Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Front Microbiol ; 15: 1423995, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39035445

RESUMEN

Streptococcus pneumoniae is a major pathogen responsible for severe complications in patients with prior influenza A virus (IAV) infection. We have previously demonstrated that S. pneumoniae exhibits increased intracellular survival within IAV-infected cells. Fluoroquinolones (FQs) are widely used to treat pneumococcal infections. However, our prior work has shown that S. pneumoniae can develop intracellular FQ persistence, a phenomenon triggered by oxidative stress within host cells. This persistence allows the bacteria to withstand high FQ concentrations. In this study, we show that IAV infection enhances pneumococcal FQ persistence during intracellular survival within pneumocytes, macrophages, and neutrophils. This enhancement is partly due to increased oxidative stress induced by the viral infection. We find that this phenotype is particularly pronounced in autophagy-proficient host cells, potentially resulting from IAV-induced blockage of autophagosome-lysosome fusion. Moreover, we identified several S. pneumoniae genes involved in oxidative stress response that contribute to FQ persistence, including sodA (superoxide dismutase), clpL (chaperone), nrdH (glutaredoxin), and psaB (Mn+2 transporter component). Our findings reveal a novel mechanism of antibiotic persistence promoted by viral infection within host cells. This underscores the importance of considering this phenomenon when using FQs to treat pneumococcal infections, especially in patients with concurrent influenza A infection.

2.
Front Microbiol ; 14: 1269843, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37789846

RESUMEN

Streptococcus pneumoniae is a gram-positive, aerotolerant bacterium that naturally colonizes the human nasopharynx, but also causes invasive infections and is a major cause of morbidity and mortality worldwide. This pathogen produces high levels of H2O2 to eliminate other microorganisms that belong to the microbiota of the respiratory tract. However, it also induces an oxidative stress response to survive under this stressful condition. Furthermore, this self-defense mechanism is advantageous in tolerating oxidative stress imposed by the host's immune response. This review provides a comprehensive overview of the strategies employed by the pneumococcus to survive oxidative stress. These strategies encompass the utilization of H2O2 scavengers and thioredoxins, the adaptive response to antimicrobial host oxidants, the regulation of manganese and iron homeostasis, and the intricate regulatory networks that control the stress response. Here, we have also summarized less explored aspects such as the involvement of reparation systems and polyamine metabolism. A particular emphasis is put on the role of the oxidative stress response during the transient intracellular life of Streptococcus pneumoniae, including coinfection with influenza A and the induction of antibiotic persistence in host cells.

3.
Microbiol Spectr ; 10(6): e0436422, 2022 12 21.
Artículo en Inglés | MEDLINE | ID: mdl-36445159

RESUMEN

Bacterial persisters represent a small subpopulation that tolerates high antibiotic concentrations without acquiring heritable resistance, and it may be generated by environmental factors. Here, we report the first antibiotic persistence mechanism in Streptococcus pneumoniae, which is induced by oxidative stress conditions and allows the pneumococcus to survive in the presence of fluoroquinolones. We demonstrated that fluoroquinolone persistence is prompted by both the impact of growth arrest and the oxidative stress response induced by H2O2 in bacterial cells. This process protected pneumococci against the deleterious effects of high ROS levels induced by fluoroquinolones. Importantly, S. pneumoniae develops persistence during infection, and is dependent on the oxidative stress status of the host cells, indicating that its transient intracellular life contributes to this mechanism. Furthermore, our findings suggest persistence may influence the outcome of antibiotic therapy and be part of a multistep mechanism in the evolution of fluoroquinolone resistance. IMPORTANCE In S. pneumoniae, different mechanisms that counteract antibiotic effects have been described, such as vancomycin tolerance, heteroresistance to penicillin and fluoroquinolone resistance, which critically affect the therapeutic efficacy. Antibiotic persistence is a type of antibiotic tolerance that allows a bacterial subpopulation to survive lethal antimicrobial concentrations. In this work, we used a host-cell infection model to reveal fluoroquinolone persistence in S. pneumoniae. This mechanism is induced by oxidative stress that the pneumococcus must overcome to survive in host cells. Many fluoroquinolones, such as levofloxacin and moxifloxacin, have a broad spectrum of activity against bacterial pathogens of community-acquired pneumonia, and they are used to treat pneumococcal diseases. However, the emergence of fluoroquinolone-resistant strains complicates antibiotic treatment of invasive infections. Consequently, antibiotic persistence in S. pneumoniae is clinically relevant due to prolonged exposure to fluoroquinolones likely favors the acquisition of mutations that generate antibiotic resistance in persisters. In addition, this work contributes to the knowledge of antibiotic persistence mechanisms in bacteria.


Asunto(s)
Fluoroquinolonas , Streptococcus pneumoniae , Streptococcus pneumoniae/genética , Fluoroquinolonas/farmacología , Peróxido de Hidrógeno/farmacología , Antibacterianos/farmacología , Antibacterianos/uso terapéutico , Levofloxacino/farmacología , Bacterias , Pruebas de Sensibilidad Microbiana
4.
BMC Genomics ; 23(1): 510, 2022 Jul 14.
Artículo en Inglés | MEDLINE | ID: mdl-35836127

RESUMEN

BACKGROUND: The SARS-CoV-2 virus is responsible for the COVID-19 pandemic. To better understand the evolution of SARS-CoV-2 early in the pandemic in the Province of Cordoba, Argentina, we performed a comparative genomic analysis of SARS-CoV-2 strains detected in survivors and non-survivors of COVID-19. We also carried out an epidemiological study to find a possible association between the symptoms and comorbidities of these patients with their clinical outcomes. RESULTS: A representative sampling was performed in different cities in the Province of Cordoba. Ten and nine complete SARS-CoV-2 genomes were obtained by next-generation sequencing of nasopharyngeal specimens from non-survivors and survivors, respectively. Phylogenetic and phylodynamic analyses revealed multiple introductions of the most common lineages in South America, including B.1, B.1.1.1, B.1.499, and N.3. Fifty-six mutations were identified, with 14% of those in common between the non-survivor and survivor groups. Specific SARS-CoV-2 mutations for survivors constituted 25% whereas for non-survivors they were 41% of the repertoire, indicating partial selectivity. The non-survivors' variants showed higher diversity in 9 genes, with a majority in Nsp3, while the survivors' variants were detected in 5 genes, with a higher incidence in the Spike protein. At least one comorbidity was present in 60% of non-survivor patients and 33% of survivors. Age 75-85 years (p = 0.018) and hospitalization (p = 0.019) were associated with non-survivor patients. Related to the most common symptoms, the prevalence of fever was similar in both groups, while dyspnea was more frequent among non-survivors and cough among survivors. CONCLUSIONS: This study describes the association of clinical characteristics with the clinical outcomes of survivors and non-survivors of COVID-19 patients, and the specific mutations found in the genome sequences of SARS-CoV-2 in each patient group. Future research on the functional characterization of novel mutations should be performed to understand the role of these variations in SARS-CoV-2 pathogenesis and COVID-19 disease outcomes. These results add new genomic data to better understand the evolution of the SARS-CoV-2 variants that spread in Argentina during the first wave of the COVID-19 pandemic.


Asunto(s)
COVID-19 , SARS-CoV-2 , Anciano , Anciano de 80 o más Años , Argentina/epidemiología , COVID-19/epidemiología , Genoma Viral , Genómica , Humanos , Pandemias , Filogenia , SARS-CoV-2/genética
5.
PLoS Pathog ; 16(8): e1008761, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32790758

RESUMEN

The virus-bacterial synergism implicated in secondary bacterial infections caused by Streptococcus pneumoniae following infection with epidemic or pandemic influenza A virus (IAV) is well documented. However, the molecular mechanisms behind such synergism remain largely ill-defined. In pneumocytes infected with influenza A virus, subsequent infection with S. pneumoniae leads to enhanced pneumococcal intracellular survival. The pneumococcal two-component system SirRH appears essential for such enhanced survival. Through comparative transcriptomic analysis between the ΔsirR and wt strains, a list of 179 differentially expressed genes was defined. Among those, the clpL protein chaperone gene and the psaB Mn+2 transporter gene, which are involved in the stress response, are important in enhancing S. pneumoniae survival in influenza-infected cells. The ΔsirR, ΔclpL and ΔpsaB deletion mutants display increased susceptibility to acidic and oxidative stress and no enhancement of intracellular survival in IAV-infected pneumocyte cells. These results suggest that the SirRH two-component system senses IAV-induced stress conditions and controls adaptive responses that allow survival of S. pneumoniae in IAV-infected pneumocytes.


Asunto(s)
Proteínas Bacterianas/metabolismo , Coinfección/mortalidad , Virus de la Influenza A/patogenicidad , Gripe Humana/mortalidad , Pulmón/patología , Infecciones Neumocócicas/mortalidad , Streptococcus pneumoniae/patogenicidad , Proteínas Bacterianas/genética , Supervivencia Celular , Coinfección/epidemiología , Humanos , Gripe Humana/microbiología , Gripe Humana/patología , Gripe Humana/virología , Pulmón/microbiología , Pulmón/virología , Infecciones Neumocócicas/microbiología , Infecciones Neumocócicas/patología , Infecciones Neumocócicas/virología , Proteínas Quinasas/genética , Proteínas Quinasas/metabolismo , Streptococcus pneumoniae/metabolismo , Estrés Fisiológico , Virulencia
6.
PLoS Pathog ; 14(6): e1007118, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29883472

RESUMEN

Streptococcus pneumoniae is an opportunistic human bacterial pathogen that usually colonizes the upper respiratory tract, but the invasion and survival mechanism in respiratory epithelial cells remains elusive. Previously, we described that acidic stress-induced lysis (ASIL) and intracellular survival are controlled by ComE through a yet unknown activation mechanism under acidic conditions, which is independent of the ComD histidine kinase that activates this response regulator for competence development at pH 7.8. Here, we demonstrate that the serine/threonine kinase StkP is essential for ASIL, and show that StkP phosphorylates ComE at Thr128. Molecular dynamic simulations predicted that Thr128-phosphorylation induces conformational changes on ComE's DNA-binding domain. Using nonphosphorylatable (ComET128A) and phosphomimetic (ComET128E) proteins, we confirmed that Thr128-phosphorylation increased the DNA-binding affinity of ComE. The non-phosphorylated form of ComE interacted more strongly with StkP than the phosphomimetic form at acidic pH, suggesting that pH facilitated crosstalk. To identify the ComE-regulated genes under acidic conditions, a comparative transcriptomic analysis was performed between the comET128A and wt strains, and differential expression of 104 genes involved in different cellular processes was detected, suggesting that the StkP/ComE pathway induced global changes in response to acidic stress. In the comET128A mutant, the repression of spxB and sodA correlated with decreased H2O2 production, whereas the reduced expression of murN correlated with an increased resistance to cell wall antibiotic-induced lysis, compatible with cell wall alterations. In the comET128A mutant, ASIL was blocked and acid tolerance response was higher compared to the wt strain. These phenotypes, accompanied with low H2O2 production, are likely responsible for the increased survival in pneumocytes of the comET128A mutant. We propose that the StkP/ComE pathway controls the stress response, thus affecting the intracellular survival of S. pneumoniae in pneumocytes, one of the first barriers that this pathogen must cross to establish an infection.


Asunto(s)
Ácidos/farmacología , Proteínas Bacterianas/metabolismo , Regulación Bacteriana de la Expresión Génica , Proteínas Serina-Treonina Quinasas/metabolismo , Streptococcus pneumoniae/crecimiento & desarrollo , Estrés Fisiológico , Células A549 , Proteínas Bacterianas/química , Proteínas Bacterianas/genética , Humanos , Estrés Oxidativo/efectos de los fármacos , Proteínas Serina-Treonina Quinasas/química , Proteínas Serina-Treonina Quinasas/genética , Streptococcus pneumoniae/efectos de los fármacos
8.
Int J Med Microbiol ; 305(1): 157-69, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25543170

RESUMEN

Streptococcus pneumoniae is a major human pathogen that can survive to stress conditions, such as the acidic environment of inflammatory foci, and tolerates lethal pH through a mechanism known as the acid tolerance response. We previously described that S. pneumoniae activates acidic-stress induced lysis in response to acidified environments, favoring the release of cell wall compounds, DNA and virulence factors. Here, we demonstrate that F(0)F(1)-ATPase is involved in the response to acidic stress. Chemical inhibitors (DCCD, optochin) of this proton pump repressed the ATR induction, but caused an increased ASIL. Confirming these findings, mutants of the subunit c of this enzyme showed the same phenotypes as inhibitors. Importantly, we demonstrated that F(0)F(1)-ATPase and ATR are necessary for the intracellular survival of the pneumococcus in macrophages. Alternatively, a screening of two-component system (TCS) mutants showed that ATR and survival in pneumocytes were controlled in contrasting ways by ComDE and CiaRH, which had been involved in the ASIL mechanism. Briefly, CiaRH was essential for ATR (ComE represses activation) whereas ComE was necessary for ASIL (CiaRH protects against induction). They did not regulate F0F1-ATPase expression, but control LytA expression on the pneumococcal surface. These results suggest that both TCSs and F(0)F(1)-ATPase control a stress response and decide between a survival or a suicide mechanism by independent pathways, either in vitro or in pneumocyte cultures. This biological model contributes to the current knowledge about bacterial response under stress conditions in host tissues, where pathogens need to survive in order to establish infections.


Asunto(s)
Viabilidad Microbiana , ATPasas de Translocación de Protón/metabolismo , Transducción de Señal , Streptococcus pneumoniae/fisiología , Estrés Fisiológico , Ácidos/toxicidad , Células Epiteliales Alveolares/microbiología , Animales , Línea Celular , Técnicas de Inactivación de Genes , Humanos , Concentración de Iones de Hidrógeno , Macrófagos/microbiología , Ratones , ATPasas de Translocación de Protón/genética , Streptococcus pneumoniae/genética
9.
Diagn Microbiol Infect Dis ; 70(1): 145-9, 2011 May.
Artículo en Inglés | MEDLINE | ID: mdl-21513850

RESUMEN

Dermatophytic mycetoma is an extremely rare subcutaneous mycosis. Here, we report the case of a 6-year-old girl with clinical, histologic, and mycologic findings consistent with a mycetoma of the scalp caused by Microsporum canis. To our knowledge, this is the first report showing the immunologic and immunogenetic features of a patient with a recalcitrant dermatophytic mycetoma.


Asunto(s)
Microsporum/aislamiento & purificación , Micetoma/diagnóstico , Micetoma/patología , Cuero Cabelludo/microbiología , Cuero Cabelludo/patología , Antifúngicos/uso terapéutico , Niño , Femenino , Genotipo , Histocitoquímica , Humanos , Microscopía , Microsporum/genética , Tipificación Molecular , Micetoma/microbiología , Micetoma/terapia , Técnicas de Tipificación Micológica , Reacción en Cadena de la Polimerasa
10.
PLoS Pathog ; 7(2): e1002000, 2011 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-21379570

RESUMEN

The prevalence of antibiotic resistance genes in pathogenic bacteria is a major challenge to treating many infectious diseases. The spread of these genes is driven by the strong selection imposed by the use of antibacterial drugs. However, in the absence of drug selection, antibiotic resistance genes impose a fitness cost, which can be ameliorated by compensatory mutations. In Streptococcus pneumoniae, ß-lactam resistance is caused by mutations in three penicillin-binding proteins, PBP1a, PBP2x, and PBP2b, all of which are implicated in cell wall synthesis and the cell division cycle. We found that the fitness cost and cell division defects conferred by pbp2b mutations (as determined by fitness competitive assays in vitro and in vivo and fluorescence microscopy) were fully compensated by the acquisition of pbp2x and pbp1a mutations, apparently by means of an increased stability and a consequent mislocalization of these protein mutants. Thus, these compensatory combinations of pbp mutant alleles resulted in an increase in the level and spectrum of ß-lactam resistance. This report describes a direct correlation between antibiotic resistance increase and fitness cost compensation, both caused by the same gene mutations acquired by horizontal transfer. The clinical origin of the pbp mutations suggests that this intergenic compensatory process is involved in the persistence of ß-lactam resistance among circulating strains. We propose that this compensatory mechanism is relevant for ß-lactam resistance evolution in Streptococcus pneumoniae.


Asunto(s)
Aminoaciltransferasas/genética , Proteínas Bacterianas/genética , Evolución Molecular , Mutación/genética , Proteínas de Unión a las Penicilinas/genética , Peptidil Transferasas/genética , Streptococcus pneumoniae/efectos de los fármacos , Streptococcus pneumoniae/genética , Resistencia betalactámica/genética , Aminoaciltransferasas/química , Aminoaciltransferasas/metabolismo , Proteínas Bacterianas/química , Proteínas Bacterianas/metabolismo , División Celular , ADN Bacteriano/genética , Citometría de Flujo , Humanos , Nasofaringe/metabolismo , Proteínas de Unión a las Penicilinas/química , Proteínas de Unión a las Penicilinas/metabolismo , Peptidil Transferasas/química , Peptidil Transferasas/metabolismo , Infecciones Neumocócicas/tratamiento farmacológico , Infecciones Neumocócicas/genética , Infecciones Neumocócicas/microbiología , Reacción en Cadena de la Polimerasa , Estabilidad Proteica , Streptococcus pneumoniae/aislamiento & purificación , beta-Lactamas/farmacología
11.
J Antimicrob Chemother ; 62(5): 973-7, 2008 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-18684700

RESUMEN

OBJECTIVES: The aim of this work was to study the effect of subinhibitory concentrations of penicillin, chloramphenicol and erythromycin on the mutation rate of Streptococcus pneumoniae. METHODS: The mutation rate to rifampicin and optochin resistance was estimated using fluctuation analysis in three capsulated S. pneumoniae strains, cultured both with and without different subinhibitory antibiotic concentrations. The atpAC and rpoB mutations that conferred optochin and rifampicin resistance, respectively, were identified by DNA sequencing. RESULTS: The exposure to subinhibitory concentrations of penicillin increased the mutation rate (expressed as mutation per cell division) to optochin resistance between 2.1- and 3.1-fold for all three strains studied. In contrast, the rifampicin resistance assay showed no significant variations. To analyse the putative cause of the different responses between the optochin and rifampicin tests, mutations that conferred resistance in both cases were analysed. The difference may be explained by the genetic nature of the atpAC mutations, mostly transversions, which are not efficiently repaired by the HexAB mismatch repair system. CONCLUSIONS: We demonstrated that subinhibitory concentrations of penicillin significantly increased the mutation rate of S. pneumoniae, suggesting that exposure to this antibiotic could help this pathogen to acquire mutations that confer resistance to other antibiotics. The optochin test was useful to detect this phenomenon and it should be considered for further mutability analysis in S. pneumoniae.


Asunto(s)
Antibacterianos/farmacología , Farmacorresistencia Bacteriana , Mutágenos/farmacología , Mutación Missense , Penicilinas/farmacología , Quinina/análogos & derivados , Streptococcus pneumoniae/efectos de los fármacos , Proteínas Bacterianas/genética , Análisis Mutacional de ADN , ADN Bacteriano/química , ADN Bacteriano/genética , ARN Polimerasas Dirigidas por ADN/genética , Humanos , Datos de Secuencia Molecular , ATPasas de Translocación de Protón/genética , Quinina/farmacología , Análisis de Secuencia de ADN , Streptococcus pneumoniae/genética
12.
J Med Microbiol ; 57(Pt 8): 992-999, 2008 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-18628501

RESUMEN

The penicillin-resistant Spain(9V)-3 clone of Streptococcus pneumoniae is widespread and presents different serotype variants originating from recombination of the capsular genes. In this work, the genetic relatedness of 29 invasive pneumococci isolated from the central region of Argentina (Cordoba, Buenos Aires, Santa Fe and La Pampa provinces) was assessed by multilocus sequence typing (MLST). All of the penicillin-non-susceptible isolates studied (21/29) belonged to a serotype 14 variant of the Spain(9V)-3 clone. This clone was predominant, suggesting that it was responsible for the penicillin resistance spread in this region. Interestingly, this serotype 14 variant (named Cordoba S14V) could be differentiated from the European one by its pbp1a gene, suggesting a different recombinational replacement of the capsular genes. The putative recombination sites were analysed, resulting in the proximal crossover point being clearly localized in the spr0309 gene, with the distal site restricted to the recU gene, confirming a different recombination event. Analysis of the dexB, cpsB, aliA and pbp1a genes from these strains showed a high similarity with the corresponding genes of the Spain(14)-5 clone, suggesting that the capsular genes were provided by this international clone. Analysis of the genetic polymorphisms of the pbp1a (nt 1473-1922) and spr0309 (nt 1-790) genes is proposed as an epidemiological tool to help recognize the Cordoba S14V of the Spain(9V)-3 clone. On the other hand, BOX-repeat-based PCR and MLST analyses of serotype 14 strains revealed a divergent epidemiology of the Cordoba S14V, suggesting a non-recent dissemination in the paediatric population. It is suggested that this molecular epidemiology work will be a reference for monitoring the evolution of S14Vs of Spain(9V)-3, the emergence of new clones and the impact of pneumococcal vaccination programmes in Argentina.


Asunto(s)
Variación Genética , Streptococcus pneumoniae/clasificación , Streptococcus pneumoniae/genética , Argentina , Secuencia de Bases , Sangre/microbiología , Clonación Molecular , Cartilla de ADN , Demografía , Humanos , Pruebas de Sensibilidad Microbiana , Datos de Secuencia Molecular , Reacción en Cadena de la Polimerasa , Polimorfismo de Longitud del Fragmento de Restricción , Serotipificación , Streptococcus pneumoniae/aislamiento & purificación
13.
Microbiology (Reading) ; 154(Pt 5): 1300-1308, 2008 May.
Artículo en Inglés | MEDLINE | ID: mdl-18451038

RESUMEN

In Streptococcus pneumoniae, autolysis is considered a programmed cell-death process executed principally by the major autolysin (LytA), and the underlying mechanism causing its activation is not completely understood. It is known that autolysis is triggered by competence development at alkaline pH and regulated by a two-component system, ComDE, which senses a competence-stimulating peptide (CSP) and behaves as a quorum-sensing mechanism. In this work, we found that acidic stress triggered a LytA-mediated autolysis and, curiously, this phenomenon was regulated by a CSP-independent ComE pathway. A further analysis of a hyperactive ComD mutant revealed that ComE needs to be phosphorylated to activate acidic stress-induced lysis (ASIL). The comE transcripts were induced by acidic culture conditions, suggesting that ComE could be sensing acidic stress. We also investigated CiaRH, a two-component system whose null mutants show a comE derepression and a CSP-dependent autolysis induction at alkaline pH. By analysis of cia comE double mutants, we demonstrated that CiaRH protected cells from ASIL by a ComE-independent pathway. Here, we propose that ComE is the principal route of the signalling pathway that determines a global stress response, and clearly regulates the induction of the LytA-mediated programmed cell death in S. pneumoniae. Acidic stress may represent for S. pneumoniae an alternative condition, in addition to competence and antibiotics, to assure the release of virulence factors, DNA and cell-wall compounds by autolysis, favouring genetic exchange and contributing to its pathogenesis.


Asunto(s)
Ácidos/farmacología , Antibacterianos/farmacología , Proteínas Bacterianas/metabolismo , Bacteriólisis , Streptococcus pneumoniae/efectos de los fármacos , Proteínas Bacterianas/genética , Eliminación de Gen , Perfilación de la Expresión Génica , Fosforilación , Proteínas Quinasas/genética , ARN Bacteriano/biosíntesis , ARN Mensajero/biosíntesis
14.
J Clin Microbiol ; 46(6): 1930-4, 2008 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-18417665

RESUMEN

Optochin susceptibility is a key test used for pneumococcal diagnosis, but optochin-resistant (Opt(r)) pneumococci have been reported in the last 2 decades. In this work, we characterized eight Opt(r) clinical strains which presented a new mutation, G47V, a predominant A49S mutation (recently reported in Brazil) and A49T. These mutations were found in the c subunit of the F(0)F(1) ATPase encoded by the atpC gene, and W206C was found in the a subunit encoded by the atpA gene. The Opt(r) clinical isolates were analyzed by BOX PCR, multilocus sequence typing, and serotype and antimicrobial resistance profiles, and they showed no epidemiological relationship. To characterize the Opt(r) mutations that could emerge among clinical strains, we studied a pool of spontaneous Opt(r) colonies obtained in vitro from the virulent D39 strain. We compared the atpAC mutations of these Opt(r) pneumococci (with or without passage through C57BL/6 mice) with those described in the clinical isolates. This analysis revealed three new mutations, G47V and L26M in the c subunit and L184S in the a subunit. Most of the mutations identified in the laboratory-generated Opt(r) strains were also found in clinical strains, with the exception of the L26M and L184S mutations, and we suppose that both mutations could emerge among invasive strains in the future. Considering that atpAC are essential genes, we propose that all spontaneous mutations that confer in vitro optochin resistance would not present severe physiological alterations in S. pneumoniae and may be carried by circulating pneumococcal strains.


Asunto(s)
Farmacorresistencia Bacteriana , Quinina/análogos & derivados , Pase Seriado , Streptococcus pneumoniae , Animales , Antibacterianos/farmacología , Argentina , Proteínas Bacterianas/genética , Niño , Preescolar , Farmacorresistencia Bacteriana/genética , Femenino , Humanos , Ratones , Ratones Endogámicos C57BL , Pruebas de Sensibilidad Microbiana , Datos de Secuencia Molecular , Mutación , Infecciones Neumocócicas/microbiología , ATPasas de Translocación de Protón/genética , Quinina/farmacología , Análisis de Secuencia de ADN , Streptococcus pneumoniae/clasificación , Streptococcus pneumoniae/efectos de los fármacos , Streptococcus pneumoniae/aislamiento & purificación , Streptococcus pneumoniae/patogenicidad , Virulencia
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA