Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 56
1.
Microbiol Spectr ; 10(3): e0082822, 2022 06 29.
Article En | MEDLINE | ID: mdl-35647694

Carbapenem-resistant Enterobacteriaceae (CRE) are an urgent threat to public health requiring the development of novel therapies. TP0586532 is a novel non-hydroxamate LpxC inhibitor that inhibits the synthesis of lipopolysaccharides, which are components of the outer membranes of Gram-negative bacteria. Based on the mechanism of action of TP0586532, we hypothesized that it might enhance the antibacterial activity of other antibiotics by increasing the permeability of the outer bacterial membrane. The combination of TP0586532 with meropenem, amikacin, cefepime, piperacillin, and tigecycline showed synergistic and additive effects against carbapenem-susceptible Klebsiella pneumoniae and Escherichia coli. Checkerboard experiments against 21 carbapenem-resistant K. pneumoniae and E. coli strains (13 blaKPC+, 5 blaNDM-1+, 2 blaVIM+, and 1 blaIMP+) showed that the combination of TP0586532 with meropenem yielded synergistic and additive effects against 9 and 12 strains, respectively. In a time-kill assay examining 12 CRE strains, synergistic effects were observed when TP0586532 was combined with meropenem against many of the strains. A membrane permeability assay using ethidium bromide (EtBr) was performed to investigate the mechanism of the potentiating effect. TP0586532 increased the influx of EtBr into a CRE strain, suggesting that TP0586532 increased membrane permeability and facilitated intracellular access for the antibiotics. Our study demonstrates that TP0586532 potentiates the in vitro antibacterial activity of meropenem against CRE. Combination therapy consisting of TP0586532 and meropenem has potential as a treatment for CRE infections. IMPORTANCE Carbapenem-resistant Enterobacteriaceae (CRE) are an urgent public health threat, as therapeutic options are limited. TP0586532 is a novel LpxC inhibitor that inhibits the synthesis of lipopolysaccharides in the outer membranes of Gram-negative bacteria. Here, we demonstrated the potentiating effects of TP0586532 on the antibacterial activity of meropenem against CRE harboring various types of carbapenemase genes (blaKPC+, blaNDM-1+ blaVIM+, and blaIMP+). TP0586532 also augmented the bactericidal effects of meropenem against CRE strains, even against those with a high level of resistance to meropenem. The potentiating effects were suggested to be mediated by an increase in bacterial membrane permeability. Our study revealed that a combination therapy consisting of TP0586532 and meropenem has the potential to be a novel therapeutic option for CRE infections.


Carbapenem-Resistant Enterobacteriaceae , Enterobacteriaceae Infections , Humans , Anti-Bacterial Agents/pharmacology , Bacterial Proteins/genetics , beta-Lactamases/genetics , Butanols/pharmacology , Carbapenem-Resistant Enterobacteriaceae/genetics , Carbapenems/pharmacology , Enterobacteriaceae Infections/drug therapy , Enterobacteriaceae Infections/microbiology , Escherichia coli/genetics , Gram-Negative Bacteria , Imidazoles/pharmacology , Klebsiella pneumoniae/genetics , Meropenem/pharmacology , Microbial Sensitivity Tests
2.
J Infect Chemother ; 28(5): 635-642, 2022 May.
Article En | MEDLINE | ID: mdl-35131156

INTRODUCTION: TP0586532 is a novel non-hydroxamate UDP-3-O-acyl-N-acetylglucosamine deacetylase (LpxC) inhibitor. Pharmacokinetic/pharmacodynamic (PK/PD) indices and magnitude of index that correlated with the efficacy of TP0586532 were determined and used to estimate the clinically effective doses of TP0586532. METHODS: Dose-fractionation studies were conducted using a murine neutropenic lung infection model caused by carbapenem-resistant Enterobacteriaceae. The relationships between the efficacy and the PK/PD index (the maximum unbound plasma concentration divided by the MIC [fCmax/MIC], the area under the unbound plasma concentration-time curve from 0 to 24 h divided by the MIC, and the cumulative percentage of a 24-h period that the unbound plasma concentration exceeds the MIC) were determined using an inhibitory sigmoid maximum-effect model. In addition, the magnitudes of fCmax/MIC were evaluated using the dose-response relationships for each of the seven carbapenem-resistant strains of Enterobacteriaceae. Furthermore, the clinically effective doses of TP0586532 were estimated using the predicted human PK parameters, the geometric mean of fCmax/MIC, and the MIC90 for carbapenem-resistant Klebsiella pneumoniae. RESULTS: The PK/PD index that best correlated with the efficacy was the fCmax/MIC. The geometric means of the fCmax/MIC associated with the net stasis and 1-log reduction endpoints were 2.30 and 3.28, respectively. The clinically effective doses of TP0586532 were estimated to be 1.24-2.74 g/day. CONCLUSION: These results indicate the potential for TP0586532 to have clinical efficacy at reasonable doses against infections caused by carbapenem-resistant Enterobacteriaceae. This study provided helpful information for a clinically effective dosing regimen of TP0586532.


Anti-Bacterial Agents , Klebsiella pneumoniae , Animals , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/therapeutic use , Enterobacteriaceae , Humans , Lung , Mice , Microbial Sensitivity Tests
3.
J Antibiot (Tokyo) ; 75(3): 136-145, 2022 03.
Article En | MEDLINE | ID: mdl-34987187

UDP-3-O-acyl-N-acetylglucosamine deacetylase (LpxC) is an essential enzyme in the biosynthesis of Lipid A, an active component of lipopolysaccharide (LPS), from UDP-3-O-acyl-N-acetylglicosamine. LPS is a major component of the cell surface of Gram-negative bacteria. LPS is known to be one of causative factors of sepsis and has been associated with high mortality in septic shock. TP0586532 is a novel non-hydroxamate LpxC enzyme inhibitor. In this study, we examined the inhibitory effect of TP0586532 on the LPS release from Klebsiella pneumoniae both in vitro and in vivo. Our results confirmed the inhibitory effect of TP0586532 on LPS release from the pathogenic bacterial species. On the other hand, meropenem and ciprofloxacin increase the level of LPS release. Furthermore, the effects of TP0586532 on LPS release and interleukin (IL)-6 production in the lung were determined using a murine model of pneumonia caused by K. pneumoniae. As observed in the in vitro study, TP0586532 showed the marked inhibitory effect on LPS release in the lungs, whereas meropenem- and ciprofloxacin-treated mice showed higher levels of LPS release and IL-6 production in the lungs as compared to those in the lungs of vehicle-treated mice. Moreover, TP0586532 used in combination with meropenem and ciprofloxacin attenuated the LPS release and IL-6 production induced by meropenem and ciprofloxacin in the lung. These results indicate that the inhibitory effect of TP0586532 on LPS release from pathogenic bacteria might be of benefit in patients with sepsis.


Amidohydrolases/metabolism , Anti-Bacterial Agents/pharmacology , Enzyme Inhibitors/pharmacology , Hydroxamic Acids/metabolism , Interleukin-6/metabolism , Lipopolysaccharides/metabolism , Animals , Ciprofloxacin/pharmacology , Female , Klebsiella pneumoniae/drug effects , Klebsiella pneumoniae/metabolism , Meropenem/metabolism , Mice , Mice, Inbred ICR , Microbial Sensitivity Tests/methods
4.
Nihon Yakurigaku Zasshi ; 157(1): 26, 2022.
Article Ja | MEDLINE | ID: mdl-34980806
5.
Nihon Yakurigaku Zasshi ; 157(1): 27-30, 2022.
Article Ja | MEDLINE | ID: mdl-34980807

The new coronavirus (SARS-CoV-2) spread throughout the world and caused a pandemic with COVID-19, an infection caused by SARS-CoV-2. Even today, an increase in the number of cases has also been observed in Japan. Since the drugs used in drug repositioning have already been tested for safety and pharmacokinetics in humans, it is possible to skip some development tests, and since the manufacturing method of the drug has already been established, it is possible to shorten the development period and reduce R&D costs. Therefore, the drug repositioning method is one of the methods that should be tried in order to achieve the initial control of a pandemic. In Japan, it has been announced that research and development using drug repositioning has been conducted to date. The following are some of the candidates that have already been identified as COVID-19 therapeutic agents in Japan and are expected to be identified in the future.


COVID-19 , Pharmaceutical Preparations , Antiviral Agents/pharmacology , Antiviral Agents/therapeutic use , Drug Repositioning , Humans , Pandemics , SARS-CoV-2
6.
J Antibiot (Tokyo) ; 75(2): 98-107, 2022 02.
Article En | MEDLINE | ID: mdl-34837061

The emergence of multi-drug resistant pathogenic bacteria, especially Gram-negative bacteria, is a worldwide health problem. New antibiotics directed at previously unexplored targets are urgently needed to overcome resistance to existing antibiotic classes. UDP-3-O-acyl-N-acetylglucosamine deacetylase (LpxC) is an attractive target for a new antibacterial agent. Although a number of LpxC inhibitors have been identified, none have been approved as antibacterial agents. These LpxC inhibitors contain a hydroxamate moiety, which is a robust zinc ion chelator. The nonspecific inhibition of metalloenzymes through zinc ion chelation is one of possibilities leading to unwanted side effects. Herein, we report that TP0586532, a non-hydroxamate LpxC inhibitor, has a broad spectrum of antibacterial activity against carbapenem-resistant Enterobacteriaceae. The MIC90 of TP0586532 against clinical isolates of carbapenem-resistant Klebsiella pneumoniae was 4 µg ml-1. TP0586532 also showed an in vivo efficacy against murine systemic, urinary tract and lung infection models caused by meropenem- or ciprofloxacin-resistant strains. The estimated maximum unbound plasma concentration value at the effective dose of TP0586532 in murine infection models was around 13 µg ml-1. TP0586532 is predicted to exhibit a in vivo efficacy without cardiovascular toxicity and showed the potential of non-hydroxamate LpxC inhibitors as antibacterial agents against carbapenem-resistant Enterobacteriaceae.


Amidohydrolases , Anti-Bacterial Agents , Enterobacteriaceae , Animals , Mice , Amidohydrolases/antagonists & inhibitors , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/toxicity , Chelating Agents/chemistry , Chelating Agents/pharmacology , Ciprofloxacin/pharmacology , Drug Resistance, Multiple, Bacterial/drug effects , Enterobacteriaceae/drug effects , Klebsiella Infections/drug therapy , Klebsiella Infections/microbiology , Klebsiella pneumoniae/drug effects , Meropenem/pharmacology , Microbial Sensitivity Tests , Zinc/chemistry
7.
Article En | MEDLINE | ID: mdl-33558293

Gonorrhea is a common, sexually transmitted disease caused by Neisseria gonorrhoeae Multidrug-resistant N. gonorrhoeae is an urgent threat, and the development of a new antimicrobial agent that functions via a new mechanism is strongly desired. We evaluated the in vitro and in vivo activities of a DNA gyrase/topoisomerase IV inhibitor, TP0480066, which is a novel 8-(methylamino)-2-oxo-1,2-dihydroquinoline derivative. The MICs of TP0480066 were substantially lower than those of other currently or previously used antimicrobials against gonococcal strains demonstrating resistance to fluoroquinolones, macrolides, ß-lactams, and aminoglycosides (MICs, ≤0.0005 µg/ml). Additionally, no cross-resistance was observed between TP0480066 and ciprofloxacin. The frequencies of spontaneous resistance to TP0480066 for N. gonorrhoeae ATCC 49226 were below the detection limit (<2.4 × 10-10) at concentrations equivalent to 32× MIC. TP0480066 also showed potent in vitro bactericidal activity and in vivo efficacy in a mouse model of N. gonorrhoeae infection. These data suggest that TP0480066 is a candidate antimicrobial agent for gonococcal infections.


Gonorrhea , Neisseria gonorrhoeae , Animals , Anti-Bacterial Agents/pharmacology , DNA Gyrase/genetics , DNA Topoisomerase IV/genetics , Drug Resistance, Bacterial , Fluoroquinolones , Gonorrhea/drug therapy , Mice , Microbial Sensitivity Tests
8.
Bioorg Med Chem ; 30: 115964, 2021 01 15.
Article En | MEDLINE | ID: mdl-33385955

Infectious diseases caused by resistant Gram-negative bacteria have become a serious problem, and the development of therapeutic drugs with a novel mechanism of action and that do not exhibit cross-resistance with existing drugs has been earnestly desired. UDP-3-O-acyl-N-acetylglucosamine deacetylase (LpxC) is a drug target that has been studied for a long time. However, no LpxC inhibitors are available on the market at present. In this study, we sought to create a new antibacterial agent without a hydroxamate moiety, which is a common component of the major LpxC inhibitors that have been reported to date and that may cause toxicity. As a result, a development candidate, TP0586532, was created that is effective against carbapenem-resistant Klebsiella pneumoniae and does not pose a cardiovascular risk.


Amidohydrolases/antagonists & inhibitors , Anti-Bacterial Agents/pharmacology , Drug Discovery , Enzyme Inhibitors/pharmacology , Escherichia coli/drug effects , Imidazoles/pharmacology , Klebsiella pneumoniae/drug effects , Amidohydrolases/metabolism , Anti-Bacterial Agents/chemical synthesis , Anti-Bacterial Agents/chemistry , Crystallography, X-Ray , Dose-Response Relationship, Drug , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/chemistry , Escherichia coli/enzymology , Imidazoles/chemical synthesis , Imidazoles/chemistry , Klebsiella pneumoniae/enzymology , Microbial Sensitivity Tests , Models, Molecular , Molecular Structure , Structure-Activity Relationship
9.
J Med Chem ; 63(23): 14805-14820, 2020 12 10.
Article En | MEDLINE | ID: mdl-33210531

UDP-3-O-acyl-N-acetylglucosamine deacetylase (LpxC) is a zinc metalloenzyme that catalyzes the first committed step in the biosynthesis of Lipid A, an essential component of the cell envelope of Gram-negative bacteria. The most advanced, disclosed LpxC inhibitors showing antibacterial activity coordinate zinc through a hydroxamate moiety with concerns about binding to other metalloenzymes. Here, we describe the discovery, optimization, and efficacy of two series of compounds derived from fragments with differing modes of zinc chelation. A series was evolved from a fragment where a glycine moiety complexes zinc, which achieved low nanomolar potency in an enzyme functional assay but poor antibacterial activity on cell cultures. A second series was based on a fragment that chelated zinc through an imidazole moiety. Structure-guided design led to a 2-(1S-hydroxyethyl)-imidazole derivative exhibiting low nanomolar inhibition of LpxC and a minimum inhibitory concentration (MIC) of 4 µg/mL against Pseudomonas aeruginosa, which is little affected by the presence of albumin.


Amidohydrolases/antagonists & inhibitors , Anti-Bacterial Agents/pharmacology , Bacterial Proteins/antagonists & inhibitors , Chelating Agents/pharmacology , Enzyme Inhibitors/pharmacology , Anilides/pharmacology , Anti-Bacterial Agents/chemical synthesis , Chelating Agents/chemical synthesis , Drug Discovery , Enzyme Inhibitors/chemical synthesis , Escherichia coli/drug effects , Escherichia coli/enzymology , Imidazoles/pharmacology , Microbial Sensitivity Tests , Molecular Structure , Piperidines/pharmacology , Pseudomonas aeruginosa/drug effects , Pseudomonas aeruginosa/enzymology , Structure-Activity Relationship , Zinc/chemistry
10.
Bioorg Med Chem ; 28(24): 115818, 2020 12 15.
Article En | MEDLINE | ID: mdl-33190073

The development of effective respiratory syncytial virus (RSV) fusion glycoprotein (F protein) inhibitors against both wild-type and the D486N-mutant F protein is urgently required. We recently reported a 15-membered macrocyclic pyrazolo[1,5-a]pyrimidine derivative 4 that exhibited potent anti-RSV activities against not only wild-type, but also D486N-mutant F protein. However, NMR studies revealed that the 15-membered derivative 4 existed as a mixture of atropisomers. An optimization study of the linker moiety between the 2-position of the benzoyl moiety and the 7-position of the pyrazolo[1,5-a]pyrimidine scaffold identified a 16-membered derivative 42c with an amide linker that showed a rapid interconversion of atropisomers. Subsequent optimization of the 5-position of the pyrazolo[1,5-a]pyrimidine scaffold and the 5-position of the benzoyl moiety resulted in the discovery of a potent clinical candidate 60b for the treatment of RSV infections.


Antiviral Agents/chemistry , Respiratory Syncytial Virus, Human/metabolism , Viral Fusion Proteins/antagonists & inhibitors , Animals , Antiviral Agents/metabolism , Antiviral Agents/pharmacology , Binding Sites , Cell Line , Cell Membrane Permeability/drug effects , Drug Evaluation, Preclinical , Half-Life , Humans , Isomerism , Macrocyclic Compounds/chemical synthesis , Macrocyclic Compounds/chemistry , Mice , Molecular Dynamics Simulation , Mutation , Pyrazoles/chemistry , Pyrazoles/metabolism , Pyrazoles/pharmacology , Pyrimidines/chemistry , Pyrimidines/metabolism , Pyrimidines/pharmacology , Structure-Activity Relationship , Viral Fusion Proteins/genetics , Viral Fusion Proteins/metabolism , Virus Internalization/drug effects
11.
Article En | MEDLINE | ID: mdl-33046486

Human respiratory syncytial virus (RSV) is a major cause of lower respiratory tract infections in early childhood. However, no vaccines have yet been approved for prevention of RSV infection, and the treatment options are limited. Therefore, development of effective and safe anti-RSV drugs is needed. In this study, we evaluated the antiviral activity and mechanism of action of a novel macrocyclic anti-RSV compound, TP0591816. TP0591816 showed significant antiviral activities against both subgroup A and subgroup B RSV, while exerting no cytotoxicity. Notably, the antiviral activity of TP0591816 was maintained against a known fusion inhibitor-resistant RSV strain with a mutation in the cysteine-rich region or in heptad repeat B. Results of a time-of-addition assay and a temperature shift assay indicated that TP0591816 inhibited fusion of RSV with the cell membrane during viral entry. In addition, TP0591816 added after cell infection also inhibited cell-cell fusion. A TP0591816-resistant virus strain selected by serial passage had an L141F mutation, but no mutation in the cysteine-rich region or in heptad repeat B in the fusion (F) protein. Treatment with TP0591816 reduced lung virus titers in a dose-dependent manner in a mouse model of RSV infection. Furthermore, the estimated effective dose of TP0591816 for use against F protein mutants was thought to be clinically realistic and potentially tolerable. Taken together, these findings suggest that TP0591816 is a promising novel candidate for the treatment of resistant RSV infection.


Respiratory Syncytial Virus Infections , Respiratory Syncytial Virus, Human , Antibodies, Viral , Antiviral Agents/pharmacology , Antiviral Agents/therapeutic use , Child, Preschool , Humans , Respiratory Syncytial Virus Infections/drug therapy , Respiratory Syncytial Virus, Human/genetics , Viral Fusion Proteins/genetics , Virus Internalization
12.
Bioorg Med Chem ; 28(22): 115776, 2020 11 15.
Article En | MEDLINE | ID: mdl-33032189

The global increase in multidrug-resistant pathogens has caused severe problems in the treatment of infections. To overcome these difficulties, the advent of a new chemical class of antibacterial drug is eagerly desired. We aimed at creating novel antibacterial agents against bacterial type II topoisomerases, which are well-validated targets. TP0480066 (compound 32) has been identified by using structure-based optimization originated from lead compound 1, which was obtained as a result of our previous lead identification studies. The MIC90 values of TP0480066 against methicillin-resistant Staphylococcus aureus (MRSA), vancomycin-resistant Enterococci (VRE), and genotype penicillin-resistant Streptococcus pneumoniae (gPRSP) were 0.25, 0.015, and 0.06 µg/mL, respectively. Hence, TP0480066 can be regarded as a promising antibacterial drug candidate of this chemical class.


Anti-Bacterial Agents/pharmacology , DNA Topoisomerases, Type II/metabolism , Quinolines/pharmacology , Topoisomerase II Inhibitors/pharmacology , Anti-Bacterial Agents/chemical synthesis , Anti-Bacterial Agents/chemistry , Cell Survival/drug effects , Dose-Response Relationship, Drug , Drug Resistance, Bacterial/drug effects , Hep G2 Cells , Humans , Methicillin-Resistant Staphylococcus aureus/drug effects , Methicillin-Resistant Staphylococcus aureus/enzymology , Microbial Sensitivity Tests , Models, Molecular , Molecular Structure , Quinolines/chemical synthesis , Quinolines/chemistry , Streptococcus pneumoniae/drug effects , Streptococcus pneumoniae/enzymology , Structure-Activity Relationship , Topoisomerase II Inhibitors/chemical synthesis , Topoisomerase II Inhibitors/chemistry , Transcriptional Regulator ERG/antagonists & inhibitors , Transcriptional Regulator ERG/metabolism , Vancomycin-Resistant Enterococci/drug effects , Vancomycin-Resistant Enterococci/enzymology
13.
ACS Med Chem Lett ; 11(6): 1145-1151, 2020 Jun 11.
Article En | MEDLINE | ID: mdl-32550994

A novel series of macrocyclic pyrazolo[1,5-a]pyrimidine derivatives as respiratory syncytial virus (RSV) fusion glycoprotein (F protein) inhibitors were designed and synthesized based on docking studies of acyclic inhibitors. This effort resulted in the discovery of several macrocyclic compounds, such as 12b, 12f, and 12h, with low nanomolar to subnanomolar activities against the wild-type RSV F protein A2. In addition, 12h showed a single-digit nanomolar potency against the previously reported drug-resistant mutant D486N. Molecular modeling and computational analyses suggested that 12h binds to the D486N mutant while maintaining a rigid bioactive conformation via macrocyclization and that it interacts with a hydrophobic cavity of the mutant using a new interaction surface of 12h. This report describes the rational design of macrocyclic compounds with dual inhibitory activities against wild-type and mutant RSV F proteins.

14.
ACS Omega ; 5(17): 10145-10159, 2020 May 05.
Article En | MEDLINE | ID: mdl-32391502

DNA gyrase and topoisomerase IV are well-validated pharmacological targets, and quinolone antibacterial drugs are marketed as their representative inhibitors. However, in recent years, resistance to these existing drugs has become a problem, and new chemical classes of antibiotics that can combat resistant strains of bacteria are strongly needed. In this study, we applied our hit-to-lead (H2L) chemistry for the identification of a new chemical class of GyrB/ParE inhibitors by efficient use of thermodynamic parameters. Investigation of the core fragments obtained by fragmentation of high-throughput screening hit compounds and subsequent expansion of the hit fragment was performed using isothermal titration calorimetry (ITC). The 8-(methylamino)-2-oxo-1,2-dihydroquinoline derivative 13e showed potent activity against Escherichia coli DNA gyrase with an IC50 value of 0.0017 µM. In this study, we demonstrated the use of ITC for primary fragment screening, followed by structural optimization to obtain lead compounds, which advanced into further optimization for creating novel antibacterial agents.

15.
Chem Pharm Bull (Tokyo) ; 68(4): 345-362, 2020.
Article En | MEDLINE | ID: mdl-32238652

Respiratory syncytial virus (RSV) is one of the most common causes of lower respiratory tract infections and a significant pathogen for both adults and children. Although two drugs have been approved for the treatment of RSV infections, the low therapeutic index of these drugs have led pharmaceutical companies to develop safe and effective small-molecule anti-RSV drugs. The pyrazolo[1,5-a]pyrimidine series of compounds containing a piperidine ring at the 2-position of the pyrazolo[1,5-a]pyrimidine scaffold are known as candidate RSV fusion (F) protein inhibitor drugs, such as presatovir and P3. The piperidine ring has been revealed to facilitate the formation of an appropriate dihedral angle between the pyrazolo[1,5-a]pyrimidine scaffold and the plane of the amide bond for exertion of anti-RSV activity. A molecular-dynamic study on newly designed compounds with an acyclic chain instead of the piperidine ring proposed and demonstrated a new series of pyrazolo[1,5-a]pyrimidine derivatives, such as 9c with a 1-methyaminopropyl moiety, showing similar dihedral angle distributions to those in presatovir. Compound 9c exhibited potent anti-RSV activity with an EC50 value of below 1 nM, which was similar to that of presatovir. A subsequent optimization study on the benzene ring of 9c led to the potent RSV F protein inhibitor 14f with an EC50 value of 0.15 nM. The possibility of improving the biological properties of anti-RSV agents by modification at the 7-position of pyrazolo[1,5-a]pyrimidine is also discussed.


Antiviral Agents/pharmacology , Drug Design , Pyrazoles/pharmacology , Pyrimidines/pharmacology , Respiratory Syncytial Virus, Human/drug effects , Antiviral Agents/chemical synthesis , Antiviral Agents/chemistry , Dose-Response Relationship, Drug , Humans , Microbial Sensitivity Tests , Molecular Structure , Pyrazoles/chemical synthesis , Pyrazoles/chemistry , Pyrimidines/chemical synthesis , Pyrimidines/chemistry , Stereoisomerism , Structure-Activity Relationship
16.
BMC Microbiol ; 20(1): 69, 2020 03 30.
Article En | MEDLINE | ID: mdl-32228455

BACKGROUND: Vibrio vulnificus hemolysin (VVH) is a pore-forming toxin secreted by Vibrio vulnificus. Cellular cholesterol was believed to be the receptor for VVH, because cholesterol could bind to VVH and preincubation with cholesterol inhibited cytotoxicity. It has been reported that specific glycans such as N-acetyl-D-galactosamine and N-acetyl-D-lactosamine bind to VVH, however, it has not been known whether these glycans could inhibit the cytotoxicity of VVH without oligomer formation. Thus, to date, binding mechanisms of VVH to cellular membrane, including specific receptors have not been elucidated. RESULTS: We show here that VVH associates with ganglioside GM1a, Fucosyl-GM1, GD1a, GT1c, and GD1b by glycan array. Among them, GM1a could pulldown VVH. Moreover, the GD1a inhibited the cytotoxicity of VVH without the formation of oligomers. CONCLUSION: This is the first report of a molecule able to inhibit the binding of VVH to target cells without oligomerization of VVH.


Cell Membrane/metabolism , Gangliosides/pharmacology , Hemolysin Proteins/metabolism , Vibrio vulnificus/pathogenicity , Animals , Bacterial Proteins/chemistry , Bacterial Proteins/metabolism , Binding Sites/drug effects , CHO Cells , Cholesterol/metabolism , Cricetulus , Glycomics/methods , Hemolysin Proteins/chemistry , Microarray Analysis , Protein Binding/drug effects , Protein Conformation , Protein Multimerization/drug effects , Vibrio vulnificus/metabolism
17.
Anaerobe ; 57: 86-89, 2019 Jun.
Article En | MEDLINE | ID: mdl-30951830

Aspiration pneumonia is an infectious disease of the lungs caused by inhalation of saliva or foods, associated with swallowing dysfunction. Therefore, the major causative organisms are oral or gastric bacteria. In this study, we evaluated the antimicrobial susceptibility patterns of the anaerobic bacteria which can cause aspiration pneumonia, Fusobacterium spp., Finegoldia magna, Bacteroides fragilis, Peptostreptococcus spp., Prevotella spp., and Streptococcus milleri group to ceftriaxone, cefmetazole, flomoxef, ampicillin/sulbactam, and ampicillin. We also tested the ß-lactamase activities of each of the bacterial strains. Fusobacterium spp. and Finegoldia magna were susceptible to all of the tested antimicrobial drugs, except ampicillin, and showed no ß-lactamase activity. The Streptococcus milleri group, Bacteroides fragilis, and Peptostreptococcus spp. showed decreased susceptibility to cefmetazole or flomoxef as compared to the susceptibility levels documented in a previous report. There was one strain of Peptostreptococcus anaerobius which was not susceptible to ampicillin/sulbactam, but also showed no ß-lactamase activity, suggesting that this strain harbored a mechanism of resistance other than the production of ß-lactamase. The susceptibility of Prevotella spp. to ceftriaxone was also decreased as compared to the susceptibility level documented in a previous report. Furthermore, ß-lactamase-positive strains were found even among ceftriaxone-susceptible strains. Elderly persons with swallowing dysfunction carry a risk of recurrent episodes of aspiration pneumonia and repeated use of antibiotics increases the risk of development of antibiotic resistance. In the present study, the antibiotic susceptibilities of some of organisms which can cause aspiration pneumonia were found to be decreased as compared to the susceptibility levels documented in a previous report. Therefore, surveillance of the antimicrobial susceptibility patterns of these bacteria is recommended to prevent the development of resistance.


Anti-Bacterial Agents/pharmacology , Bacteria, Anaerobic/drug effects , Bacteria, Anaerobic/isolation & purification , Bacterial Infections/microbiology , Microbial Sensitivity Tests , Pneumonia, Aspiration/microbiology , Humans
18.
Calcif Tissue Int ; 103(3): 289-297, 2018 09.
Article En | MEDLINE | ID: mdl-29725706

Abaloparatide (ABL) is a novel synthetic peptide analog of parathyroid hormone-related protein. In previous reports, intermittent ABL administration showed robust bone mineral density (BMD) increase and reduced the incidence of fractures in patients with osteoporosis, while its calcemic effect was reduced, as compared with teriparatide (TPTD), a parathyroid hormone N-terminal fragment. The present study aimed to elucidate the effects of ABL on bone anabolism and bone turnover as compared with TPTD. In ovariectomized (OVX) rats, ABL increased the bone strength and BMD of lumbar spine by intermittent administration similar to TPTD. Both ABL and TPTD increased the bone formation marker serum P1NP with little effect on the bone resorption maker urine DPD/Cr, suggesting anabolic effects on bone. In human osteoblastic cells, both peptides increased the expression of bone resorption-related factors such as RANKL/OPG and M-CSF, and the effects of ABL were significantly attenuated as compared with those of TPTD under transient 6-h treatment, although no significant differences were found under continuous treatment. In contrast, ABL and TPTD similarly promoted the expression of bone formation-related factors, IGF-1 and osteocalcin. In addition, there were no significant differences in the effects on WNT signaling inhibitors such as sclerostin and dickkopf-related protein 1 (DKK1) between the two peptides. These results demonstrate that ABL exerts bone anabolic effects in OVX rats. It is also indicated that ABL stimulates the expression of RANKL/OPG and M-CSF less than TPTD, while showing similar effects on bone formation-related factors and WNT signaling inhibitors in vitro. The profile of ABL indicates that it would be a suitable bone anabolic agent for osteoporosis.


Bone Density Conservation Agents/pharmacology , Bone Remodeling/drug effects , Bone and Bones/drug effects , Parathyroid Hormone-Related Protein/pharmacology , Teriparatide/pharmacology , Anabolic Agents/pharmacology , Animals , Cell Line , Female , Humans , Rats , Rats, Sprague-Dawley
19.
J Obstet Gynaecol ; 38(2): 226-230, 2018 Feb.
Article En | MEDLINE | ID: mdl-28920516

To investigate the involvement of ICAM-1 in the adhesion of Candida to the genitourinary epithelial cells in high glucose, we examined the adhesion of Candida albicans or Candida glabrata to human vaginal epithelial cells (VK2/E6E7) or human vulvovaginal epidermal cells (A431). These cells were cultured in 100, 500 or 3000 mg/dL glucose for three days and inoculated with Candida for 60 minutes. Followed by, adhering of Candida to the cells, which were counted. While the adhesion of Candida albicans to VK2/E6E7 significantly increased in the high glucose, A431 did not. We next examined the expression of ICAM-1 as a ligand on the epithelial cells. ICAM-1 expression was increased in VK2/E6E7 cultured in the high glucose; however, the expression level in A431 was not high compared with VK2/E6E7. This data suggested that ICAM-1 functions as one of ligands in the adhesion of Candida albicans to the vaginal epithelial cells in a high glucose environment. Impact statement What is already known on the subject: Candida's complement receptor is involved in the adhesion to epithelial cells. The expression of this receptor has been reported to increase as glucose concentration increases. This is considered as a contributing factor to the high risk for vulvovaginal candidiasis (VVC) in diabetes. On the host side, diabetic patients have a factor that facilitates adhesion of Candida to epithelial cells. This factor has been unknown until recently. What the results of this study add: In this study, we used a vaginal epithelial cell line and showed that the adhesion of C. albicans to cells increased at higher glucose concentrations. At the same time, ICAM-1 expression of cells also increased. Thereby, it is suggested that the expression of ICAM-1 in vaginal epithelial cells is increased by glucose such as urinary sugar in diabetic patients and is a condition for facilitating adhesion of Candida. What the implications are of these findings for clinical practice and/or further research: We expect not only host immune dysfunction but also alteration in epithelial cells will be focussed on as a cause of VVC in diabetic patients.


Candida albicans/metabolism , Candida glabrata/metabolism , Candidiasis, Vulvovaginal/microbiology , Epithelial Cells/microbiology , Glucose/pharmacology , Intercellular Adhesion Molecule-1/metabolism , Vagina/microbiology , Blotting, Western , Candida albicans/isolation & purification , Candida glabrata/isolation & purification , Candidiasis, Vulvovaginal/etiology , Candidiasis, Vulvovaginal/genetics , Cell Culture Techniques , Diabetes Complications/genetics , Diabetes Complications/microbiology , Female , Gene Expression Regulation , Glucose/metabolism , Humans , Risk Factors , Tissue Adhesions/metabolism
20.
J Antibiot (Tokyo) ; 70(3): 264-271, 2017 Mar.
Article En | MEDLINE | ID: mdl-28074049

We characterized in vitro activities of α-methoxyimino acylides against macrolide-resistant clinical isolates of Streptococcus pneumoniae, Streptococcus pyogenes and Mycoplasma pneumoniae with ribosome modification or substitution and selected acylide-resistant mutants to clarify the binding point of the acylides. The acylides had low MICs against erm(B) gene-containing S. pneumoniae and S. pyogenes (MIC90s, 1-4 µg ml-1). For M. pneumoniae, although they had poor potencies against macrolide-resistant strains with the A2058G (Escherichia coli numbering) mutation in 23S rRNA (MICs, >32 µg ml-1), one of them showed in vitro activities against macrolide-resistant strains with the A2058U or A2059G mutations (MICs, 0.5-1 µg ml-1). These A2058U and A2059G mutant strains were used for the selection of acylide-resistant mutants. A genetic analysis showed that new point mutations in acylide-resistant mutants were found at G2576 in domain V of 23S rRNA and at Lys90 in L22 ribosomal protein. Furthermore, a molecular modeling study revealed that G2505/C2610, which enables stacking with G2576, might interact with a pyridyl moiety or an α-methoxyimino group at the 3-position of acylides. The α-methoxyimino acylides were shown to possess a tertiary binding point at G2505/C2610 in 23S rRNA. Our results suggest that α-methoxyimino acylides represent significant progress in macrolide antimicrobials.


Anti-Bacterial Agents/pharmacology , Drug Resistance, Bacterial/drug effects , Drug Resistance, Bacterial/genetics , RNA, Ribosomal, 23S/genetics , Macrolides/pharmacology , Microbial Sensitivity Tests , Models, Molecular , Mutation , Mycoplasma pneumoniae/drug effects , Mycoplasma pneumoniae/genetics , Streptococcus pneumoniae/drug effects , Streptococcus pneumoniae/genetics , Streptococcus pyogenes/drug effects , Streptococcus pyogenes/genetics
...