Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
1.
Nature ; 597(7878): 698-702, 2021 09.
Artigo em Inglês | MEDLINE | ID: mdl-34526714

RESUMO

The development of new antibiotics to treat infections caused by drug-resistant Gram-negative pathogens is of paramount importance as antibiotic resistance continues to increase worldwide1. Here we describe a strategy for the rational design of diazabicyclooctane inhibitors of penicillin-binding proteins from Gram-negative bacteria to overcome multiple mechanisms of resistance, including ß-lactamase enzymes, stringent response and outer membrane permeation. Diazabicyclooctane inhibitors retain activity in the presence of ß-lactamases, the primary resistance mechanism associated with ß-lactam therapy in Gram-negative bacteria2,3. Although the target spectrum of an initial lead was successfully re-engineered to gain in vivo efficacy, its ability to permeate across bacterial outer membranes was insufficient for further development. Notably, the features that enhanced target potency were found to preclude compound uptake. An improved optimization strategy leveraged porin permeation properties concomitant with biochemical potency in the lead-optimization stage. This resulted in ETX0462, which has potent in vitro and in vivo activity against Pseudomonas aeruginosa plus all other Gram-negative ESKAPE pathogens, Stenotrophomonas maltophilia and biothreat pathogens. These attributes, along with a favourable preclinical safety profile, hold promise for the successful clinical development of the first novel Gram-negative chemotype to treat life-threatening antibiotic-resistant infections in more than 25 years.


Assuntos
Antibacterianos/farmacologia , Desenho de Fármacos , Farmacorresistência Bacteriana Múltipla , Bactérias Gram-Negativas/efeitos dos fármacos , Animais , Antibacterianos/química , Compostos Aza/química , Compostos Aza/farmacologia , Ciclo-Octanos/química , Ciclo-Octanos/farmacologia , Feminino , Camundongos , Camundongos Endogâmicos BALB C , Estrutura Molecular , Proteínas de Ligação às Penicilinas/antagonistas & inibidores , Pseudomonas aeruginosa/efeitos dos fármacos , beta-Lactamases
2.
Antimicrob Agents Chemother ; : e0011224, 2024 Jun 18.
Artigo em Inglês | MEDLINE | ID: mdl-38888319

RESUMO

Inhalation anthrax is the most severe form of Bacillus anthracis infection, often progressing to fatal conditions if left untreated. While recommended antibiotics can effectively treat anthrax when promptly administered, strains engineered for antibiotic resistance could render these drugs ineffective. Telavancin, a semisynthetic lipoglycopeptide antibiotic, was evaluated in this study as a novel therapeutic against anthrax disease. Specifically, the aims were to (i) assess in vitro potency of telavancin against 17 B. anthracis isolates by minimum inhibitory concentration (MIC) testing and (ii) evaluate protective efficacy in rabbits infected with a lethal dose of aerosolized anthrax spores and treated with human-equivalent intravenous telavancin doses (30 mg/kg every 12 hours) for 5 days post-antigen detection versus a humanized dose of levofloxacin and vehicle control. Blood samples were collected at various times post-infection to assess the level of bacteremia and antibody production, and tissues were collected to determine bacterial load. The animals' body temperatures were also recorded. Telavancin demonstrated potent bactericidal activity against all strains tested (MICs 0.06-0.125 µg/mL). Further, telavancin conveyed 100% survival in this model and cleared B. anthracis from the bloodstream and organ tissues more effectively than a humanized dose of levofloxacin. Collectively, the low MICs against all strains tested and rapid bactericidal in vivo activity demonstrate that telavancin has the potential to be an effective alternative for the treatment or prophylaxis of anthrax infection.

3.
Mol Microbiol ; 117(6): 1419-1433, 2022 06.
Artigo em Inglês | MEDLINE | ID: mdl-35526138

RESUMO

Toxin-antitoxin loci regulate adaptive responses to stresses associated with the host environment and drug exposure. Phylogenomic studies have shown that Mycobacterium tuberculosis encodes a naturally expanded type II toxin-antitoxin system, including ParDE/RelBE superfamily members. Type II toxins are presumably regulated exclusively through protein-protein interactions with type II antitoxins. However, experimental observations in M. tuberculosis indicated that additional control mechanisms regulate RelBE2 type II loci under host-associated stress conditions. Herein, we describe for the first time a novel antisense RNA, termed asRelE2, that co-regulates RelE2 production via targeted processing by the Mtb RNase III, Rnc. We find that convergent expression of this coding-antisense hybrid TA locus, relBE2-asrelE2, is controlled in a cAMP-dependent manner by the essential cAMP receptor protein transcription factor, Crp, in response to the host-associated stresses of low pH and nutrient limitation. Ex vivo survival studies with relE2 and asrelE2 knockout strains showed that RelE2 contributes to Mtb survival in activated macrophages and low pH to nutrient limitation. To our knowledge, this is the first report of a novel tripartite type IIb TA loci and antisense post-transcriptional regulation of a type II TA loci.


Assuntos
Antitoxinas , Toxinas Bacterianas , Mycobacterium tuberculosis , Sistemas Toxina-Antitoxina , Antitoxinas/genética , Antitoxinas/metabolismo , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Toxinas Bacterianas/genética , Toxinas Bacterianas/metabolismo , Mycobacterium tuberculosis/genética , Mycobacterium tuberculosis/metabolismo , RNA Antissenso/genética , RNA Antissenso/metabolismo , Sistemas Toxina-Antitoxina/genética
4.
Biochemistry ; 56(13): 1865-1878, 2017 04 04.
Artigo em Inglês | MEDLINE | ID: mdl-28225601

RESUMO

There is growing awareness of the link between drug-target residence time and in vivo drug activity, and there are increasing efforts to determine the molecular factors that control the lifetime of a drug-target complex. Rational alterations in the drug-target residence time require knowledge of both the ground and transition states on the inhibition reaction coordinate, and we have determined the structure-kinetic relationship for 22 ethyl- or hexyl-substituted diphenyl ethers that are slow-binding inhibitors of bpFabI1, the enoyl-ACP reductase FabI1 from Burkholderia pseudomallei. Analysis of enzyme inhibition using a two-dimensional kinetic map demonstrates that the ethyl and hexyl diphenyl ethers fall into two distinct clusters. Modifications to the ethyl diphenyl ether B ring result in changes to both on and off rates, where residence times of up to ∼700 min (∼11 h) are achieved by either ground state stabilization (PT444) or transition state destabilization (slower on rate) (PT404). By contrast, modifications to the hexyl diphenyl ether B ring result in residence times of 300 min (∼5 h) through changes in only ground state stabilization (PT119). Structural analysis of nine enzyme:inhibitor complexes reveals that the variation in structure-kinetic relationships can be rationalized by structural rearrangements of bpFabI1 and subtle changes to the orientation of the inhibitor in the binding pocket. Finally, we demonstrate that three compounds with residence times on bpFabI1 from 118 min (∼2 h) to 670 min (∼11 h) have in vivo efficacy in an acute B. pseudomallei murine infection model using the virulent B. pseudomallei strain Bp400.


Assuntos
Antibacterianos/química , Proteínas de Bactérias/antagonistas & inibidores , Burkholderia pseudomallei/efeitos dos fármacos , Enoil-(Proteína de Transporte de Acila) Redutase (NADH)/antagonistas & inibidores , Inibidores Enzimáticos/química , Melioidose/dietoterapia , Éteres Fenílicos/química , Animais , Antibacterianos/farmacologia , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Sítios de Ligação , Burkholderia pseudomallei/enzimologia , Burkholderia pseudomallei/genética , Burkholderia pseudomallei/crescimento & desenvolvimento , Contagem de Colônia Microbiana , Cristalografia por Raios X , Enoil-(Proteína de Transporte de Acila) Redutase (NADH)/genética , Enoil-(Proteína de Transporte de Acila) Redutase (NADH)/metabolismo , Inibidores Enzimáticos/farmacologia , Feminino , Expressão Gênica , Cinética , Pulmão/efeitos dos fármacos , Pulmão/microbiologia , Melioidose/tratamento farmacológico , Melioidose/microbiologia , Camundongos , Camundongos Endogâmicos BALB C , Testes de Sensibilidade Microbiana , Simulação de Dinâmica Molecular , Éteres Fenílicos/farmacologia , Ligação Proteica , Estrutura Secundária de Proteína , Baço/efeitos dos fármacos , Baço/microbiologia , Relação Estrutura-Atividade
5.
J Biol Chem ; 289(23): 15987-6005, 2014 Jun 06.
Artigo em Inglês | MEDLINE | ID: mdl-24739388

RESUMO

Determining the molecular basis for target selectivity is of particular importance in drug discovery. The ideal antibiotic should be active against a broad spectrum of pathogenic organisms with a minimal effect on human targets. CG400549, a Staphylococcus-specific 2-pyridone compound that inhibits the enoyl-acyl carrier protein reductase (FabI), has recently been shown to possess human efficacy for the treatment of methicillin-resistant Staphylococcus aureus infections, which constitute a serious threat to human health. In this study, we solved the structures of three different FabI homologues in complex with several pyridone inhibitors, including CG400549. Based on these structures, we rationalize the 65-fold reduced affinity of CG400549 toward Escherichia coli versus S. aureus FabI and implement concepts to improve the spectrum of antibacterial activity. The identification of different conformational states along the reaction coordinate of the enzymatic hydride transfer provides an elegant visual depiction of the relationship between catalysis and inhibition, which facilitates rational inhibitor design. Ultimately, we developed the novel 4-pyridone-based FabI inhibitor PT166 that retained favorable pharmacokinetics and efficacy in a mouse model of S. aureus infection with extended activity against Gram-negative and mycobacterial organisms.


Assuntos
Antibacterianos/farmacologia , Desenho de Fármacos , Enoil-(Proteína de Transporte de Acila) Redutase (NADH)/antagonistas & inibidores , Inibidores Enzimáticos/farmacologia , Piridonas/farmacologia , Animais , Antibacterianos/química , Antibacterianos/farmacocinética , Sequência de Bases , Cristalografia por Raios X , Primers do DNA , Inibidores Enzimáticos/química , Inibidores Enzimáticos/farmacocinética , Feminino , Camundongos , Camundongos Endogâmicos ICR , Testes de Sensibilidade Microbiana , Estrutura Molecular , Reação em Cadeia da Polimerase , Piridonas/química , Staphylococcus aureus/efeitos dos fármacos , Staphylococcus aureus/crescimento & desenvolvimento
6.
Antimicrob Agents Chemother ; 58(2): 931-5, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24277048

RESUMO

The bacterial fatty acid biosynthesis pathway is a validated target for the development of novel chemotherapeutics. However, since Burkholderia pseudomallei carries genes that encode both FabI and FabV enoyl-acyl carrier protein (ACP) reductase homologues, the enoyl-ACP reductase that is essential for in vivo growth needs to be defined so that the correct drug target can be chosen for development. Accordingly, ΔfabI1, ΔfabI2, and ΔfabV knockout strains were constructed and tested in a mouse model of infection. Mice infected with a ΔfabI1 strain did not show signs of morbidity, mortality, or dissemination after 30 days of infection compared to the wild-type and ΔfabI2 and ΔfabV mutant strains that had times to mortality of 60 to 84 h. Although signs of morbidity and mortality of ΔfabI2 and ΔfabV strains were not significantly different from those of the wild-type strain, a slight delay was observed. A FabI1-specific inhibitor was used to confirm that inhibition of FabI1 results in reduced bacterial burden and efficacy in an acute B. pseudomallei murine model of infection. This work establishes that FabI1 is required for growth of Burkholderia pseudomallei in vivo and is a potential molecular target for drug development.


Assuntos
Antibacterianos/farmacologia , Proteínas de Bactérias/genética , Burkholderia pseudomallei/genética , Enoil-(Proteína de Transporte de Acila) Redutase (NADPH, B-Específica)/genética , Inibidores Enzimáticos/farmacologia , Melioidose/tratamento farmacológico , Animais , Antibacterianos/química , Proteínas de Bactérias/antagonistas & inibidores , Proteínas de Bactérias/metabolismo , Burkholderia pseudomallei/efeitos dos fármacos , Burkholderia pseudomallei/enzimologia , Burkholderia pseudomallei/patogenicidade , Enoil-(Proteína de Transporte de Acila) Redutase (NADPH, B-Específica)/antagonistas & inibidores , Enoil-(Proteína de Transporte de Acila) Redutase (NADPH, B-Específica)/metabolismo , Inibidores Enzimáticos/química , Feminino , Técnicas de Inativação de Genes , Isoenzimas/antagonistas & inibidores , Isoenzimas/genética , Isoenzimas/metabolismo , Melioidose/microbiologia , Melioidose/mortalidade , Camundongos , Mutação , Análise de Sobrevida , Resultado do Tratamento
7.
Antimicrob Agents Chemother ; 58(3): 1646-51, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24379198

RESUMO

Identification of a novel class of anti-Burkholderia compounds is key in addressing antimicrobial resistance to current therapies as well as naturally occurring resistance. The FabI enoyl-ACP reductase in Burkholderia is an underexploited target that presents an opportunity for development of a new class of inhibitors. A library of substituted diphenyl ethers was used to identify FabI1-specific inhibitors for assessment in Burkholderia pseudomallei ex vivo and murine efficacy models. Active FabI1 inhibitors were identified in a two-stage format consisting of percent inhibition screening and MIC determination by the broth microdilution method. Each compound was evaluated against the B. pseudomallei 1026b (efflux-proficient) and Bp400 (efflux-compromised) strains. In vitro screening identified candidate substituted diphenyl ethers that exhibited MICs of less than 1 µg/ml, and enzyme kinetic assays were used to assess potency and specificity against the FabI1 enzyme. These compounds demonstrated activity in a Burkholderia ex vivo efficacy model, and two demonstrated efficacy in an acute B. pseudomallei mouse infection model. This work establishes substituted diphenyl ethers as a suitable platform for development of novel anti-Burkholderia compounds that can be used for treatment of melioidosis.


Assuntos
Antibacterianos/farmacologia , Burkholderia pseudomallei/efeitos dos fármacos , Éteres Fenílicos/farmacologia , Animais , Burkholderia pseudomallei/enzimologia , Modelos Animais de Doenças , Enoil-(Proteína de Transporte de Acila) Redutase (NADH)/antagonistas & inibidores , Enoil-(Proteína de Transporte de Acila) Redutase (NADH)/metabolismo , Feminino , Melioidose/tratamento farmacológico , Camundongos , Camundongos Endogâmicos BALB C , Testes de Sensibilidade Microbiana , Células Vero/efeitos dos fármacos
8.
Microbiol Spectr ; 12(5): e0353423, 2024 May 02.
Artigo em Inglês | MEDLINE | ID: mdl-38534149

RESUMO

To address intracellular mycobacterial infections, we developed a cocktail of four enzymes that catalytically attack three layers of the mycobacterial envelope. This cocktail is delivered to macrophages, through a targeted liposome presented here as ENTX_001. Endolytix Cocktail 1 (EC1) leverages mycobacteriophage lysin enzymes LysA and LysB, while also including α-amylase and isoamylase for degradation of the mycobacterial envelope from outside of the cell. The LysA family of proteins from mycobacteriophages has been shown to cleave the peptidoglycan layer, whereas LysB is an esterase that hydrolyzes the linkage between arabinogalactan and mycolic acids of the mycomembrane. The challenge of gaining access to the substrates of LysA and LysB provided exogenously was addressed by adding amylase enzymes that degrade the extracellular capsule shown to be present in Mycobacterium tuberculosis. This enzybiotic approach avoids antimicrobial resistance, specific receptor-mediated binding, and intracellular DNA surveillance pathways that limit many bacteriophage applications. We show this cocktail of enzymes is bactericidal in vitro against both rapid- and slow-growing nontuberculous mycobacteria (NTM) as well as M. tuberculosis strains. The EC1 cocktail shows superior killing activity when compared to previously characterized LysB alone. EC1 is also powerfully synergistic with standard-of-care antibiotics. In addition to in vitro killing of NTM, ENTX_001 demonstrates the rescue of infected macrophages from necrotic death by Mycobacteroides abscessus and Mycobacterium avium. Here, we demonstrate shredding of mycobacterial cells by EC1 into cellular debris as a mechanism of bactericide.IMPORTANCEThe world needs entirely new forms of antibiotics as resistance to chemical antibiotics is a critical problem facing society. We addressed this need by developing a targeted enzyme therapy for a broad range of species and strains within mycobacteria and highly related genera including nontuberculous mycobacteria such as Mycobacteroides abscessus, Mycobacterium avium, Mycobacterium intracellulare, as well as Mycobacterium tuberculosis. One advantage of this approach is the ability to drive our lytic enzymes through encapsulation into macrophage-targeted liposomes resulting in attack of mycobacteria in the cells that harbor them where they hide from the adaptive immune system and grow. Furthermore, this approach shreds mycobacteria independent of cell physiology as the drug targets the mycobacterial envelope while sidestepping the host range limitations observed with phage therapy and resistance to chemical antibiotics.


Assuntos
Galactanos , Macrófagos , Micobacteriófagos , Mycobacterium tuberculosis , Micobactérias não Tuberculosas , Mycobacterium tuberculosis/efeitos dos fármacos , Mycobacterium tuberculosis/enzimologia , Micobacteriófagos/genética , Micobacteriófagos/enzimologia , Macrófagos/microbiologia , Macrófagos/virologia , Humanos , Micobactérias não Tuberculosas/efeitos dos fármacos , Lipossomos/química , Antibacterianos/farmacologia , Peptidoglicano/metabolismo , Testes de Sensibilidade Microbiana , Endopeptidases/metabolismo , Endopeptidases/farmacologia , Endopeptidases/genética
9.
J Bacteriol ; 195(2): 351-8, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23144254

RESUMO

Francisella tularensis is classified as a category A priority pathogen and causes fatal disseminated disease in humans upon inhalation of less than 50 bacteria. Although drugs are available for treatment, they are not ideal because of toxicity and route of delivery, and in some cases patients relapse upon withdrawal. We have an ongoing program to develop novel FAS-II FabI enoyl-ACP reductase enzyme inhibitors for Francisella and other select agents. To establish F. tularensis FabI (FtFabI) as a clinically relevant drug target, we demonstrated that fatty acid biosynthesis and FabI activity are essential for growth even in the presence of exogenous long-chain lipids and that FtfabI is not transcriptionally altered in the presence of exogenous long-chain lipids. Inhibition of FtFabI or fatty acid synthesis results in loss of viability that is not rescued by exogenous long-chain lipid supplementation. Importantly, whole-genome transcriptional profiling of F. tularensis with DNA microarrays from infected tissues revealed that FtfabI and de novo fatty acid biosynthetic genes are transcriptionally active during infection. This is the first demonstration that the FabI enoyl-ACP-reductase enzyme encoded by F. tularensis is essential and not bypassed by exogenous fatty acids and that de novo fatty acid biosynthetic components encoded in F. tularensis are transcriptionally active during infection in the mouse model of tularemia.


Assuntos
Enoil-(Proteína de Transporte de Acila) Redutase (NADH)/biossíntese , Francisella tularensis/enzimologia , Regulação Bacteriana da Expressão Gênica , Genes Bacterianos , Genes Essenciais , Viabilidade Microbiana , Tularemia/microbiologia , Animais , Modelos Animais de Doenças , Enoil-(Proteína de Transporte de Acila) Redutase (NADH)/genética , Ácidos Graxos/biossíntese , Francisella tularensis/genética , Francisella tularensis/crescimento & desenvolvimento , Perfilação da Expressão Gênica , Humanos , Camundongos , Análise em Microsséries , Análise de Sequência com Séries de Oligonucleotídeos
10.
Bioorg Med Chem ; 21(11): 3318-26, 2013 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-23623254

RESUMO

Francisella tularensis is a highly virulent pathogenic bacterium. In order to identify novel potential antibacterial agents against F. tularensis, libraries of trisubstituted benzimidazoles were screened against F. tularensis LVS strain. In a preliminary screening assay, remarkably, 23 of 2,5,6- and 2,5,7-trisubstituted benzimidazoles showed excellent activity exhibiting greater than 90% growth inhibition at 1 µg/mL. Among those hits, 21 compounds showed MIC90 values in the range of 0.35-48.6 µg/mL after accurate MIC determination. In ex vivo efficacy assays, four of these compounds exhibited 2-3log reduction in colony forming units (CFU) per mL at concentrations of 10 and 50 µg/mL.


Assuntos
Antibacterianos/farmacologia , Benzimidazóis/farmacologia , Francisella tularensis/efeitos dos fármacos , Macrófagos/efeitos dos fármacos , Bibliotecas de Moléculas Pequenas/farmacologia , Animais , Antibacterianos/síntese química , Benzimidazóis/síntese química , Linhagem Celular , Francisella tularensis/crescimento & desenvolvimento , Ensaios de Triagem em Larga Escala , Macrófagos/microbiologia , Camundongos , Testes de Sensibilidade Microbiana , Bibliotecas de Moléculas Pequenas/química , Relação Estrutura-Atividade
11.
PLoS Negl Trop Dis ; 17(11): e0011795, 2023 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-38011278

RESUMO

Burkholderia pseudomallei is the causative agent of melioidosis, which is increasingly being reported worldwide. Mortality rates as high as 40% have been reported based on clinical patient outcomes in the endemic areas of Australia and Thailand. Novel therapies are needed to reduce treatment duration and adverse effects and improve treatment outcomes. Epetraborole, a novel antibiotic, targets leucyl-tRNA synthetase (LeuRS), an essential enzyme that catalyzes the attachment of leucine to transfer RNA. Epetraborole was evaluated for in vitro activity and efficacy in a murine model to assess clinical relevance against Burkholderia pseudomallei infections for possible treatment of melioidosis. Epetraborole was tested against 13 clinically derived and three reference B. pseudomallei strains that have a broad spectrum of susceptibilities to the standard-of-care (SoC) drugs for melioidosis, which showed that epetraborole exhibited minimal inhibitory concentrations of 0.25-4 µg/mL. Ex vivo studies using THP-1 macrophages confirmed the potency of epetraborole and demonstrated synergy between epetraborole and ceftazidime. In the acute pulmonary murine infection model of melioidosis, epetraborole demonstrated equivalent efficacy when delivered orally or subcutaneously, which compared well with the standard-of-care drug ceftazidime. In addition, adding epetraborole to ceftazidime significantly improved antimicrobial activity in this animal model. This work warrants further exploration of epetraborole as a candidate for treating melioidosis and substantiates LeuRS as a clinically relevant drug target in B. pseudomallei.


Assuntos
Aminoacil-tRNA Sintetases , Burkholderia pseudomallei , Melioidose , Animais , Camundongos , Humanos , Ceftazidima/farmacologia , Ceftazidima/uso terapêutico , Melioidose/tratamento farmacológico , Antibacterianos/farmacologia , Antibacterianos/uso terapêutico , Aminoacil-tRNA Sintetases/farmacologia , Aminoacil-tRNA Sintetases/uso terapêutico
12.
JAC Antimicrob Resist ; 4(2): dlac028, 2022 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-35350133

RESUMO

Background: NIAID has a programme for testing drug candidates against biodefense and emerging bacterial pathogens that uses defined strain panels consisting of standard laboratory reference strains and strains of clinical origin. Objectives: The current studies were performed to assess the activity of standard-of-care drugs, determine benchmark criteria for new investigational antibacterial candidate prioritization and identify reduced non-redundant strain panels for candidate performance classification. Methods: The susceptibilities of each strain in the screening panels to 40 standard-of-care drugs and clinical drug combinations were determined by percentage growth inhibition using multiple concentrations, a method commonly used in efficient high-throughput screening efforts. The drug susceptibility of each strain was categorized based on interpretive criteria to benchmark the activity of each standard-of-care drug and drug combination, followed by confirmation of select active drugs. Exact match and clustering analyses defined focused non-redundant species and pan-species screening panels. Results: This process revealed a broad spectrum of susceptibilities among strains in each species, with important differences between the standard laboratory reference strains and strains of clinical origin. Exact match and clustering analyses identified subsets of non-redundant strains that can more efficiently classify drug activity resulting in individual species screening panels, a pan-species screening panel and a pan-species maximum resistance panel. Conclusions: This study resulted in improved non-redundant species screening panels for benchmarking the performance of new investigational antibacterial candidates with the greatest potential for efficacy against clinically relevant Category A and B priority and emerging pathogens.

13.
Front Immunol ; 13: 811430, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35250984

RESUMO

Despite significant research efforts, treatment options for severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) remain limited. This is due in part to a lack of therapeutics that increase host defense to the virus. Replication of SARS-CoV-2 in lung tissue is associated with marked infiltration of macrophages and activation of innate immune inflammatory responses that amplify tissue injury. Antagonists of the androgen (AR) and glucocorticoid (GR) receptors have shown efficacy in models of COVID-19 and in clinical studies because the cell surface proteins required for viral entry, angiotensin converting enzyme 2 (ACE2) and the transmembrane protease, serine 2 (TMPRSS2), are transcriptionally regulated by these receptors. We postulated that the GR and AR modulator, PT150, would reduce infectivity of SARS-CoV-2 and prevent inflammatory lung injury in the Syrian golden hamster model of COVID-19 by down-regulating expression of critical genes regulated through these receptors. Animals were infected intranasally with 2.5 × 104 TCID50/ml equivalents of SARS-CoV-2 (strain 2019-nCoV/USA-WA1/2020) and PT150 was administered by oral gavage at 30 and 100 mg/Kg/day for a total of 7 days. Animals were examined at 3, 5 and 7 days post-infection (DPI) for lung histopathology, viral load and production of proteins regulating the progression of SARS-CoV-2 infection. Results indicated that oral administration of PT150 caused a dose-dependent decrease in replication of SARS-CoV-2 in lung, as well as in expression of ACE2 and TMPRSS2. Lung hypercellularity and infiltration of macrophages and CD4+ T-cells were dramatically decreased in PT150-treated animals, as was tissue damage and expression of IL-6. Molecular docking studies suggest that PT150 binds to the co-activator interface of the ligand-binding domain of both AR and GR, thereby acting as an allosteric modulator and transcriptional repressor of these receptors. Phylogenetic analysis of AR and GR revealed a high degree of sequence identity maintained across multiple species, including humans, suggesting that the mechanism of action and therapeutic efficacy observed in Syrian hamsters would likely be predictive of positive outcomes in patients. PT150 is therefore a strong candidate for further clinical development for the treatment of COVID-19 across variants of SARS-CoV-2.


Assuntos
Antivirais/farmacologia , Tratamento Farmacológico da COVID-19 , Glucocorticoides/metabolismo , Imunidade Inata/efeitos dos fármacos , Inflamação/tratamento farmacológico , Receptores Androgênicos/metabolismo , Internalização do Vírus/efeitos dos fármacos , Animais , COVID-19/metabolismo , Modelos Animais de Doenças , Feminino , Inflamação/metabolismo , Inflamação/virologia , Pulmão/virologia , Masculino , Mesocricetus , SARS-CoV-2/efeitos dos fármacos , SARS-CoV-2/metabolismo , Serina Endopeptidases/metabolismo , Glicoproteína da Espícula de Coronavírus/metabolismo , Carga Viral/efeitos dos fármacos
14.
J Antimicrob Chemother ; 66(3): 564-73, 2011 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-21393229

RESUMO

OBJECTIVES: As an initial step in developing novel antibacterials against Burkholderia pseudomallei, we have characterized the FabI enoyl-ACP reductase homologues in the type II fatty acid biosynthesis pathway from this organism and performed an initial enzyme inhibition study. METHODS: A BLAST analysis identified two FabI enoyl-ACP reductase homologues, bpmFabI-1 and bpmFabI-2, in the B. pseudomallei genome, which were cloned, overexpressed in Escherichia coli and purified. Steady-state kinetics was used to determine the reaction mechanism and the sensitivity of bpmFabI-1 to four diphenyl ether FabI inhibitors. The antibacterial activity of the inhibitors was assessed using a wild-type strain of Burkholderia thailandensis (E264) and an efflux pump mutant (Bt38). RESULTS: Consistent with its annotation as an enoyl-ACP reductase, bpmFabI-1 catalysed the NADH-dependent reduction of 2-trans-dodecenoyl-CoA via a sequential Bi Bi mechanism. In contrast, bpmFabI-2 was inactive with all substrates tested and only bpmfabI-1 was transcriptionally active under the growth conditions employed. The sensitivity of bpmFabI-1 to four diphenyl ethers was evaluated and in each case the compounds were slow-onset inhibitors with K(i) values of 0.5-2 nM. In addition, triclosan and PT01 had MIC values of 30 and 70 mg/L for B. pseudomallei as well as a wild-type strain of B. thailandensis (E264), but MIC values of <1 mg/L for the efflux pump mutant Bt38. A reduction in MIC values was also observed for the pump mutant strain with the other diphenyl ethers. CONCLUSIONS: Provided that efflux can be circumvented, bpmFabI-1 is a suitable target for drug discovery.


Assuntos
Proteínas de Bactérias/antagonistas & inibidores , Burkholderia pseudomallei/efeitos dos fármacos , Burkholderia pseudomallei/enzimologia , Enoil-(Proteína de Transporte de Acila) Redutase (NADH)/antagonistas & inibidores , Proteínas de Bactérias/genética , Burkholderia pseudomallei/genética , Clonagem Molecular , Enoil-(Proteína de Transporte de Acila) Redutase (NADH)/genética , Escherichia coli/genética , Expressão Gênica , Cinética , Testes de Sensibilidade Microbiana , Proteínas Recombinantes/antagonistas & inibidores , Proteínas Recombinantes/genética , Proteínas Recombinantes/isolamento & purificação , Homologia de Sequência de Aminoácidos
15.
JAC Antimicrob Resist ; 3(2): dlab058, 2021 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-34223120

RESUMO

BACKGROUND: Francisella tularensis is a highly virulent and contagious Gram-negative intracellular bacterium that causes the disease tularaemia in mammals and is classified as a Category A priority pathogen. METHODS: We utilized a systematic analysis of antibacterial potency, extent of dissemination by analysis of bacterial burden in a secondary vital organ, and survival rates to assess the efficacy of a novel rifampicin derivative, TPR1. The efficacy of TPR1 was evaluated alone and in combination with the standard of care drug, doxycycline, against type A F. tularensis Schu S4 using a lethal pulmonary model of infection in mice. RESULTS: TPR1 has an MIC value range of 0.125-4 mg/L against reference laboratory strain Schu S4 and a panel of clinical strains. TPR1 alone reduced the bacterial burden in the lungs and spleen at 40 mg/kg and 80 mg/kg, and no antagonism was observed when co-administered with doxycycline. Dosing at 40 mg/kg doxycycline reduced the bacterial burden by 1 log10 cfu in the lungs and 4 log10 cfu in the spleen in comparison to untreated controls. Co-administration of TPR1 and doxycycline demonstrated efficacy upon treatment withdrawal after 4 days of treatment, and 100% survival. CONCLUSIONS: Significantly, TPR1 demonstrated efficacy when delivered alone and in combination with doxycycline, which provides compelling evidence of a superior treatment strategy that would normally rely on a single chemotherapeutic for efficacy. In addition, this work substantiates the use of rifampicin derivatives as a platform for the development of novel treatments to other bacterial agents in addition to tularaemia.

16.
PLoS One ; 16(1): e0245171, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33493177

RESUMO

Infection with Influenza A virus can lead to the development of encephalitis and subsequent neurological deficits ranging from headaches to neurodegeneration. Post-encephalitic parkinsonism has been reported in surviving patients of H1N1 infections, but not all cases of encephalitic H1N1 infection present with these neurological symptoms, suggesting that interactions with an environmental neurotoxin could promote more severe neurological damage. The heavy metal, manganese (Mn), is a potential interacting factor with H1N1 because excessive exposure early in life can induce long-lasting effects on neurological function through inflammatory activation of glial cells. In the current study, we used a two-hit model of neurotoxin-pathogen exposure to examine whether exposure to Mn during juvenile development would induce a more severe neuropathological response following infection with H1N1 in adulthood. To test this hypothesis, C57BL/6 mice were exposed to MnCl2 in drinking water (50 mg/kg/day) for 30 days from days 21-51 postnatal, then infected intranasally with H1N1 three weeks later. Analyses of dopaminergic neurons, microglia and astrocytes in basal ganglia indicated that although there was no significant loss of dopaminergic neurons within the substantia nigra pars compacta, there was more pronounced activation of microglia and astrocytes in animals sequentially exposed to Mn and H1N1, as well as altered patterns of histone acetylation. Whole transcriptome Next Generation Sequencing (RNASeq) analysis was performed on the substantia nigra and revealed unique patterns of gene expression in the dual-exposed group, including genes involved in antioxidant activation, mitophagy and neurodegeneration. Taken together, these results suggest that exposure to elevated levels of Mn during juvenile development could sensitize glial cells to more severe neuro-immune responses to influenza infection later in life through persistent epigenetic changes.


Assuntos
Regulação da Expressão Gênica , Vírus da Influenza A Subtipo H1N1/metabolismo , Manganês/farmacologia , Meningite Viral/metabolismo , Neuroglia/metabolismo , Infecções por Orthomyxoviridae/metabolismo , Substância Negra/metabolismo , Animais , Feminino , Masculino , Meningite Viral/patologia , Camundongos , Neuroglia/patologia , Neuroglia/virologia , Infecções por Orthomyxoviridae/patologia , RNA-Seq , Substância Negra/patologia , Substância Negra/virologia
17.
J Med Chem ; 64(20): 15214-15249, 2021 10 28.
Artigo em Inglês | MEDLINE | ID: mdl-34614347

RESUMO

Novel bacterial topoisomerase inhibitors (NBTIs) are among the most promising new antibiotics in preclinical/clinical development. We previously reported dioxane-linked NBTIs with potent antistaphylococcal activity and reduced hERG inhibition, a key safety liability. Herein, polarity-focused optimization enabled the delineation of clear structure-property relationships for both microsomal metabolic stability and hERG inhibition, resulting in the identification of lead compound 79. This molecule demonstrates potent antibacterial activity against diverse Gram-positive pathogens, inhibition of both DNA gyrase and topoisomerase IV, a low frequency of resistance, a favorable in vitro cardiovascular safety profile, and in vivo efficacy in a murine model of methicillin-resistant Staphylococcus aureus infection.


Assuntos
Antibacterianos/farmacologia , Dioxanos/farmacologia , Inibidores Enzimáticos/farmacologia , Canais de Potássio Éter-A-Go-Go/antagonistas & inibidores , Staphylococcus aureus Resistente à Meticilina/efeitos dos fármacos , Antibacterianos/síntese química , Antibacterianos/química , DNA Girase/metabolismo , DNA Topoisomerase IV/antagonistas & inibidores , DNA Topoisomerase IV/metabolismo , Dioxanos/síntese química , Dioxanos/química , Relação Dose-Resposta a Droga , Inibidores Enzimáticos/síntese química , Inibidores Enzimáticos/química , Canais de Potássio Éter-A-Go-Go/metabolismo , Humanos , Testes de Sensibilidade Microbiana , Estrutura Molecular , Relação Estrutura-Atividade
18.
Biochim Biophys Acta ; 1768(7): 1796-804, 2007 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-17531950

RESUMO

We show that cryptdin-4 (Crp4), an antimicrobial peptide found in mice, induces the aggregation and hemi-fusion of charged phospholipid vesicles constructed of the anionic lipid POPG and the zwitterionic lipid POPC. Hemi-fusion is confirmed with positive total lipid-mixing assay results, negative inner monolayer lipid-mixing assay results, and negative results from contents-mixing assays. Aggregation, as quantified by absorbance and dynamic light scattering, is self-limiting, creating finite-sized vesicle assemblies. The rate limiting step in the formation process is the mixing of juxtaposed membrane leaflets, which is regulated by bound peptide concentration as well as vesicle radius (with larger vesicles less prone to hemi-fusion). Bound peptide concentration is readily controlled by total peptide concentration and the fraction of anionic lipid in the vesicles. As little as 1% PEGylated lipid significantly reduces aggregate size by providing a steric barrier for membrane apposition. Finally, as stable hemi-fusion is a rare occurrence, we compare properties of Crp4 to those of many peptides known to induce complete fusion and lend insight into conditions necessary for this unusual type of membrane merger.


Assuntos
Lipossomas Unilamelares/química , alfa-Defensinas/farmacologia , 4-Cloro-7-nitrobenzofurazano/análogos & derivados , 4-Cloro-7-nitrobenzofurazano/química , Animais , Luz , Camundongos , Fosfatidiletanolaminas/química , Fosfatidilgliceróis/química , Polietilenoglicóis/química , Rodaminas/química , Espalhamento de Radiação
19.
Pathog Dis ; 76(4)2018 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-29788125

RESUMO

There has been a significant reduction in annual tuberculosis incidence since the World Health Organization declared tuberculosis a global health threat. However, treatment of M. tuberculosis infections requires lengthy multidrug therapeutic regimens to achieve a durable cure. The development of new drugs that are active against resistant strains and phenotypically diverse organisms continues to present the greatest challenge in the future. Numerous phylogenomic analyses have revealed that the Mtb genome encodes a significantly expanded repertoire of toxin-antitoxin (TA) loci that makes up the Mtb TA system. A TA loci is a two-gene operon encoding a 'toxin' protein that inhibits bacterial growth and an interacting 'antitoxin' partner that neutralizes the inhibitory activity of the toxin. The presence of multiple chromosomally encoded TA loci in Mtb raises important questions in regard to expansion, regulation and function. Thus, the functional roles of TA loci in Mtb pathogenesis have received considerable attention over the last decade. The cumulative results indicate that they are involved in regulating adaptive responses to stresses associated with the host environment and drug treatment. Here we review the TA families encoded in Mtb, discuss the duplication of TA loci in Mtb, regulatory mechanism of TA loci, and phenotypic heterogeneity and pathogenesis.


Assuntos
Antitoxinas/genética , Toxinas Bacterianas/genética , Regulação Bacteriana da Expressão Gênica , Genoma Bacteriano , Mycobacterium tuberculosis/genética , Sistemas Toxina-Antitoxina , Antitoxinas/biossíntese , Toxinas Bacterianas/biossíntese , Duplicação Cromossômica , Heterogeneidade Genética , Loci Gênicos , Interações Hospedeiro-Patógeno , Humanos , Mycobacterium tuberculosis/classificação , Mycobacterium tuberculosis/crescimento & desenvolvimento , Mycobacterium tuberculosis/patogenicidade , Óperon , Filogenia , Transdução de Sinais , Tuberculose/metabolismo , Tuberculose/microbiologia
20.
Colloids Surf B Biointerfaces ; 60(2): 236-42, 2007 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-17686617

RESUMO

Cryptdin-4 is a beta-sheet antimicrobial peptide of the defensin family that is found in the immune system of mice. Several structure-activity studies of this peptide have previously been conducted, but none have been based on residue-membrane interactions as part of an overall hypothesis on the peptide's orientation in the membrane. We pursue this valuable approach by first using previously reported NMR structural data to propose a membrane-bound orientation of the peptide. Four mutants are then strategically designed to modulate membrane perturbative activity in a manner consistent with the proposed binding orientation. Membrane perturbation is evaluated using a simple fluorescence-based vesicle leakage assay using POPG to form the model membrane. Effects of peptide mutations are found to be consistent with the suggested binding orientation. This approach is successfully used to create synthetic peptides with enhanced or diminished ability to perturb membranes and also yields insights on the nature of peptide-membrane interactions.


Assuntos
Peptídeos Catiônicos Antimicrobianos/química , alfa-Defensinas/química , Animais , Peptídeos Catiônicos Antimicrobianos/síntese química , Peptídeos Catiônicos Antimicrobianos/genética , Sítios de Ligação , Espectroscopia de Ressonância Magnética/métodos , Membranas Artificiais , Camundongos , Modelos Moleculares , Mutagênese Sítio-Dirigida , Fosfatidilgliceróis/química , Estrutura Secundária de Proteína , Propriedades de Superfície , alfa-Defensinas/síntese química , alfa-Defensinas/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA