Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 1.022
Filtrar
Más filtros

Intervalo de año de publicación
1.
Clin Microbiol Rev ; 37(1): e0010122, 2024 03 14.
Artículo en Inglés | MEDLINE | ID: mdl-38235979

RESUMEN

Pneumocystis jirovecii is a ubiquitous opportunistic fungus that can cause life-threatening pneumonia. People with HIV (PWH) who have low CD4 counts are one of the populations at the greatest risk of Pneumocystis jirovecii pneumonia (PCP). While guidelines have approached the diagnosis, prophylaxis, and management of PCP, the numerous studies of PCP in PWH are dominated by the 1980s and 1990s. As such, most studies have included younger male populations, despite PCP affecting both sexes and a broad age range. Many studies have been small and observational in nature, with an overall lack of randomized controlled trials. In many jurisdictions, and especially in low- and middle-income countries, the diagnosis can be challenging due to lack of access to advanced and/or invasive diagnostics. Worldwide, most patients will be treated with 21 days of high-dose trimethoprim sulfamethoxazole, although both the dose and the duration are primarily based on historical practice. Whether treatment with a lower dose is as effective and less toxic is gaining interest based on observational studies. Similarly, a 21-day tapering regimen of prednisone is used for patients with more severe disease, yet other doses, other steroids, or shorter durations of treatment with corticosteroids have not been evaluated. Now with the widespread availability of antiretroviral therapy, improved and less invasive PCP diagnostic techniques, and interest in novel treatment strategies, this review consolidates the scientific body of literature on the diagnosis and management of PCP in PWH, as well as identifies areas in need of more study and thoughtfully designed clinical trials.


Asunto(s)
Infecciones por VIH , Pneumocystis carinii , Neumonía por Pneumocystis , Femenino , Humanos , Masculino , Neumonía por Pneumocystis/diagnóstico , Neumonía por Pneumocystis/tratamiento farmacológico , Neumonía por Pneumocystis/prevención & control , Infecciones por VIH/complicaciones , Infecciones por VIH/tratamiento farmacológico , Combinación Trimetoprim y Sulfametoxazol/uso terapéutico , Combinación Trimetoprim y Sulfametoxazol/farmacología
2.
Antimicrob Agents Chemother ; 68(5): e0005724, 2024 May 02.
Artículo en Inglés | MEDLINE | ID: mdl-38526080

RESUMEN

Enterohemorrhagic Escherichia coli causes watery to bloody diarrhea, which may progress to hemorrhagic colitis and hemolytic-uremic syndrome. While early studies suggested that antibiotic treatment may worsen the pathology of an enterohemorrhagic Escherichia coli (EHEC) infection, recent work has shown that certain non-Shiga toxin-inducing antibiotics avert disease progression. Unfortunately, both intestinal bacterial infections and antibiotic treatment are associated with dysbiosis. This can alleviate colonization resistance, facilitate secondary infections, and potentially lead to more severe illness. To address the consequences in the context of an EHEC infection, we used the established mouse infection model organism Citrobacter rodentium ϕstx2dact and monitored changes in fecal microbiota composition during infection and antibiotic treatment. C. rodentium ϕstx2dact infection resulted in minor changes compared to antibiotic treatment. The infection caused clear alterations in the microbial community, leading mainly to a reduction of Muribaculaceae and a transient increase in Enterobacteriaceae distinct from Citrobacter. Antibiotic treatments of the infection resulted in marked and distinct variations in microbiota composition, diversity, and dispersion. Enrofloxacin and trimethoprim/sulfamethoxazole, which did not prevent Shiga toxin-mediated organ damage, had the least disruptive effects on the intestinal microbiota, while kanamycin and tetracycline, which rapidly cleared the infection without causing organ damage, caused a severe reduction in diversity. Kanamycin treatment resulted in the depletion of all but Bacteroidetes genera, whereas tetracycline effects on Clostridia were less severe. Together, these data highlight the need to address the impact of individual antibiotics in the clinical care of life-threatening infections and consider microbiota-regenerating therapies.IMPORTANCEUnderstanding the impact of antibiotic treatment on EHEC infections is crucial for appropriate clinical care. While discouraged by early studies, recent findings suggest certain antibiotics can impede disease progression. Here, we investigated the impact of individual antibiotics on the fecal microbiota in the context of an established EHEC mouse model using C. rodentium ϕstx2dact. The infection caused significant variations in the microbiota, leading to a transient increase in Enterobacteriaceae distinct from Citrobacter. However, these effects were minor compared to those observed for antibiotic treatments. Indeed, antibiotics that most efficiently cleared the infection also had the most detrimental effect on the fecal microbiota, causing a substantial reduction in microbial diversity. Conversely, antibiotics showing adverse effects or incomplete bacterial clearance had a reduced impact on microbiota composition and diversity. Taken together, our findings emphasize the delicate balance required to weigh the harmful effects of infection and antibiosis in treatment.


Asunto(s)
Antibacterianos , Citrobacter rodentium , Infecciones por Enterobacteriaceae , Heces , Microbioma Gastrointestinal , Ratones Endogámicos C57BL , Animales , Citrobacter rodentium/efectos de los fármacos , Ratones , Antibacterianos/farmacología , Antibacterianos/uso terapéutico , Antibacterianos/efectos adversos , Heces/microbiología , Infecciones por Enterobacteriaceae/tratamiento farmacológico , Infecciones por Enterobacteriaceae/microbiología , Microbioma Gastrointestinal/efectos de los fármacos , Combinación Trimetoprim y Sulfametoxazol/uso terapéutico , Combinación Trimetoprim y Sulfametoxazol/farmacología , Escherichia coli Enterohemorrágica/efectos de los fármacos , Enrofloxacina/farmacología , Enrofloxacina/uso terapéutico , Femenino , Modelos Animales de Enfermedad , Disbiosis/microbiología
3.
J Antimicrob Chemother ; 79(3): 559-563, 2024 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-38217846

RESUMEN

BACKGROUND: Risk factors for ciprofloxacin or MDR in primary care urine specimens are not well defined. OBJECTIVES: We created a primary care-specific antibiogram for Escherichia coli isolates from cases with complicated and uncomplicated urinary tract infection (UTI) and evaluated risk factors for ciprofloxacin, trimethoprim/sulfamethoxazole and MDR among Enterobacterales. METHODS: We conducted a cross-sectional study to determine resistance and risk factors by collecting urine cultures from all patients (≥18 years) presenting with provider-suspected UTI at two primary care, safety-net clinics in Houston, TX, USA between November 2018 and March 2020. RESULTS: Among 1262 cultures, 308 cultures grew 339 uropathogens. Patients with Enterobacterales (n = 199) were mostly female (93.5%) with a mean age of 48.5 years. E. coli was the predominant uropathogen isolated (n = 187/339; 55%) and had elevated trimethoprim/sulfamethoxazole (43.6%) and ciprofloxacin (29.5%) resistance, low nitrofurantoin (1.8%) resistance, and no fosfomycin resistance. Among E. coli, 10.6% were ESBL positive and 24.9% had MDR. Birth outside the U.S.A., prior (2 year) trimethoprim/sulfamethoxazole resistance, and diabetes mellitus were associated with trimethoprim/sulfamethoxazole resistance. Prior (60 day) fluoroquinolone use, prior ciprofloxacin resistance and both diabetes mellitus and hypertension were strongly associated with ciprofloxacin resistance. Prior fluoroquinolone use and a history of resistance to any studied antibiotic were associated with MDR, while pregnancy was protective. CONCLUSIONS: We found elevated resistance to UTI-relevant antimicrobials and novel factors associated with resistance; these data can be incorporated into clinical decision tools to improve organism and drug concordance.


Asunto(s)
Diabetes Mellitus , Gammaproteobacteria , Embarazo , Humanos , Femenino , Persona de Mediana Edad , Masculino , Ciprofloxacina/farmacología , Estudios Transversales , Escherichia coli , Combinación Trimetoprim y Sulfametoxazol/farmacología , Factores de Riesgo , Fluoroquinolonas , Pruebas de Sensibilidad Microbiana , Resistencia a Múltiples Medicamentos , Atención Primaria de Salud
4.
BMC Microbiol ; 24(1): 212, 2024 Jun 14.
Artículo en Inglés | MEDLINE | ID: mdl-38877418

RESUMEN

BACKGROUND: Long-term treatment with trimethoprim-sulfamethoxazole (SXT) can lead to the formation of small-colony variants (SCVs) of Staphylococcus aureus. However, the mechanism behind SCVs formation remains poorly understood. In this study, we explored the phenotype and omics-based characterization of S. aureus SCVs induced by SXT and shed light on the potential causes of SCV formation. METHODS: Stable SCVs were obtained by continuously treating S. aureus isolates using 12/238 µg/ml of SXT, characterized by growth kinetics, antibiotic susceptibility testing, and auxotrophism test. Subsequently, a pair of representative strains (SCV and its parental strain) were selected for genomic, transcriptomic and metabolomic analysis. RESULTS: Three stable S. aureus SCVs were successfully screened and proven to be homologous to their corresponding parental strains. Phenotypic tests showed that all SCVs were non-classical mechanisms associated with impaired utilization of menadione, heme and thymine, and exhibited slower growth and higher antibiotic minimum inhibitory concentrations (MICs), compared to their corresponding parental strains. Genomic data revealed 15 missense mutations in 13 genes in the representative SCV, which were involved in adhesion, intramolecular phosphate transfer on ribose, transport pathways, and phage-encoded proteins. The combination analysis of transcriptome and metabolome identified 35 overlapping pathways possible associated with the phenotype switching of S. aureus. These pathways mainly included changes in metabolism, such as purine metabolism, pyruvate metabolism, amino acid metabolism, and ABC transporters, which could play a crucial role in promoting SCVs development by affecting nucleic acid synthesis and energy metabolism in bacteria. CONCLUSION: This study provides profound insights into the causes of S. aureus SCV formation induced by SXT. The findings may offer valuable clues for developing new strategies to combat S. aureus SCV infections.


Asunto(s)
Antibacterianos , Pruebas de Sensibilidad Microbiana , Staphylococcus aureus , Combinación Trimetoprim y Sulfametoxazol , Staphylococcus aureus/genética , Staphylococcus aureus/efectos de los fármacos , Staphylococcus aureus/crecimiento & desarrollo , Staphylococcus aureus/metabolismo , Combinación Trimetoprim y Sulfametoxazol/farmacología , Antibacterianos/farmacología , Metabolómica , Humanos , Genómica , Fenotipo , Infecciones Estafilocócicas/microbiología , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Transcriptoma , Perfilación de la Expresión Génica , Multiómica
5.
Antimicrob Agents Chemother ; 67(1): e0135222, 2023 01 24.
Artículo en Inglés | MEDLINE | ID: mdl-36507667

RESUMEN

Burkholderia cepacia complex (Bcc) and Burkholderia gladioli are opportunistic human pathogens that are inherently multidrug resistant, limiting treatment options for infections. Here, a novel diazabicyclooctane, ETX0462, was evaluated for activity against Bcc and B. gladioli. Ninety-eight percent of the isolates examined in this study were susceptible. ETX0462 was found to demonstrate in vitro activity superior to that of currently available treatment options (e.g., trimethoprim-sulfamethoxazole and ceftazidime).


Asunto(s)
Infecciones por Burkholderia , Complejo Burkholderia cepacia , Burkholderia , Humanos , Antibacterianos/farmacología , Antibacterianos/uso terapéutico , Ceftazidima/uso terapéutico , Combinación Trimetoprim y Sulfametoxazol/farmacología , Combinación Trimetoprim y Sulfametoxazol/uso terapéutico , Infecciones por Burkholderia/tratamiento farmacológico
6.
J Antimicrob Chemother ; 78(5): 1201-1210, 2023 05 03.
Artículo en Inglés | MEDLINE | ID: mdl-36935372

RESUMEN

OBJECTIVES: Antibacterial antifolate drugs might have a wider role in the management of staphylococcal infection. One factor that could potentially limit their use in this context is pre-existing resistance. Here we explored the prevalence and genetic basis for resistance to these drugs in a large collection (n = 1470) of multidrug-resistant (MDR) Staphylococcus aureus. METHODS: Strains were subjected to susceptibility testing to detect resistance to trimethoprim, sulfamethoxazole, co-trimoxazole and the investigational drug, iclaprim. Whole-genome sequences were interrogated to establish the genetic basis for resistance. RESULTS: According to CLSI breakpoints, 15.2% of the strains were resistant to trimethoprim, 5.2% to sulfamethoxazole and 4.1% to co-trimoxazole. Using the proposed breakpoint for iclaprim, 89% of the trimethoprim-resistant strains exhibited non-susceptibility to this agent. Sulfamethozaxole resistance was exclusively the result of mutation in the drug target (dihydropteroate synthase). Resistance to trimethoprim and iclaprim also resulted from mutation in the target (dihydrofolate reductase; DHFR) but was more commonly associated with horizontal acquisition of genes encoding drug-insensitive DHFR proteins. Among the latter, we identified a novel gene (dfrL) encoding a DHFR with ∼35% identity to native and known resistant DHFRs, which was confirmed via molecular cloning to mediate high-level resistance. CONCLUSIONS: This study provides a detailed picture of the genotypes underlying staphylococcal resistance to antifolate drugs in clinical use and in development. Prevalence estimates suggest that resistance to the diaminopyrimidines (trimethoprim/iclaprim) is not uncommon among MDR S. aureus, and considerably higher than observed for sulfamethoxazole or co-trimoxazole.


Asunto(s)
Antagonistas del Ácido Fólico , Staphylococcus aureus Resistente a Meticilina , Infecciones Estafilocócicas , Humanos , Antagonistas del Ácido Fólico/farmacología , Staphylococcus aureus/genética , Staphylococcus aureus/metabolismo , Staphylococcus aureus Resistente a Meticilina/genética , Combinación Trimetoprim y Sulfametoxazol/farmacología , Prevalencia , Antibacterianos/farmacología , Antibacterianos/metabolismo , Infecciones Estafilocócicas/epidemiología , Infecciones Estafilocócicas/microbiología , Tetrahidrofolato Deshidrogenasa/genética , Pruebas de Sensibilidad Microbiana
7.
Int J Med Microbiol ; 313(2): 151577, 2023 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-36841056

RESUMEN

Staphylococcus aureus (S. aureus) is one of the critical clinical pathogens which can cause multiple diseases ranging from skin infections to fatal sepsis. S. aureus is generally considered to be an extracellular pathogen. However, more and more evidence has shown that S. aureus can survive inside various cells. Folate plays an essential role in multiple life activities, including the conversion of serine and glycine, the remethylation of homocysteine to methionine, and the de novo synthesis of purine /dTMP, et al. More and more studies reported that S. aureus intracellular infection requires the involvement of folate metabolism. This review focused on the mechanisms of folate metabolism and related substances affecting S. aureus infection. Loss of tetrahydrofolic acid (THF)-dependent dTMP directly inhibits the nucleotide synthesis pathway of the S. aureus due to pabA deficiency. Besides, trimethoprim-sulfamethoxazole (TMP/SMX), a potent antibiotic that treats S. aureus infections, interferes in the process of the folate mechanism and leads to the production of thymidine-dependent small-colony variants (TD-SCVs). In addition, S. aureus is resistant to lysostaphin in the presence of serine hydroxymethyltransferase (SHMT). We provide new insights for understanding the molecular pathogenesis of S. aureus infection.


Asunto(s)
Infecciones Estafilocócicas , Staphylococcus aureus , Humanos , Staphylococcus aureus/metabolismo , Timidina Monofosfato/metabolismo , Timidina Monofosfato/uso terapéutico , Infecciones Estafilocócicas/tratamiento farmacológico , Combinación Trimetoprim y Sulfametoxazol/farmacología , Combinación Trimetoprim y Sulfametoxazol/uso terapéutico , Ácido Fólico/metabolismo , Ácido Fólico/uso terapéutico
8.
BMC Microbiol ; 23(1): 200, 2023 07 28.
Artículo en Inglés | MEDLINE | ID: mdl-37507660

RESUMEN

While trimethoprim-sulfamethoxazole (TMP-SMX) is the first-line therapy of Stenotrophomonas maltophilia infections, colistin is one of the therapeutic options in cases of allergy or resistance to TMP-SMX. However, understanding the global status of resistance to colistin amongst S. maltophilia isolates could be helpful for appropriate antibiotic prescription. This study aimed to conduct a systematic review and meta-analysis to examine the prevalence of colistin resistance in clinical S. maltophilia isolates worldwide. According to eligibility criteria, a total of 61 studies were included in the analysis. The pooled prevalence for colistin resistance was 42% (95% CI: 35-49%), ranging from 0.1 to 97%. Subgroups analysis indicated that, the pooled prevalence of colistin resistance was 44% (95% CI: 29-60%) in 15 studies during 2000-2010, and it was estimated to be 41% (95% CI: 33-50%) in 46 articles from 2011 to 2021. It was 46% (95% CI: 35-58%) in the studies that used broth microdilution method, and 39% (95% CI: 30-49%) in the studies with other used methods. The resistance rate in Asian countries was 45% (95% CI: 31-60%), in European countries was 45% (95% CI: 34-56%) and in the countries of North and South America was 33% (95% CI: 20-46%). Our review showed notable resistance to colistin in clinical S. maltophilia isolates. Given the estimated resistance rates, alternative antibiotics could be preferred to treat serious infections due to S. maltophilia.


Asunto(s)
Infecciones por Bacterias Gramnegativas , Stenotrophomonas maltophilia , Humanos , Colistina/farmacología , Combinación Trimetoprim y Sulfametoxazol/farmacología , Combinación Trimetoprim y Sulfametoxazol/uso terapéutico , Prevalencia , Infecciones por Bacterias Gramnegativas/tratamiento farmacológico , Infecciones por Bacterias Gramnegativas/epidemiología , Pruebas de Sensibilidad Microbiana , Antibacterianos/farmacología , Antibacterianos/uso terapéutico
9.
BMC Microbiol ; 23(1): 178, 2023 07 05.
Artículo en Inglés | MEDLINE | ID: mdl-37407940

RESUMEN

BACKGROUND: Non-typeable Haemophilus influenzae (NTHi) has become the major cause of invasive H. influenzae diseases in the post-H. influenzae type b vaccine era. The emergence of multidrug-resistant (MDR) NTHi is a growing public health problem. Herein, we investigated the molecular basis of MDR in NTHi. The isolated NTHi were subjected to antimicrobial susceptibility testing for 12 agents. Whole genome and plasmid sequencing were conducted and analyzed to identify significant genetic variations and plasmid-encoded genes conferred antibiotic resistance. RESULTS: Thirteen (50%) MDR NTHi isolates were obtained; of these, 92.3% were non-susceptible to ampicillin, 30.8% to amoxicillin-clavulanate, 61.5% to cefuroxime, 61.5% to ciprofloxacin/levofloxacin, 92.3% to trimethoprim-sulfamethoxazole, 30.8% to tetracycline, and 7.7% to azithromycin. Eight ampicillin-resistant isolates were ß-lactamase positive; of these, 6 carried blaTEM-1 and 2 carried blaROB-1, whereas 4 were ß-lactamase negative. Genetic variations in mrdA, mepA, and pbpG were correlated with amoxicillin-clavulanate non-susceptibility, whereas variations in ftsI and lpoA conferred cefuroxime resistance. Five variations in gyrA, 2 in gyrB, 3 in parC, 1 in parE, and 1 in the parC-parE intergenic region were associated with levofloxacin/ciprofloxacin non-susceptibility. Among these genes, 8 variations were linked to high-level levofloxacin resistance. Six variations in folA were associated with trimethoprim-sulfamethoxazole resistance. Plasmid-bearing tet(B) and mef(A) genes were responsible for tetracycline and azithromycin resistance in 4 and 1 MDR isolates, respectively. CONCLUSIONS: This study clarified the molecular epidemiology of MDR in NTHi. This can benefit the monitoring of drug resistance trends in NTHi and the adequate medical management of patients with NTHi infection.


Asunto(s)
Infecciones por Haemophilus , Haemophilus influenzae , Humanos , Haemophilus influenzae/genética , Cefuroxima/farmacología , Levofloxacino/farmacología , Combinación Trimetoprim y Sulfametoxazol/farmacología , Azitromicina , Pruebas de Sensibilidad Microbiana , Antibacterianos/farmacología , Antibacterianos/uso terapéutico , Ampicilina , Infecciones por Haemophilus/tratamiento farmacológico , Combinación Amoxicilina-Clavulanato de Potasio , Tetraciclina , Ciprofloxacina , beta-Lactamasas/genética
10.
Microb Pathog ; 177: 106051, 2023 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-36858185

RESUMEN

OBJECTIVE: Carbapenem-resistant Enterobacterales (CRE) infections result in higher treatment costs and mortality rates. Integrons play important roles in emergence and spread of antibiotic resistant genes. To get a better understand on the effects of integron on CRE resistance, distribution of common carbapenemase genes and class 1 integron in clinical CRE isolates were investigated. METHOD: Carbapenemase genes, including blaKPC, blaVIM, blaIMP, blaNDM, blaGES, blaVEB and blaOXA-23, were screened in 161 CRE isolates and subtypes of these genes were confirmed through sequence analysis. Class 1 integron was screened and common promoter and gene cassette arrays were determined by sequencing. The resistant rates to clinical commonly used antibiotics between integron positive and integron negative CRE isolates were compared. RESULTS: Of 161 CRE isolates, the most prevalent carbapenemase gene was blaKPC-2, which was detected in 139 isolates, including 99 Klebsiella pneumoniae. Class 1 integron was detected in 78 isolates. Twenty different gene cassettes, including two carbapenemase genes blaVEB-1 and blaIMP-4, and nine different gene cassette arrays, including blaVEB-1-aadB-arr-2-cmlA5-blaOXA-10-aadA1, aadB-catB8-blaOXA-10-aadA1-dfrA1-aacA4 and blaIMP-4-qacG-aacA4-catB3, were detected. Five types of common promoters were identified. Relative weak promoter PcH1 was the dominant type. Resistant rates of CRE isolates containing class 1 integrons to ceftazidime, amikacin, trimethoprim/sulfamethoxazole and gentamicin were higher than those without class 1 integrons (P < 0.05). CONCLUSION: Class 1 integrons play important roles in the emergence and spread of CRE resistance. To the best of our knowledge, this is the first report of aadB-catB8-blaOXA-10-aadA1-dfrA1-aacA4 and blaIMP-4-qacG-aacA4-catB3 in the same Providencia rettgeri isolate and blaVEB-1-aadB-arr-2-cmlA5-blaOXA-10-aadA1 in P. rettgeri.


Asunto(s)
Carbapenémicos , Integrones , Carbapenémicos/farmacología , Integrones/genética , Antibacterianos/farmacología , Combinación Trimetoprim y Sulfametoxazol/farmacología , Amicacina , Pruebas de Sensibilidad Microbiana
11.
Foodborne Pathog Dis ; 20(7): 252-260, 2023 07.
Artículo en Inglés | MEDLINE | ID: mdl-37384919

RESUMEN

Multidrug resistant (MDR) Escherichia coli threaten the preservation of antimicrobials to treat infections in humans and livestock. Thus, it is important to understand where antimicrobial-resistant E. coli persist and factors that contribute to its their development. Crossbred cattle (n = 249; body weight = 244 kg ±25 kg standard deviation) were blocked by arrival date and assigned metaphylactic antimicrobial treatments of sterile saline control, tulathromycin (TUL), ceftiofur, or florfenicol at random. Trimethoprim-sulfamethoxazole (COTR) and third-generation cephalosporin (CTXR)-resistant E. coli were isolated from fecal samples on days 0, 28, 56, 112, 182, and study END (day 252 for block 1 and day 242 for block 2). Then, susceptibility testing was conducted on all confirmed isolates. MDR was detected in both COTR and CTXR E. coli isolates. In COTR isolates, the number of antimicrobials each isolate was resistant to and the minimum inhibitory concentration (MIC) for amoxicillin-clavulanic acid, ceftriaxone, and gentamicin was greatest on day 28 compared with all other days (p ≤ 0.04). Similarly, chloramphenicol MIC was greater on day 28 than on day 0 (p < 0.01). Overall, sulfisoxazole MIC was less for TUL than all other treatments (p ≤ 0.02), and trimethoprim-sulfamethoxazole MIC was greater for TUL than all other treatments (p ≤ 0.03). Finally, there was no effect of treatment, day, or treatment × day for tetracycline or meropenem MIC (p ≥ 0.07). In CTXR isolates, there was an effect of day for all antimicrobials tested except ampicillin and meropenem (p ≤ 0.06). In conclusion, administering a metaphylactic antimicrobial at feedlot arrival did influence the susceptibility of COTR and CTXR E. coli. However, MDR E. coli are widely distributed, and the MIC for most antimicrobials was not different from the initial value upon completion of the feeding period.


Asunto(s)
Infecciones por Escherichia coli , Escherichia coli , Animales , Bovinos , Antibacterianos/farmacología , Antibacterianos/uso terapéutico , Cefalosporinas/farmacología , Farmacorresistencia Bacteriana , Infecciones por Escherichia coli/tratamiento farmacológico , Infecciones por Escherichia coli/veterinaria , Meropenem/farmacología , Meropenem/uso terapéutico , Pruebas de Sensibilidad Microbiana , Combinación Trimetoprim y Sulfametoxazol/farmacología , Combinación Trimetoprim y Sulfametoxazol/uso terapéutico , Masculino
12.
J Am Pharm Assoc (2003) ; 63(5): 1461-1471, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37414282

RESUMEN

BACKGROUND: The 2011 Infectious Diseases Society of America and European Society of Clinical Microbiology and Infectious Diseases guidelines recommend ciprofloxacin or sulfamethoxazole-trimethoprim (SMX-TMP) as first-line agents to treat uncomplicated acute pyelonephritis (APN). OBJECTIVE: With increasing antimicrobial resistance rates and recent changes in practice patterns, the objective of this systematic review was to describe the effectiveness of cephalosporins for uncomplicated APN in more recently published literature. METHODS: Preferred Reporting Items for Systematic Reviews and Meta-Analyses guidelines were used for reporting. We searched PubMed, Embase, and Scopus for publications between January 2010 and September 2022. Eligible articles detailed patients with uncomplicated APN, treated with first- to fourth-generation cephalosporins, and identified a clinical, microbiological, or health care utilization outcome. Studies with more than 30% of complicated APN patients, non-English-language studies, case reports, case series, pharmacodynamic or pharmacokinetic studies, and in vitro laboratory or animal studies were excluded. Screening, review, and extraction were performed independently by 2 researchers, plus a third for conflict resolution. Critical appraisal of studies was performed using Joanna Briggs Institute checklists. RESULTS: Eight studies met inclusion, including 5 cohort studies (62.5%), 2 randomized controlled trials (25%), and 1 nonrandomized experimental study (12.5%). Cephalosporins most used across the studies included cefazolin, cephalexin, cefuroxime, cefotaxime, cefdinir, cefditoren, and ceftriaxone. Outcomes assessed were diverse, including clinical or microbiological success and time to defervescence or symptom resolution. Cephalosporins displayed effectiveness for the treatment of acute uncomplicated APN regardless of study design or the presence of a comparison group. No trials reported inferiority of clinical treatment outcomes compared with a fluoroquinolone or SMX-TMP. CONCLUSION: Cephalosporins may be viable treatment options for the management of uncomplicated APN.


Asunto(s)
Enfermedades Transmisibles , Pielonefritis , Humanos , Antibacterianos/uso terapéutico , Antibacterianos/farmacología , Cefalosporinas/uso terapéutico , Enfermedades Transmisibles/tratamiento farmacológico , Pielonefritis/tratamiento farmacológico , Pielonefritis/microbiología , Combinación Trimetoprim y Sulfametoxazol/farmacología , Combinación Trimetoprim y Sulfametoxazol/uso terapéutico
13.
Mikrobiyol Bul ; 57(1): 108-118, 2023 Jan.
Artículo en Turco | MEDLINE | ID: mdl-36636850

RESUMEN

Nocardia species are low virulence bacteria found in nature. They can be an infectious agent, especially in patients with risk factors such as underlying immunosuppression, chronic lung disease, and malignancy. They can be easily overlooked because they are not seen frequently and has no pathognomonic symptoms. With this study, it was aimed to draw attention to the importance of microscopic examination of Gram-stained smears in the diagnosis of Nocardia infections in routine microbiology laboratories. Cases in which Nocardia spp. were detected in their clinical samples between November 2014-December 2015 in Hacettepe University Medical Faculty Hospital were included in the study. In the direct microscopic examination of Gram-stained smears of the samples arriving to the laboratory, the incubation periods of the cultures of the samples compatible with Nocardia spp. were extended. Then relevant colonies were identified by conventional microbiological methods and also by matrix-assisted laser desorption ionization-time of flight mass spectrometry (MALDI-TOF MS, bioMerieux, France) automated system. Species-level identification of Nocardia isolates was performed by 16S rRNA gene sequence analysis. To demonstrate the genetic relationship between Nocardia isolates, pulsed-field gel electrophoresis (PFGE) was performed. In vitro susceptibility of the isolates against amoxicillin-clavulanate (AMC), linezolid, moxifloxacin, trimethoprim-sulfamethoxazole (TMP-SXT), amikacin, imipenem, clarithromycin, cefepime, cefotaxime, ceftriaxone, and ciprofloxacin was determined using the gradient strip method (E-test). A total of 19 Nocardia spp. strains were isolated from eight patients. Four cases exhibited repeated growth of Nocardia spp. up to a period of nine months. The most frequently isolated species was N.cyriacigeorgica, which was identified in four cases. Other species isolated from patients were N.asteroides, N.transvalensis, N.farcinicia, and N.asiatica/arthritidis. When the results obtained with DNA sequence analysis and MALDI-TOF MS were compared, 16 (84.2%) of 19 isolates were correctly identified to the genus level and 9 (47.4%) to the species level with MALDI-TOF MS, while three (15.8%) isolates could not be identified, and seven (36.8%) isolates were misidentified. According to the PFGE results, it was determined that the strains isolated from the same patient were genetically identical. All isolates were susceptible to amikacin, cefepime, cefotaxime, ceftriaxone, imipenem, linezolid, and except one isolate to TMP-SXT. Among the study isolates, the most common resistance was against ciprofloxacin (62.5%), followed by clarithromycin (37.5%). N.cyriacigeorgica was determined as the most frequently detected and the most resistant species to antibiotics in the study population. Direct microscopic examination of clinical specimens is one of the most valuable methods for the identification of Nocardia-type bacteria, which is difficult to isolate in microbiology laboratories. With this study, the importance of examining Gram-stained clinical samples was emphasized in the identification of Nocardia species, which can emerge with a wide variety of clinical forms and can be easily overlooked. In addition, antibiotic susceptibility profiles of the isolated bacteria were determinedto contribute to species-specific susceptibility profiles. Accurate identification of Nocardia species will contribute to clinical and epidemiological studies.


Asunto(s)
Nocardiosis , Nocardia , Humanos , Amicacina , Linezolid , Claritromicina , Cefepima , Ceftriaxona , ARN Ribosómico 16S/genética , Nocardiosis/diagnóstico , Nocardiosis/tratamiento farmacológico , Nocardiosis/microbiología , Antibacterianos/farmacología , Antibacterianos/uso terapéutico , Nocardia/genética , Combinación Trimetoprim y Sulfametoxazol/farmacología , Combinación Trimetoprim y Sulfametoxazol/uso terapéutico , Imipenem , Ciprofloxacina , Cefotaxima
14.
Mikrobiyol Bul ; 57(4): 535-552, 2023 Oct.
Artículo en Turco | MEDLINE | ID: mdl-37885384

RESUMEN

Urinary tract infection (UTI) caused by Escherichia coli is a significant health issue in children. Today especially E.coli O25b/ST131, defined as a pandemic clone, is a serious public health problem due to its high virulence and antimicrobial resistance rates. In this study, a total of 200 (100 first and 100 recurrent UTI-causing) E.coli isolates from urine samples sent to the Ankara University School of Medicine Cebeci Training and Research Hospital Central Laboratory between January and September 2021 with the preliminary diagnosis of UTI in pediatric patients aged three to 18 years were analyzed for antimicrobial resistance rates, phylogenetic group distributions, virulence factor frequencies and whether they belong to the O25b/ST131 clone. It is aimed in this study that, the obtained data will shed light on new studies for diagnosis, treatment and prophylaxis options that can be developed for more effective UTI management by contributing to the surveillance studies in our country. Antimicrobial susceptibility of E.coli isolates identified by conventional methods was evaluated by Kirby-Bauer disc diffusion method and extended spectrum beta-lactamase (ESBL) production was evaluated by double disc synergy test. Polymerase chain reaction (PCR) was used for the investigation of phylogenetic grouping, the O25b/ST131 clone, virulence genes and the molecular level classification of the isolates detected as uropathogenic E.coli (UPEC). Pulsed-field gel electrophoresis (PFGE) was performed with the isolates collected at different times from the same patient. The highest antimicrobial resistance rates observed were against ampicillin (n= 100, 50%), cefazolin (n= 99, 49.5%), trimethoprim-sulfamethoxazole (n= 55, 27.5%), amoxicillin-clavulanic acid (n= 43, 21.5%) and cefotaxime (n= 43, 21.5%). In recurrent UTI agents, resistance rates were higher for cefotaxime (n= 29, 29%), trimethoprim-sulfamethoxazole (n= 35, 35%) and cefepime (n= 25, 25%) and in O25b/ST131 isolates (n= 67) the rates were higher for amikacin (n= 3, 4.5%), gentamicin (n= 10, 14.9%) and ciprofloxacin (n= 17, 25.4%) when compared to the first UTI agents and non-O25b/ ST131 isolates (p< 0.05). It was found that 29% (n = 58) of the isolates were multidrug resistant (MDR) and 19% (n = 38) produced ESBL.The rate of recurrent UTI agents was found to be higher among ESBL producing isolates and/or MDR isolates (n= 36, 62% and n= 27, 71%, respectively, p< 0.05). It was found that 45.5% (n= 91) of the isolates were in D, 37.5% (n= 75) in B2, 12.5% (n= 25) in A, and 4.5% (n= 9) in B1 phylogenetic groups and isolates belonging to B2 and D phylogenetic groups had higher antibiotic resistance rates and carried more virulence genes (p< 0.05). Of the isolates, 33.5% (n= 67) were found to belong to the O25b/ST131 clone, no significant difference was found between the O25b/ST131 rates among the first and recurrent UTI agents (p> 0.05). It was determined that the isolates most frequently carry virulence genes for adhesion [fimH 97% (n= 194), papA 57% (n= 114), yfcV 49.5% (n= 99)] and iron uptake systems [fyuA 85.5% (n= 171), chuA 78% (n= 156), iutA 73% (n= 146)]. All virulence factors were detected more frequently in isolates belonging to the O25b/ST131 clone (p< 0.05). Of the isolates, 97% (n= 65) belonging to the O25b/ST131 clone and 27.1% (n= 36) not belonging to this clone were defined as UPEC with molecular analysis (p< 0.0001). Thirty-three isolates belonging to 15 patients were evaluated with PFGE, and it was observed that the latter isolate and the first isolate of eight patients (53%) had the same band profile. Focusing on surveillance, diagnostic testing, treatment algorithms, and preventive measures for E.coli and especially for ST131 clone, which is frequently observed as causative agent in childhood UTIs, will help to manage challenging E.coli infections.


Asunto(s)
Antiinfecciosos , Infecciones por Escherichia coli , Infecciones Urinarias , Humanos , Niño , Escherichia coli/genética , Filogenia , Factores de Virulencia/genética , Combinación Trimetoprim y Sulfametoxazol/farmacología , Infecciones por Escherichia coli/tratamiento farmacológico , Infecciones por Escherichia coli/epidemiología , Infecciones Urinarias/tratamiento farmacológico , Infecciones Urinarias/epidemiología , Infecciones Urinarias/diagnóstico , Cefotaxima/farmacología , beta-Lactamasas/genética , Células Clonales , Antiinfecciosos/farmacología , Antibacterianos/farmacología , Antibacterianos/uso terapéutico
15.
Clin Infect Dis ; 75(1): 131-140, 2022 08 24.
Artículo en Inglés | MEDLINE | ID: mdl-34599811

RESUMEN

BACKGROUND: Pneumococcal serotypes differ in antimicrobial susceptibility. However, patterns and causes of this variation are not comprehensively understood. METHODS: We undertook a systematic review of epidemiologic studies of pneumococci isolated from carriage or invasive disease among children globally from 2000-2019. We evaluated associations of each serotype with nonsusceptibility to penicillin, macrolides, and trimethoprim/sulfamethoxazole. We evaluated differences in the prevalence of nonsusceptibility to major antibiotic classes across serotypes using random-effects meta-regression models and assessed changes in prevalence of nonsusceptibility after implementation of pneumococcal conjugate vaccines (PCVs). We also evaluated associations between biological characteristics of serotypes and their likelihood of nonsusceptibility to each drug. RESULTS: We included data from 129 studies representing 32 187 isolates across 52 countries. Within serotypes, the proportion of nonsusceptible isolates varied geographically and over time, in settings using and those not using PCVs. Factors predicting enhanced fitness of serotypes in colonization as well as enhanced pathogenicity were each associated with higher likelihood of nonsusceptibility to penicillin, macrolides, and trimethoprim/sulfamethoxazole. Increases in prevalence of nonsusceptibility following PCV implementation were evident among non-PCV serotypes, including 6A, 6C, 15A, 15B/C, 19A, and 35B; however, this pattern was not universally evident among non-PCV serotypes. Postvaccination increases in nonsusceptibility for serotypes 6A and 19A were attenuated in settings that implemented PCV13. CONCLUSIONS: In pneumococci, nonsusceptibility to penicillin, macrolides, and trimethoprim/sulfamethoxazole is associated with more frequent opportunities for antibiotic exposure during both prolonged carriage episodes and when serotypes cause disease. These findings suggest multiple pathways leading to resistance selection in pneumococci.


Asunto(s)
Infecciones Neumocócicas , Antibacterianos/farmacología , Antibacterianos/uso terapéutico , Niño , Humanos , Lactante , Macrólidos/farmacología , Nasofaringe , Penicilinas/farmacología , Penicilinas/uso terapéutico , Infecciones Neumocócicas/epidemiología , Infecciones Neumocócicas/prevención & control , Vacunas Neumococicas , Serogrupo , Streptococcus pneumoniae , Combinación Trimetoprim y Sulfametoxazol/farmacología , Combinación Trimetoprim y Sulfametoxazol/uso terapéutico , Vacunas Conjugadas
16.
Emerg Infect Dis ; 28(6): 1110-1116, 2022 06.
Artículo en Inglés | MEDLINE | ID: mdl-35608550

RESUMEN

Annually, Shigella spp. cause ≈188 million cases of diarrheal disease globally, including 500,000 cases in the United States; rates of antimicrobial resistance are increasing. To determine antimicrobial resistance and risk factors in San Diego, California, USA, we retrospectively reviewed cases of diarrheal disease caused by Shigella flexneri and S. sonnei diagnosed during 2017-2020. Of 128 evaluable cases, S. flexneri was slightly more common than S. sonnei; most cases were in persons who were gay or bisexual cisgender men, were living with HIV, were unhoused, or used methamphetamines. Overall, rates of resistance to azithromycin, fluoroquinolones, ampicillin, and trimethoprim/sulfamethoxazole (TMP/SMX) were comparable to the most recent national data reported from the Centers for Disease Control and Prevention; 55% of isolates were resistant to azithromycin, 23% to fluoroquinolones, 70% to ampicillin, and 83% to TMP/SMX. The rates that we found for TMP/SMX were slightly higher than those in national data.


Asunto(s)
Antiinfecciosos , Disentería Bacilar , Shigella , Ampicilina/farmacología , Ampicilina/uso terapéutico , Antibacterianos/farmacología , Antiinfecciosos/farmacología , Azitromicina/farmacología , Azitromicina/uso terapéutico , California/epidemiología , Diarrea , Farmacorresistencia Bacteriana , Disentería Bacilar/epidemiología , Fluoroquinolonas/farmacología , Fluoroquinolonas/uso terapéutico , Humanos , Masculino , Pruebas de Sensibilidad Microbiana , Estudios Retrospectivos , Shigella sonnei , Combinación Trimetoprim y Sulfametoxazol/farmacología , Estados Unidos
17.
Mol Microbiol ; 115(4): 610-622, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33053234

RESUMEN

One of the most commonly prescribed antibiotics against Burkholderia infections is co-trimoxazole, a cocktail of trimethoprim and sulfamethoxazole. Trimethoprim elicits an upregulation of the mal gene cluster, which encodes proteins involved in synthesis of the cytotoxic polyketide malleilactone; trimethoprim does so by increasing expression of the malR gene, which encodes the activator MalR. We report that B. thailandensis grown on trimethoprim exhibited increased virulence against Caenorhabditis elegans. This enhanced virulence correlated with an increase in expression of the mal gene cluster. Notably, inhibition of xanthine dehydrogenase by addition of allopurinol led to similar upregulation of malA and malR, with addition of trimethoprim or allopurinol also resulting in an equivalent intracellular accumulation of xanthine. Xanthine is a ligand for the transcription factor MftR that leads to attenuated DNA binding, and we show using chromatin immunoprecipitation that MftR binds directly to malR. Our gene expression data suggest that malR expression is repressed by both MftR and by a separate transcription factor, which also responds to a metabolite that accumulates on exposure to trimethoprim. Since allopurinol elicits a similar increase in malR/malA expression as trimethoprim, we suggest that impaired purine homeostasis plays a primary role in trimethoprim-mediated induction of malR and in turn malA.


Asunto(s)
Proteínas Bacterianas/fisiología , Burkholderia/efectos de los fármacos , Burkholderia/fisiología , Caenorhabditis elegans/microbiología , Regulación Bacteriana de la Expresión Génica , Purinas/metabolismo , Proteínas Represoras/fisiología , Trimetoprim/farmacología , Animales , Antibacterianos/farmacología , Burkholderia/patogenicidad , Infecciones por Burkholderia/microbiología , Homeostasis , Familia de Multigenes , Sulfametoxazol/farmacología , Factores de Transcripción/metabolismo , Combinación Trimetoprim y Sulfametoxazol/farmacología , Virulencia , Xantina/metabolismo
18.
Antimicrob Agents Chemother ; 66(3): e0216721, 2022 03 15.
Artículo en Inglés | MEDLINE | ID: mdl-35007135

RESUMEN

Trimethoprim/sulfamethoxazole (TMP/SMZ) is considered the treatment of choice for infections caused by Stenotrophomonas maltophilia, but limited pharmacodynamic data are available to support current susceptibility breakpoints or guide optimal dosing. Time-kill studies using a TMP/SMZ concentration of 4/40 µg/mL were conducted to compare 4 S. maltophilia with 4 Escherichia coli isolates having the same MICs (0.25/4.75 to 4/76 µg/mL) in cation-adjusted Mueller-Hinton broth (CAMHB) and ISO-Sensitest broth (ISO broth). With the exception of the resistant isolates (4/76 µg/mL), which resulted in regrowth approaching the growth of the control, TMP/SMZ displayed significantly greater killing for E. coli than for S. maltophilia at each MIC. Against E. coli, the mean changes at 24 h were -4.49, -1.73, -1.59, and +1.83 log10 CFU for isolates with MICs of 0.25/4.75, 1/19, 2/39, and 4/74 µg/mL, respectively. The area under the concentration-time curve for the free, unbound fraction of the drug (fAUC)/MIC ratio required for stasis and 1-log10 and 2-log10 CFU reductions were 40.7, 59.5, and 86.3, respectively. In contrast, TMP/SMZ displayed no stasis or CFU reductions against any S. maltophilia isolate regardless of the MIC, and no pharmacodynamic thresholds were quantifiable. Observations were consistent in both CAMHB and ISO broth. These data add increasing evidence that current TMP/SMZ susceptibility breakpoints against S. maltophilia should be reassessed.


Asunto(s)
Stenotrophomonas maltophilia , Antibacterianos/farmacología , Antibacterianos/uso terapéutico , Cationes , Escherichia coli , Pruebas de Sensibilidad Microbiana , Combinación Trimetoprim y Sulfametoxazol/farmacología
19.
J Antimicrob Chemother ; 77(3): 767-770, 2022 02 23.
Artículo en Inglés | MEDLINE | ID: mdl-35195694

RESUMEN

OBJECTIVES: In some countries, third-generation cephalosporins (3GCs) serve as first-line therapy in children with urinary tract infections (UTIs). However, their use may contribute to the emergence of antibiotic resistance, notably among Gram-negative bacteria (GNB). Integrons are bacterial genetic elements involved in antibiotic resistance in GNB. Their absence is associated with >97% susceptibility to trimethoprim/sulfamethoxazole in adults infected with GNB. The objective of this study was to examine the value of integron detection directly from urine samples as a predictive marker of resistance to trimethoprim/sulfamethoxazole in children with GNB-related UTIs. METHODS: Children admitted to the Limoges University Hospital's paediatric emergency department between February 2018 and March 2019 with a suspicion of UTI were eligible for the study. Only confirmed cases presenting a positive urine culture with unique GNB were retained for further study analyses. Integrons were detected directly from urines using real-time PCR. RESULTS: The data of 72 patients were analysed and integrons were detected in 15 urine samples. The negative predictive value of integron detection for resistance to trimethoprim/sulfamethoxazole was 100% as all of the GNB (all were Enterobacterales) isolated from patients with no integrons detected in their urine samples were susceptible to trimethoprim/sulfamethoxazole. CONCLUSIONS: The detection of integrons in cases of paediatric patients with suspected UTI could help limit 3GC empirical use and empower an empirical first-line strategy better tailored to the needs of each patient.


Asunto(s)
Integrones , Infecciones Urinarias , Adulto , Niño , Farmacorresistencia Microbiana , Humanos , Pruebas de Sensibilidad Microbiana , Combinación Trimetoprim y Sulfametoxazol/farmacología , Combinación Trimetoprim y Sulfametoxazol/uso terapéutico , Infecciones Urinarias/tratamiento farmacológico , Infecciones Urinarias/microbiología
20.
J Antimicrob Chemother ; 77(11): 3187-3193, 2022 10 28.
Artículo en Inglés | MEDLINE | ID: mdl-36101486

RESUMEN

BACKGROUND: Trimethoprim/sulfamethoxazole has historically been the treatment of choice for infection caused by Stenotrophomonas maltophilia. This study sought to define the pharmacodynamic indices and magnitude of exposure required for stasis and 1 log10 cfu reductions. METHODS: Pharmacodynamic studies were conducted using the in vitro chemostat model over 24 h against three trimethoprim/sulfamethoxazole-susceptible S. maltophilia isolates with MICs from 0.25/4.75 to 2/38 mg/L. The primary endpoint was the change in cfu at 24 h relative to baseline. The log ratio of the area under the cfu curve (LR AUcfu) was a secondary endpoint. Trimethoprim and sulfamethoxazole exposures required for stasis and 1 log10 cfu/mL reduction were determined. RESULTS: Trimethoprim/sulfamethoxazole exposures achieved stasis and 1 log10 cfu/mL reductions in 9/16 (56%) and 2/16 (13%) of experiments. Both the fAUC/MIC and fCmax/MIC were identified as equivalent pharmacodynamic drivers, with stasis achieved at an fAUC/MIC of 67.4 and 30.0 for trimethoprim and sulfamethoxazole, respectively. Clinically meaningful exposures required to achieve 1 log10 cfu/mL reductions were not quantifiable. The LR AUcfu analysis supported the lack of overall bacterial burden reduction against S. maltophilia. CONCLUSIONS: In this in vitro chemostat model, trimethoprim/sulfamethoxazole monotherapy, even at higher doses, achieved limited activity against susceptible S. maltophilia.


Asunto(s)
Infecciones por Bacterias Gramnegativas , Stenotrophomonas maltophilia , Humanos , Antibacterianos/farmacología , Antibacterianos/uso terapéutico , Combinación Trimetoprim y Sulfametoxazol/farmacología , Combinación Trimetoprim y Sulfametoxazol/uso terapéutico , Pruebas de Sensibilidad Microbiana , Infecciones por Bacterias Gramnegativas/tratamiento farmacológico , Infecciones por Bacterias Gramnegativas/microbiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA