Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 6.013
Filtrar
Mais filtros

Intervalo de ano de publicação
1.
Cell ; 158(5): 1060-1071, 2014 Aug 28.
Artigo em Inglês | MEDLINE | ID: mdl-25171407

RESUMO

Antibiotic resistance is a key medical concern, with antibiotic use likely being an important cause. However, here we describe an alternative route to clinically relevant antibiotic resistance that occurs solely due to competitive interactions among bacterial cells. We consistently observe that isolates of Methicillin-resistant Staphylococcus aureus diversify spontaneously into two distinct, sequentially arising strains. The first evolved strain outgrows the parent strain via secretion of surfactants and a toxic bacteriocin. The second is resistant to the bacteriocin. Importantly, this second strain is also resistant to intermediate levels of vancomycin. This so-called VISA (vancomycin-intermediate S. aureus) phenotype is seen in many hard-to-treat clinical isolates. This strain diversification also occurs during in vivo infection in a mouse model, which is consistent with the fact that both coevolved phenotypes resemble strains commonly found in clinic. Our study shows how competition between coevolving bacterial strains can generate antibiotic resistance and recapitulate key clinical phenotypes.


Assuntos
Staphylococcus aureus Resistente à Meticilina/classificação , Staphylococcus aureus Resistente à Meticilina/genética , Infecções Estafilocócicas/microbiologia , Sequência de Aminoácidos , Animais , Antibacterianos/farmacologia , Bacteriocinas/genética , Bacteriocinas/metabolismo , Biofilmes/efeitos dos fármacos , Evolução Biológica , Feminino , Staphylococcus aureus Resistente à Meticilina/efeitos dos fármacos , Staphylococcus aureus Resistente à Meticilina/fisiologia , Camundongos Endogâmicos BALB C , Fenômenos Microbiológicos , Dados de Sequência Molecular , Pigmentação , Alinhamento de Sequência , Infecções Estafilocócicas/tratamento farmacológico , Staphylococcus aureus/classificação , Staphylococcus aureus/efeitos dos fármacos , Staphylococcus aureus/genética , Staphylococcus aureus/fisiologia , Vancomicina/farmacologia
2.
Proc Natl Acad Sci U S A ; 121(29): e2315310121, 2024 Jul 16.
Artigo em Inglês | MEDLINE | ID: mdl-38990944

RESUMO

Bacitracin is a macrocyclic peptide antibiotic that is widely used as a topical treatment for infections caused by gram-positive bacteria. Mechanistically, bacitracin targets bacteria by specifically binding to the phospholipid undecaprenyl pyrophosphate (C55PP), which plays a key role in the bacterial lipid II cycle. Recent crystallographic studies have shown that when bound to C55PP, bacitracin adopts a highly ordered amphipathic conformation. In doing so, all hydrophobic side chains align on one face of the bacitracin-C55PP complex, presumably interacting with the bacterial cell membrane. These insights led us to undertake structure-activity investigations into the individual contribution of the nonpolar amino acids found in bacitracin. To achieve this we designed, synthesized, and evaluated a series of bacitracin analogues, a number of which were found to exhibit significantly enhanced antibacterial activity against clinically relevant, drug-resistant pathogens. As for the natural product, these next-generation bacitracins were found to form stable complexes with C55PP. The structure-activity insights thus obtained serve to inform the design of C55PP-targeting antibiotics, a key and underexploited antibacterial strategy.


Assuntos
Antibacterianos , Bacitracina , Testes de Sensibilidade Microbiana , Antibacterianos/farmacologia , Antibacterianos/química , Bacitracina/farmacologia , Bacitracina/química , Relação Estrutura-Atividade , Farmacorresistência Bacteriana/efeitos dos fármacos , Vancomicina/farmacologia , Vancomicina/química , Vancomicina/análogos & derivados , Desenho de Fármacos , Fosfatos de Poli-Isoprenil/metabolismo , Fosfatos de Poli-Isoprenil/química , Fosfatos de Poli-Isoprenil/farmacologia
3.
Nat Chem Biol ; 20(7): 924-933, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-38942968

RESUMO

Keratinicyclins and keratinimicins are recently discovered glycopeptide antibiotics. Keratinimicins show broad-spectrum activity against Gram-positive bacteria, while keratinicyclins form a new chemotype by virtue of an unusual oxazolidinone moiety and exhibit specific antibiosis against Clostridioides difficile. Here we report the mechanism of action of keratinicyclin B (KCB). We find that steric constraints preclude KCB from binding peptidoglycan termini. Instead, KCB inhibits C. difficile growth by binding wall teichoic acids (WTAs) and interfering with cell wall remodeling. A computational model, guided by biochemical studies, provides an image of the interaction of KCB with C. difficile WTAs and shows that the same H-bonding framework used by glycopeptide antibiotics to bind peptidoglycan termini is used by KCB for interacting with WTAs. Analysis of KCB in combination with vancomycin (VAN) shows highly synergistic and specific antimicrobial activity, and that nanomolar combinations of the two drugs are sufficient for complete growth inhibition of C. difficile, while leaving common commensal strains unaffected.


Assuntos
Antibacterianos , Clostridioides difficile , Testes de Sensibilidade Microbiana , Clostridioides difficile/efeitos dos fármacos , Antibacterianos/farmacologia , Antibacterianos/química , Vancomicina/farmacologia , Vancomicina/química , Parede Celular/efeitos dos fármacos , Parede Celular/metabolismo , Ácidos Teicoicos/metabolismo , Peptidoglicano/metabolismo , Peptidoglicano/química , Quimioterapia Combinada , Peptídeos Cíclicos , Lipopeptídeos
4.
Proc Natl Acad Sci U S A ; 120(15): e2208737120, 2023 04 11.
Artigo em Inglês | MEDLINE | ID: mdl-37011186

RESUMO

The alarming rise in superbugs that are resistant to drugs of last resort, including vancomycin-resistant enterococci and staphylococci, has become a significant global health hazard. Here, we report the click chemistry synthesis of an unprecedented class of shapeshifting vancomycin dimers (SVDs) that display potent activity against bacteria that are resistant to the parent drug, including the ESKAPE pathogens, vancomycin-resistant Enterococcus (VRE), methicillin-resistant Staphylococcus aureus (MRSA), as well as vancomycin-resistant S. aureus (VRSA). The shapeshifting modality of the dimers is powered by a triazole-linked bullvalene core, exploiting the dynamic covalent rearrangements of the fluxional carbon cage and creating ligands with the capacity to inhibit bacterial cell wall biosynthesis. The new shapeshifting antibiotics are not disadvantaged by the common mechanism of vancomycin resistance resulting from the alteration of the C-terminal dipeptide with the corresponding d-Ala-d-Lac depsipeptide. Further, evidence suggests that the shapeshifting ligands destabilize the complex formed between the flippase MurJ and lipid II, implying the potential for a new mode of action for polyvalent glycopeptides. The SVDs show little propensity for acquired resistance by enterococci, suggesting that this new class of shapeshifting antibiotic will display durable antimicrobial activity not prone to rapidly acquired clinical resistance.


Assuntos
Staphylococcus aureus Resistente à Meticilina , Enterococos Resistentes à Vancomicina , Vancomicina/farmacologia , Antibacterianos/uso terapêutico , Testes de Sensibilidade Microbiana
5.
Mol Microbiol ; 121(6): 1182-1199, 2024 06.
Artigo em Inglês | MEDLINE | ID: mdl-38690761

RESUMO

The dipeptide D-Ala-D-Ala is an essential component of peptidoglycan and the target of vancomycin. Most Clostridioides difficile strains possess the vanG operon responsible for the synthesis of D-Ala-D-Ser, which can replace D-Ala-D-Ala in peptidoglycan. The C. difficile vanG operon is regulated by a two-component system, VanRS, but is not induced sufficiently by vancomycin to confer resistance to this antibiotic. Surprisingly, in the absence of the VanS histidine kinase (HK), the vanG operon is still induced by vancomycin and also by another antibiotic, ramoplanin, in a VanR-dependent manner. This suggested the cross-regulation of VanR by another HK or kinases that are activated in the presence of certain lipid II-targeting antibiotics. We identified these HKs as CD35990 and CD22880. However, mutations in either or both HKs did not affect the regulation of the vanG operon in wild-type cells suggesting that intact VanS prevents the cross-activation of VanR by non-cognate HKs. Overproduction of VanR in the absence of VanS, CD35990, and CD22880 led to high expression of the vanG operon indicating that VanR can potentially utilize at least one more phosphate donor for its activation. Candidate targets of CD35990- and CD22880-mediated regulation in the presence of vancomycin or ramoplanin were identified by RNA-Seq.


Assuntos
Antibacterianos , Proteínas de Bactérias , Clostridioides difficile , Regulação Bacteriana da Expressão Gênica , Histidina Quinase , Óperon , Resistência a Vancomicina , Vancomicina , Óperon/genética , Clostridioides difficile/genética , Clostridioides difficile/efeitos dos fármacos , Clostridioides difficile/metabolismo , Histidina Quinase/metabolismo , Histidina Quinase/genética , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Vancomicina/farmacologia , Resistência a Vancomicina/genética , Antibacterianos/farmacologia , Depsipeptídeos/farmacologia , Fatores de Transcrição
6.
J Immunol ; 211(7): 1099-1107, 2023 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-37624046

RESUMO

The gut microbiome is an important modulator of the host immune system. In this study, we found that altering the gut microbiome by oral vancomycin increases liver invariant NKT (iNKT) cell function. Enhanced iNKT cytokine production and activation marker expression were observed in vancomycin-treated mice following both Ag-specific and Ag-independent in vivo iNKT stimulations, with a more prominent effect in the liver than in the spleen. Fecal transplantation studies demonstrated that the iNKT functional regulation is mediated by altering the gut microbiome but uncoupled from the modulation of iNKT cell population size. Interestingly, when stimulated in vitro, iNKT cells from vancomycin-treated mice did not show increased activation, suggesting an indirect regulation. iNKT cells expressed high levels of IL-18 receptor, and vancomycin increased the expression of IL-18 in the liver. Blocking IL-18 by neutralizing Ab or using genetically deficient mice attenuated the enhanced iNKT activation. Liver macrophages were identified as a major source of IL-18. General macrophage depletion by clodronate abolished this iNKT activation. Using anti-CSF-1R depletion or LyzCrexCSF-1RLsL-DTR mice identified CSF-1R+ macrophages as a critical modulator of iNKT function. Vancomycin treatment had no effect on iNKT cell function in vivo in IL-18 knockout macrophage reconstituted mice. Together, our results demonstrate that the gut microbiome controls liver iNKT function via regulating CSF-1R+ macrophages to produce IL-18.


Assuntos
Microbioma Gastrointestinal , Camundongos , Animais , Interleucina-18 , Vancomicina/farmacologia , Macrófagos , Fígado , Camundongos Knockout , Receptores Proteína Tirosina Quinases
7.
Nature ; 572(7771): 665-669, 2019 08.
Artigo em Inglês | MEDLINE | ID: mdl-31435014

RESUMO

Intestinal commensal bacteria can inhibit dense colonization of the gut by vancomycin-resistant Enterococcus faecium (VRE), a leading cause of hospital-acquired infections1,2. A four-strained consortium of commensal bacteria that contains Blautia producta BPSCSK can reverse antibiotic-induced susceptibility to VRE infection3. Here we show that BPSCSK reduces growth of VRE by secreting a lantibiotic that is similar to the nisin-A produced by Lactococcus lactis. Although the growth of VRE is inhibited by BPSCSK and L. lactis in vitro, only BPSCSK colonizes the colon and reduces VRE density in vivo. In comparison to nisin-A, the BPSCSK lantibiotic has reduced activity against intestinal commensal bacteria. In patients at high risk of VRE infection, high abundance of the lantibiotic gene is associated with reduced density of E. faecium. In germ-free mice transplanted with patient-derived faeces, resistance to VRE colonization correlates with abundance of the lantibiotic gene. Lantibiotic-producing commensal strains of the gastrointestinal tract reduce colonization by VRE and represent potential probiotic agents to re-establish resistance to VRE.


Assuntos
Bacteriocinas/metabolismo , Bacteriocinas/farmacologia , Enterococcus faecium/efeitos dos fármacos , Lactococcus lactis/metabolismo , Probióticos , Resistência a Vancomicina/efeitos dos fármacos , Enterococos Resistentes à Vancomicina/efeitos dos fármacos , Animais , Antibacterianos/biossíntese , Antibacterianos/isolamento & purificação , Antibacterianos/metabolismo , Antibacterianos/farmacologia , Bacteriocinas/genética , Bacteriocinas/isolamento & purificação , Enterococcus faecium/crescimento & desenvolvimento , Enterococcus faecium/isolamento & purificação , Fezes/microbiologia , Feminino , Trato Gastrointestinal/efeitos dos fármacos , Trato Gastrointestinal/microbiologia , Vida Livre de Germes , Bactérias Gram-Positivas/efeitos dos fármacos , Bactérias Gram-Positivas/crescimento & desenvolvimento , Humanos , Lactococcus lactis/química , Lactococcus lactis/crescimento & desenvolvimento , Lactococcus lactis/fisiologia , Camundongos , Testes de Sensibilidade Microbiana , Microbiota/genética , Nisina/química , Nisina/farmacologia , Simbiose/efeitos dos fármacos , Vancomicina/farmacologia , Enterococos Resistentes à Vancomicina/crescimento & desenvolvimento , Enterococos Resistentes à Vancomicina/isolamento & purificação
8.
Proc Natl Acad Sci U S A ; 119(43): e2210912119, 2022 10 25.
Artigo em Inglês | MEDLINE | ID: mdl-36252016

RESUMO

The alarming rise of multidrug-resistant Gram-positive bacteria has precipitated a healthcare crisis, necessitating the development of new antimicrobial therapies. Here we describe a new class of antibiotics based on a ring-fused 2-pyridone backbone, which are active against vancomycin-resistant enterococci (VRE), a serious threat as classified by the Centers for Disease Control and Prevention, and other multidrug-resistant Gram-positive bacteria. Ring-fused 2-pyridone antibiotics have bacteriostatic activity against actively dividing exponential phase enterococcal cells and bactericidal activity against nondividing stationary phase enterococcal cells. The molecular mechanism of drug-induced killing of stationary phase cells mimics aspects of fratricide observed in enterococcal biofilms, where both are mediated by the Atn autolysin and the GelE protease. In addition, combinations of sublethal concentrations of ring-fused 2-pyridones and standard-of-care antibiotics, such as vancomycin, were found to synergize to kill clinical strains of VRE. Furthermore, a broad range of antibiotic resistant Gram-positive pathogens, including those responsible for the increasing incidence of antibiotic resistant healthcare-associated infections, are susceptible to this new class of 2-pyridone antibiotics. Given the broad antibacterial activities of ring-fused 2-pyridone compounds against Gram-positive (GmP) bacteria we term these compounds GmPcides, which hold promise in combating the rising tide of antibiotic resistant Gram-positive pathogens.


Assuntos
Bactérias Gram-Positivas , Piridonas , Enterococos Resistentes à Vancomicina , Antibacterianos/farmacologia , Farmacorresistência Bacteriana Múltipla , Bactérias Gram-Positivas/efeitos dos fármacos , Testes de Sensibilidade Microbiana , N-Acetil-Muramil-L-Alanina Amidase/farmacologia , Piridonas/farmacologia , Vancomicina/farmacologia , Enterococos Resistentes à Vancomicina/efeitos dos fármacos
9.
Proc Natl Acad Sci U S A ; 119(30): e2118262119, 2022 07 26.
Artigo em Inglês | MEDLINE | ID: mdl-35858453

RESUMO

Human infections with methicillin-resistant Staphylococcus aureus (MRSA) are commonly treated with vancomycin, and strains with decreased susceptibility, designated as vancomycin-intermediate S. aureus (VISA), are associated with treatment failure. Here, we profiled the phenotypic, mutational, and transcriptional landscape of 10 VISA strains adapted by laboratory evolution from one common MRSA ancestor, the USA300 strain JE2. Using functional and independent component analysis, we found that: 1) despite the common genetic background and environmental conditions, the mutational landscape diverged between evolved strains and included mutations previously associated with vancomycin resistance (in vraT, graS, vraFG, walKR, and rpoBCD) as well as novel adaptive mutations (SAUSA300_RS04225, ssaA, pitAR, and sagB); 2) the first wave of mutations affected transcriptional regulators and the second affected genes involved in membrane biosynthesis; 3) expression profiles were predominantly strain-specific except for sceD and lukG, which were the only two genes significantly differentially expressed in all clones; 4) three independent virulence systems (φSa3, SaeR, and T7SS) featured as the most transcriptionally perturbed gene sets across clones; 5) there was a striking variation in oxacillin susceptibility across the evolved lineages (from a 10-fold increase to a 63-fold decrease) that also arose in clinical MRSA isolates exposed to vancomycin and correlated with susceptibility to teichoic acid inhibitors; and 6) constitutive expression of the VraR regulon explained cross-susceptibility, while mutations in walK were associated with cross-resistance. Our results show that adaptation to vancomycin involves a surprising breadth of mutational and transcriptional pathways that affect antibiotic susceptibility and possibly the clinical outcome of infections.


Assuntos
Antibacterianos , Staphylococcus aureus Resistente à Meticilina , Infecções Estafilocócicas , Staphylococcus aureus , Resistência a Vancomicina , Vancomicina , Antibacterianos/química , Antibacterianos/farmacologia , Antibacterianos/uso terapêutico , Evolução Molecular , Humanos , Staphylococcus aureus Resistente à Meticilina/metabolismo , Testes de Sensibilidade Microbiana , Oxacilina/química , Oxacilina/farmacologia , Infecções Estafilocócicas/tratamento farmacológico , Staphylococcus aureus/efeitos dos fármacos , Staphylococcus aureus/genética , Staphylococcus aureus/patogenicidade , Vancomicina/química , Vancomicina/farmacologia , Vancomicina/uso terapêutico , Resistência a Vancomicina/genética , Virulência/genética
10.
Clin Microbiol Rev ; 36(2): e0005922, 2023 06 21.
Artigo em Inglês | MEDLINE | ID: mdl-37067406

RESUMO

Vancomycin-resistant enterococci (VRE) are common causes of bloodstream infections (BSIs) with high morbidity and mortality rates. They are pathogens of global concern with a limited treatment pipeline. Significant challenges exist in the management of VRE BSI, including drug dosing, the emergence of resistance, and the optimal treatment for persistent bacteremia and infective endocarditis. Therapeutic drug monitoring (TDM) for antimicrobial therapy is evolving for VRE-active agents; however, there are significant gaps in the literature for predicting antimicrobial efficacy for VRE BSIs. To date, TDM has the greatest evidence for predicting drug toxicity for the three main VRE-active antimicrobial agents daptomycin, linezolid, and teicoplanin. This article presents an overview of the treatment options for VRE BSIs, the role of antimicrobial dose optimization through TDM in supporting clinical infection management, and challenges and perspectives for the future.


Assuntos
Anti-Infecciosos , Bacteriemia , Infecções por Bactérias Gram-Positivas , Sepse , Enterococos Resistentes à Vancomicina , Humanos , Antibacterianos/farmacologia , Antibacterianos/uso terapêutico , Vancomicina/farmacologia , Vancomicina/uso terapêutico , Linezolida/uso terapêutico , Bacteriemia/tratamento farmacológico , Anti-Infecciosos/uso terapêutico , Infecções por Bactérias Gram-Positivas/tratamento farmacológico
11.
J Infect Dis ; 229(6): 1648-1657, 2024 Jun 14.
Artigo em Inglês | MEDLINE | ID: mdl-38297970

RESUMO

BACKGROUND: Staphylococcus aureus is the most common cause of life-threatening endovascular infections, including infective endocarditis (IE). These infections, especially when caused by methicillin-resistant strains (MRSA), feature limited therapeutic options and high morbidity and mortality rates. METHODS: Herein, we investigated the role of the purine biosynthesis repressor, PurR, in virulence factor expression and vancomycin (VAN) treatment outcomes in experimental IE due to MRSA. RESULTS: The PurR-mediated repression of purine biosynthesis was confirmed by enhanced purF expression and production of an intermediate purine metabolite in purR mutant strain. In addition, enhanced expression of the transcriptional regulators, sigB and sarA, and their key downstream virulence genes (eg, fnbA, and hla) was demonstrated in the purR mutant in vitro and within infected cardiac vegetations. Furthermore, purR deficiency enhanced fnbA/fnbB transcription, translating to increased fibronectin adhesion versus the wild type and purR-complemented strains. Notably, the purR mutant was refractory to significant reduction in target tissues MRSA burden following VAN treatment in the IE model. CONCLUSIONS: These findings suggest that the purine biosynthetic pathway intersects the coordination of virulence factor expression and in vivo persistence during VAN treatment, and may represent an avenue for novel antimicrobial development targeting MRSA.


Assuntos
Antibacterianos , Proteínas de Bactérias , Endocardite Bacteriana , Staphylococcus aureus Resistente à Meticilina , Purinas , Proteínas Repressoras , Infecções Estafilocócicas , Vancomicina , Staphylococcus aureus Resistente à Meticilina/efeitos dos fármacos , Staphylococcus aureus Resistente à Meticilina/genética , Staphylococcus aureus Resistente à Meticilina/metabolismo , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Animais , Infecções Estafilocócicas/microbiologia , Infecções Estafilocócicas/tratamento farmacológico , Purinas/biossíntese , Antibacterianos/farmacologia , Vancomicina/farmacologia , Proteínas Repressoras/genética , Proteínas Repressoras/metabolismo , Endocardite Bacteriana/microbiologia , Endocardite Bacteriana/tratamento farmacológico , Fatores de Virulência/genética , Fatores de Virulência/metabolismo , Camundongos , Regulação Bacteriana da Expressão Gênica , Modelos Animais de Doenças , Testes de Sensibilidade Microbiana , Humanos
12.
Clin Infect Dis ; 79(1): 15-21, 2024 Jul 19.
Artigo em Inglês | MEDLINE | ID: mdl-38382090

RESUMO

BACKGROUND: Epidemiologic studies have shown decreasing vancomycin susceptibility among clinical Clostridioides difficile isolates, but the impact on patient outcomes is unknown. We hypothesized that reduced vancomycin susceptibility would be associated with decreased rates of sustained clinical response (SCR). METHODS: This multicenter cohort study included adults with C. difficile infection (CDI) treated with oral vancomycin between 2016 and 2021. Clostridioides difficile isolates underwent agar dilution vancomycin susceptibility testing, ribotyping, and Sanger sequencing of the vancomycin resistance vanR gene. Reduced susceptibility was defined as vancomycin minimum inhibitory concentration (MIC) >2 µg/mL. The primary outcome was 30-day SCR; secondary outcomes were 14-day initial cure, 30-day recurrence, and 30-day mortality. Exploratory analysis assessed the association between the VanR Thr115Ala polymorphism, susceptibility, and outcomes. RESULTS: A high proportion (34% [102/300]) of C. difficile isolates exhibited reduced vancomycin susceptibility (range, 0.5-16 µg/mL; MIC50/90 = 2/4 µg/mL). Ribotype 027 accounted for the highest proportion (77.4% [41/53]) of isolates with reduced vancomycin susceptibility. Overall, 83% (249) of patients achieved 30-day SCR. Reduced vancomycin susceptibility was associated with lower rates of 30-day SCR (76% [78/102]) than vancomycin-susceptible strains (86% [171/198]; P = .031). A significantly lower rate of 14-day initial cure was also observed among individuals infected with strains with reduced vancomycin susceptibility (89% vs 96%; P = .04). Reduced susceptibility remained an independent predictor of 30-day SCR in multivariable modeling (odds ratio, 0.52 [95% confidence interval, .28-.97]; P = .04). CONCLUSIONS: Reduced vancomycin susceptibility in C. difficile was associated with decreased odds of 30-day SCR and lower 14-day initial cure rates in the studied patient cohort.


Assuntos
Antibacterianos , Clostridioides difficile , Infecções por Clostridium , Testes de Sensibilidade Microbiana , Vancomicina , Humanos , Clostridioides difficile/efeitos dos fármacos , Clostridioides difficile/genética , Vancomicina/uso terapêutico , Vancomicina/farmacologia , Infecções por Clostridium/tratamento farmacológico , Infecções por Clostridium/microbiologia , Masculino , Feminino , Antibacterianos/uso terapêutico , Antibacterianos/farmacologia , Pessoa de Meia-Idade , Idoso , Resistência a Vancomicina/genética , Estudos de Coortes , Resultado do Tratamento , Adulto , Ribotipagem , Idoso de 80 Anos ou mais
13.
Antimicrob Agents Chemother ; 68(1): e0090323, 2024 Jan 10.
Artigo em Inglês | MEDLINE | ID: mdl-38095427

RESUMO

Vancomycin taper and pulse regimens are commonly used to treat recurrent Clostridioides difficile infections, but the mechanism by which these regimens might reduce recurrences is unclear. Here, we used a mouse model to test the hypothesis that pulse dosing of vancomycin after a 10-day treatment course enhances clearance of C. difficile from the intestinal tract. Mice with C. difficile colonization received 10 days of once-daily oral vancomycin followed by 20 days of treatment with saline (controls), daily vancomycin, or pulse dosing of vancomycin every 2 or 3 days. Stool samples were collected to measure the concentration of C. difficile during and after treatment, vancomycin concentrations, and growth of vegetative C. difficile during every 3 days dosing. Pulse dosing of vancomycin was not effective in maintaining suppression of C. difficile (P > 0.05 in comparison to saline controls); growth of vegetative C. difficile occurred between pulse doses when vancomycin decreased to undetectable levels. Daily dosing of vancomycin suppressed C. difficile during treatment, but recurrent colonization occurred after treatment in more than 75% of mice, and by post-treatment day 14, there was no significant difference among the control, pulse dosing, and daily dosing groups (P > 0.05). These findings demonstrate that pulse dosing of vancomycin every 2 or 3 days does not facilitate the clearance of C. difficile spores in mice. Studies are needed to examine the impact of vancomycin taper and pulsed regimens in patients.


Assuntos
Clostridioides difficile , Infecções por Clostridium , Humanos , Animais , Camundongos , Vancomicina/farmacologia , Antibacterianos/farmacologia , Infecções por Clostridium/tratamento farmacológico , Modelos Animais de Doenças
14.
Antimicrob Agents Chemother ; 68(1): e0099223, 2024 Jan 10.
Artigo em Inglês | MEDLINE | ID: mdl-38059634

RESUMO

Vancomycin is the first-line agent to treat pulmonary infections caused by methicillin-resistant Staphylococcus aureus (MRSA) in people with cystic fibrosis (PwCF). However, there is no consensus on vancomycin initial dosing in this population among health institutions, and there is a large variability in initial dosing across the United States. In this study, we characterized the pharmacokinetics (PK) of vancomycin in PwCF using a population PK approach. The clinical PK data to develop the population PK model were obtained from vancomycin therapeutic monitoring data from PwCF undergoing treatment for infections due to MRSA. The population PK model was then used to perform comprehensive Monte Carlo simulations to evaluate the probability of target attainment (PTA) of 12 different initial dosing scenarios. The area under the curve to minimum inhibitory concentration (MIC) ratio ≥400 mg*h/L and <650 mg*h/L were used as efficacy and toxicity targets for PTA analysis. A total of 181 vancomycin plasma concentrations were included in the analysis. A one-compartment model with first-order elimination best described the data. Weight significantly influenced the vancomycin PK (P < 0.05). In the final model, clearance was estimated as 5.52 L/h/70 kg, and the volume of distribution was 31.5 L/70 kg. The PTA analysis showed that at MIC = 1 µg/mL, doses 1,500 q8h and 2,000 q12h showed the highest %PTA in achieving both efficacy and toxicity targets. The PTA results from this study may potentially inform the initial dosing regimens of vancomycin to treat pulmonary infections due to MRSA in PwCF.


Assuntos
Fibrose Cística , Staphylococcus aureus Resistente à Meticilina , Adulto , Humanos , Vancomicina/farmacologia , Antibacterianos/farmacologia , Fibrose Cística/tratamento farmacológico , Testes de Sensibilidade Microbiana , Método de Monte Carlo
15.
Antimicrob Agents Chemother ; 68(3): e0122223, 2024 Mar 06.
Artigo em Inglês | MEDLINE | ID: mdl-38265216

RESUMO

Clostridioides difficile infection (CDI) is a leading cause of hospital-acquired diarrhea, which often stems from disruption of the gut microbiota by broad-spectrum antibiotics. The increasing prevalence of antibiotic-resistant C. difficile strains, combined with disappointing clinical trial results for recent antibiotic candidates, underscores the urgent need for novel CDI antibiotics. To this end, we investigated C. difficile enoyl ACP reductase (CdFabK), a crucial enzyme in de novo fatty acid synthesis, as a drug target for microbiome-sparing antibiotics. To test this concept, we evaluated the efficacy and in vivo spectrum of activity of the phenylimidazole analog 296, which is validated to inhibit intracellular CdFabK. Against major CDI-associated ribotypes 296 had an Minimum inhibitory concentration (MIC90) of 2 µg/mL, which was comparable to vancomycin (1 µg/mL), a standard of care antibiotic. In addition, 296 achieved high colonic concentrations and displayed dosed-dependent efficacy in mice with colitis CDI. Mice that were given 296 retained colonization resistance to C. difficile and had microbiomes that resembled the untreated mice. Conversely, both vancomycin and fidaxomicin induced significant changes to mice microbiomes, in a manner consistent with prior reports. CdFabK, therefore, represents a potential target for microbiome-sparing CDI antibiotics, with phenylimidazoles providing a good chemical starting point for designing such agents.


Assuntos
Clostridioides difficile , Infecções por Clostridium , Animais , Camundongos , Vancomicina/farmacologia , Oxirredutases , Antibacterianos/farmacologia , Antibacterianos/uso terapêutico , Fidaxomicina/farmacologia , Infecções por Clostridium/tratamento farmacológico
16.
Antimicrob Agents Chemother ; 68(3): e0089323, 2024 Mar 06.
Artigo em Inglês | MEDLINE | ID: mdl-38259090

RESUMO

Staphylococcus aureus is an important human pathogen and vancomycin is widely used for the treatment of S. aureus infections. The global regulator agr is known as a well-described virulence regulator. Previous studies have found that agr-dysfunction strains are more likely to develop into vancomycin-resistant strains, but the mechanism for this phenomenon remains unknown. VraSR is a two-component regulatory system related to vancomycin resistance. In this study, we found that the expression levels of vraR were higher in agr-dysfunction clinical strains than in the agr-functional strains. We knocked out agr in a clinical strain, and quantitative reverse transcription PCR and ß-galactosidase activity assays revealed that agr repressed transcription of vraR. After vancomycin exposures, population analysis revealed larger subpopulations displaying reduced susceptibility in agr knockout strain compared with wild-type strain, and this pattern was also observed in agr-dysfunction clinical strains compared with the agr-functional strains. Electrophoretic mobility experiment demonstrated binding of purified AgrA to the promoter region of vraR. In conclusion, our results indicated that the loss of agr function in S. aureus may contribute to the evolution of reduced vancomycin susceptibility through the downregulation of vraSR.


Assuntos
Infecções Estafilocócicas , Staphylococcus aureus , Humanos , Vancomicina/farmacologia , Antibacterianos/farmacologia , Infecções Estafilocócicas/tratamento farmacológico , Infecções Estafilocócicas/epidemiologia , Regiões Promotoras Genéticas/genética , Proteínas de Bactérias/metabolismo
17.
Antimicrob Agents Chemother ; 68(5): e0115923, 2024 May 02.
Artigo em Inglês | MEDLINE | ID: mdl-38506549

RESUMO

Vancomycin heteroresistance is prone to missed detection and poses a risk of clinical treatment failure. We encountered one clinical Enterococcus faecium strain, SRR12, that carried a complete vanM gene cluster but was determined as susceptible to vancomycin using the broth microdilution method. However, distinct subcolonies appeared within the clear zone of inhibition in the E-test assay, one of which, named SRR12-v1, showed high-level resistance to vancomycin. SRR12 was confirmed as heteroresistant to vancomycin using population analysis profiling and displayed "revive" growth curves with a lengthy lag phase of over 13 hours when exposed to 2-32 mg/L vancomycin. The resistant subcolony SRR12-v1 was found to carry an identical vanM gene cluster to that of SRR12 but a significantly increased vanM copy number in the genome. Long-read whole genome sequencing revealed that a one-copy vanM gene cluster was located on a pELF1-like linear plasmid in SRR12. In comparison, tandem amplification of the vanM gene cluster jointed with IS1216E was seated on a linear plasmid in the genome of SRR12-v1. These amplifications of the vanM gene cluster were demonstrated as unstable and would decrease accompanied by fitness reversion after serial passaging for 50 generations under increasing vancomycin pressure or without antibiotic pressure but were relatively stable under constant vancomycin pressure. Further, vanM resistance in resistant variants was verified to be carried by conjugative plasmids with variable sizes using conjugation assays and S1-pulsed field gel electrophoresis blotting, suggesting the instability/flexibility of vanM cluster amplification in the genome and an increased risk of vanM resistance dissemination.


Assuntos
Antibacterianos , Enterococcus faecium , Testes de Sensibilidade Microbiana , Família Multigênica , Plasmídeos , Resistência a Vancomicina , Vancomicina , Enterococcus faecium/genética , Enterococcus faecium/efeitos dos fármacos , Plasmídeos/genética , Vancomicina/farmacologia , Resistência a Vancomicina/genética , Antibacterianos/farmacologia , Proteínas de Bactérias/genética , Humanos , Sequenciamento Completo do Genoma
18.
Antimicrob Agents Chemother ; 68(5): e0143923, 2024 May 02.
Artigo em Inglês | MEDLINE | ID: mdl-38591854

RESUMO

Phage therapy has (re)emerged as a serious possibility for combating multidrug-resistant bacterial infections, including those caused by vancomycin-resistant Enterococcus faecium strains. These opportunistic pathogens belong to a specific clonal complex 17, against which relatively few phages have been screened. We isolated a collection of 21 virulent phages growing on these vancomycin-resistant isolates. Each of these phages harbored a typical narrow plaquing host range, lysing at most 5 strains and covering together 10 strains of our panel of 14 clinical isolates. To enlarge the host spectrum of our phages, the Appelmans protocol was used. We mixed four out of our most complementary phages in a cocktail that we iteratively grew on eight naive strains from our panel, of which six were initially refractory to at least three of the combined phages. Fifteen successive passages permitted to significantly improve the lytic activity of the cocktail, from which phages with extended host ranges within the E. faecium species could be isolated. A single evolved phage able to kill up to 10 of the 14 initial E. faecium strains was obtained, and it barely infected nearby species. All evolved phages had acquired point mutations or a recombination event in the tail fiber genetic region, suggesting these genes might have driven phage evolution by contributing to their extended host spectra.


Assuntos
Bacteriófagos , Enterococcus faecium , Especificidade de Hospedeiro , Enterococos Resistentes à Vancomicina , Enterococcus faecium/efeitos dos fármacos , Bacteriófagos/genética , Enterococos Resistentes à Vancomicina/efeitos dos fármacos , Terapia por Fagos/métodos , Infecções por Bactérias Gram-Positivas/microbiologia , Resistência a Vancomicina , Vancomicina/farmacologia , Humanos , Antibacterianos/farmacologia
19.
Antimicrob Agents Chemother ; 68(7): e0056124, 2024 Jul 09.
Artigo em Inglês | MEDLINE | ID: mdl-38899926

RESUMO

Staphylococcus aureus is a pathogenic bacterium responsible for a broad spectrum of infections, including cutaneous, respiratory, osteoarticular, and systemic infections. It poses a significant clinical challenge due to its ability to develop antibiotic resistance. This resistance limits therapeutic options, increases the risk of severe complications, and underscores the urgent need for new strategies to address this threat, including the investigation of treatments complementary to antibiotics. The evaluation of novel antimicrobial agents often employs animal models, with the zebrafish embryo model being particularly interesting for studying host-pathogen interactions, establishing itself as a crucial tool in this field. For the first time, this study presents a zebrafish embryo model for the in vivo assessment of bacteriophage efficacy against S. aureus infection. A localized infection was induced by microinjecting either methicillin-resistant S. aureus (MRSA) or methicillin-susceptible S. aureus (MSSA). Subsequent treatments involved administering either bacteriophage, vancomycin (the reference antibiotic for MRSA), or a combination of both via the same route to explore potential synergistic effects. Our findings indicate that the bacteriophage was as effective as vancomycin in enhancing survival rates, whether used alone or in combination. Moreover, bacteriophage treatment appears to be even more effective in reducing the bacterial load in S. aureus-infected embryos post-treatment than the antibiotic. Our study validates the use of the zebrafish embryo model and highlights its potential as a valuable tool in assessing bacteriophage efficacy treatments in vivo.


Assuntos
Antibacterianos , Staphylococcus aureus Resistente à Meticilina , Terapia por Fagos , Infecções Estafilocócicas , Vancomicina , Peixe-Zebra , Animais , Peixe-Zebra/microbiologia , Staphylococcus aureus Resistente à Meticilina/efeitos dos fármacos , Staphylococcus aureus Resistente à Meticilina/virologia , Terapia por Fagos/métodos , Vancomicina/farmacologia , Vancomicina/uso terapêutico , Infecções Estafilocócicas/terapia , Infecções Estafilocócicas/microbiologia , Infecções Estafilocócicas/tratamento farmacológico , Antibacterianos/farmacologia , Antibacterianos/uso terapêutico , Modelos Animais de Doenças , Embrião não Mamífero/microbiologia , Testes de Sensibilidade Microbiana
20.
Antimicrob Agents Chemother ; 68(3): e0162123, 2024 Mar 06.
Artigo em Inglês | MEDLINE | ID: mdl-38364016

RESUMO

Antimicrobial resistance is emerging in clinical strains of Clostridioides difficile. Ibezapolstat (IBZ) is a DNA polymerase IIIC inhibitor that has completed phase II clinical trials. IBZ has potent in vitro activity against wild-type, susceptible strains but its effect on C. difficile strains with reduced susceptibility to metronidazole (MTZ), vancomycin (VAN), or fidaxomicin (FDX) has not been tested. The primary objective of this study was to test the antibacterial properties of IBZ against multidrug-resistant C. difficile strains. The in vitro activity, bactericidal, and time-kill activity of IBZ versus comparators were evaluated against 100 clinical strains of which 59 had reduced susceptibility to other C. difficile antibiotics. Morphologic changes against a multidrug resistance strain were visualized by light and scanning electron microscopy. The overall IBZ MIC50/90 values (µg/mL) for evaluated C. difficile strains were 4/8, compared with 2/4 for VAN, 0.5/1 for FDX, and 0.25/4 for MTZ. IBZ MIC50/90 values did not differ based on non-susceptibility to antibiotic class or number of classes to which strains were non-susceptible. IBZ bactericidal activity was similar to the minimum inhibitory concentration (MIC) and maintained in wild-type and non-susceptible strains. Time-kill assays against two laboratory wild-type and two clinical non-susceptible strains demonstrated sustained IBZ activity despite reduced killing by comparator antibiotics for IBZ and VAN non-susceptible strains. Microscopy visualized increased cell lengthening and cellular damage in multidrug-resistant strains exposed to IBZ sub-MIC concentrations. This study demonstrated the potent antibacterial activity of IBZ against a large collection of C. difficile strains including multidrug-resistant strains. This study highlights the therapeutic potential of IBZ against multidrug-resistant strains of C. difficile.


Assuntos
Anti-Infecciosos , Clostridioides difficile , Infecções por Clostridium , Nucleosídeos de Purina , Humanos , Clostridioides , Infecções por Clostridium/tratamento farmacológico , Infecções por Clostridium/microbiologia , Antibacterianos/farmacologia , Antibacterianos/uso terapêutico , Anti-Infecciosos/farmacologia , Vancomicina/farmacologia , Vancomicina/uso terapêutico , Metronidazol/farmacologia , Metronidazol/uso terapêutico , Fidaxomicina/farmacologia , Fidaxomicina/uso terapêutico , Testes de Sensibilidade Microbiana
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA