Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 108
Filtrar
1.
ACS Appl Bio Mater ; 7(3): 1947-1957, 2024 Mar 18.
Artículo en Inglés | MEDLINE | ID: mdl-38394042

RESUMEN

Methicillin-resistant Staphylococcus aureus (MRSA) is associated with high levels of morbidity and is considered a difficult-to-treat infection, often requiring nonstandard treatment regimens and antibiotics. Since over 40% of the emerging antibiotic compounds have insufficient solubility that limits their bioavailability and thus efficacy through oral or intravenous administration, it is crucial that alternative drug delivery products be developed for wound care applications. Existing effective treatments for soft tissue MRSA infections, such as fusidic acid (FA), which is typically administered orally, could also benefit from alternative routes of administration to improve local efficacy and bioavailability while reducing the required therapeutic dose. Herein, we report an antimicrobial poly(oligoethylene glycol methacrylate) (POEGMA)-based composite hydrogel loaded with fusidic acid-encapsulating self-assembled polylactic acid-b-poly(oligo(ethylene glycol) methyl ether methacrylate) (PLA-POEGMA) nanoparticles for the treatment of MRSA-infected skin wounds. The inclusion of the self-assembled nanoparticles (380 nm diameter when loaded with fusidic acid) does not alter the favorable mechanical properties and stability of the hydrogel in the context of its use as a wound dressing, while fusidic acid (FA) can be released from the hydrogel over ∼10 h via a diffusion-controlled mechanism. The antimicrobial studies demonstrate a clear zone of inhibition in vitro and a 1-2 order of magnitude inhibition of bacterial growth in vivo in an MRSA-infected full-thickness excisional murine wound model even at very low antibiotic doses. Our approach thus can both circumvent challenges in the local delivery of hydrophobic antimicrobial compounds and directly deliver antimicrobials into the wound to effectively combat methicillin-resistant infections using a fraction of the drug dose required using other clinically relevant strategies.


Asunto(s)
Antibacterianos , Staphylococcus aureus Resistente a Meticilina , Polietilenglicoles , Animales , Ratones , Antibacterianos/farmacología , Antibacterianos/uso terapéutico , Ácido Fusídico/farmacología , Ácido Fusídico/uso terapéutico , Hidrogeles/química
2.
RSC Chem Biol ; 4(8): 600-612, 2023 Aug 03.
Artículo en Inglés | MEDLINE | ID: mdl-37547457

RESUMEN

Bloodstream infections caused by invasive, non-typhoidal Salmonella (iNTS) are a major global health concern, particularly in Africa where the pathogenic variant of Salmonella Typhimurium sequence type (ST) 313 is dominant. Unlike S. Typhimurium strains that cause gastroenteritis, iNTS strains cause bloodstream infections and are resistant to multiple first-line antibiotics, thus limiting current treatment options. Here, we developed and implemented multiple small molecule screens under physiological, infection-relevant conditions to reveal chemical sensitivities in ST313 and to identify host-directed therapeutics as entry points to drug discovery to combat the clinical burden of iNTS. Screening ST313 iNTS under host-mimicking growth conditions identified 92 compounds with antimicrobial activity despite inherent multidrug resistance. We characterized the antimicrobial activity of the nucleoside analog 3'-azido-3'-deoxythymidine as an exemplary compound from this screen, which depended on bacterial thymidine kinase activity for antimicrobial activity. In a companion macrophage-based screening platform designed to enrich for host-directed therapeutics, we identified three compounds (amodiaquine, berbamine, and indatraline) as actives that required the presence of host cells for antibacterial activity. These three compounds had antimicrobial activity only in the presence of host cells that significantly inhibited intracellular ST313 iNTS replication in macrophages. This work provides evidence that despite high invasiveness and multidrug resistance, ST313 iNTS remains susceptible to unconventional drug discovery approaches.

3.
J Med Chem ; 66(13): 9006-9022, 2023 07 13.
Artículo en Inglés | MEDLINE | ID: mdl-37315221

RESUMEN

The continued efficacy of glycopeptide antibiotics (GPAs) against Gram-positive bacteria is challenged by the emergence and spread of GPA-resistant pathogens, particularly vancomycin-resistant enterococci (VRE). The growing frequency of GPA resistance propels the need for innovative development of more effective antibiotics. Unlike canonical GPAs like vancomycin, Type V GPAs adopt a distinct mode of action by binding peptidoglycan and blocking the activity of autolysins essential for cell division, rendering them a promising class of antibiotics for further development. In this study, the Type V GPA, rimomycin A, was modified to generate 32 new analogues. Compound 17, derived from rimomycin A through N-terminal acylation and C-terminal amidation, exhibited improved anti-VRE activity and solubility. In a VRE-A neutropenic thigh infection mouse model, compound 17 significantly lowered the bacterial load by 3-4 orders of magnitude. This study sets the stage to develop next-generation GPAs in response to growing VRE infections.


Asunto(s)
Infecciones por Bacterias Grampositivas , Enterococos Resistentes a la Vancomicina , Animales , Ratones , Antibacterianos/farmacología , Antibacterianos/uso terapéutico , Antibacterianos/química , Glicopéptidos/farmacología , Glicopéptidos/uso terapéutico , Glicopéptidos/química , Infecciones por Bacterias Grampositivas/tratamiento farmacológico , Infecciones por Bacterias Grampositivas/microbiología , Pruebas de Sensibilidad Microbiana , Biología Sintética , Vancomicina/farmacología , Vancomicina/química
4.
Nat Chem Biol ; 19(11): 1342-1350, 2023 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-37231267

RESUMEN

Acinetobacter baumannii is a nosocomial Gram-negative pathogen that often displays multidrug resistance. Discovering new antibiotics against A. baumannii has proven challenging through conventional screening approaches. Fortunately, machine learning methods allow for the rapid exploration of chemical space, increasing the probability of discovering new antibacterial molecules. Here we screened ~7,500 molecules for those that inhibited the growth of A. baumannii in vitro. We trained a neural network with this growth inhibition dataset and performed in silico predictions for structurally new molecules with activity against A. baumannii. Through this approach, we discovered abaucin, an antibacterial compound with narrow-spectrum activity against A. baumannii. Further investigations revealed that abaucin perturbs lipoprotein trafficking through a mechanism involving LolE. Moreover, abaucin could control an A. baumannii infection in a mouse wound model. This work highlights the utility of machine learning in antibiotic discovery and describes a promising lead with targeted activity against a challenging Gram-negative pathogen.


Asunto(s)
Acinetobacter baumannii , Aprendizaje Profundo , Animales , Ratones , Antibacterianos/farmacología , Farmacorresistencia Bacteriana Múltiple , Pruebas de Sensibilidad Microbiana
5.
EcoSal Plus ; 11(1): eesp00182022, 2023 Dec 12.
Artículo en Inglés | MEDLINE | ID: mdl-37220071

RESUMEN

The microbiome of Crohn's disease (CD) patients is composed of a microbial community that is considered dysbiotic and proinflammatory in nature. The overrepresentation of Enterobacteriaceae species is a common feature of the CD microbiome, and much attention has been given to understanding the pathogenic role this feature plays in disease activity. Over 2 decades ago, a new Escherichia coli subtype called adherent-invasive E. coli (AIEC) was isolated and linked to ileal Crohn's disease. Since the isolation of the first AIEC strain, additional AIEC strains have been isolated from both inflammatory bowel disease (IBD) patients and non-IBD individuals using the original in vitro phenotypic characterization methods. Identification of a definitive molecular marker of the AIEC pathotype has been elusive; however, significant advancements have been made in understanding the genetic, metabolic, and virulence determinants of AIEC infection biology. Here, we review the current knowledge of AIEC pathogenesis to provide additional, objective measures that could be considered in defining AIEC and their pathogenic potential.


Asunto(s)
Enfermedad de Crohn , Infecciones por Escherichia coli , Enfermedades Inflamatorias del Intestino , Humanos , Enfermedad de Crohn/metabolismo , Enfermedad de Crohn/patología , Escherichia coli/genética , Enfermedades Inflamatorias del Intestino/metabolismo , Enfermedades Inflamatorias del Intestino/patología , Íleon/metabolismo , Íleon/patología
7.
Infect Immun ; 91(1): e0050522, 2023 01 24.
Artículo en Inglés | MEDLINE | ID: mdl-36511702

RESUMEN

The NleGs are the largest family of type 3 secreted effectors in attaching and effacing (A/E) pathogens, such as enterohemorrhagic Escherichia coli (EHEC), enteropathogenic E. coli, and Citrobacter rodentium. NleG effectors contain a conserved C-terminal U-box domain acting as a ubiquitin protein ligase and target host proteins via a variable N-terminal portion. The specific roles of these effectors during infection remain uncertain. Here, we demonstrate that the three NleG effectors-NleG1Cr, NleG7Cr, and NleG8Cr-encoded by C. rodentium DBS100 play distinct roles during infection in mice. Using individual nleGCr knockout strains, we show that NleG7Cr contributes to bacterial survival during enteric infection while NleG1Cr promotes the expression of diarrheal symptoms and NleG8Cr contributes to accelerated lethality in susceptible mice. Furthermore, the NleG8Cr effector contains a C-terminal PDZ domain binding motif that enables interaction with the host protein GOPC. Both the PDZ domain binding motif and the ability to engage with host ubiquitination machinery via the intact U-box domain proved to be necessary for NleG8Cr function, contributing to the observed phenotype during infection. We also establish that the PTZ binding motif in the EHEC NleG8 (NleG8Ec) effector, which shares 60% identity with NleG8Cr, is engaged in interactions with human GOPC. The crystal structure of the NleG8Ec C-terminal peptide in complex with the GOPC PDZ domain, determined to 1.85 Å, revealed a conserved interaction mode similar to that observed between GOPC and eukaryotic PDZ domain binding motifs. Despite these common features, nleG8Ec does not complement the ΔnleG8Cr phenotype during infection, revealing functional diversification between these NleG effectors.


Asunto(s)
Infecciones por Enterobacteriaceae , Escherichia coli Enterohemorrágica , Escherichia coli Enteropatógena , Proteínas de Escherichia coli , Humanos , Animales , Ratones , Citrobacter rodentium/genética , Infecciones por Enterobacteriaceae/microbiología , Transporte Biológico , Proteínas de Escherichia coli/genética , Escherichia coli Enteropatógena/genética , Escherichia coli Enterohemorrágica/genética , Proteínas de la Matriz de Golgi/metabolismo , Proteínas Adaptadoras Transductoras de Señales/metabolismo
8.
ACS Infect Dis ; 8(10): 2187-2197, 2022 10 14.
Artículo en Inglés | MEDLINE | ID: mdl-36098580

RESUMEN

Gram-negative bacteria are intrinsically resistant to a plethora of antibiotics that effectively inhibit the growth of Gram-positive bacteria. The intrinsic resistance of Gram-negative bacteria to classes of antibiotics, including rifamycins, aminocoumarins, macrolides, glycopeptides, and oxazolidinones, has largely been attributed to their lack of accumulation within cells due to poor permeability across the outer membrane, susceptibility to efflux pumps, or a combination of these factors. Due to the difficulty in discovering antibiotics that can bypass these barriers, finding targets and compounds that increase the activity of these ineffective antibiotics against Gram-negative bacteria has the potential to expand the antibiotic spectrum. In this study, we investigated the genetic determinants for resistance to rifampicin, novobiocin, erythromycin, vancomycin, and linezolid to determine potential targets of antibiotic-potentiating compounds. We subsequently performed a high-throughput screen of ∼50,000 diverse, synthetic compounds to uncover molecules that potentiate the activity of at least one of the five Gram-positive-targeting antibiotics. This led to the discovery of two membrane active compounds capable of potentiating linezolid and an inhibitor of lipid A biosynthesis capable of potentiating rifampicin and vancomycin. Furthermore, we characterized the ability of known inhibitors of lipid A biosynthesis to potentiate the activity of rifampicin against Gram-negative pathogens.


Asunto(s)
Antibacterianos , Oxazolidinonas , Antibacterianos/química , Antibacterianos/farmacología , Eritromicina/farmacología , Bacterias Gramnegativas/genética , Linezolid , Lípido A , Novobiocina/farmacología , Oxazolidinonas/farmacología , Rifampin/farmacología , Vancomicina/farmacología
9.
Nat Commun ; 12(1): 6664, 2021 11 18.
Artículo en Inglés | MEDLINE | ID: mdl-34795263

RESUMEN

Crohn's disease is an inflammatory disease of the gastrointestinal tract characterized by an aberrant response to microbial and environmental triggers. This includes an altered microbiome dominated by Enterobacteriaceae and in particular adherent-invasive E. coli (AIEC). Clinical evidence implicates periods of psychological stress in Crohn's disease exacerbation, and disturbances in the gut microbiome might contribute to the pathogenic mechanism. Here we show that stress-exposed mice develop ileal dysbiosis, dominated by the expansion of Enterobacteriaceae. In an AIEC colonisation model, stress-induced glucocorticoids promote apoptosis of CD45+CD90+ cells that normally produce IL-22, a cytokine that is essential for the maintenance of ileal mucosal barrier integrity. Blockade of glucocorticoid signaling or administration of recombinant IL-22 restores mucosal immunity, prevents ileal dysbiosis, and blocks AIEC expansion. We conclude that psychological stress impairs IL-22-driven protective immunity in the gut, which creates a favorable niche for the expansion of pathobionts that have been implicated in Crohn's disease. Importantly, this work also shows that immunomodulation can counteract the negative effects of psychological stress on gut immunity and hence disease-associated dysbiosis.


Asunto(s)
Disbiosis/inmunología , Microbioma Gastrointestinal/inmunología , Inmunidad Mucosa/inmunología , Interleucinas/inmunología , Mucosa Intestinal/inmunología , Estrés Psicológico/inmunología , Animales , Adhesión Bacteriana/inmunología , Enfermedad de Crohn/inmunología , Enfermedad de Crohn/microbiología , Disbiosis/microbiología , Enterobacteriaceae/clasificación , Enterobacteriaceae/genética , Enterobacteriaceae/inmunología , Escherichia coli/inmunología , Escherichia coli/fisiología , Infecciones por Escherichia coli/inmunología , Infecciones por Escherichia coli/microbiología , Microbioma Gastrointestinal/genética , Humanos , Íleon/inmunología , Íleon/microbiología , Íleon/patología , Interleucinas/metabolismo , Masculino , Ratones Endogámicos C57BL , Receptores de Glucocorticoides/inmunología , Receptores de Glucocorticoides/metabolismo , Antígenos Thy-1/inmunología , Antígenos Thy-1/metabolismo , Interleucina-22
10.
Am J Physiol Endocrinol Metab ; 321(3): E338-E350, 2021 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-34280051

RESUMEN

Obesity is associated with metabolic, immunological, and infectious disease comorbidities, including an increased risk of enteric infection and inflammatory bowel disease such as Crohn's disease (CD). Expansion of intestinal pathobionts such as adherent-invasive Escherichia coli (AIEC) is a common dysbiotic feature of CD, which is amplified by prior use of oral antibiotics. Although high-fat, high-sugar diets are associated with dysbiotic expansion of E. coli, it is unknown if the content of fat or another dietary component in obesogenic diets is sufficient to promote AIEC expansion. Here, we found that administration of an antibiotic combined with feeding mice an obesogenic low-fiber, high-sucrose, high-fat diet (HFD) that is typically used in rodent-obesity studies promoted AIEC intestinal expansion. Even a short-term (i.e., 1 day) pulse of HFD feeding before infection was sufficient to promote AIEC expansion, indicating that the magnitude of obesity was not the main driver of AIEC expansion. Controlled-diet experiments demonstrated that neither dietary fat nor sugar were the key determinants of AIEC colonization, but that lowering dietary fiber from approximately 13% to 5%-6% was sufficient to promote the intestinal expansion of AIEC when combined with antibiotics in mice. When combined with antibiotics, lowering fiber promoted AIEC intestinal expansion to a similar extent as widely used HFDs in mice. However, lowering dietary fiber was sufficient to promote AIEC intestinal expansion without affecting body mass. Our results show that low dietary fiber combined with oral antibiotics are environmental factors that promote the expansion of Crohn's disease-associated pathobionts in the gut.NEW & NOTEWORTHY It is commonly thought that obesity or a high-fat diet alters pathogenic bacteria and promotes inflammatory gut diseases. We found that lower dietary fiber is a key factor that expands a gut pathobiont linked to Crohn's disease, independent of obesity status in mice.


Asunto(s)
Enfermedad de Crohn/microbiología , Fibras de la Dieta/administración & dosificación , Intestinos/microbiología , Obesidad/microbiología , Animales , Escherichia coli/fisiología , Masculino , Ratones , Ratones Endogámicos C57BL
11.
PLoS Pathog ; 17(5): e1009532, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33984072

RESUMEN

Bacteria inhabit diverse environmental niches and consequently must modulate their metabolism to adapt to stress. The nucleotide second messengers guanosine tetraphosphate (ppGpp) and guanosine pentaphosphate (pppGpp) (collectively referred to as (p)ppGpp) are essential for survival during nutrient starvation. (p)ppGpp is synthesized by the RelA-SpoT homologue (RSH) protein family and coordinates the control of cellular metabolism through its combined effect on over 50 proteins. While the role of (p)ppGpp has largely been associated with nutrient limitation, recent studies have shown that (p)ppGpp and related nucleotides have a previously underappreciated effect on different aspects of bacterial physiology, such as maintaining cellular homeostasis and regulating bacterial interactions with a host, other bacteria, or phages. (p)ppGpp produced by pathogenic bacteria facilitates the evasion of host defenses such as reactive nitrogen intermediates, acidic pH, and the complement system. Additionally, (p)ppGpp and pyrophosphorylated derivatives of canonical adenosine nucleotides called (p)ppApp are emerging as effectors of bacterial toxin proteins. Here, we review the RSH protein family with a focus on its unconventional roles during host infection and bacterial competition.


Asunto(s)
Bacterias/metabolismo , Infecciones Bacterianas/microbiología , Fenómenos Fisiológicos Bacterianos , Proteínas Bacterianas/metabolismo , Difosfatos/metabolismo , Nucleótidos/metabolismo , Estrés Fisiológico , Animales , Infecciones Bacterianas/metabolismo , Infecciones Bacterianas/patología , Proteínas Bacterianas/genética , Regulación Bacteriana de la Expresión Génica , Humanos , Fosforilación
12.
Nat Commun ; 12(1): 2032, 2021 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-33795670

RESUMEN

Adherent-invasive Escherichia coli (AIEC) are pathogenic bacteria frequently isolated from patients who have Crohn's disease (CD). Despite the phenotypic differences between AIEC and commensal E. coli, comparative genomic approaches have been unable to differentiate these two groups, making the identification of key virulence factors a challenge. Here, we conduct a high-resolution, in vivo genetic screen to map AIEC genes required for intestinal colonization of mice. In addition, we use in vivo RNA-sequencing to define the host-associated AIEC transcriptome. We identify diverse metabolic pathways required for efficient gut colonization by AIEC and show that a type IV secretion system (T4SS) is required to form biofilms on the surface of epithelial cells, thereby promoting AIEC persistence in the gut. E. coli isolated from CD patients are enriched for a T4SS, suggesting a possible connection to disease activity. Our findings establish the T4SS as a principal AIEC colonization factor and highlight the use of genome-wide screens in decoding the infection biology of CD-associated bacteria that otherwise lack a defined genetic signature.


Asunto(s)
Enfermedad de Crohn/patología , Escherichia coli/genética , Perfilación de la Expresión Génica/métodos , Ensayos Analíticos de Alto Rendimiento/métodos , Sistemas de Secreción Tipo IV/genética , Animales , Adhesión Bacteriana/genética , Biopelículas , Células CACO-2 , Enfermedad de Crohn/microbiología , Células Epiteliales/metabolismo , Células Epiteliales/microbiología , Escherichia coli/clasificación , Escherichia coli/fisiología , Infecciones por Escherichia coli/microbiología , Infecciones por Escherichia coli/patología , Femenino , Humanos , Mucosa Intestinal/metabolismo , Mucosa Intestinal/microbiología , Ratones Endogámicos C57BL , Factores de Virulencia/genética
14.
Infect Immun ; 89(2)2021 01 19.
Artículo en Inglés | MEDLINE | ID: mdl-33139383

RESUMEN

The stringent response is an essential mechanism of metabolic reprogramming during environmental stress that is mediated by the nucleotide alarmones guanosine tetraphosphate and pentaphosphate [(p)ppGpp]. In addition to physiological adaptations, (p)ppGpp also regulates virulence programs in pathogenic bacteria, including Salmonella enterica serovar Typhimurium. S Typhimurium is a common cause of acute gastroenteritis, but it may also spread to systemic tissues, resulting in severe clinical outcomes. During infection, S Typhimurium encounters a broad repertoire of immune defenses that it must evade for successful host infection. Here, we examined the role of the stringent response in S Typhimurium resistance to complement-mediated killing and found that the (p)ppGpp synthetase-hydrolase, SpoT, is required for bacterial survival in human serum. We identified the nucleotide hydrolase, PpnN, as a target of the stringent response that is required to promote bacterial fitness in serum. Using chromatography and mass spectrometry, we show that PpnN hydrolyzes purine and pyrimidine monophosphates to generate free nucleobases and ribose 5'-phosphate, and that this metabolic activity is required for conferring resistance to complement killing. In addition to PpnN, we show that (p)ppGpp is required for the biosynthesis of the very long and long O-antigen in the outer membrane, known to be important for complement resistance. Our results provide new insights into the role of the stringent response in mediating evasion of the innate immune system by pathogenic bacteria.


Asunto(s)
Resistencia a la Enfermedad/inmunología , Ligasas/inmunología , N-Glicosil Hidrolasas/inmunología , Salmonella typhimurium/inmunología , Salmonella typhimurium/patogenicidad , Virulencia/genética , Virulencia/inmunología , Regulación Bacteriana de la Expresión Génica , Variación Genética , Humanos , Inmunidad Innata , Ligasas/genética , N-Glicosil Hidrolasas/genética , Serogrupo
15.
STAR Protoc ; 1(2): 100057, 2020 09 18.
Artículo en Inglés | MEDLINE | ID: mdl-33111100

RESUMEN

Anti-virulence therapies are under active investigation as antibiotic alternatives; however, their identification from large-scale chemical libraries poses a unique challenge. The dispensability of virulence factors for growth in vitro precludes conventional, optical density-based screening methods. Here, we provide a protocol for high-throughput screening with a cell-based, promoter reporter platform. We describe the use of this method for the identification of anti-SPI-2 inhibitors specific to Salmonella Typhimurium, which may be modified to investigate other virulence factors. For complete details on the use and execution of this protocol, please refer to Tsai et al. (2020).


Asunto(s)
Proteínas Bacterianas , Técnicas Bacteriológicas/métodos , Ensayos Analíticos de Alto Rendimiento/métodos , Proteínas de la Membrana , Salmonella typhimurium/genética , Salmonella typhimurium/patogenicidad , Antibacterianos/farmacología , Proteínas Bacterianas/antagonistas & inhibidores , Proteínas Bacterianas/genética , Proteínas de la Membrana/antagonistas & inhibidores , Proteínas de la Membrana/genética , Salmonella typhimurium/efectos de los fármacos
16.
Cell Rep ; 32(3): 107927, 2020 07 21.
Artículo en Inglés | MEDLINE | ID: mdl-32698013

RESUMEN

Antibiotics halt the growth of bacteria by targeting core, essential physiology that is required for life on standard microbiological media. Many more biochemical and virulence processes, however, are required for bacteria to cause infection in a host. Indeed, chemical inhibitors of the latter processes are overlooked using conventional antibiotic drug discovery approaches. Here, we use human blood serum as an alternative growth medium to explore new targets and compounds. High-throughput screening of genetic and chemical libraries identified compounds targeting biological activities required by Klebsiella pneumoniae to grow in serum, such as nucleobase biosynthesis and iron acquisition, and showed that serum can chemically transform compounds to reveal cryptic antibacterial activity. One of these compounds, ruthenium red, was effective in a rat bloodstream infection model. Our data demonstrate that human serum is an effective tool to find new chemical matter to address the current antibiotic resistance crisis.


Asunto(s)
Antibacterianos/análisis , Antibacterianos/farmacología , Pruebas Genéticas , Klebsiella pneumoniae/genética , Suero/microbiología , Bibliotecas de Moléculas Pequeñas/análisis , Animales , Antibacterianos/química , Daño del ADN , Modelos Animales de Enfermedad , Aprobación de Drogas , Femenino , Humanos , Hidrólisis , Indoles/farmacología , Hierro/metabolismo , Infecciones por Klebsiella/sangre , Infecciones por Klebsiella/microbiología , Klebsiella pneumoniae/efectos de los fármacos , Klebsiella pneumoniae/crecimiento & desarrollo , Fenotipo , Ratas Wistar , Rojo de Rutenio/farmacología , Bibliotecas de Moléculas Pequeñas/química , Triptófano/biosíntesis , Uracilo/biosíntesis
17.
Cell Chem Biol ; 27(7): 793-805.e7, 2020 07 16.
Artículo en Inglés | MEDLINE | ID: mdl-32413287

RESUMEN

Salmonella serovars are leading causes of gastrointestinal disease and have become increasingly resistant to fluoroquinolone and cephalosporin antibiotics. Overcoming this healthcare crisis requires new approaches in antibiotic discovery and the identification of unique bacterial targets. In this work, we describe a chemical genomics approach to identify inhibitors of Salmonella virulence. From a cell-based, promoter reporter screen of ∼50,000 small molecules, we identified dephostatin as a non-antibiotic compound that inhibits intracellular virulence factors and polymyxin resistance genes. Dephostatin disrupts signaling through both the SsrA-SsrB and PmrB-PmrA two-component regulatory systems and restores sensitivity to the last-resort antibiotic, colistin. Cell-based experiments and mouse models of infection demonstrate that dephostatin attenuates Salmonella virulence in vitro and in vivo, suggesting that perturbing regulatory networks is a promising strategy for the development of anti-infectives.


Asunto(s)
Antibacterianos/farmacología , Salmonella/patogenicidad , Bibliotecas de Moléculas Pequeñas/farmacología , Virulencia/efectos de los fármacos , Animales , Antibacterianos/química , Antibacterianos/uso terapéutico , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Colistina/farmacología , Colistina/uso terapéutico , Sinergismo Farmacológico , Femenino , Histidina Quinasa/genética , Histidina Quinasa/metabolismo , Hidroquinonas/farmacología , Hidroquinonas/uso terapéutico , Ratones , Ratones Endogámicos C57BL , Polimixina B/farmacología , Salmonella/metabolismo , Salmonelosis Animal/tratamiento farmacológico , Salmonelosis Animal/mortalidad , Bibliotecas de Moléculas Pequeñas/química , Bibliotecas de Moléculas Pequeñas/uso terapéutico , Tasa de Supervivencia , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Virulencia/genética
18.
Nature ; 578(7796): 582-587, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-32051588

RESUMEN

Addressing the ongoing antibiotic crisis requires the discovery of compounds with novel mechanisms of action that are capable of treating drug-resistant infections1. Many antibiotics are sourced from specialized metabolites produced by bacteria, particularly those of the Actinomycetes family2. Although actinomycete extracts have traditionally been screened using activity-based platforms, this approach has become unfavourable owing to the frequent rediscovery of known compounds. Genome sequencing of actinomycetes reveals an untapped reservoir of biosynthetic gene clusters, but prioritization is required to predict which gene clusters may yield promising new chemical matter2. Here we make use of the phylogeny of biosynthetic genes along with the lack of known resistance determinants to predict divergent members of the glycopeptide family of antibiotics that are likely to possess new biological activities. Using these predictions, we uncovered two members of a new functional class of glycopeptide antibiotics-the known glycopeptide antibiotic complestatin and a newly discovered compound we call corbomycin-that have a novel mode of action. We show that by binding to peptidoglycan, complestatin and corbomycin block the action of autolysins-essential peptidoglycan hydrolases that are required for remodelling of the cell wall during growth. Corbomycin and complestatin have low levels of resistance development and are effective in reducing bacterial burden in a mouse model of skin MRSA infection.


Asunto(s)
Antibacterianos , Descubrimiento de Drogas , Péptidos Cíclicos , Peptidoglicano/efectos de los fármacos , Peptidoglicano/metabolismo , Actinobacteria/química , Actinobacteria/genética , Actinobacteria/metabolismo , Animales , Antibacterianos/química , Antibacterianos/metabolismo , Antibacterianos/farmacología , Vías Biosintéticas/genética , Pared Celular/metabolismo , Clorofenoles/química , Clorofenoles/metabolismo , Clorofenoles/farmacología , Modelos Animales de Enfermedad , Farmacorresistencia Microbiana/efectos de los fármacos , Farmacorresistencia Microbiana/genética , Femenino , Staphylococcus aureus Resistente a Meticilina/efectos de los fármacos , Ratones , Pruebas de Sensibilidad Microbiana , Familia de Multigenes , N-Acetil Muramoil-L-Alanina Amidasa/antagonistas & inhibidores , Péptidos Cíclicos/química , Péptidos Cíclicos/metabolismo , Péptidos Cíclicos/farmacología , Filogenia , Piel/microbiología , Infecciones Estafilocócicas/microbiología
19.
Nat Microbiol ; 5(1): 93-101, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31659298

RESUMEN

To revitalize the antibiotic pipeline, it is critical to identify and validate new antimicrobial targets1. In Mycobacteria tuberculosis and Francisella tularensis, biotin biosynthesis is a key fitness determinant during infection2-5, making it a high-priority target. However, biotin biosynthesis has been overlooked for priority pathogens such as Acinetobacter baumannii, Klebsiella pneumoniae and Pseudomonas aeruginosa. This can be attributed to the lack of attenuation observed for biotin biosynthesis genes during transposon mutagenesis studies in mouse infection models6-9. Previous studies did not consider the 40-fold higher concentration of biotin in mouse plasma compared to human plasma. Here, we leveraged the unique affinity of streptavidin to develop a mouse infection model with human levels of biotin. Our model suggests that biotin biosynthesis is essential during infection with A. baumannii, K. pneumoniae and P. aeruginosa. Encouragingly, we establish the capacity of our model to uncover in vivo activity for the biotin biosynthesis inhibitor MAC13772. Our model addresses the disconnect in biotin levels between humans and mice, and explains the failure of potent biotin biosynthesis inhibitors in standard mouse infection models.


Asunto(s)
Antibacterianos/farmacología , Bacterias/efectos de los fármacos , Infecciones Bacterianas/tratamiento farmacológico , Biotina/biosíntesis , Farmacorresistencia Bacteriana/efectos de los fármacos , Animales , Antibacterianos/química , Antibacterianos/uso terapéutico , Bacterias/genética , Bacterias/crecimiento & desarrollo , Infecciones Bacterianas/sangre , Proteínas Bacterianas/antagonistas & inhibidores , Proteínas Bacterianas/química , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Biotina/sangre , Modelos Animales de Enfermedad , Farmacorresistencia Bacteriana/genética , Humanos , Ratones , Pruebas de Sensibilidad Microbiana , Modelos Moleculares , Mutación , Especificidad de la Especie , Estreptavidina/administración & dosificación , Transaminasas/antagonistas & inhibidores , Transaminasas/química , Transaminasas/genética , Transaminasas/metabolismo
20.
Microbiol Resour Announc ; 8(24)2019 Jun 13.
Artículo en Inglés | MEDLINE | ID: mdl-31196922

RESUMEN

Citrobacter rodentium strain DBS100 causes an infection of the intestines in mice. It provides an important model for human gastrointestinal pathogens, such as enteropathogenic and enterohemorrhagic Escherichia coli, which cause life-threatening infections. To identify the genetic determinants that are common across the enteropathogenic bacteria, we sequenced the DBS100 genome.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...