Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 52
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
Nat Commun ; 15(1): 4036, 2024 May 13.
Artigo em Inglês | MEDLINE | ID: mdl-38740750

RESUMO

Microbial Ni2+ homeostasis underpins the virulence of several clinical pathogens. Ni2+ is an essential cofactor in urease and [NiFe]-hydrogenases involved in colonization and persistence. Many microbes produce metallophores to sequester metals necessary for their metabolism and starve competing neighboring organisms. The fungal metallophore aspergillomarasmine A (AMA) shows narrow specificity for Zn2+, Ni2+, and Co2+. Here, we show that this specificity allows AMA to block the uptake of Ni2+ and attenuate bacterial Ni-dependent enzymes, offering a potential strategy for reducing virulence. Bacterial exposure to AMA perturbs H2 metabolism, ureolysis, struvite crystallization, and biofilm formation and shows efficacy in a Galleria mellonella animal infection model. The inhibition of Ni-dependent enzymes was aided by Zn2+, which complexes with AMA and competes with the native nickelophore for the uptake of Ni2+. Biochemical analyses demonstrated high-affinity binding of AMA-metal complexes to NikA, the periplasmic substrate-binding protein of the Ni2+ uptake system. Structural examination of NikA in complex with Ni-AMA revealed that the coordination geometry of Ni-AMA mimics the native ligand, Ni-(L-His)2, providing a structural basis for binding AMA-metal complexes. Structure-activity relationship studies of AMA identified regions of the molecule that improve NikA affinity and offer potential routes for further developing this compound as an anti-virulence agent.


Assuntos
Proteínas de Bactérias , Níquel , Níquel/metabolismo , Níquel/química , Animais , Virulência/efeitos dos fármacos , Proteínas de Bactérias/metabolismo , Biofilmes/efeitos dos fármacos , Zinco/metabolismo , Zinco/química , Mariposas/microbiologia , Urease/metabolismo , Urease/antagonistas & inibidores , Transporte Biológico
2.
Nat Chem Biol ; 20(2): 234-242, 2024 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-37973888

RESUMO

The efficacy of aminoglycoside antibiotics is waning due to the acquisition of diverse resistance mechanisms by bacteria. Among the most prevalent are aminoglycoside acetyltransferases (AACs) that inactivate the antibiotics through acetyl coenzyme A-mediated modification. Most AACs are members of the GCN5 superfamily of acyltransferases which lack conserved active site residues that participate in catalysis. ApmA is the first reported AAC belonging to the left-handed ß-helix superfamily. These enzymes are characterized by an essential active site histidine that acts as an active site base. Here we show that ApmA confers broad-spectrum aminoglycoside resistance with a molecular mechanism that diverges from other detoxifying left-handed ß-helix superfamily enzymes and canonical GCN5 AACs. We find that the active site histidine plays different functions depending on the acetyl-accepting aminoglycoside substrate. This flexibility in the mechanism of a single enzyme underscores the plasticity of antibiotic resistance elements to co-opt protein catalysts in the evolution of drug detoxification.


Assuntos
Aminoglicosídeos , Histidina , Aminoglicosídeos/farmacologia , Antibacterianos/farmacologia , Antibacterianos/química , Acetiltransferases/genética , Acetiltransferases/metabolismo , Bactérias/metabolismo
3.
J Ind Microbiol Biotechnol ; 50(1)2023 Feb 17.
Artigo em Inglês | MEDLINE | ID: mdl-38052426

RESUMO

Microbial natural products are specialized metabolites that are sources of many bioactive compounds including antibiotics, antifungals, antiparasitics, anticancer agents, and probes of biology. The assembly of libraries of producers of natural products has traditionally been the province of the pharmaceutical industry. This sector has gathered significant historical collections of bacteria and fungi to identify new drug leads with outstanding outcomes-upwards of 60% of drug scaffolds originate from such libraries. Despite this success, the repeated rediscovery of known compounds and the resultant diminishing chemical novelty contributed to a pivot from this source of bioactive compounds toward more tractable synthetic compounds in the drug industry. The advent of advanced mass spectrometry tools, along with rapid whole genome sequencing and in silico identification of biosynthetic gene clusters that encode the machinery necessary for the synthesis of specialized metabolites, offers the opportunity to revisit microbial natural product libraries with renewed vigor. Assembling a suitable library of microbes and extracts for screening requires the investment of resources and the development of methods that have customarily been the proprietary purview of large pharmaceutical companies. Here, we report a perspective on our efforts to assemble a library of natural product-producing microbes and the establishment of methods to extract and fractionate bioactive compounds using resources available to most academic labs. We validate the library and approach through a series of screens for antimicrobial and cytotoxic agents. This work serves as a blueprint for establishing libraries of microbial natural product producers and bioactive extract fractions suitable for screens of bioactive compounds. ONE-SENTENCE SUMMARY: Natural products are key to discovery of novel antimicrobial agents: Here, we describe our experience and lessons learned in constructing a microbial natural product and pre-fractionated extract library.


Assuntos
Antineoplásicos , Produtos Biológicos , Produtos Biológicos/química , Biblioteca Gênica , Fungos/genética , Indústria Farmacêutica
4.
J Med Chem ; 66(13): 9006-9022, 2023 07 13.
Artigo em Inglês | MEDLINE | ID: mdl-37315221

RESUMO

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.


Assuntos
Infecções por Bactérias Gram-Positivas , Enterococos Resistentes à Vancomicina , Animais , Camundongos , Antibacterianos/farmacologia , Antibacterianos/uso terapêutico , Antibacterianos/química , Glicopeptídeos/farmacologia , Glicopeptídeos/uso terapêutico , Glicopeptídeos/química , Infecções por Bactérias Gram-Positivas/tratamento farmacológico , Infecções por Bactérias Gram-Positivas/microbiologia , Testes de Sensibilidade Microbiana , Biologia Sintética , Vancomicina/farmacologia , Vancomicina/química
5.
Proc Natl Acad Sci U S A ; 120(16): e2221253120, 2023 04 18.
Artigo em Inglês | MEDLINE | ID: mdl-37043535

RESUMO

The outer membrane of gram-negative bacteria prevents many antibiotics from reaching intracellular targets. However, some antimicrobials can take advantage of iron import transporters to cross this barrier. We showed previously that the thiopeptide antibiotic thiocillin exploits the nocardamine xenosiderophore transporter, FoxA, of the opportunistic pathogen Pseudomonas aeruginosa for uptake. Here, we show that FoxA also transports the xenosiderophore bisucaberin and describe at 2.5 Å resolution the crystal structure of bisucaberin bound to FoxA. Bisucaberin is distinct from other siderophores because it forms a 3:2 rather than 1:1 siderophore-iron complex. Mutations in a single extracellular loop of FoxA differentially affected nocardamine, thiocillin, and bisucaberin binding, uptake, and signal transduction. These results show that in addition to modulating ligand binding, the extracellular loops of siderophore transporters are of fundamental importance for controlling ligand uptake and its regulatory consequences, which have implications for the development of siderophore-antibiotic conjugates to treat difficult infections.


Assuntos
Antibacterianos , Sideróforos , Sideróforos/metabolismo , Antibacterianos/farmacologia , Antibacterianos/metabolismo , Ligantes , Proteínas de Membrana Transportadoras/genética , Proteínas de Membrana Transportadoras/metabolismo , Ferro/metabolismo , Transdução de Sinais , Pseudomonas aeruginosa/genética , Proteínas da Membrana Bacteriana Externa/metabolismo , Proteínas de Bactérias/metabolismo
6.
Mol Cell ; 82(17): 3151-3165.e9, 2022 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-35907401

RESUMO

Rifamycin antibiotics such as rifampin are potent inhibitors of prokaryotic RNA polymerase (RNAP) used to treat tuberculosis and other bacterial infections. Although resistance arises in the clinic principally through mutations in RNAP, many bacteria possess highly specific enzyme-mediated resistance mechanisms that modify and inactivate rifamycins. The expression of these enzymes is controlled by a 19-bp cis-acting rifamycin-associated element (RAE). Guided by the presence of RAE sequences, we identify a helicase-like protein, HelR, in Streptomyces venezuelae that confers broad-spectrum rifamycin resistance. We show that HelR also promotes tolerance to rifamycins, enabling bacterial evasion of the toxic properties of these antibiotics. HelR forms a complex with RNAP and rescues transcription inhibition by displacing rifamycins from RNAP, thereby providing resistance by target protection . Furthermore, HelRs are broadly distributed in Actinobacteria, including several opportunistic Mycobacterial pathogens, offering yet another challenge for developing new rifamycin antibiotics.


Assuntos
Rifamicinas , Tuberculose , Antibacterianos/farmacologia , RNA Polimerases Dirigidas por DNA/genética , RNA Polimerases Dirigidas por DNA/metabolismo , Humanos , Rifampina/metabolismo , Rifampina/farmacologia , Rifamicinas/farmacologia , Streptomyces/enzimologia
7.
ACS Omega ; 7(5): 4170-4184, 2022 Feb 08.
Artigo em Inglês | MEDLINE | ID: mdl-35155911

RESUMO

The aminopolycarboxylic acid aspergillomarasmine A (AMA) is a natural Zn2+ metallophore and inhibitor of metallo-ß-lactamases (MBLs) which reverses ß-lactam resistance. The first crystal structure of an AMA coordination complex is reported and reveals a pentadentate ligand with distorted octahedral geometry. We report the solid-phase synthesis of 23 novel analogs of AMA involving structural diversification of each subunit (l-Asp, l-APA1, and l-APA2). Inhibitory activity was evaluated in vitro using five strains of Escherichia coli producing globally prevalent MBLs. Further in vitro assessment was performed with purified recombinant enzymes and intracellular accumulation studies. Highly constrained structure-activity relationships were demonstrated, but three analogs revealed favorable characteristics where either Zn2+ affinity or the binding mode to MBLs were improved. This study identifies compounds that can further be developed to produce more potent and broader-spectrum MBL inhibitors with improved pharmacodynamic/pharmacokinetic properties.

8.
Cell Chem Biol ; 28(10): 1394-1406.e10, 2021 10 21.
Artigo em Inglês | MEDLINE | ID: mdl-33979648

RESUMO

Natural products (NPs) encompass a rich source of bioactive chemical entities. Here, we used human cancer stem cells (CSCs) in a chemical genomics campaign with NP chemical space to interrogate extracts from diverse strains of actinomycete for anti-cancer properties. We identified a compound (McM25044) capable of selectively inhibiting human CSC function versus normal stem cell counterparts. Biochemical and molecular studies revealed that McM025044 exerts inhibition on human CSCs through the small ubiquitin-like modifier (SUMO) cascade, found to be hyperactive in a variety of human cancers. McM025044 impedes the SUMOylation pathway via direct targeting of the SAE1/2 complex. Treatment of patient-derived CSCs resulted in reduced levels of SUMOylated proteins and suppression of progenitor and stem cell capacity measured in vitro and in vivo. Our study overcomes a barrier in chemically inhibiting oncogenic SUMOylation activity and uncovers a unique role for SAE2 in the biology of human cancers.


Assuntos
Células-Tronco Neoplásicas/metabolismo , Enzimas Ativadoras de Ubiquitina/metabolismo , Animais , Antineoplásicos/química , Antineoplásicos/metabolismo , Antineoplásicos/farmacologia , Antineoplásicos/uso terapêutico , Sítios de Ligação , Produtos Biológicos/química , Produtos Biológicos/metabolismo , Produtos Biológicos/farmacologia , Produtos Biológicos/uso terapêutico , Linhagem Celular Tumoral , Autorrenovação Celular , Sobrevivência Celular/efeitos dos fármacos , Humanos , Leucemia Mieloide Aguda/tratamento farmacológico , Leucemia Mieloide Aguda/patologia , Camundongos , Simulação de Acoplamento Molecular , Células-Tronco Neoplásicas/citologia , Interferência de RNA , RNA Interferente Pequeno/metabolismo , Sumoilação/efeitos dos fármacos , Enzimas Ativadoras de Ubiquitina/química , Enzimas Ativadoras de Ubiquitina/genética
9.
mBio ; 12(1)2021 02 09.
Artigo em Inglês | MEDLINE | ID: mdl-33563840

RESUMO

Apramycin is an aminoglycoside antibiotic with the potential to be developed to combat multidrug-resistant pathogens. Its unique structure evades the clinically widespread mechanisms of aminoglycoside resistance that currently compromise the efficacy of other members in this drug class. Of the aminoglycoside-modifying enzymes that chemically alter these antibiotics, only AAC(3)-IVa has been demonstrated to confer resistance to apramycin through N-acetylation. Knowledge of other modification mechanisms is important to successfully develop apramycin for clinical use. Here, we show that ApmA is structurally unique among the previously described aminoglycoside-modifying enzymes and capable of conferring a high level of resistance to apramycin. In vitro experiments indicated ApmA to be an N-acetyltransferase, but in contrast to AAC(3)-IVa, ApmA has a unique regiospecificity of the acetyl transfer to the N2' position of apramycin. Crystallographic analysis of ApmA conclusively showed that this enzyme is an acetyltransferase from the left-handed ß-helix protein superfamily (LßH) with a conserved active site architecture. The success of apramycin will be dependent on consideration of the impact of this potential form of clinical resistance.IMPORTANCE Apramycin is an aminoglycoside antibiotic that has been traditionally used in veterinary medicine. Recently, it has become an attractive candidate to repurpose in the fight against multidrug-resistant pathogens prioritized by the World Health Organization. Its atypical structure circumvents most of the clinically relevant mechanisms of resistance that impact this class of antibiotics. Prior to repurposing apramycin, it is important to understand the resistance mechanisms that could be a liability. Our study characterizes the most recently identified apramycin resistance element, apmA We show ApmA does not belong to the protein families typically associated with aminoglycoside resistance and is responsible for modifying a different site on the molecule. The data presented will be critical in the development of apramycin derivatives that will evade apmA in the event it becomes prevalent in the clinic.


Assuntos
Acetiltransferases/química , Acetiltransferases/metabolismo , Antibacterianos/química , Antibacterianos/metabolismo , Escherichia coli/efeitos dos fármacos , Nebramicina/análogos & derivados , Acetilação , Aminoglicosídeos/química , Cristalização , Farmacorresistência Bacteriana/genética , Escherichia coli/enzimologia , Escherichia coli/genética , Proteínas de Escherichia coli/química , Proteínas de Escherichia coli/metabolismo , Humanos , Testes de Sensibilidade Microbiana , Nebramicina/química , Nebramicina/metabolismo
10.
ACS Infect Dis ; 6(12): 3163-3173, 2020 12 11.
Artigo em Inglês | MEDLINE | ID: mdl-33164482

RESUMO

The rise of bacterial antibiotic resistance coupled with a diminished antibiotic drug pipeline underlines the importance of developing rational strategies to discover new antimicrobials. Microbially derived natural products are the basis for most of the antibiotic arsenal available to modern medicine. Here, we demonstrate a resistance-based approach to identify producers of elfamycins, an under-explored class of natural product antibiotics that target the essential translation factor EF-Tu. Antibiotic producers carry self-resistance genes to avoid suicide. These genes are often found within the same biosynthetic gene cluster (BGC) responsible for making the antibiotic, and we exploited this trait to identify members of the kirromycin class of elfamycin producers. Genome mining of Streptomyces spp. led to the identification of three isolates that harbor kirromycin-resistant EF-Tu (EF-TuKirR) within predicted natural product BGCs. Activity-guided purification on extracts of one of the Streptomyces isolates, which was not known to produce an elfamycin, identified it as a producer of phenelfamycin B, a linear polyketide. Phenelfamycin B demonstrates impressive antibacterial activity (MIC ∼ 1 µg/mL) against multidrug-resistant Neisseria gonorrhoeae, a clinically important Gram negative pathogen. The antigonococcal activity of phenelfamycin was shown to be the result of inhibition of protein biosynthesis by binding to EF-Tu. These results indicate that a resistance-based approach of identifying elfamycin producers is translatable to other antibiotic classes that can identify new and overlooked antibiotics necessary to address the antibiotic crisis.


Assuntos
Antibacterianos , Farmacorresistência Bacteriana , Streptomyces , Antibacterianos/farmacologia , Farmacorresistência Bacteriana/efeitos dos fármacos , Humanos , Fator Tu de Elongação de Peptídeos/genética , Fator Tu de Elongação de Peptídeos/metabolismo , Biossíntese de Proteínas/efeitos dos fármacos , Streptomyces/genética
11.
Nature ; 578(7796): 582-587, 2020 02.
Artigo em Inglês | MEDLINE | ID: mdl-32051588

RESUMO

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.


Assuntos
Antibacterianos , Descoberta de Drogas , Peptídeos Cíclicos , Peptidoglicano/efeitos dos fármacos , Peptidoglicano/metabolismo , Actinobacteria/química , Actinobacteria/genética , Actinobacteria/metabolismo , Animais , Antibacterianos/química , Antibacterianos/metabolismo , Antibacterianos/farmacologia , Vias Biossintéticas/genética , Parede Celular/metabolismo , Clorofenóis/química , Clorofenóis/metabolismo , Clorofenóis/farmacologia , Modelos Animais de Doenças , Resistência Microbiana a Medicamentos/efeitos dos fármacos , Resistência Microbiana a Medicamentos/genética , Feminino , Staphylococcus aureus Resistente à Meticilina/efeitos dos fármacos , Camundongos , Testes de Sensibilidade Microbiana , Família Multigênica , N-Acetil-Muramil-L-Alanina Amidase/antagonistas & inibidores , Peptídeos Cíclicos/química , Peptídeos Cíclicos/metabolismo , Peptídeos Cíclicos/farmacologia , Filogenia , Pele/microbiologia , Infecções Estafilocócicas/microbiologia
12.
Artigo em Inglês | MEDLINE | ID: mdl-31932375

RESUMO

The rise of Gram-negative pathogens expressing metallo-ß-lactamases (MBLs) is a growing concern, threatening the efficacy of ß-lactam antibiotics, in particular, the carbapenems. There are no inhibitors of MBLs in current clinical use. Aspergillomarasmine A (AMA) is an MBL inhibitor isolated from Aspergillus versicolor with the ability to rescue meropenem activity in MBL-producing bacteria both in vitro and in vivo Here, we systematically explored the pairing of AMA with six ß-lactam antibiotic partners against 19 MBLs from three subclasses (B1, B2, and B3). Cell-based assays performed with Escherichia coli and Klebsiella pneumoniae showed that bacteria producing NDM-1 and VIM-2 of subclass B1 were the most susceptible to AMA inhibition, whereas bacteria producing CphA2 and AIM-1 of subclasses B2 and B3, respectively, were the least sensitive. Intracellular antibiotic accumulation assays and in vitro enzyme assays demonstrated that the efficacy of AMA/ß-lactam combinations did not correlate with outer membrane permeability or drug efflux. We determined that the optimal ß-lactam partners for AMA are the carbapenem antibiotics and that the efficacy of AMA is linked to the Zn2+ affinity of specific MBLs.


Assuntos
Antibacterianos/farmacologia , Ácido Aspártico/análogos & derivados , Carbapenêmicos/farmacologia , Escherichia coli/efeitos dos fármacos , Klebsiella pneumoniae/efeitos dos fármacos , Inibidores de beta-Lactamases/farmacologia , Ácido Aspártico/farmacologia , Aspergillus/genética , Permeabilidade da Membrana Celular/fisiologia , Escherichia coli/genética , Klebsiella pneumoniae/genética , Testes de Sensibilidade Microbiana , Resistência beta-Lactâmica/genética , beta-Lactamases/genética , beta-Lactamases/metabolismo
13.
ACS Infect Dis ; 4(6): 980-987, 2018 06 08.
Artigo em Inglês | MEDLINE | ID: mdl-29634241

RESUMO

Plazomicin is a next-generation, semisynthetic aminoglycoside antibiotic currently under development for the treatment of infections due to multidrug-resistant Enterobacteriaceae. The compound was designed by chemical modification of the natural product sisomicin to provide protection from common aminoglycoside modifying enzymes that chemically alter these drugs via N-acetylation, O-adenylylation, or O-phosphorylation. In this study, plazomicin was profiled against a panel of isogenic strains of Escherichia coli individually expressing twenty-one aminoglycoside resistance enzymes. Plazomicin retained antibacterial activity against 15 of the 17 modifying enzyme-expressing strains tested. Expression of only two of the modifying enzymes, aac(2')-Ia and aph(2″)-IVa, decreased plazomicin potency. On the other hand, expression of 16S rRNA ribosomal methyltransferases results in a complete lack of plazomicin potency. In vitro enzymatic assessment confirmed that AAC(2')-Ia and APH(2'')-IVa (aminoglycoside acetyltransferase, AAC; aminoglycoside phosphotransferase, APH) were able to utilize plazomicin as a substrate. AAC(2')-Ia and APH(2'')-IVa are limited in their distribution to Providencia stuartii and Enterococci, respectively. These data demonstrate that plazomicin is not modified by a broad spectrum of common aminoglycoside modifying enzymes including those commonly found in Enterobacteriaceae. However, plazomicin is inactive in the presence of 16S rRNA ribosomal methyltransferases, which should be monitored in future surveillance programs.


Assuntos
Aminoglicosídeos/metabolismo , Antibacterianos/química , Antibacterianos/farmacologia , Sisomicina/análogos & derivados , Acetiltransferases/antagonistas & inibidores , Aminoglicosídeos/química , Aminoglicosídeos/farmacologia , Escherichia coli/efeitos dos fármacos , Escherichia coli/enzimologia , Humanos , Espectrometria de Massas , Testes de Sensibilidade Microbiana , Modelos Moleculares , Conformação Molecular , Estrutura Molecular , Sisomicina/química , Sisomicina/farmacologia , Relação Estrutura-Atividade
14.
Cell Chem Biol ; 25(4): 403-412.e5, 2018 04 19.
Artigo em Inglês | MEDLINE | ID: mdl-29398560

RESUMO

Rifamycin monooxygenases (Rox) are present in a variety of environmental bacteria and are associated with decomposition of the clinically utilized antibiotic rifampin. Here we report the structure and function of a drug-inducible rox gene from Streptomyces venezuelae, which encodes a class A flavoprotein monooxygenase that inactivates a broad range of rifamycin antibiotics. Our findings describe a mechanism of rifamycin inactivation initiated by monooxygenation of the 2-position of the naphthyl group, which subsequently results in ring opening and linearization of the antibiotic. The result is an antibiotic that no longer adopts the basket-like structure essential for binding to the RNA exit tunnel of the target RpoB, thereby providing the molecular logic of resistance. This unique mechanism of enzymatic inactivation underpins the broad spectrum of rifamycin resistance mediated by Rox enzymes and presents a new antibiotic resistance mechanism not yet seen in microbial antibiotic detoxification.


Assuntos
Antibacterianos/metabolismo , Proteínas de Bactérias/metabolismo , Farmacorresistência Bacteriana , Oxigenases de Função Mista/metabolismo , Rifamicinas/metabolismo , Streptomyces/enzimologia , Antibacterianos/química , Antibacterianos/farmacologia , Proteínas de Bactérias/química , Oxigenases de Função Mista/química , Simulação de Acoplamento Molecular , Conformação Proteica , Rifamicinas/química , Rifamicinas/farmacologia , Streptomyces/química , Streptomyces/efeitos dos fármacos , Streptomyces/metabolismo
15.
Nat Commun ; 9(1): 112, 2018 01 09.
Artigo em Inglês | MEDLINE | ID: mdl-29317655

RESUMO

The production of antibiotics by microbes in the environment and their use in medicine and agriculture select for existing and emerging resistance. To address this inevitability, prudent development of antibiotic drugs requires careful consideration of resistance evolution. Here, we identify the molecular basis for expanded substrate specificity in MphI, a macrolide kinase (Mph) that does not confer resistance to erythromycin, in contrast to other known Mphs. Using a combination of phylogenetics, drug-resistance phenotypes, and in vitro enzyme assays, we find that MphI and MphK phosphorylate erythromycin poorly resulting in an antibiotic-sensitive phenotype. Using likelihood reconstruction of ancestral sequences and site-saturation combinatorial mutagenesis, supported by Mph crystal structures, we determine that two non-obvious mutations in combination expand the substrate range. This approach should be applicable for studying the functional evolution of any antibiotic resistance enzyme and for evaluating the evolvability of resistance enzymes to new generations of antibiotic scaffolds.


Assuntos
Proteínas de Bactérias/metabolismo , Farmacorresistência Bacteriana , Macrolídeos/metabolismo , Fosfotransferases/metabolismo , Antibacterianos/química , Antibacterianos/metabolismo , Antibacterianos/farmacologia , Proteínas de Bactérias/química , Proteínas de Bactérias/genética , Eritromicina/química , Eritromicina/metabolismo , Eritromicina/farmacologia , Escherichia coli/efeitos dos fármacos , Escherichia coli/genética , Macrolídeos/química , Macrolídeos/farmacologia , Modelos Moleculares , Estrutura Molecular , Fosfotransferases/classificação , Fosfotransferases/genética , Filogenia , Domínios Proteicos , Especificidade por Substrato
16.
ISME J ; 12(3): 885-897, 2018 03.
Artigo em Inglês | MEDLINE | ID: mdl-29259290

RESUMO

The ecology of antibiotic resistance involves the interplay of a long natural history of antibiotic production in the environment, and the modern selection of resistance in pathogens through human use of these drugs. Important components of the resistome are intrinsic resistance genes of environmental bacteria, evolved and acquired over millennia, and their mobilization, which drives dissemination in pathogens. Understanding the dynamics and evolution of resistance across bacterial taxa is essential to address the current crisis in drug-resistant infections. Here we report the exploration of antibiotic resistance in the Paenibacillaceae prompted by our discovery of an ancient intrinsic resistome in Paenibacillus sp. LC231, recovered from the isolated Lechuguilla cave environment. Using biochemical and gene expression analysis, we have mined the resistome of the second member of the Paenibacillaceae family, Brevibacillus brevis VM4, which produces several antimicrobial secondary metabolites. Using phylogenomics, we show that Paenibacillaceae resistomes are in flux, evolve mostly independent of secondary metabolite biosynthetic diversity, and are characterized by cryptic, redundant, pseudoparalogous, and orthologous genes. We find that in contrast to pathogens, mobile genetic elements are not significantly responsible for resistome remodeling. This offers divergent modes of resistome development in pathogens and environmental bacteria.


Assuntos
Antibacterianos/farmacologia , Farmacorresistência Bacteriana/genética , Paenibacillus , Brevibacillus/efeitos dos fármacos , Brevibacillus/genética , Cavernas , Ecologia , Perfilação da Expressão Gênica , Humanos , Paenibacillus/efeitos dos fármacos , Paenibacillus/genética
17.
Cell Chem Biol ; 24(1): 98-109, 2017 Jan 19.
Artigo em Inglês | MEDLINE | ID: mdl-28017602

RESUMO

Solving the antibiotic resistance crisis requires the discovery of new antimicrobial drugs and the preservation of existing ones. The discovery of inhibitors of antibiotic resistance, antibiotic adjuvants, is a proven example of the latter. A major difficulty in identifying new antibiotics is the frequent rediscovery of known compounds, necessitating laborious "dereplication" to identify novel chemical entities. We have developed an antibiotic resistance platform (ARP) that can be used for both the identification of antibiotic adjuvants and for antibiotic dereplication. The ARP is a cell-based array of mechanistically distinct individual resistance elements in an identical genetic background. In dereplication mode, we demonstrate the rapid identification, and thus discrimination, of common antibiotics. In adjuvant discovery mode, we show that the ARP can be harnessed in screens to identify inhibitors of resistance. The ARP is therefore a powerful tool that has broad application in confronting the resistance crisis.


Assuntos
Adjuvantes Farmacêuticos/farmacologia , Antibacterianos/farmacologia , Bactérias/efeitos dos fármacos , Descoberta de Drogas , Farmacorresistência Bacteriana/efeitos dos fármacos , Adjuvantes Farmacêuticos/química , Antibacterianos/química , Ensaios de Triagem em Larga Escala , Estrutura Molecular
18.
Nat Commun ; 7: 13803, 2016 12 08.
Artigo em Inglês | MEDLINE | ID: mdl-27929110

RESUMO

Antibiotic resistance is ancient and widespread in environmental bacteria. These are therefore reservoirs of resistance elements and reflective of the natural history of antibiotics and resistance. In a previous study, we discovered that multi-drug resistance is common in bacteria isolated from Lechuguilla Cave, an underground ecosystem that has been isolated from the surface for over 4 Myr. Here we use whole-genome sequencing, functional genomics and biochemical assays to reveal the intrinsic resistome of Paenibacillus sp. LC231, a cave bacterial isolate that is resistant to most clinically used antibiotics. We systematically link resistance phenotype to genotype and in doing so, identify 18 chromosomal resistance elements, including five determinants without characterized homologues and three mechanisms not previously shown to be involved in antibiotic resistance. A resistome comparison across related surface Paenibacillus affirms the conservation of resistance over millions of years and establishes the longevity of these genes in this genus.


Assuntos
Cavernas/microbiologia , Farmacorresistência Bacteriana Múltipla/genética , Paenibacillus/fisiologia , Acetiltransferases/genética , Amidoidrolases/genética , Proteínas de Bactérias/genética , Macrolídeos/metabolismo , Fosfotransferases/genética , Fosfotransferases/metabolismo , Especificidade por Substrato , Sequenciamento Completo do Genoma
19.
Angew Chem Int Ed Engl ; 55(42): 13259-13262, 2016 10 10.
Artigo em Inglês | MEDLINE | ID: mdl-27633338

RESUMO

The fungal secondary metabolite aspergillomarasmine A (AMA) has recently been identified as an inhibitor of metallo-ß-lactamases NDM-1 and VIM-2. Described herein is an efficient and practical route to AMA and its related compounds by a sulfamidate approach. In addition, a series of derivatives has been prepared and tested for biological activity in an effort to explore preliminary structure activity relationships. While it was determined that natural LLL isomer of AMA remains the most effective inactivator of NDM-1 enzyme activity both in vitro and in cells, the structure is highly tolerant of the changes in the stereochemistry at positions 3, 6, and 9.


Assuntos
Amidas/farmacologia , Antibacterianos/farmacologia , Ácido Aspártico/análogos & derivados , Inibidores Enzimáticos/farmacologia , beta-Lactamases/metabolismo , Acinetobacter/efeitos dos fármacos , Acinetobacter/enzimologia , Amidas/química , Antibacterianos/síntese química , Antibacterianos/química , Ácido Aspártico/síntese química , Ácido Aspártico/química , Ácido Aspártico/farmacologia , Relação Dose-Resposta a Droga , Enterobacteriaceae/efeitos dos fármacos , Enterobacteriaceae/enzimologia , Inibidores Enzimáticos/síntese química , Inibidores Enzimáticos/química , Testes de Sensibilidade Microbiana , Estrutura Molecular , Pseudomonas/efeitos dos fármacos , Pseudomonas/enzimologia , Relação Estrutura-Atividade
20.
Methods Mol Biol ; 1440: 99-108, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27311667

RESUMO

Sortase enzymes have specific endopeptidase activity, cleaving within a defined pentapeptide sequence at the C-terminal end of their protein substrates. Here, we describe how monitoring sortase cleavage activity can be achieved using peptide substrates. Peptide cleavage can be readily analyzed by liquid chromatography/tandem mass spectrometry (LC/MS/MS), which allows for the precise definition of cleavage sites. This technique could be used to analyze the peptidase activity of any enzyme, and identify sites of cleavage within any peptide.


Assuntos
Cromatografia Líquida/métodos , Cisteína Endopeptidases/metabolismo , Espectrometria de Massas em Tandem/métodos , Peptídeos/química , Proteólise
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA