional level, our analysis revealed associations with post-COVID-19 in homolactic fermentation, pentose phosphate, NAD salvage, and flavin biosynthesis. These findings highlight the intricate interplay between the gut microbiota, their metabolites, and systemic cytokines in shaping post-COVID symptoms. Unraveling the gut microbiome's role in post-infectious complications opens avenues for new treatments for those patients with prolonged symptoms.


Asunto(s)
COVID-19 , Citocinas , Microbioma Gastrointestinal , SARS-CoV-2 , Humanos , COVID-19/inmunología , COVID-19/microbiología , COVID-19/complicaciones , COVID-19/sangre , Citocinas/sangre , Masculino , Femenino , Persona de Mediana Edad , Anciano , Adulto , ARN Ribosómico 16S/genética , Síndrome Post Agudo de COVID-19 , Heces/microbiología , Heces/virología
6.
Mol Biol Rep ; 51(1): 665, 2024 May 22.
Artículo en Inglés | MEDLINE | ID: mdl-38777940

RESUMEN

BACKGROUND: Staphylococcus aureus (S. aureus) associated with COVID-19 has not been well documented. This cross-sectional study evaluated the association between nasal S. aureus carriage and COVID-19. METHODS AND RESULTS: Nasopharyngeal samples were collected from 391 participants presenting for COVID-19 test in Lagos, Nigeria, and S. aureus was isolated from the samples. Antimicrobial susceptibility test was done by disc diffusion method. All S. aureus isolates were screened for the presence of mecA, panton-valentine leucocidin (PVL) and toxic shock syndrome toxin (TSST) virulence genes by polymerase chain reaction. Staphylococcal protein A (spa) typing was conducted for all the isolates. Participants with COVID-19 had double the prevalence of S. aureus (42.86%) compared to those who tested negative (20.54%). A significant association was seen between S. aureus nasal carriage and COVID-19 (p = 0.004). Antimicrobial sensitivity results showed resistance to oxacillin (100%), cefoxitin (53%), and vancomycin (98.7%). However, only 41% of the isolates harbored the mecA gene, with SCCmecV being the most common SCCmec type. There was no association between the carriage of virulence genes and COVID-19. A total of 23 Spa types were detected, with t13249 and t095 being the two most common spa types. CONCLUSION: This study examined the association between nasal S. aureus carriage and SARS-COV-2 infection. Further research is required to fully explore the implications of S. aureus co-infection with COVID-19.


Asunto(s)
COVID-19 , SARS-CoV-2 , Infecciones Estafilocócicas , Staphylococcus aureus , Humanos , COVID-19/microbiología , COVID-19/epidemiología , COVID-19/virología , Infecciones Estafilocócicas/epidemiología , Infecciones Estafilocócicas/microbiología , Estudios Transversales , Masculino , Femenino , Staphylococcus aureus/genética , Staphylococcus aureus/efectos de los fármacos , Staphylococcus aureus/patogenicidad , Staphylococcus aureus/aislamiento & purificación , Adulto , SARS-CoV-2/genética , SARS-CoV-2/aislamiento & purificación , Persona de Mediana Edad , Toxinas Bacterianas/genética , Staphylococcus aureus Resistente a Meticilina/genética , Staphylococcus aureus Resistente a Meticilina/aislamiento & purificación , Staphylococcus aureus Resistente a Meticilina/patogenicidad , Staphylococcus aureus Resistente a Meticilina/efectos de los fármacos , Comorbilidad , Proteínas Bacterianas/genética , Virulencia/genética , Nigeria/epidemiología , Farmacorresistencia Bacteriana Múltiple/genética , Antibacterianos/farmacología , Portador Sano/epidemiología , Portador Sano/microbiología , Pruebas de Sensibilidad Microbiana , Proteínas de Unión a las Penicilinas/genética , Leucocidinas/genética , Exotoxinas/genética , Factores de Virulencia/genética , Adulto Joven
7.
mSystems ; 9(6): e0018524, 2024 Jun 18.
Artículo en Inglés | MEDLINE | ID: mdl-38700338

RESUMEN

Acute ischemic stroke (AIS) patients with active COVID-19 infection often have more severe symptoms and worse recovery. COVID-19 infection can cause gut microbiota dysbiosis, which is also a risk factor for poor outcomes in AIS patients. However, the association between gut microbiota and functional outcomes among AIS patients with COVID-19 infection has not been fully clarified yet. In this study, we performed 16S rRNA gene sequencing to characterize the gut microbial community among AIS patients with acute COVID-19 infection, AIS patients with post-acute COVID-19 infection, and AIS patients without COVID-19 infection. We found that AIS patients with acute COVID-19 experienced poorer recovery and significant gut dysbiosis, characterized by higher levels of Enterobacteriaceae and lower levels of Ruminococcaceae and Lachnospiraceae. Furthermore, a shorter time window (less than 28 days) between COVID-19 infection and stroke was identified as a risk factor for poor functional outcomes in AIS patients with COVID-19, and the enrichment of Enterobacteriaceae was indicated as a mediator in the relationship between infection time window and poor stroke outcomes. Our findings highlight the importance of early intervention after COVID-19 infection, especially by regulating the gut microbiota, which plays a role in the prognosis of AIS patients with COVID-19 infection.IMPORTANCEThe gut microbiota plays an important role in the association between respiratory system and cerebrovascular system through the gut-lung axis and gut-brain axis. However, the specific connection between gut bacteria and the functional outcomes of acute ischemic stroke (AIS) patients with COVID-19 is not fully understood yet. In our study, we observed a significant decrease in bacterial diversity and shifts in the abundance of key bacterial families in AIS patients with acute COVID-19 infection. Furthermore, we identified that the time window was a critical influence factor for stroke outcomes, and the enrichment of Enterobacteriaceae acted as a mediator in the relationship between the infection time window and poor stroke outcomes. Our research provides a new perspective on the complex interplay among AIS, COVID-19 infection, and gut microbiota dysbiosis. Moreover, recognizing Enterobacteriaceae as a potential mediator of poor stroke prognosis offers a novel avenue for future exploration and therapeutic interventions.


Asunto(s)
COVID-19 , Disbiosis , Microbioma Gastrointestinal , Accidente Cerebrovascular Isquémico , Humanos , COVID-19/complicaciones , COVID-19/microbiología , COVID-19/epidemiología , Microbioma Gastrointestinal/fisiología , Masculino , Femenino , Accidente Cerebrovascular Isquémico/microbiología , Persona de Mediana Edad , Anciano , SARS-CoV-2 , ARN Ribosómico 16S/genética , Factores de Riesgo
8.
Front Cell Infect Microbiol ; 14: 1297312, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38690325

RESUMEN

Background: During the coronavirus disease 2019 (COVID-19) pandemic, in patients treated for SARS-CoV-2 infection, infections with the Klebsiella pneumoniae bacteria producing New Delhi metallo-B-lactamase (NDM) carbapenemase in the USA, Brazil, Mexico, and Italy were observed, especially in intensive care units (ICUs). This study aimed to assess the impact of Klebsiella pneumoniae NDM infection and other bacterial infections on mortality in patients treated in ICUs due to COVID-19. Methods: The 160 patients who qualified for the study were hospitalized in ICUs due to COVID-19. Three groups were distinguished: patients with COVID-19 infection only (N = 72), patients with COVID-19 infection and infection caused by Klebsiella pneumoniae NDM (N = 30), and patients with COVID-19 infection and infection of bacterial etiology other than Klebsiella pneumoniae NDM (N = 58). Mortality in the groups and chosen demographic data; biochemical parameters analyzed on days 1, 3, 5, and 7; comorbidities; and ICU scores were analyzed. Results: Bacterial infection, including with Klebsiella pneumoniae NDM type, did not elevate mortality rates. In the group of patients who survived the acute phase of COVID-19 the prolonged survival time was demonstrated: the median overall survival time was 13 days in the NDM bacterial infection group, 14 days in the other bacterial infection group, and 7 days in the COVID-19 only group. Comparing the COVID-19 with NDM infection and COVID-19 only groups, the adjusted model estimated a statistically significant hazard ratio of 0.28 (p = 0.002). Multivariate analysis revealed that age, APACHE II score, and CRP were predictors of mortality in all the patient groups. Conclusion: In patients treated for SARS-CoV-2 infection acquiring a bacterial infection due to prolonged hospitalization associated with the treatment of COVID-19 did not elevate mortality rates. The data suggests that in severe COVID-19 patients who survived beyond the first week of hospitalization, bacterial infections, particularly Klebsiella pneumoniae NDM, do not significantly impact mortality. Multivariate analysis revealed that age, APACHE II score, and CRP were predictors of mortality in all the patient groups.


Asunto(s)
COVID-19 , Farmacorresistencia Bacteriana Múltiple , Unidades de Cuidados Intensivos , Infecciones por Klebsiella , Klebsiella pneumoniae , SARS-CoV-2 , beta-Lactamasas , Humanos , COVID-19/mortalidad , COVID-19/microbiología , Klebsiella pneumoniae/efectos de los fármacos , Klebsiella pneumoniae/enzimología , Masculino , Femenino , Infecciones por Klebsiella/mortalidad , Infecciones por Klebsiella/tratamiento farmacológico , Infecciones por Klebsiella/microbiología , Infecciones por Klebsiella/epidemiología , beta-Lactamasas/metabolismo , beta-Lactamasas/genética , Persona de Mediana Edad , Anciano , Adulto , Antibacterianos/uso terapéutico , Antibacterianos/farmacología , Anciano de 80 o más Años
9.
Front Cell Infect Microbiol ; 14: 1295841, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38707510

RESUMEN

Introduction: Although the existence of Candida species in the respiratory tract is often considered commensal, it is crucial to recognize the significance of Candida colonization in immunocompromised or COVID-19 patients. The emergence of Candida auris as an emerging pathogen further emphasizes the importance of monitoring yeast infection/colonization, particularly in COVID-19 patients. Methods: In this study, respiratory samples mainly from COVID-19 patients, primarily those suspected of having a fungal infection, were cultured on Sabouraud dextrose agar plates and the yeast colonies were identified using a two-step multiplex PCR method. The samples suspected of C. auris underwent specific nested PCR followed by sequence analysis. Results: A total of 199 respiratory samples were collected from 73 women and 126 men, ranging in age from 1.6 to 88 years. Among the patients, 141 had COVID-19, 32 had cancer, 5 were hospitalized in ICU, 2 had chronic obstructive pulmonary disease)COPD(, and others were patients with combination diseases. From these samples, a total of 334 yeast strains were identified. C. albicans (n=132, 39.52%) was the most common species, followed by C. tropicalis (n=67, 20%), C. glabrata (n=56, 16.76%), C. krusei (n=18, 5.4%), C. parapsilosis (n=17, 5.08%), Saccharomyces cerevisiae (n=10, 3%), C. kefyr (n=9, 2.6%), C. dubliniensis (n=7, 2.1%), C. lusitaniae (n=5, 1.5%), C. auris (n=3, 0.9%), C. guilliermondii (n=2, 0.6%), C. rugosa (n=1, 0.3%), C. intermedia (n=1, 0.3%), and Trichosporon spp. (n=1, 0.3%). C. auris was detected in a patient in ICU and two COVID-19 patients. While its presence was confirmed through sequence analysis, our extensive efforts to isolate C. auris were unsuccessful. Conclusion: While C. albicans colonization remains prevalent, our study found no evidence of Candida lung infection. Since the role of Candida colonization in airway secretions remains ambiguous due to limited research, further studies are imperative to shed light on this matter.


Asunto(s)
COVID-19 , Candida auris , Candidiasis , SARS-CoV-2 , Humanos , COVID-19/microbiología , Anciano , Persona de Mediana Edad , Femenino , Masculino , Anciano de 80 o más Años , Adulto , Preescolar , Candidiasis/microbiología , Niño , Adolescente , Adulto Joven , SARS-CoV-2/genética , SARS-CoV-2/aislamiento & purificación , Lactante , Candida auris/genética , Candida auris/aislamiento & purificación , Candida/aislamiento & purificación , Candida/clasificación , Candida/genética , Sistema Respiratorio/microbiología , Sistema Respiratorio/virología , Reacción en Cadena de la Polimerasa Multiplex
10.
BMC Infect Dis ; 24(1): 422, 2024 Apr 22.
Artículo en Inglés | MEDLINE | ID: mdl-38649899

RESUMEN

Cat scratch disease (CSD) is caused by Bartonella henselae (B. henselae) and presents as lymphadenopathy following close contact with cats. However, in context of the global COVID-19 pandemic, clinical manifestations of CSD may vary, posing new challenges for healthcare professionals. Here we describe a case of a 54-year-old male with painful left upper arm mass, which gradually resolved until he was infected with COVID-19. The mass then rapidly progressed before admission. Meanwhile, pulmonary symptoms including pleural effusion emerged simultaneously. The cause was undetermined with routine blood culture and pathological test until the next generation sequencing (NGS) confirmed the presence of B. henselae. We believe this case is the first to report localized aggravation of CSD after COVID-19 infection and hopefully, offers treatment experience for clinicians worldwide.


Asunto(s)
Bartonella henselae , COVID-19 , Enfermedad por Rasguño de Gato , Humanos , Masculino , COVID-19/complicaciones , COVID-19/microbiología , Bartonella henselae/genética , Bartonella henselae/aislamiento & purificación , Enfermedad por Rasguño de Gato/microbiología , Enfermedad por Rasguño de Gato/complicaciones , Enfermedad por Rasguño de Gato/tratamiento farmacológico , Persona de Mediana Edad , Infección Latente , SARS-CoV-2
11.
J Microbiol Immunol Infect ; 57(3): 457-469, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38584042

RESUMEN

INTRODUCTION: Aim of the study was the molecular characterization of 21 ceftazidime/avibactam resistant (CZA-R) Klebsiella pneumoniae strains, collected in the period October 2021-March 2022 from an Intensive Care COVID Unit in a Northern Italian Hospital. METHODS: After growth on selective/chromogenic culture media and susceptibility tests assessment, resistance genes content was ascertained for all the isolates by the HybriSpot 12 multiplexing, PCR and Whole-Genome Sequencing (WGS). Clonality was assessed by PFGE and MLST according to the Pasteur scheme. A SNPs-based phylogenetic tree was obtained comparing representative isolates and global genomes. The blaKPC gene horizontal transmission was evaluated by conjugation experiments. blaKPC-166 was cloned in a pCR2.1 vector and transformed in chemically competent TOP10 cells. RESULTS: Sixteen inpatients resulted positive for colonization and/or infection by KPC-producing K. pneumoniae (KPC-Kp) strains. The 21 CZA-R KPC-Kp isolates obtained showed MDR phenotype; susceptibility to meropenem was always retained. All the CZA-R KPC-Kp presented a novel blaKPC variant, named blaKPC-166, showing a single nucleotide substitution (T811C) compared to the blaKPC-94; but related to blaKPC-2. TWO DIFFERENT PULSOTYPES WERE DETECTED: A in 18/21 and B in 1/21 cases, two strains from the same patient being untypable by PFGE. Interestingly, the outbreak was sustained by the high-risk clone ST307, although the ST22, ST6342, ST6418 and ST6811 have also been identified and associated to KPC-166. Worryingly, blaKPC-166 could be transferred horizontally and, after cloning, it conferred resistance to CZA. DISCUSSION: This novel variant confers CZA-resistance and carbapenems susceptibility restoration. As KPC-166 was found expressed by multiple Kp clones, greater efforts should be made to prevent the further dissemination of such strains in Italian clinical settings.


Asunto(s)
Antibacterianos , Compuestos de Azabiciclo , Ceftazidima , Brotes de Enfermedades , Combinación de Medicamentos , Unidades de Cuidados Intensivos , Infecciones por Klebsiella , Klebsiella pneumoniae , Pruebas de Sensibilidad Microbiana , beta-Lactamasas , Klebsiella pneumoniae/genética , Klebsiella pneumoniae/efectos de los fármacos , Klebsiella pneumoniae/aislamiento & purificación , Humanos , Ceftazidima/farmacología , Ceftazidima/uso terapéutico , Italia/epidemiología , Infecciones por Klebsiella/epidemiología , Infecciones por Klebsiella/microbiología , Infecciones por Klebsiella/tratamiento farmacológico , Compuestos de Azabiciclo/farmacología , beta-Lactamasas/genética , Antibacterianos/farmacología , Farmacorresistencia Bacteriana Múltiple/genética , COVID-19/epidemiología , COVID-19/virología , COVID-19/microbiología , Filogenia , Proteínas Bacterianas/genética , Secuenciación Completa del Genoma , Masculino , Tipificación de Secuencias Multilocus , Femenino
12.
mBio ; 15(6): e0198223, 2024 Jun 12.
Artículo en Inglés | MEDLINE | ID: mdl-38651925

RESUMEN

Invasive pulmonary aspergillosis is a severe fungal infection primarily affecting immunocompromised patients. Individuals with severe viral infections have recently been identified as vulnerable to developing invasive fungal infections. Both influenza-associated pulmonary aspergillosis (IAPA) and COVID-19-associated pulmonary aspergillosis (CAPA) are linked to high mortality rates, emphasizing the urgent need for an improved understanding of disease pathogenesis to unveil new molecular targets with diagnostic and therapeutic potential. The recent establishment of animal models replicating the co-infection context has offered crucial insights into the mechanisms that underlie susceptibility to disease. However, the development and progression of human viral-fungal co-infections exhibit a significant degree of interindividual variability, even among patients with similar clinical conditions. This observation implies a significant role for host genetics, but information regarding the genetic basis for viral-fungal co-infections is currently limited. In this review, we discuss how genetic factors known to affect either antiviral or antifungal immunity could potentially reveal pathogenetic mechanisms that predispose to IAPA or CAPA and influence the overall disease course. These insights are anticipated to foster further research in both pre-clinical models and human patients, aiming to elucidate the complex pathophysiology of viral-associated pulmonary aspergillosis and contributing to the identification of new diagnostic and therapeutic targets to improve the management of these co-infections.


Asunto(s)
COVID-19 , Coinfección , Humanos , Coinfección/microbiología , Coinfección/inmunología , Coinfección/virología , COVID-19/inmunología , COVID-19/complicaciones , COVID-19/microbiología , COVID-19/virología , Animales , Aspergilosis Pulmonar/inmunología , SARS-CoV-2/genética , SARS-CoV-2/inmunología , Aspergilosis Pulmonar Invasiva/inmunología , Aspergilosis Pulmonar Invasiva/microbiología
13.
Crit Care ; 28(1): 133, 2024 04 22.
Artículo en Inglés | MEDLINE | ID: mdl-38649970

RESUMEN

BACKGROUND: Acute respiratory distress syndrome (ARDS) is responsible for 400,000 deaths annually worldwide. Few improvements have been made despite five decades of research, partially because ARDS is a highly heterogeneous syndrome including various types of aetiologies. Lower airway microbiota is involved in chronic inflammatory diseases and recent data suggest that it could also play a role in ARDS. Nevertheless, whether the lower airway microbiota composition varies between the aetiologies of ARDS remain unknown. The aim of this study is to compare lower airway microbiota composition between ARDS aetiologies, i.e. pulmonary ARDS due to influenza, SARS-CoV-2 or bacterial infection. METHODS: Consecutive ARDS patients according to Berlin's classification requiring invasive ventilation with PCR-confirmed influenza or SARS-CoV-2 infections and bacterial infections (> 105 CFU/mL on endotracheal aspirate) were included. Endotracheal aspirate was collected at admission, V3-V4 and ITS2 regions amplified by PCR, deep-sequencing performed on MiSeq sequencer (Illumina®) and data analysed using DADA2 pipeline. RESULTS: Fifty-three patients were included, 24 COVID-19, 18 influenza, and 11 bacterial CAP-related ARDS. The lower airway bacteriobiota and mycobiota compositions (ß-diversity) were dissimilar between the three groups (p = 0.05 and p = 0.01, respectively). The bacterial α-diversity was significantly lower in the bacterial CAP-related ARDS group compared to the COVID-19 ARDS group (p = 0.04). In contrast, influenza-related ARDS patients had higher lung mycobiota α-diversity than the COVID-19-related ARDS (p = 0 < 01). CONCLUSION: Composition of lower airway microbiota (both microbiota and mycobiota) differs between influenza, COVID-19 and bacterial CAP-related ARDS. Future studies investigating the role of lung microbiota in ARDS pathophysiology should take aetiology into account.


Asunto(s)
COVID-19 , Gripe Humana , Microbiota , Síndrome de Dificultad Respiratoria , Humanos , COVID-19/microbiología , COVID-19/complicaciones , COVID-19/fisiopatología , Síndrome de Dificultad Respiratoria/microbiología , Síndrome de Dificultad Respiratoria/virología , Síndrome de Dificultad Respiratoria/fisiopatología , Masculino , Femenino , Persona de Mediana Edad , Gripe Humana/microbiología , Gripe Humana/fisiopatología , Gripe Humana/complicaciones , Microbiota/fisiología , Anciano , Infecciones Bacterianas/microbiología
14.
Med Mycol ; 62(5)2024 May 03.
Artículo en Inglés | MEDLINE | ID: mdl-38592959

RESUMEN

Candida species are the primary cause of fungal infections in intensive care units (ICUs). Despite the increasing prevalence of Candida-related infections, monitoring the progression of these infections from colonization in COVID-19 ICU patients lacks sufficient information. This study aims prospectively to compare 62 COVID-19 and 60 non-COVID-19 ICU patients from admission to discharge in terms of colonization development, rates, isolated Candida species, risk factors, and Candida infections during hospitalization. A total of 1464 samples were collected at specific time intervals from various body sites [mouth, skin (axilla), rectal, and urine]. All samples were inoculated onto CHROMagar Candida and CHROMagar Candida Plus media, and isolates identified using MALDI-TOF MS. COVID-19 patients exhibited significantly higher colonization rates in oral, rectal, and urine samples compared to non-COVID-19 patients, (p < 0.05). Among the Candida species, non-albicans Candida was more frequently detected in COVID-19 patients, particularly in oral (75.8%-25%; p < 0.001) and rectal regions (74.19% - 46.66%; p < 0.05). Colonization with mixed Candida species was also more prevalent in the oropharyngeal region (p < 0.05). Mechanical ventilation and corticosteroid use emerged as elevated risk factors among COVID-19 patients (p < 0.05). Despite the colonization prevalence, both COVID-19-positive and negative patients exhibited low incidences of Candida infections, with rates of 9.67% (n = 6/62) and 6.67% (n = 3/60), respectively. Consequently, although Candida colonization rates were higher in COVID-19 ICU patients, there was no significant difference in Candida infection development compared to the non-COVID-19 group. However, the elevated rate of non-albicans Candida isolates highlights potential future infections, particularly given their intrinsic resistance in prophylactic or empirical treatments if needed. Additionally, the high rate of mixed colonization emphasizes the importance of using chromogenic media for routine evaluation.


This is the first prospective cohort study comparing Candida colonization features including species and body sites from the time of admission to the externalization in intensive care unit patients with and without COVID-19. It provides key points that can be referenced for fungal approaches in future disasters.


Asunto(s)
COVID-19 , Candida , Candidiasis , Unidades de Cuidados Intensivos , Humanos , Unidades de Cuidados Intensivos/estadística & datos numéricos , COVID-19/microbiología , COVID-19/epidemiología , Masculino , Estudios Prospectivos , Femenino , Persona de Mediana Edad , Candida/aislamiento & purificación , Candida/clasificación , Anciano , Candidiasis/epidemiología , Candidiasis/microbiología , Turquía/epidemiología , Adulto , Factores de Riesgo , SARS-CoV-2/aislamiento & purificación , Prevalencia , Boca/microbiología , Boca/virología
15.
Cell Host Microbe ; 32(5): 651-660.e4, 2024 May 08.
Artículo en Inglés | MEDLINE | ID: mdl-38657605

RESUMEN

The mechanisms underlying the many phenotypic manifestations of post-acute COVID-19 syndrome (PACS) are poorly understood. Herein, we characterized the gut microbiome in heterogeneous cohorts of subjects with PACS and developed a multi-label machine learning model for using the microbiome to predict specific symptoms. Our processed data covered 585 bacterial species and 500 microbial pathways, explaining 12.7% of the inter-individual variability in PACS. Three gut-microbiome-based enterotypes were identified in subjects with PACS and associated with different phenotypic manifestations. The trained model showed an accuracy of 0.89 in predicting individual symptoms of PACS in the test set and maintained a sensitivity of 86% and a specificity of 82% in predicting upcoming symptoms in an independent longitudinal cohort of subjects before they developed PACS. This study demonstrates that the gut microbiome is associated with phenotypic manifestations of PACS, which has potential clinical utility for the prediction and diagnosis of PACS.


Asunto(s)
COVID-19 , Microbioma Gastrointestinal , Aprendizaje Automático , Fenotipo , Síndrome Post Agudo de COVID-19 , SARS-CoV-2 , Humanos , COVID-19/microbiología , Masculino , Femenino , Persona de Mediana Edad , Adulto , Bacterias/clasificación , Bacterias/aislamiento & purificación , Bacterias/genética , Anciano , Heces/microbiología , Heces/virología , Estudios de Cohortes , Estudios Longitudinales
16.
mBio ; 15(5): e0053924, 2024 May 08.
Artículo en Inglés | MEDLINE | ID: mdl-38591881

RESUMEN

A recent study published in mBio by Cao et al. revealed the crucial roles of bacteria in benefitting SARS-CoV-2 mutations (B. Cao, X. Wang, W. Yin, Z. Gao, and B. Xia, mBio e3187-23, 2024, https://doi.org/10.1128/mbio.03187-23). Understanding the underlying mechanisms driving the evolution of SARS-CoV-2 is crucial for predicting the future trajectory of the COVID-19 pandemic and developing preventive and treatment strategies. This study provides important insights into the rapid and complex evolution of viruses facilitated by bacterial-virus interactions.


Asunto(s)
Bacterias , COVID-19 , Mutación , SARS-CoV-2 , SARS-CoV-2/genética , COVID-19/virología , COVID-19/microbiología , Humanos , Bacterias/genética , Bacterias/clasificación , Evolución Molecular
17.
BMC Microbiol ; 24(1): 138, 2024 Apr 24.
Artículo en Inglés | MEDLINE | ID: mdl-38658823

RESUMEN

BACKGROUND: Co-infection with other pathogens in coronavirus disease 2019 (COVID-19) patients exacerbates disease severity and impacts patient prognosis. Clarifying the exact pathogens co-infected with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is premise of the precise treatment for COVID-19 patients. METHODS: Sputum samples were collected from 17 patients in the COVID-19 positive group and 18 patients in the COVID-19 negative group. DNA extraction was performed to obtain the total DNA. Sequencing analysis using 16S and ITS rRNA gene was carried out to analyze the composition of bacterial and fungal communities. Meanwhile, all the samples were inoculated for culture. RESULTS: We did not observe significant differences in bacterial composition between the COVID-19 positive and negative groups. However, a significantly higher abundance of Candida albicans was observed in the upper respiratory tract samples from the COVID-19 positive group compared to the COVID-19 negative group. Moreover, the Candida albicans strains isolated from COVID-19 positive group exhibited impaired secretion of aspartyl proteinases. CONCLUSION: COVID-19 positive patients demonstrate a notable increase in the abundance of Candida albicans, along with a decrease in the levels of aspartyl proteinases, indicating the alteration of microbiota composition of upper respiratory tract.


Asunto(s)
Bacterias , COVID-19 , Candida albicans , Microbiota , Sistema Respiratorio , SARS-CoV-2 , Esputo , Humanos , COVID-19/microbiología , COVID-19/virología , Microbiota/genética , Masculino , Candida albicans/aislamiento & purificación , Candida albicans/genética , Femenino , Esputo/microbiología , Esputo/virología , Persona de Mediana Edad , SARS-CoV-2/genética , SARS-CoV-2/aislamiento & purificación , Bacterias/clasificación , Bacterias/aislamiento & purificación , Bacterias/genética , Sistema Respiratorio/microbiología , Sistema Respiratorio/virología , Anciano , ARN Ribosómico 16S/genética , Adulto , Coinfección/microbiología , Coinfección/virología
18.
Microbiol Spectr ; 12(5): e0416623, 2024 May 02.
Artículo en Inglés | MEDLINE | ID: mdl-38557102

RESUMEN

Throughout the COVID-19 pandemic, extensive research has been conducted on SARS-COV-2 to elucidate its genome, prognosis, and possible treatments. However, few looked at the microbial markers that could be explored in infected patients and that could predict possible disease severity. The aim of this study is to compare the nasopharyngeal microbiota of healthy subjects, moderate, under medication, and recovered SARS-COV-2 patients. In 2020, 38 nasopharyngeal swabs were collected from 6 healthy subjects, 14 moderates, 10 under medication and 8 recovered SARS-COV-2 patients at the Prince Mohammed Bin Abdulaziz Hospital Riyadh. Metatranscriptomic sequencing was performed using Minion Oxford nanopore sequencing. No significant difference in alpha as well as beta diversity was observed among all four categories. Nevertheless, we have found that Streptococcus spp including Streptococcus pneumoniae and Streptococcus thermophilus were among the top 15 most abundant species detected in COVID-19 patients but not in healthy subjects. The genus Staphylococcus was found to be associated with COVID-19 patients compared to healthy subjects. Furthermore, the abundance of Leptotrichia was significantly higher in healthy subjects compared to recovered patients. Corynebacterium on the other hand, was associated with under-medication patients. Taken together, our study revealed no differences in the overall microbial composition between healthy subjects and COVID-19 patients. Significant differences were seen only at specific taxonomic level. Future studies should explore the nasopharyngeal microbiota between controls and COVID-19 patients while controlling for confounders including age, gender, and comorbidities; since these latter could affect the results and accordingly the interpretation.IMPORTANCEIn this work, no significant difference in the microbial diversity was seen between healthy subjects and COVID-19 patients. Changes in specific taxa including Leptotrichia, Staphylococcus, and Corynebacterium were only observed. Leptotrichia was significantly higher in healthy subjects, whereas Staphylococcus and Corynebacterium were mostly associated with COVID-19, and specifically with under-medication SARS-COV-2 patients, respectively. Although the COVID-19 pandemic has ended, the SARS-COV-2 virus is continuously evolving and the emergence of new variants causing more severe disease should be always kept in mind. Microbial markers in SARS-COV-2 infected patients can be useful in the early suspicion of the disease, predicting clinical outcomes, framing hospital and intensive care unit admission as well as, risk stratification. Data on which microbial marker to tackle is still controversial and more work is needed, hence the importance of this study.


Asunto(s)
COVID-19 , Secuenciación de Nucleótidos de Alto Rendimiento , Microbiota , Nasofaringe , SARS-CoV-2 , Humanos , COVID-19/microbiología , COVID-19/virología , COVID-19/epidemiología , COVID-19/diagnóstico , Nasofaringe/microbiología , Nasofaringe/virología , Microbiota/genética , SARS-CoV-2/genética , SARS-CoV-2/aislamiento & purificación , Masculino , Femenino , Persona de Mediana Edad , Adulto , Metagenómica/métodos , Metagenoma , Anciano , Bacterias/clasificación , Bacterias/genética , Bacterias/aislamiento & purificación , Índice de Severidad de la Enfermedad , Streptococcus pneumoniae/genética , Streptococcus pneumoniae/aislamiento & purificación , Streptococcus pneumoniae/clasificación
19.
Curr Opin Infect Dis ; 37(3): 170-175, 2024 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-38437245

RESUMEN

PURPOSE OF REVIEW: Prevention of acute respiratory illnesses (ARI) in children is a global health priority, as these remain a leading cause of pediatric morbidity and mortality throughout the world. As new products and strategies to prevent respiratory infections caused by important pathogens such as severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), influenza, respiratory syncytial virus and pneumococcus are advancing, increasing evidence suggests that these and other respiratory viruses and pneumococci may exhibit interactions that are associated with altered colonization and disease dynamics. We aim to review recent data evaluating interactions between respiratory viruses and pneumococci in the upper respiratory tract and their potential impact on pneumococcal colonization patterns and disease outcomes. RECENT FINDINGS: While interactions between influenza infection and subsequent increased susceptibility and transmissibility of colonizing pneumococci have been widely reported in the literature, emerging evidence suggests that human rhinovirus, SARS-CoV-2, and other viruses may also exhibit interactions with pneumococci and alter pneumococcal colonization patterns. Additionally, colonizing pneumococci may play a role in modifying outcomes associated with respiratory viral infections. Recent evidence suggests that vaccination with pneumococcal conjugate vaccines, and prevention of colonization with pneumococcal serotypes included in these vaccines, may be associated with reducing the risk of subsequent viral infection and the severity of the associated illnesses. SUMMARY: Understanding the direction and dynamics of viral-pneumococcal interactions may elucidate the potential effects of existing and emerging viral and bacterial vaccines and other preventive strategies on the health impact of these important respiratory pathogens.


Asunto(s)
Nasofaringe , Infecciones Neumocócicas , Infecciones del Sistema Respiratorio , Streptococcus pneumoniae , Humanos , Infecciones del Sistema Respiratorio/microbiología , Infecciones del Sistema Respiratorio/virología , Niño , Infecciones Neumocócicas/prevención & control , Infecciones Neumocócicas/microbiología , Nasofaringe/microbiología , Nasofaringe/virología , COVID-19/microbiología , SARS-CoV-2 , Vacunas Neumococicas , Preescolar , Coinfección/microbiología , Virosis
20.
J Glob Antimicrob Resist ; 37: 165-167, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38458537

RESUMEN

PURPOSE: The impact of the COVID-19 pandemic on antimicrobial resistance (AMR) is largely studied in healthcare settings. There is a need to understand the fluctuations in AMR during pandemic at the community level. With urinary tract infection (UTI) being one of the most common infections in the community, the AMR profile of community-acquired UTI (CA-UTI) is considered representative AMR at the community level. METHODS: The study was taken in a cohort of patients with a clinical diagnosis of CA-UTI. The four study sites represented different community health centres in India. Escherichia coli isolates were analysed phenotypically and genotypically for AMR pre-COVID (October 2019-February 2020) and in the first (March 2020-February 2021) and second waves of COVID-19 (March 2021-December 2021). RESULTS: E. coli was the predominant uropathogen (229, 82%). Increased susceptibility to nitrofurantoin was observed during the pandemic. Reduced susceptibility to first-line oral antibiotics and carbapenems was seen during the second wave, and an increased minimum inhibitory concentration (MIC50) to beta-lactams and fluoroquinolones was seen during the pandemic. Genomic analysis of E. coli isolates showed some AMR genes (aacC1, aacC4, SHV, QepA) only during the second wave. CONCLUSION: One good outcome of the pandemic was increased susceptibility to nitrofurantoin, while drawback was a significant decrease in susceptibility to oral antibiotics during the second wave and increased MIC50 of some antibiotics. Decreased susceptibility to last-resort carbapenems and the occurrence of various AMR genes during the second wave of the pandemic are of great concern.


Asunto(s)
Antibacterianos , COVID-19 , Infecciones Comunitarias Adquiridas , Infecciones por Escherichia coli , Escherichia coli , Pruebas de Sensibilidad Microbiana , Infecciones Urinarias , Humanos , Infecciones Urinarias/microbiología , Infecciones Urinarias/epidemiología , India/epidemiología , COVID-19/epidemiología , COVID-19/microbiología , Infecciones Comunitarias Adquiridas/microbiología , Infecciones Comunitarias Adquiridas/epidemiología , Escherichia coli/efectos de los fármacos , Escherichia coli/genética , Escherichia coli/aislamiento & purificación , Infecciones por Escherichia coli/microbiología , Infecciones por Escherichia coli/epidemiología , Antibacterianos/farmacología , SARS-CoV-2/efectos de los fármacos , SARS-CoV-2/genética , Farmacorresistencia Bacteriana , Pandemias , Femenino
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA