Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 3.394
Filter
1.
World J Microbiol Biotechnol ; 40(10): 316, 2024 Sep 09.
Article in English | MEDLINE | ID: mdl-39249607

ABSTRACT

Istamycins (ISMs) are 2-deoxyfortamine-containing aminoglycoside antibiotics (AGAs) produced by Streptomyces tenjimariensis ATCC 31603 with broad-spectrum bactericidal activities against most of the clinically relevant pathogens. Therefore, this study aimed to statistically optimize the environmental conditions affecting ISMs production using the central composite design (CCD). Both the effect of culture media composition and incubation time and agitation rate were studied as one factor at the time (OFAT). The results showed that both the aminoglycoside production medium and the protoplast regeneration medium gave the highest specific productivity. Results also showed that 6 days incubation time and 200 rpm agitation were optimum for their production. A CCD quadratic model of 17 runs was employed to test three key variables: initial pH, incubation temperature, and concentration of calcium carbonate. A significant statistical model was obtained including, an initial pH of 6.38, incubation temperature of 30 ˚C, and 5.3% CaCO3 concentration. This model was verified experimentally in the lab and resulted in a 31-fold increase as compared to the unoptimized conditions and a threefold increase to that generated by using the optimized culture media. To our knowledge, this is the first report about studying environmental conditions affecting ISM production as OFAT and through CCD design of the response surface methodology (RSM) employed for statistical optimization. In conclusion, the CCD design is an effective tool for optimizing ISMs at the shake flask level. However, the optimized conditions generated using the CCD model in this study should be scaled up in a fermenter for industrial production of ISMs by S. tenjimariensis ATCC 31603 considering the studied environmental conditions that significantly influence the production proces.


Subject(s)
Anti-Bacterial Agents , Culture Media , Fermentation , Streptomyces , Temperature , Streptomyces/metabolism , Streptomyces/growth & development , Culture Media/chemistry , Hydrogen-Ion Concentration , Anti-Bacterial Agents/biosynthesis , Anti-Bacterial Agents/pharmacology , Calcium Carbonate/metabolism , Aminoglycosides/pharmacology , Industrial Microbiology , Bioreactors/microbiology
2.
Sci Rep ; 14(1): 20713, 2024 09 05.
Article in English | MEDLINE | ID: mdl-39237684

ABSTRACT

Lidamycin (LDM) has been confirmed to have a strong anti-pancreatic cancer effect and can affect the mitochondrial function of pancreatic cancer cells. Mitofusin-2 (Mfn2) is located in the outer membrane of mitochondria, and Mfn2 is currently believed to play a role in cancer inhibition in pancreatic cancer. In order to explore whether the anti-pancreatic cancer effect of LDM is related to Mfn2-mediated mitophagy, Bioinformatics and in vitro cell experiments are used for experimental research. The experimental results demonstrated that Mfn2 is correlated with mitochondrial autophagy in pancreatic cancer. Lidamycin can increase the expression of Mfn2 in pancreatic cancer and affect the process of EMT, affect the level of reactive oxygen species and mitochondrial membrane potential, and increase the expression of mitochondrial autophagy marker proteins BNIP3L and Beclin1. These results demonstrate that Mfn2 affects mitophagy in pancreatic cancer cells by regulating the expression of Mfn2.


Subject(s)
GTP Phosphohydrolases , Membrane Proteins , Mitochondrial Proteins , Mitophagy , Pancreatic Neoplasms , Humans , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/genetics , Mitophagy/drug effects , GTP Phosphohydrolases/metabolism , GTP Phosphohydrolases/genetics , Cell Line, Tumor , Mitochondrial Proteins/metabolism , Mitochondrial Proteins/genetics , Membrane Proteins/metabolism , Membrane Proteins/genetics , Aminoglycosides/pharmacology , Gene Expression Regulation, Neoplastic/drug effects , Reactive Oxygen Species/metabolism , Mitochondria/metabolism , Mitochondria/drug effects , Membrane Potential, Mitochondrial/drug effects , Beclin-1/metabolism , Beclin-1/genetics , Proto-Oncogene Proteins/metabolism , Tumor Suppressor Proteins
3.
Microbiol Spectr ; 12(10): e0135624, 2024 Oct 03.
Article in English | MEDLINE | ID: mdl-39162491

ABSTRACT

We evaluated the in vivo activity of nikkomycin Z against central nervous system coccidioidomycosis. Mice were inoculated intracranially with arthroconidia of Coccidoides immitis, and treatment with nikkomycin Z (50, 100, or 300 mg/kg orally TID) or fluconazole (25 mg/kg orally BID) began 2 days later. Each dose of nikkomycin Z and fluconazole significantly improved survival and reduced brain fungal burden compared with vehicle control. Further studies of nikkomycin Z against coccidioidomycosis are warranted. IMPORTANCE: Coccidioides species are endemic fungi that are capable of causing disease in patients with various comorbidities, as well as in otherwise healthy individuals. Treatment options for coccidioidomycosis are suboptimal, as azole antifungals may be limited by drug interactions and adverse effects due to interactions with enzymes found in humans and other mammals. Nikkomycin Z is an investigational agent that works against a target specific to the fungal cell wall (chitin), which is not present in the cells of humans or other mammals. In this study, we show that frequent oral administration of nikkomycin Z is effective in an experimental model of central nervous system coccidioidomycosis. Further studies of nikkomycin Z against coccidioidomycosis may be warranted.


Subject(s)
Aminoglycosides , Antifungal Agents , Coccidioides , Coccidioidomycosis , Disease Models, Animal , Animals , Mice , Aminoglycosides/administration & dosage , Aminoglycosides/pharmacology , Administration, Oral , Antifungal Agents/administration & dosage , Antifungal Agents/pharmacology , Coccidioidomycosis/drug therapy , Coccidioidomycosis/microbiology , Coccidioides/drug effects , Fluconazole/administration & dosage , Fluconazole/pharmacology , Brain/microbiology , Central Nervous System Fungal Infections/drug therapy , Central Nervous System Fungal Infections/microbiology , Humans
4.
Microbiol Spectr ; 12(10): e0387623, 2024 Oct 03.
Article in English | MEDLINE | ID: mdl-39162554

ABSTRACT

Metallo-beta-lactamase (MBL)-producing carbapenem-resistant Enterobacteriaceae (CRE) infections continue to pose a serious threat to healthcare. Due to their unique active site, MBLs evade the activity of many novel beta-lactam/beta-lactamase inhibitor combinations, which have been specifically targeted toward those carbapenemases with serine active sites. Furthermore, resistance to most, if not all, other clinically relevant antimicrobial classes leaves few reliable therapeutic options. Combination therapy has thus played a vital role in the treatment of MBL-producing CRE infections. In this study, we utilized the static time-kill assay to investigate clinically relevant concentrations of cefepime, piperacillin-tazobactam, and meropenem alone and in combination with either amikacin or the novel plazomicin to determine if combinations of routinely used beta-lactam therapy with an aminoglycoside would achieve bactericidal activity against eight clinically isolated Verona integron-encoded MBL (VIM)-producing CRE. Furthermore, we compared this activity to the combination of aztreonam/avibactam, which has shown potent activity against MBL-producing CRE. Both aztreonam/avibactam and meropenem with either aminoglycoside were rapidly bactericidal within 4 hours and remained bactericidal through 24 hours against all isolates with few exceptions. Combinations including cefepime and piperacillin-tazobactam were also rapidly bactericidal, but activity after 24 hours was inconsistent depending upon the partner aminoglycoside and isolate. Further investigation is warranted to elucidate optimal antibiotic exposures against MBL-producing CRE, including novel agents in the pipeline.IMPORTANCECarbapenem-resistant Enterobacterales (CRE) are one of the most pressing antimicrobial-resistant threats at present. In addition to exhibiting resistance to many, if not all, commonly used antimicrobial agents, CRE achieves these resistant phenotypes through a variety of mechanisms, each of which can uniquely affect available treatment options. The present study is an in vitro investigation of several Verona integron-encoded metallo-beta-lactamase (VIM)-producing CRE isolated from patients at our academic medical center. Because metallo-beta-lactamases (MBLs) are inherently resistant to many of the novel treatments designed to treat CRE due to their different active site composition, we tested several antimicrobial combinations containing routinely utilized broad-spectrum beta-lactams and aminoglycosides. Our results further our understanding of combination therapy options against VIM-producing CRE, including with non-carbapenem-beta-lactams cefepime and piperacillin. By optimizing combinations of existing antimicrobial agents, we hope to expand the available armamentarium against these resistant pathogens.


Subject(s)
Aminoglycosides , Anti-Bacterial Agents , Microbial Sensitivity Tests , beta-Lactamases , beta-Lactams , Anti-Bacterial Agents/pharmacology , beta-Lactamases/metabolism , beta-Lactamases/genetics , beta-Lactams/pharmacology , Humans , Aminoglycosides/pharmacology , Enterobacteriaceae/drug effects , Enterobacteriaceae/enzymology , Enterobacteriaceae/genetics , Enterobacteriaceae Infections/microbiology , Enterobacteriaceae Infections/drug therapy , Carbapenem-Resistant Enterobacteriaceae/drug effects , Carbapenem-Resistant Enterobacteriaceae/genetics , Meropenem/pharmacology , Piperacillin, Tazobactam Drug Combination/pharmacology , Cefepime/pharmacology , Amikacin/pharmacology , beta-Lactamase Inhibitors/pharmacology , Sisomicin/analogs & derivatives , Sisomicin/pharmacology , Bacterial Proteins/metabolism , Bacterial Proteins/genetics
5.
Microbiol Spectr ; 12(10): e0064724, 2024 Oct 03.
Article in English | MEDLINE | ID: mdl-39191399

ABSTRACT

Bone and soft tissue infections caused by biofilm-forming bacteria, such as methicillin-resistant Staphylococcus aureus (MRSA), remain a significant clinical challenge. While the control of local infection is necessary, systemic treatment is also required, and biofilm eradication is a critical target for successful management. Topical antibiotic treatments, such as antibiotic-loaded bone cement (ALBC), have been used for some time, and continuous local antibiotic perfusion therapy, a less invasive method, has been developed by our group. However, the optimal antibiotics and concentrations for biofilms of clinical isolates are still not well understood. We examined the efficacy of high concentrations of gentamicin against MRSA biofilms and the role of gentamicin resistance genes in biofilm eradication. We collected 101 MRSA samples from a hospital in Japan and analyzed their gene properties, including methicillin and gentamicin resistance, and their minimum biofilm eradication concentration (MBEC) values. Our results showed that high concentrations of gentamicin are effective against MRSA biofilms and that even concentrations lower than the MBEC value could eliminate biofilms after prolonged exposure. We also identified three aminoglycoside/gentamicin resistance genes [aac(6')-aph(2″), aph(3')-III, and ant(4')-IA] and found that the presence or absence of these genes may inform the selection of treatments. It was also found that possession of the aac(6')-aph(2″) gene correlated with the minimum inhibitory concentration/MBEC values of gentamicin. Although this study provides insight into the efficacy of gentamicin against MRSA biofilms and the role of gentamicin resistance genes, careful selection of the optimal treatment strategy is needed for clinical application. IMPORTANCE: Our analysis of 101 MRSA clinical isolates has provided valuable insights that could enhance treatment selection for biofilm infections in orthopedics. We found that high concentrations of gentamicin were effective against MRSA biofilms, and even prolonged exposure to concentrations lower than the minimum biofilm eradication concentration (MBEC) value could eliminate biofilms. The presence of the aac(6')-aph(2″) gene, an aminoglycoside resistance gene, was found to correlate with the minimum inhibitory concentration (MIC) and MBEC values of gentamicin, providing a potential predictive tool for treatment susceptibility. These results suggest that extended high concentrations of local gentamicin treatment could effectively eliminate MRSA biofilms in orthopedic infections. Furthermore, testing for gentamicin MIC or the possession of the aac(6')-aph(2″) gene could help select treatment, including topical gentamicin administration and surgical debridement.


Subject(s)
Aminoglycosides , Anti-Bacterial Agents , Biofilms , Gentamicins , Methicillin-Resistant Staphylococcus aureus , Microbial Sensitivity Tests , Staphylococcal Infections , Biofilms/drug effects , Biofilms/growth & development , Gentamicins/pharmacology , Methicillin-Resistant Staphylococcus aureus/drug effects , Methicillin-Resistant Staphylococcus aureus/genetics , Methicillin-Resistant Staphylococcus aureus/isolation & purification , Humans , Anti-Bacterial Agents/pharmacology , Staphylococcal Infections/microbiology , Staphylococcal Infections/drug therapy , Aminoglycosides/pharmacology , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Drug Resistance, Bacterial/genetics , Japan
6.
Biomedica ; 44(2): 182-190, 2024 05 30.
Article in English, Spanish | MEDLINE | ID: mdl-39088528

ABSTRACT

Introduction: The Mycobacterium chelonae species and the M. avium and M. abscessus complexes are emerging pathogens that cause mycobacteriosis. Treatment depends on the species and subspecies identified. The drugs of choice are macrolides and aminoglycosides. However, due to the resistance identified to these drugs, determining the microbe's sensitivity profile will allow clinicians to improve the understanding of the prognosis and evolution of these pathologies. Objective: To describe the macrolide and aminoglycoside susceptibility profile of cultures identified by Colombia's Laboratorio Nacional de Referencia de Mycobacteria from 2018 to 2022, as Mycobacterium avium complex, M. abscessus complex, and M. chelonae. Materials and methods. This descriptive study exposes the susceptibility profile to macrolides and aminoglycosides of cultures identified as M. avium complex, M. abscessus complex, and M. chelonae using the GenoType® NTM-DR method. Materials and methods: This descriptive study exposes the susceptibility profile to macrolides and aminoglycosides of cultures identified as M. avium complex, M. abscessus complex, and M. chelonae using the GenoType® NTM-DR method. Results: We identified 159 (47.3 %) cultures as M. avium complex, of which 154 (96.9 %) were sensitive to macrolides, and 5 (3.1 %) were resistant; all were sensitive to aminoglycosides. From the 125 (37.2 %) cultures identified as M. abscessus complex, 68 (54.4 %) were sensitive to macrolides, 57 (45.6 %) were resistant to aminoglycosides, and just one (0.8 %) showed resistance to aminoglycosides. The 52 cultures (15.5 %) identified as M. chelonae were sensitive to macrolides and aminoglycosides. Conclusions: The three studied species of mycobacteria have the least resistance to Amikacin. Subspecies identification and their susceptibility profiles allow the establishment of appropriate treatment schemes, especially against M. abscessus.


Introducción. Mycobacterium chelonae y los complejos Mycobacterium avium y M. abscessus, son agentes patógenos emergentes causantes de micobacteriosis. El tratamiento de esta infección depende de la especie y la subespecie identificadas. Los fármacos de elección son los macrólidos y aminoglucósidos, contra los cuales se ha reportado resistencia; por esta razón, el determinar el perfil de sensibilidad le permite al médico tratante comprender mejor el pronóstico y la evolución de estas infecciones. Objetivo. Describir los perfiles de sensibilidad ante macrólidos y aminoglucósidos, de los cultivos identificados como complejo Mycobacterium avium, complejo M. abscessus o especie M. chelonae, en el Laboratorio Nacional de Referencia de Micobacterias durante los años 2018 a 2022. Materiales y métodos. Se llevó a cabo un estudio descriptivo del perfil de sensibilidad a macrólidos y aminoglucósidos, de los cultivos identificados como complejo M. avium, complejo M. abscessus o M. chelonae, mediante la metodología GenoType® NTM-DR. Resultados. Los cultivos del complejo M. avium fueron 159 (47,3 %), de los cuales, 154 (96,9 %) fueron sensibles y 5 (3,1 %) resistentes a los macrólidos; todos fueron sensibles a los aminoglucósidos. Del complejo M. abscessus se estudiaron 125 (37,2 %) cultivos, 68 (54,4 %) resultaron sensibles y 57 (45,6 %) resistentes a los macrólidos; solo un cultivo (0,8 %) fue resistente a los aminoglucósidos. De M. chelonae se analizaron 52 cultivos (15,5 %), todos sensibles a los macrólidos y aminoglucósidos. Conclusiones. En las tres especies de micobacterias estudiadas, la resistencia contra la amikacina fue la menos frecuente. La identificación de las subespecies y los perfiles de sensibilidad permiten instaurar esquemas de tratamiento adecuados, especialmente en las micobacteriosis causadas por M. abscessus.


Subject(s)
Aminoglycosides , Macrolides , Microbial Sensitivity Tests , Mycobacterium Infections, Nontuberculous , Mycobacterium abscessus , Mycobacterium avium Complex , Mycobacterium chelonae , Macrolides/pharmacology , Mycobacterium abscessus/drug effects , Mycobacterium abscessus/genetics , Mycobacterium abscessus/isolation & purification , Colombia/epidemiology , Mycobacterium chelonae/drug effects , Mycobacterium chelonae/genetics , Mycobacterium chelonae/isolation & purification , Aminoglycosides/pharmacology , Humans , Mycobacterium avium Complex/drug effects , Mycobacterium avium Complex/genetics , Mycobacterium avium Complex/isolation & purification , Mycobacterium Infections, Nontuberculous/microbiology , Mycobacterium Infections, Nontuberculous/epidemiology , Mycobacterium Infections, Nontuberculous/drug therapy , Anti-Bacterial Agents/pharmacology , Drug Resistance, Bacterial , Prevalence , Drug Resistance, Multiple, Bacterial
7.
Cells ; 13(15)2024 Jul 29.
Article in English | MEDLINE | ID: mdl-39120305

ABSTRACT

Transient receptor potential (TRP) channels are broadly implicated in the developmental programs of most tissues. Amongst these tissues, skeletal muscle and adipose are noteworthy for being essential in establishing systemic metabolic balance. TRP channels respond to environmental stimuli by supplying intracellular calcium that instigates enzymatic cascades of developmental consequence and often impinge on mitochondrial function and biogenesis. Critically, aminoglycoside antibiotics (AGAs) have been shown to block the capacity of TRP channels to conduct calcium entry into the cell in response to a wide range of developmental stimuli of a biophysical nature, including mechanical, electromagnetic, thermal, and chemical. Paradoxically, in vitro paradigms commonly used to understand organismal muscle and adipose development may have been led astray by the conventional use of streptomycin, an AGA, to help prevent bacterial contamination. Accordingly, streptomycin has been shown to disrupt both in vitro and in vivo myogenesis, as well as the phenotypic switch of white adipose into beige thermogenic status. In vivo, streptomycin has been shown to disrupt TRP-mediated calcium-dependent exercise adaptations of importance to systemic metabolism. Alternatively, streptomycin has also been used to curb detrimental levels of calcium leakage into dystrophic skeletal muscle through aberrantly gated TRPC1 channels that have been shown to be involved in the etiology of X-linked muscular dystrophies. TRP channels susceptible to AGA antagonism are critically involved in modulating the development of muscle and adipose tissues that, if administered to behaving animals, may translate to systemwide metabolic disruption. Regenerative medicine and clinical communities need to be made aware of this caveat of AGA usage and seek viable alternatives, to prevent contamination or infection in in vitro and in vivo paradigms, respectively.


Subject(s)
Aminoglycosides , Anti-Bacterial Agents , Transient Receptor Potential Channels , Humans , Animals , Anti-Bacterial Agents/pharmacology , Transient Receptor Potential Channels/metabolism , Aminoglycosides/pharmacology , Muscle, Skeletal/metabolism , Muscle, Skeletal/drug effects , Adipose Tissue/metabolism , Adipose Tissue/drug effects
8.
Elife ; 132024 Aug 02.
Article in English | MEDLINE | ID: mdl-39093940

ABSTRACT

Aminoglycoside antibiotics target ribosomes and are effective against a wide range of bacteria. Here, we demonstrated that knockout strains related to energy metabolism in Escherichia coli showed increased tolerance to aminoglycosides during the mid-exponential growth phase. Contrary to expectations, these mutations did not reduce the proton motive force or aminoglycoside uptake, as there were no significant changes in metabolic indicators or intracellular gentamicin levels between wild-type and mutant strains. Our comprehensive proteomics analysis unveiled a noteworthy upregulation of proteins linked to the tricarboxylic acid (TCA) cycle in the mutant strains during the mid-exponential growth phase, suggesting that these strains compensate for the perturbation in their energy metabolism by increasing TCA cycle activity to maintain their membrane potential and ATP levels. Furthermore, our pathway enrichment analysis shed light on local network clusters displaying downregulation across all mutant strains, which were associated with both large and small ribosomal binding proteins, ribosome biogenesis, translation factor activity, and the biosynthesis of ribonucleoside monophosphates. These findings offer a plausible explanation for the observed tolerance of aminoglycosides in the mutant strains. Altogether, this research provides valuable insights into the mechanisms of aminoglycoside tolerance, paving the way for novel strategies to combat such cells.


Bacteria that are resistant to antibiotic drugs pose a significant challenge to human health around the globe. They have acquired genetic mutations that allow them to survive and grow in the presence of one or more antibiotics, making it harder for clinicians to eliminate such bacteria from human patients with life-threatening infections. Some bacteria may be able to temporarily develop tolerance to an antibiotic by altering how they grow and behave, without acquiring any new genetic mutations. Such drug-tolerant bacteria are more likely to survive long enough to gain mutations that may promote drug resistance. Recent studies suggest that genes involved in processes collectively known as energy metabolism, which convert food sources into the chemical energy cells need to survive and grow, may play a role in both tolerance and resistance. For example, Escherichia coli bacteria develop mutations in energy metabolism genes when exposed to members of a family of antibiotics known as the aminoglycosides. However, it remains unclear what exact role energy metabolism plays in antibiotic tolerance. To address this question, Shiraliyev and Orman studied how a range of E. coli strains with different genetic mutations affecting energy metabolism could survive in the presence of aminoglycosides. The experiments found that most of the mutant strains had a higher tolerance to the drugs than normal E. coli. Unexpectedly, this increased tolerance did not appear to be due to the drugs entering the mutant bacterium cells less than they enter normal cells (a common strategy of drug resistance and tolerance). Further experiments using a technique, known as proteomics, revealed that many genes involved in energy metabolism were upregulated in the mutant bacteria, suggesting these cells were compensating for the genetic abnormalities they have. Furthermore, the mutant bacteria had lower levels of the molecules the antibiotics target than normal bacteria. The findings of Shiraliyev and Orman offer critical insights into how bacteria become tolerant of aminoglycoside antibiotics. In the future, this may guide the development of new strategies to combat bacterial diseases.


Subject(s)
Aminoglycosides , Anti-Bacterial Agents , Escherichia coli , Ribosomal Proteins , Ribosomal Proteins/metabolism , Ribosomal Proteins/genetics , Aminoglycosides/pharmacology , Anti-Bacterial Agents/pharmacology , Escherichia coli/genetics , Escherichia coli/drug effects , Escherichia coli/metabolism , Energy Metabolism/drug effects , Escherichia coli Proteins/metabolism , Escherichia coli Proteins/genetics , Drug Tolerance , Proteomics , Citric Acid Cycle/drug effects
9.
Front Cell Infect Microbiol ; 14: 1435123, 2024.
Article in English | MEDLINE | ID: mdl-39139766

ABSTRACT

Background: Aminoglycoside-modifying enzymes (AMEs) play an essential role in bacterial resistance to aminoglycoside antimicrobials. With the development of sequencing techniques, more bacterial genomes have been sequenced, which has aided in the discovery of an increasing number of novel resistance mechanisms. Methods: The bacterial species was identified by 16S rRNA gene homology and average nucleotide identity (ANI) analyses. The minimum inhibitory concentration (MIC) of each antimicrobial was determined by the agar dilution method. The protein was expressed with the pCold I vector in E. coli BL21, and enzyme kinetic parameters were examined. The whole-genome sequence of the bacterium was obtained via the Illumina and PacBio sequencing platforms. Reconstruction of the phylogenetic tree, identification of conserved functional residues, and gene context analysis were performed using the corresponding bioinformatic techniques. Results: A novel aminoglycoside resistance gene, designated aph(3')-Ie, which confers resistance to ribostamycin, kanamycin, sisomicin and paromomycin, was identified in the chromosome of the animal bacterium Citrobacter gillenii DW61, which exhibited a multidrug resistance phenotype. APH(3')-Ie showed the highest amino acid identity of 74.90% with the functionally characterized enzyme APH(3')-Ia. Enzyme kinetics analysis demonstrated that it had phosphorylation activity toward four aminoglycoside substrates, exhibiting the highest affinity (K m, 4.22 ± 0.88 µM) and the highest catalytic efficiency [k cat/K m, (32.27 ± 8.14) × 104] for ribomycin. Similar to the other APH(3') proteins, APH(3')-Ie contained all the conserved functional sites of the APH family. The aph(3')-Ie homologous genes were present in C. gillenii isolates from different sources, including some of clinical significance. Conclusion: In this work, a novel chromosomal aminoglycoside resistance gene, designated aph(3')-Ie, conferring resistance to aminoglycoside antimicrobials, was identified in a rabbit isolate C. gillenii DW61. The elucidation of the novel resistance mechanism will aid in the effective treatment of infections caused by pathogens carrying such resistance genes.


Subject(s)
Aminoglycosides , Anti-Bacterial Agents , Citrobacter , Microbial Sensitivity Tests , Phylogeny , RNA, Ribosomal, 16S , Animals , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/metabolism , Citrobacter/enzymology , Citrobacter/genetics , Citrobacter/metabolism , Citrobacter/classification , Aminoglycosides/pharmacology , Aminoglycosides/metabolism , RNA, Ribosomal, 16S/genetics , Rabbits , Drug Resistance, Multiple, Bacterial/genetics , Genome, Bacterial , Whole Genome Sequencing , Sisomicin/pharmacology , Sisomicin/analogs & derivatives , Sisomicin/metabolism , Kanamycin Kinase/genetics , Kanamycin Kinase/metabolism , Ribostamycin/metabolism , Drug Resistance, Bacterial/genetics , Kanamycin/pharmacology , Escherichia coli/genetics , Escherichia coli/metabolism , Enterobacteriaceae Infections/microbiology
10.
Sci Transl Med ; 16(759): eadn2140, 2024 Aug 07.
Article in English | MEDLINE | ID: mdl-39110778

ABSTRACT

Hearing loss is a major health concern in our society, affecting more than 400 million people worldwide. Among the causes, aminoglycoside therapy can result in permanent hearing loss in 40% to 60% of patients receiving treatment, and despite these high numbers, no drug for preventing or treating this type of hearing loss has yet been approved by the US Food and Drug Administration. We have previously conducted high-throughput screenings of bioactive compounds, using zebrafish as our discovery platform, and identified piplartine as a potential therapeutic molecule. In the present study, we expanded this work and characterized piplartine's physicochemical and therapeutic properties. We showed that piplartine had a wide therapeutic window and neither induced nephrotoxicity in vivo in zebrafish nor interfered with aminoglycoside antibacterial activity. In addition, a fluorescence-based assay demonstrated that piplartine did not inhibit cytochrome C activity in microsomes. Coadministration of piplartine protected from kanamycin-induced hair cell loss in zebrafish and protected hearing function, outer hair cells, and presynaptic ribbons in a mouse model of kanamycin ototoxicity. Last, we investigated piplartine's mechanism of action by phospho-omics, immunoblotting, immunohistochemistry, and molecular dynamics experiments. We found an up-regulation of AKT1 signaling in the cochleas of mice cotreated with piplartine. Piplartine treatment normalized kanamycin-induced up-regulation of TRPV1 expression and modulated the gating properties of this receptor. Because aminoglycoside entrance to the inner ear is, in part, mediated by TRPV1, these results suggested that by regulating TRPV1 expression, piplartine blocked aminoglycoside's entrance, thereby preventing the long-term deleterious effects of aminoglycoside accumulation in the inner ear compartment.


Subject(s)
Aminoglycosides , Hearing Loss , TRPV Cation Channels , Zebrafish , Animals , TRPV Cation Channels/metabolism , Aminoglycosides/pharmacology , Hearing Loss/chemically induced , Hearing Loss/metabolism , Hearing Loss/prevention & control , Hearing Loss/pathology , Mice , Ototoxicity/metabolism , Kanamycin , Dioxolanes/pharmacology , Piperidones
11.
BMC Infect Dis ; 24(1): 680, 2024 Jul 09.
Article in English | MEDLINE | ID: mdl-38982386

ABSTRACT

BACKGROUND: Aminoglycosides have been a cornerstone of the treatment of nosocomial infections caused by Pseudomonas aeruginosa for over 80 years. However, escalating emergence of resistance poses a significant challenge. Therefore, this study aimed to investigate the prevailing patterns of aminoglycoside resistance among clinical isolates of P. aeruginosa in Iran; as well as the underlying resistance mechanisms observed in patients referred to Ardabil hospitals. METHODS: A total of 200 isolates from five hospitals were evaluated. The resistance profiles of P. aeruginosa isolates to tobramycin, amikacin, and netilmicin were determined using the disk diffusion method. The capacity of aminoglycoside-resistant isolates to form biofilms was assessed through a phenotypic assay, and the results were confirmed using the gene amplification technique. The presence of genes associated with aminoglycoside resistance was detected using polymerase chain reaction (PCR). Quantitative reverse transcription PCR (qRT-PCR) was performed to measure the expression levels of genes encoding the MexXY-OprM efflux pump and PhoPQ two-component system (TCS). RESULTS: The prevalence of aminoglycoside-resistant P. aeruginosa isolates was 48%, with 94.7% demonstrating multidrug resistance (MDR). All aminoglycoside-resistant P. aeruginosa strains exhibited biofilm-forming capabilities and harbored all the genes associated with biofilm production. Among the nine genes encoding 16S rRNA methylase and aminoglycoside-modifying enzymes, three genes were detected in these isolates: aac(6')-Ib (85.4%), ant(2'')-Ia (18.7%), and aph(3')-VI (3.1%). Additionally, all aminoglycoside-resistant P. aeruginosa isolates carried mexY and phoP genes, although the expression levels of mexY and phoP were 75% and 87.5%, respectively. CONCLUSION: Given the considerably high prevalence of aminoglycoside-resistant P. aeruginosa strains, urgent measures are warranted to transition towards the use of novel aminoglycosides and to uphold vigilant surveillance of resistance patterns.


Subject(s)
Aminoglycosides , Anti-Bacterial Agents , Biofilms , Microbial Sensitivity Tests , Pseudomonas Infections , Pseudomonas aeruginosa , Pseudomonas aeruginosa/drug effects , Pseudomonas aeruginosa/genetics , Pseudomonas aeruginosa/isolation & purification , Iran/epidemiology , Humans , Aminoglycosides/pharmacology , Anti-Bacterial Agents/pharmacology , Pseudomonas Infections/microbiology , Pseudomonas Infections/epidemiology , Biofilms/drug effects , Biofilms/growth & development , Prevalence , Drug Resistance, Multiple, Bacterial/genetics , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Drug Resistance, Bacterial/genetics , Amikacin/pharmacology , Cross Infection/microbiology , Cross Infection/epidemiology , Tobramycin/pharmacology
12.
J Appl Microbiol ; 135(7)2024 Jul 02.
Article in English | MEDLINE | ID: mdl-38955378

ABSTRACT

AIMS: This study was conducted to evaluate the in vitro activity of clinically relevant aminoglycosides and to determine the prevalence of genes encoding aminoglycoside modifying enzymes (AMEs) and 16S ribosomal RNA (rRNA) methyltransferases among aminoglycoside-resistant E. coli (n = 61) and K. pneumoniae (n = 44) clinical isolates. Associated resistances to beta-lactams and their bla genes as well as the genetic relatedness of isolates were also investigated. MATERIALS AND METHODS: A total of 105 aminoglycoside-resistant E. coli (n = 61) and K. pneumoniae (n = 44) isolates recovered between March and May 2017 from 100 patients hospitalized in different wards of Charles Nicolle Hospital of Tunis, Tunisia, were studied. Minimal inhibitory concentrations of aminoglycoside compounds were determined by broth microdilution method. Aminoglycosides resistance encoding genes [aph(3´)-Ia, aph(3') IIa, aph(3´)-VIa, ant(2″)-Ia, aac(3)-IIa, aac(3)-IVa, aac(6')-Ib, rmtA, rmtB, rmtC, armA, and npmA] and bla genes were investigated by PCR and sequencing. Genetic relatedness was examined by multilocus sequence typing (MLST) for representative isolates. RESULTS: High rates of aminoglycoside resistance were found: gentamicin (85.7%), tobramycin (87.6%), kanamycin (78.0%), netilmincin (74.3%), and amikcin (18.0%). Most common AME gene was aac(3)-IIa (42%), followed by aac(6')-Ib (36.2%) and aph(3')-VIa (32.4%). The majority of isolates were resistant to beta-lactams and blaCTX-M-15 was the most common ESBL. The blaNDM-1 and blaOXA-48 were also produced by 1 and 23 isolates, respectively. Novel sequence types have been reported among our isolates and high-risk clonal lineages have been detected, such as E. coli ST43 (ST131 in Achtman MLST scheme) and K. pneumoniae (ST11/ST13). CONCLUSIONS: The high prevalence of aminoglycoside resistance rates and the diversity of corresponding genes, with diverse ß-lactamase enzymes among genetically heterogeneous clinical isolates present a matter of concern.


Subject(s)
Aminoglycosides , Anti-Bacterial Agents , Escherichia coli , Klebsiella pneumoniae , Microbial Sensitivity Tests , Aminoglycosides/pharmacology , Tunisia , Escherichia coli/genetics , Escherichia coli/drug effects , Escherichia coli/isolation & purification , Humans , Anti-Bacterial Agents/pharmacology , Klebsiella pneumoniae/genetics , Klebsiella pneumoniae/drug effects , Klebsiella pneumoniae/isolation & purification , Klebsiella pneumoniae/enzymology , Escherichia coli Infections/microbiology , Drug Resistance, Bacterial/genetics , Methyltransferases/genetics , Methyltransferases/metabolism , Klebsiella Infections/microbiology , beta-Lactamases/genetics , beta-Lactamases/metabolism , Bacterial Proteins/genetics , Bacterial Proteins/metabolism
13.
Microb Pathog ; 194: 106822, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39047802

ABSTRACT

Multidrug-resistant pathogens are now thought to be the primary global causes of disease and death. Therefore, it is imperative to develop new effective bioactive compounds from microbial sources, such as Streptomyces species. Nevertheless, the pharmaceutical industry suffered financial losses and low-quality end products as a result of Streptomyces bacteriophage contamination. To reduce the likelihood of phage-induced issues in the medical industry, it is crucial to develop a method for finding phage-resistant strains. Hence, we aimed to isolate and characterize Streptomyces spp. and Streptomyces phages from various rhizospheric soil samples in Egypt and to investigate their antibacterial activities. Moreover, we targeted development of a Streptomyces phage-resistant strain to extract its active metabolites and further testing its antibacterial activity. Herein, the antibacterial activities of the isolated 58 Streptomyces isolates showed that 10 (17.2 %) Streptomyces isolates had antibacterial activities against the tested bacteria including Listeria monocytogenes, E. coli O157, Acinetobacter baumannii, methicillin resistant-vancomycin-intermediate Staphylococcus aureus (MRSA-VISA) and Micrococcus luteus. Three lytic bacteriophages (ϕPRSC1, ϕPRSC2, and ϕPRSC4) belonging to the families Siphoviridae and Podoviridae were obtained from the rhizospheric soil samples using the most potent S. abietis isolate as the host strain. The three isolated Streptomyces phages were thermostable, ultraviolet stable, infectious, and had a wide range of hosts against the 10 tested Streptomyces isolates with antibacterial activities. The DNA of the ϕPRSC1 and ϕPRSC4 phages were resistant to digestion by EcoRI and HindIII, but the DNA of ϕPRSC2 was resistant to digestion by EcoRI and sensitive to digestion by HindIII. Of note, we developed a S. abietis strain resistant to the three isolated phages and its antibacterial activities were twice that of the wild strain. Finally, telomycin was recognized as an antibacterial metabolite extracted from phage-resistant S. abietis strain, which was potent against the tested Gram-positive bacteria including L. monocytogenes, MRSA-VISA, and M. luteus. Thus, our findings open new horizons for researching substitute antimicrobial medications for both existing and reemerging illnesses.


Subject(s)
Anti-Bacterial Agents , Soil Microbiology , Streptomyces , Streptomyces/virology , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/isolation & purification , Egypt , Podoviridae/isolation & purification , Siphoviridae/isolation & purification , Siphoviridae/genetics , Bacteriophages/isolation & purification , Methicillin-Resistant Staphylococcus aureus/drug effects , Methicillin-Resistant Staphylococcus aureus/virology , Listeria monocytogenes/drug effects , Listeria monocytogenes/virology , Micrococcus luteus/drug effects , Acinetobacter baumannii/drug effects , Acinetobacter baumannii/virology , Aminoglycosides/pharmacology , Microbial Sensitivity Tests , Rhizosphere
14.
Nature ; 632(8023): 39-49, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39085542

ABSTRACT

In this Review, we explore natural product antibiotics that do more than simply inhibit an active site of an essential enzyme. We review these compounds to provide inspiration for the design of much-needed new antibacterial agents, and examine the complex mechanisms that have evolved to effectively target bacteria, including covalent binders, inhibitors of resistance, compounds that utilize self-promoted entry, those that evade resistance, prodrugs, target corrupters, inhibitors of 'undruggable' targets, compounds that form supramolecular complexes, and selective membrane-acting agents. These are exemplified by ß-lactams that bind covalently to inhibit transpeptidases and ß-lactamases, siderophore chimeras that hijack import mechanisms to smuggle antibiotics into the cell, compounds that are activated by bacterial enzymes to produce reactive molecules, and antibiotics such as aminoglycosides that corrupt, rather than merely inhibit, their targets. Some of these mechanisms are highly sophisticated, such as the preformed ß-strands of darobactins that target the undruggable ß-barrel chaperone BamA, or teixobactin, which binds to a precursor of peptidoglycan and then forms a supramolecular structure that damages the membrane, impeding the emergence of resistance. Many of the compounds exhibit more than one notable feature, such as resistance evasion and target corruption. Understanding the surprising complexity of the best antimicrobial compounds provides a roadmap for developing novel compounds to address the antimicrobial resistance crisis by mining for new natural products and inspiring us to design similarly sophisticated antibiotics.


Subject(s)
Anti-Bacterial Agents , Bacteria , Biological Products , Animals , Humans , Aminoglycosides/pharmacology , Aminoglycosides/chemistry , Aminoglycosides/metabolism , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/metabolism , Bacteria/drug effects , Bacteria/enzymology , Bacteria/metabolism , beta Lactam Antibiotics/chemistry , beta Lactam Antibiotics/pharmacology , beta-Lactamase Inhibitors/chemistry , beta-Lactamase Inhibitors/pharmacology , Biological Products/chemistry , Biological Products/pharmacology , Biological Products/metabolism , Drug Design , Drug Resistance, Bacterial/drug effects , Peptidyl Transferases/antagonists & inhibitors , Prodrugs/pharmacology , Prodrugs/chemistry , Prodrugs/metabolism , Siderophores/metabolism , Siderophores/chemistry , Siderophores/pharmacology
15.
BMC Infect Dis ; 24(1): 763, 2024 Jul 31.
Article in English | MEDLINE | ID: mdl-39085804

ABSTRACT

BACKGROUND: One of the most prevalent bacteria that cause nosocomial infections is Pseudomonas aeruginosa. Fluoroquinolones (FQ) and aminoglycosides are vital antipseudomonal drugs, but resistance is increasingly prevalent. The study sought to investigate the diverse mechanisms underlying FQ and aminoglycoside resistance in various P. aeruginosa strains particularly during the COVID-19 crisis. METHODS: From various clinical and environmental samples, 110 P. aeruginosa isolates were identified and their susceptibility to several antibiotic classes was evaluated. Molecular techniques were used to track target gene mutations, the presence of genes encoding for quinolone resistance, modifying enzymes for aminoglycosides and resistance methyltransferase (RMT). Efflux pump role was assessed phenotypically and genotypically. Random amplified polymorphic DNA (RAPD) analysis was used to measure clonal diversity. RESULTS: QnrS was the most frequently encountered quinolone resistance gene (37.5%) followed by qnrA (31.2%) and qnrD (25%). Among aminoglycoside resistant isolates, 94.1% harbored modifying enzymes genes, while RMT genes were found in 55.9% of isolates. The aac(6')-Ib and rmtB were the most prevalent genes (79.4% and 32.3%, respectively). Most FQ resistant isolates overexpressed mexA (87.5%). RAPD fingerprinting showed 63.2% polymorphism. CONCLUSIONS: Aminoglycosides and FQ resistance observed in this study was attributed to several mechanisms with the potential for cross-contamination existence so, strict infection control practices are crucial.


Subject(s)
Aminoglycosides , Anti-Bacterial Agents , COVID-19 , Fluoroquinolones , Genotype , Microbial Sensitivity Tests , Phenotype , Pseudomonas Infections , Pseudomonas aeruginosa , Pseudomonas aeruginosa/genetics , Pseudomonas aeruginosa/drug effects , Pseudomonas aeruginosa/isolation & purification , Humans , Aminoglycosides/pharmacology , Egypt/epidemiology , COVID-19/epidemiology , Fluoroquinolones/pharmacology , Anti-Bacterial Agents/pharmacology , Pseudomonas Infections/microbiology , Pseudomonas Infections/epidemiology , SARS-CoV-2/genetics , SARS-CoV-2/drug effects , Drug Resistance, Bacterial/genetics , Hospitals , Random Amplified Polymorphic DNA Technique , Pandemics , Drug Resistance, Multiple, Bacterial/genetics
16.
J Mol Graph Model ; 131: 108817, 2024 Sep.
Article in English | MEDLINE | ID: mdl-38976944

ABSTRACT

The global antibiotic resistance problem necessitates fast and effective approaches to finding novel inhibitors to treat bacterial infections. In this study, we propose a computational workflow to identify plausible high-affinity compounds from FDA-approved, investigational, and experimental libraries for the decoding center on the small subunit 30S of the E. coli ribosome. The workflow basically consists of two molecular docking calculations on the intact 30S, followed by molecular dynamics (MD) simulations coupled with MM-GBSA calculations on a truncated ribosome structure. The parameters used in the molecular docking suits, Glide and AutoDock Vina, as well as in the MD simulations with Desmond were carefully adjusted to obtain expected interactions for the ligand-rRNA complexes. A filtering procedure was followed, considering a fingerprint based on aminoglycoside's binding site on the 30S to obtain seven hit compounds either with different clinical usages or aminoglycoside derivatives under investigation, suggested for in vitro studies. The detailed workflow developed in this study promises an effective and fast approach for the estimation of binding free energies of large protein-RNA and ligand complexes.


Subject(s)
Aminoglycosides , Escherichia coli , Molecular Docking Simulation , Molecular Dynamics Simulation , Ribosomes , Aminoglycosides/chemistry , Aminoglycosides/pharmacology , Escherichia coli/drug effects , Ribosomes/chemistry , Ribosomes/metabolism , Binding Sites , Ligands , Workflow , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/pharmacology
17.
Int J Mycobacteriol ; 13(2): 197-205, 2024 Apr 01.
Article in English | MEDLINE | ID: mdl-38916392

ABSTRACT

BACKGROUND: Tuberculosis (TB), a global infectious threat, has seen a concerning rise in aminoglycoside-resistant Mycobacterium tuberculosis (M.tb) strains. The potential role of capsule proteins remains largely unexplored. This layer acts as the primary barrier for tubercle bacilli, attempting to infiltrate host cells and subsequent disease development. METHODS: The study aims to bridge this gap by investigating the differentially expressed capsule proteins in aminoglycoside-resistant M.tb clinical isolates compared with drug-sensitive isolates employing two-dimensional gel electrophoresis, mass spectrometry, and bioinformatic approaches. RESULTS: We identified eight proteins that exhibited significant upregulation in aminoglycoside-resistant isolates. Protein Rv3029c and Rv2110c were associated with intermediary metabolism and respiration; Rv2462c with cell wall and cell processes; Rv3804c with lipid metabolism; Rv2416c and Rv2623 with virulence and detoxification/adaptation; Rv0020c with regulatory functions; and Rv0639 with information pathways. Notably, the Group-based Prediction System for Prokaryotic Ubiquitin-like Protein (GPS-PUP) algorithm identified potential pupylation sites within all proteins except Rv3804c. Interactome analysis using the STRING 12.0 database revealed potential interactive partners for these proteins, suggesting their involvement in aminoglycoside resistance. Molecular docking studies revealed suitable binding between amikacin and kanamycin drugs with Rv2462c, Rv3804c, and Rv2623 proteins. CONCLUSION: As a result, our findings illustrate the multifaceted nature of aminoglycoside resistance in M.tb and the importance of understanding how capsule proteins play a role in counteracting drug efficacy. Identifying the role of these proteins in drug resistance is crucial for developing more effective treatments and diagnostics for TB.


Subject(s)
Aminoglycosides , Bacterial Proteins , Drug Resistance, Bacterial , Mycobacterium tuberculosis , Proteomics , Mycobacterium tuberculosis/metabolism , Mycobacterium tuberculosis/drug effects , Mycobacterium tuberculosis/genetics , Humans , Bacterial Proteins/metabolism , Bacterial Proteins/genetics , Aminoglycosides/pharmacology , Bacterial Capsules/metabolism , Antitubercular Agents/pharmacology , Microbial Sensitivity Tests , Computational Biology , Electrophoresis, Gel, Two-Dimensional , Tuberculosis/microbiology
18.
Virulence ; 15(1): 2367647, 2024 Dec.
Article in English | MEDLINE | ID: mdl-38884466

ABSTRACT

The global surge in multidrug-resistant bacteria owing to antibiotic misuse and overuse poses considerable risks to human and animal health. With existing antibiotics losing their effectiveness and the protracted process of developing new antibiotics, urgent alternatives are imperative to curb disease spread. Notably, improving the bactericidal effect of antibiotics by using non-antibiotic substances has emerged as a viable strategy. Although reduced nicotinamide adenine dinucleotide (NADH) may play a crucial role in regulating bacterial resistance, studies examining how the change of metabolic profile and bacterial resistance following by exogenous administration are scarce. Therefore, this study aimed to elucidate the metabolic changes that occur in Edwardsiella tarda (E. tarda), which exhibits resistance to various antibiotics, following the exogenous addition of NADH using metabolomics. The effects of these alterations on the bactericidal activity of neomycin were investigated. NADH enhanced the effectiveness of aminoglycoside antibiotics against E. tarda ATCC15947, achieving bacterial eradication at low doses. Metabolomic analysis revealed that NADH reprogrammed the ATCC15947 metabolic profile by promoting purine metabolism and energy metabolism, yielding increased adenosine triphosphate (ATP) levels. Increased ATP levels played a crucial role in enhancing the bactericidal effects of neomycin. Moreover, exogenous NADH promoted the bactericidal efficacy of tetracyclines and chloramphenicols. NADH in combination with neomycin was effective against other clinically resistant bacteria, including Aeromonas hydrophila, Vibrio parahaemolyticus, methicillin-resistant Staphylococcus aureus, and Listeria monocytogenes. These results may facilitate the development of effective approaches for preventing and managing E. tarda-induced infections and multidrug resistance in aquaculture and clinical settings.


Subject(s)
Aminoglycosides , Anti-Bacterial Agents , Edwardsiella tarda , NAD , Edwardsiella tarda/drug effects , Anti-Bacterial Agents/pharmacology , NAD/metabolism , Aminoglycosides/pharmacology , Animals , Fish Diseases/microbiology , Fish Diseases/drug therapy , Microbial Sensitivity Tests , Enterobacteriaceae Infections/microbiology , Enterobacteriaceae Infections/drug therapy , Adenosine Triphosphate/metabolism , Neomycin/pharmacology , Drug Synergism , Metabolomics , Drug Resistance, Multiple, Bacterial/drug effects
19.
Antimicrob Agents Chemother ; 68(7): e0011224, 2024 Jul 09.
Article in English | MEDLINE | ID: mdl-38888319

ABSTRACT

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.


Subject(s)
Aminoglycosides , Anthrax , Anti-Bacterial Agents , Bacillus anthracis , Lipoglycopeptides , Microbial Sensitivity Tests , Respiratory Tract Infections , Animals , Lipoglycopeptides/pharmacology , Rabbits , Anthrax/drug therapy , Anthrax/microbiology , Anthrax/mortality , Bacillus anthracis/drug effects , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/therapeutic use , Aminoglycosides/pharmacology , Respiratory Tract Infections/drug therapy , Respiratory Tract Infections/microbiology , Disease Models, Animal , Levofloxacin/pharmacology , Female
20.
Mol Inform ; 43(7): e202300339, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38853661

ABSTRACT

Aminoglycosides are crucial antibiotics facing challenges from bacterial resistance. This study addresses the importance of aminoglycoside modifying enzymes in the context of escalating resistance. Drawing upon over two decades of structural data in the Protein Data Bank, we focused on two key antibiotics, neomycin B and kanamycin A, to explore how the aminoglycoside structure is exploited by this family of enzymes. A systematic comparison across diverse enzymes and the RNA A-site target identified common characteristics in the recognition mode, while assessing the adaptability of neomycin B and kanamycin A in various environments.


Subject(s)
Framycetin , Kanamycin , RNA, Bacterial , RNA, Ribosomal , Kanamycin/chemistry , Kanamycin/pharmacology , Framycetin/chemistry , Framycetin/pharmacology , RNA, Bacterial/chemistry , RNA, Bacterial/metabolism , RNA, Ribosomal/chemistry , RNA, Ribosomal/metabolism , Aminoglycosides/chemistry , Aminoglycosides/pharmacology , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL