Helicobacter pylori (H. pylori) is closely associated with the diseases such as gastric sinusitis, peptic ulcers, and gastric adenocarcinoma. Its drug resistance is very severe, and new antibiotics are urgently needed. Nine comfrey compounds were screened by antimicrobial susceptibility testing, among which deoxyshikonin had the best inhibitory effect, with a minimum inhibitory concentration (MIC) of 0.5-1 µg/mL. In addition, deoxyshikonin also has a good antibacterial effect in an acidic environment, it is highly safe, and H. pylori does not readily develop drug resistance. Through in vivo experiments, it was proven that deoxyshikonin (7 mg/kg) had a beneficial therapeutic effect on acute gastritis in mice infected with the multidrug-resistant H. pylori BS001 strain. After treatment with desoxyshikonin, colonization of H. pylori in the gastric mucosa of mice was significantly reduced, gastric mucosal damage was repaired, inflammatory factors were reduced, and the treatment effect was better than that of standard triple therapy. Therefore, deoxyshikonin is a promising lead drug to solve the difficulty of drug resistance in H. pylori, and its antibacterial mechanism may be to destroy the biofilm and cause an oxidation reaction.
{"title":"Deoxyshikonin: a promising lead drug grass against drug resistance or sensitivity to <i>Helicobacter pylori</i> in an acidic environment.","authors":"Jia-Yin Xu, Hui-Hua Dong, Li-Juan Liao, Shi-Xian Yang, Lu-Yao Wang, Hao Chen, Peipei Luo, Liang Huang, Ai-Xing Guan, Yan-Qiang Huang","doi":"10.1128/aac.00959-24","DOIUrl":"10.1128/aac.00959-24","url":null,"abstract":"<p><p><i>Helicobacter pylori</i> (<i>H. pylori</i>) is closely associated with the diseases such as gastric sinusitis, peptic ulcers, and gastric adenocarcinoma. Its drug resistance is very severe, and new antibiotics are urgently needed. Nine comfrey compounds were screened by antimicrobial susceptibility testing, among which deoxyshikonin had the best inhibitory effect, with a minimum inhibitory concentration (MIC) of 0.5-1 µg/mL. In addition, deoxyshikonin also has a good antibacterial effect in an acidic environment, it is highly safe, and <i>H. pylori</i> does not readily develop drug resistance. Through <i>in vivo</i> experiments, it was proven that deoxyshikonin (7 mg/kg) had a beneficial therapeutic effect on acute gastritis in mice infected with the multidrug-resistant <i>H. pylori</i> BS001 strain. After treatment with desoxyshikonin, colonization of <i>H. pylori</i> in the gastric mucosa of mice was significantly reduced, gastric mucosal damage was repaired, inflammatory factors were reduced, and the treatment effect was better than that of standard triple therapy. Therefore, deoxyshikonin is a promising lead drug to solve the difficulty of drug resistance in <i>H. pylori</i>, and its antibacterial mechanism may be to destroy the biofilm and cause an oxidation reaction.</p>","PeriodicalId":8152,"journal":{"name":"Antimicrobial Agents and Chemotherapy","volume":" ","pages":"e0095924"},"PeriodicalIF":4.1,"publicationDate":"2024-10-08","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11460997/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142016180","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-10-08Epub Date: 2024-09-05DOI: 10.1128/aac.00870-24
Yingsi Fang, Lloyd G Clarke, Brandon J Smith, Sunish Shah
This study investigated the real-world incidence rate of serotonin syndrome in patients receiving tedizolid and concomitant serotonergic agents. A retrospective cohort of 479 adult patients was assessed between January 2015 and July 2023. Overall, a rare rate of 0.4% (2/479) of possible serotonin syndrome with tedizolid was identified. Given that concomitant serotonergic agents were commonly used, further study is warranted to determine causality.
{"title":"Incidence of serotonin syndrome in patients receiving tedizolid and concomitant serotonergic agents.","authors":"Yingsi Fang, Lloyd G Clarke, Brandon J Smith, Sunish Shah","doi":"10.1128/aac.00870-24","DOIUrl":"10.1128/aac.00870-24","url":null,"abstract":"<p><p>This study investigated the real-world incidence rate of serotonin syndrome in patients receiving tedizolid and concomitant serotonergic agents. A retrospective cohort of 479 adult patients was assessed between January 2015 and July 2023. Overall, a rare rate of 0.4% (2/479) of possible serotonin syndrome with tedizolid was identified. Given that concomitant serotonergic agents were commonly used, further study is warranted to determine causality.</p>","PeriodicalId":8152,"journal":{"name":"Antimicrobial Agents and Chemotherapy","volume":" ","pages":"e0087024"},"PeriodicalIF":4.1,"publicationDate":"2024-10-08","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11459941/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142131587","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-10-08Epub Date: 2024-08-28DOI: 10.1128/aac.00777-24
Maria Isabel Tejeda, Javier Fernández, Pablo Valledor, Cristina Almirall, José Barberán, Santiago Romero-Brufau
Errors in antibiotic prescriptions are frequent, often resulting from the inadequate coverage of the infection-causative microorganism. The efficacy of iAST, a machine-learning-based software offering empirical and organism-targeted antibiotic recommendations, was assessed. The study was conducted in a 12-hospital Spanish institution. After model fine-tuning with 27,531 historical antibiograms, 325 consecutive patients with acute infections were selected for retrospective validation. The primary endpoint was comparing each of the top three of iAST's antibiotic recommendations' success rates (confirmed by antibiogram results) with the antibiotic prescribed by the physicians. Secondary endpoints included examining the same hypothesis within specific study population subgroups and assessing antibiotic stewardship by comparing the percentage of antibiotics recommended that belonged to different World Health Organization AWaRe groups within each arm of the study. All of iAST first three recommendations were non-inferior to doctor prescription in the primary endpoint analysis population as well as the secondary endpoint. The overall success rate of doctors' empirical treatment was 68.93%, while that of the first three iAST options was 91.06% (P < 0.001), 90.63% (P < 0.001), and 91.06% (P < 001), respectively. For organism-targeted therapy, the doctor's overall success rate was 84.16%, and that of the first three ranked iAST options was 97.83% (P < 0.001), 94.09% (P < 0.001), and 91.30% (P < 0.001), respectively. In empirical therapy, compared to physician prescriptions, iAST demonstrated a greater propensity to recommend access antibiotics, fewer watch antibiotics, and higher reserve antibiotics. In organism-targeted therapy, iAST advised a higher utilization of access antibiotics. The present study demonstrates iAST accuracy in predicting antibiotic susceptibility, showcasing its potential to promote effective antibiotic stewardship.
Clinical trials: This study is registered with ClinicalTrials.gov as NCT06174519.
{"title":"Retrospective validation study of a machine learning-based software for empirical and organism-targeted antibiotic therapy selection.","authors":"Maria Isabel Tejeda, Javier Fernández, Pablo Valledor, Cristina Almirall, José Barberán, Santiago Romero-Brufau","doi":"10.1128/aac.00777-24","DOIUrl":"10.1128/aac.00777-24","url":null,"abstract":"<p><p>Errors in antibiotic prescriptions are frequent, often resulting from the inadequate coverage of the infection-causative microorganism. The efficacy of iAST, a machine-learning-based software offering empirical and organism-targeted antibiotic recommendations, was assessed. The study was conducted in a 12-hospital Spanish institution. After model fine-tuning with 27,531 historical antibiograms, 325 consecutive patients with acute infections were selected for retrospective validation. The primary endpoint was comparing each of the top three of iAST's antibiotic recommendations' success rates (confirmed by antibiogram results) with the antibiotic prescribed by the physicians. Secondary endpoints included examining the same hypothesis within specific study population subgroups and assessing antibiotic stewardship by comparing the percentage of antibiotics recommended that belonged to different World Health Organization AWaRe groups within each arm of the study. All of iAST first three recommendations were non-inferior to doctor prescription in the primary endpoint analysis population as well as the secondary endpoint. The overall success rate of doctors' empirical treatment was 68.93%, while that of the first three iAST options was 91.06% (<i>P</i> < 0.001), 90.63% (<i>P</i> < 0.001), and 91.06% (<i>P</i> < 001), respectively. For organism-targeted therapy, the doctor's overall success rate was 84.16%, and that of the first three ranked iAST options was 97.83% (<i>P</i> < 0.001), 94.09% (<i>P</i> < 0.001), and 91.30% (<i>P</i> < 0.001), respectively. In empirical therapy, compared to physician prescriptions, iAST demonstrated a greater propensity to recommend access antibiotics, fewer watch antibiotics, and higher reserve antibiotics. In organism-targeted therapy, iAST advised a higher utilization of access antibiotics. The present study demonstrates iAST accuracy in predicting antibiotic susceptibility, showcasing its potential to promote effective antibiotic stewardship.</p><p><strong>Clinical trials: </strong>This study is registered with ClinicalTrials.gov as NCT06174519.</p>","PeriodicalId":8152,"journal":{"name":"Antimicrobial Agents and Chemotherapy","volume":" ","pages":"e0077724"},"PeriodicalIF":4.1,"publicationDate":"2024-10-08","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11460031/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142078878","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-10-08Epub Date: 2024-08-28DOI: 10.1128/aac.00712-24
Marc Gaona, Jordi Corral, Susana Campoy, Jordi Barbé, María Pérez Varela, Jesús Aranda
Acinetobacter baumannii is a notorious opportunistic pathogen responsible for healthcare-associated infections worldwide. Efflux pumps play crucial roles in mediating antimicrobial resistance, motility, and virulence. In this study, we present the identification and characterization of the new A. baumannii efflux pump SxtP belonging to the MFS superfamily (major facilitator superfamily), along with its associated activator LysR-type transcriptional regulator (LTTR) SxtR, demonstrating their roles in sulfamethoxazole/trimethoprim (also known as co-trimoxazole or SXT) resistance, surface-associated motility and virulence.
{"title":"The novel MFS efflux pump SxtP, regulated by the LysR-type transcriptional activator SxtR, is involved in the susceptibility to sulfamethoxazole/trimethoprim (SXT) and the pathogenesis of <i>Acinetobacter baumannii</i>.","authors":"Marc Gaona, Jordi Corral, Susana Campoy, Jordi Barbé, María Pérez Varela, Jesús Aranda","doi":"10.1128/aac.00712-24","DOIUrl":"10.1128/aac.00712-24","url":null,"abstract":"<p><p><i>Acinetobacter baumannii</i> is a notorious opportunistic pathogen responsible for healthcare-associated infections worldwide. Efflux pumps play crucial roles in mediating antimicrobial resistance, motility, and virulence. In this study, we present the identification and characterization of the new <i>A. baumannii</i> efflux pump SxtP belonging to the MFS superfamily (<u>m</u>ajor <u>f</u>acilitator <u>s</u>uperfamily), along with its associated activator <u>L</u>ysR<u>-</u>type <u>t</u>ranscriptional <u>r</u>egulator (LTTR) SxtR, demonstrating their roles in sulfamethoxazole/trimethoprim (also known as co-trimoxazole or SXT) resistance, surface-associated motility and virulence.</p>","PeriodicalId":8152,"journal":{"name":"Antimicrobial Agents and Chemotherapy","volume":" ","pages":"e0071224"},"PeriodicalIF":4.1,"publicationDate":"2024-10-08","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11459933/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142078879","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-10-08Epub Date: 2024-08-20DOI: 10.1128/aac.00800-24
Josef D Wolf, Michael R Sirrine, Robert M Cox, Richard K Plemper
Small-molecule antivirals can be used as chemical probes to stabilize transitory conformational stages of viral target proteins, facilitating structural analyses. Here, we evaluate allosteric pneumo- and paramyxovirus polymerase inhibitors that have the potential to serve as chemical probes and aid the structural characterization of short-lived intermediate conformations of the polymerase complex. Of multiple inhibitor classes evaluated, we discuss in-depth distinct scaffolds that were selected based on well-understood structure-activity relationships, insight into resistance profiles, biochemical characterization of the mechanism of action, and photoaffinity-based target mapping. Each class is thought to block structural rearrangements of polymerase domains albeit target sites and docking poses are distinct. This review highlights validated druggable targets in the paramyxo- and pneumovirus polymerase proteins and discusses discrete structural stages of the polymerase complexes required for bioactivity.
{"title":"Structural basis of paramyxo- and pneumovirus polymerase inhibition by non-nucleoside small-molecule antivirals.","authors":"Josef D Wolf, Michael R Sirrine, Robert M Cox, Richard K Plemper","doi":"10.1128/aac.00800-24","DOIUrl":"10.1128/aac.00800-24","url":null,"abstract":"<p><p>Small-molecule antivirals can be used as chemical probes to stabilize transitory conformational stages of viral target proteins, facilitating structural analyses. Here, we evaluate allosteric pneumo- and paramyxovirus polymerase inhibitors that have the potential to serve as chemical probes and aid the structural characterization of short-lived intermediate conformations of the polymerase complex. Of multiple inhibitor classes evaluated, we discuss in-depth distinct scaffolds that were selected based on well-understood structure-activity relationships, insight into resistance profiles, biochemical characterization of the mechanism of action, and photoaffinity-based target mapping. Each class is thought to block structural rearrangements of polymerase domains albeit target sites and docking poses are distinct. This review highlights validated druggable targets in the paramyxo- and pneumovirus polymerase proteins and discusses discrete structural stages of the polymerase complexes required for bioactivity.</p>","PeriodicalId":8152,"journal":{"name":"Antimicrobial Agents and Chemotherapy","volume":" ","pages":"e0080024"},"PeriodicalIF":4.1,"publicationDate":"2024-10-08","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11459973/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142003479","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-10-08Epub Date: 2024-08-28DOI: 10.1128/aac.00909-24
Dario Akaberi, Monireh Pourghasemi Lati, Janina Krambrich, Julia Berger, Grace Neilsen, Emilia Strandback, S Pauliina Turunen, Johan Wannberg, Hjalmar Gullberg, Martin Moche, Praveen Kumar Chinthakindi, Tomas Nyman, Stefan G Sarafianos, Anja Sandström, Josef D Järhult, Kristian Sandberg, Åke Lundkvist, Oscar Verho, Johan Lennerstrand
In vitro screening of large compound libraries with automated high-throughput screening is expensive and time-consuming and requires dedicated infrastructures. Conversely, the selection of DNA-encoded chemical libraries (DECLs) can be rapidly performed with routine equipment available in most laboratories. In this study, we identified novel inhibitors of SARS-CoV-2 main protease (Mpro) through the affinity-based selection of the DELopen library (open access for academics), containing 4.2 billion compounds. The identified inhibitors were peptide-like compounds containing an N-terminal electrophilic group able to form a covalent bond with the nucleophilic Cys145 of Mpro, as confirmed by x-ray crystallography. This DECL selection campaign enabled the discovery of the unoptimized compound SLL11 (IC50 = 30 nM), proving that the rapid exploration of large chemical spaces enabled by DECL technology allows for the direct identification of potent inhibitors avoiding several rounds of iterative medicinal chemistry. As demonstrated further by x-ray crystallography, SLL11 was found to adopt a highly unique U-shaped binding conformation, which allows the N-terminal electrophilic group to loop back to the S1' subsite while the C-terminal amino acid sits in the S1 subsite. MP1, a close analog of SLL11, showed antiviral activity against SARS-CoV-2 in the low micromolar range when tested in Caco-2 and Calu-3 (EC50 = 2.3 µM) cell lines. As peptide-like compounds can suffer from low cell permeability and metabolic stability, the cyclization of the compounds will be explored in the future to improve their antiviral activity.
利用自动高通量筛选技术对大型化合物库进行体外筛选既昂贵又耗时,而且需要专用的基础设施。相反,DNA编码化学文库(DECL)的筛选可以利用大多数实验室现有的常规设备快速完成。在这项研究中,我们通过对包含 42 亿个化合物的 DELopen 库(面向学术界开放)进行基于亲和力的筛选,确定了新型 SARS-CoV-2 主要蛋白酶(Mpro)抑制剂。经 X 射线晶体学证实,确定的抑制剂是肽类化合物,含有一个 N 端亲电基团,能与 Mpro 的亲核 Cys145 形成共价键。这次 DECL 挑选活动发现了未优化的化合物 SLL11(IC50 = 30 nM),证明了 DECL 技术对大型化学空间的快速探索可以直接识别强效抑制剂,避免了几轮反复的药物化学研究。X 射线晶体学进一步证明,SLL11 采用了非常独特的 U 型结合构象,它允许 N 端亲电基团环回 S1'亚位,而 C 端氨基酸则位于 S1 亚位。MP1是SLL11的近似物,在Caco-2和Calu-3(EC50 = 2.3 µM)细胞系中的测试结果显示,MP1对SARS-CoV-2的抗病毒活性在低微摩尔范围内。由于肽类化合物的细胞渗透性和新陈代谢稳定性较低,未来将探索将这些化合物环化,以提高其抗病毒活性。
{"title":"Identification of novel and potent inhibitors of SARS-CoV-2 main protease from DNA-encoded chemical libraries.","authors":"Dario Akaberi, Monireh Pourghasemi Lati, Janina Krambrich, Julia Berger, Grace Neilsen, Emilia Strandback, S Pauliina Turunen, Johan Wannberg, Hjalmar Gullberg, Martin Moche, Praveen Kumar Chinthakindi, Tomas Nyman, Stefan G Sarafianos, Anja Sandström, Josef D Järhult, Kristian Sandberg, Åke Lundkvist, Oscar Verho, Johan Lennerstrand","doi":"10.1128/aac.00909-24","DOIUrl":"10.1128/aac.00909-24","url":null,"abstract":"<p><p><i>In vitro</i> screening of large compound libraries with automated high-throughput screening is expensive and time-consuming and requires dedicated infrastructures. Conversely, the selection of DNA-encoded chemical libraries (DECLs) can be rapidly performed with routine equipment available in most laboratories. In this study, we identified novel inhibitors of SARS-CoV-2 main protease (M<sup>pro</sup>) through the affinity-based selection of the DELopen library (open access for academics), containing 4.2 billion compounds. The identified inhibitors were peptide-like compounds containing an N-terminal electrophilic group able to form a covalent bond with the nucleophilic Cys145 of M<sup>pro</sup>, as confirmed by x-ray crystallography. This DECL selection campaign enabled the discovery of the unoptimized compound SLL11 (IC<sub>50</sub> = 30 nM), proving that the rapid exploration of large chemical spaces enabled by DECL technology allows for the direct identification of potent inhibitors avoiding several rounds of iterative medicinal chemistry. As demonstrated further by x-ray crystallography, SLL11 was found to adopt a highly unique U-shaped binding conformation, which allows the N-terminal electrophilic group to loop back to the S1' subsite while the C-terminal amino acid sits in the S1 subsite. MP1, a close analog of SLL11, showed antiviral activity against SARS-CoV-2 in the low micromolar range when tested in Caco-2 and Calu-3 (EC<sub>50</sub> = 2.3 µM) cell lines. As peptide-like compounds can suffer from low cell permeability and metabolic stability, the cyclization of the compounds will be explored in the future to improve their antiviral activity.</p>","PeriodicalId":8152,"journal":{"name":"Antimicrobial Agents and Chemotherapy","volume":" ","pages":"e0090924"},"PeriodicalIF":4.1,"publicationDate":"2024-10-08","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11459923/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142078849","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-10-08Epub Date: 2024-08-29DOI: 10.1128/aac.01006-24
Gwendolyn E Wood, Jin Woo Lee, Thilini Peramuna, Karen L Wendt, Caroline M Kim, Laarni Kendra T Aguila, Claire L Calderon, Robert H Cichewicz
Antimicrobial resistance is extremely common in Mycoplasma genitalium, a frequent cause of urethritis in men and cervicitis, vaginitis, and pelvic inflammatory disease in women. Treatment of M. genitalium infections is difficult due to intrinsic and acquired resistance to many antibiotic classes. We undertook a program to identify novel antimicrobials with activity against M. genitalium from fungal natural products. Extracts of Ramularia coccinea contained a molecule with potent activity that was subsequently identified as fusidic acid, a fusidane-type antibiotic that has been in clinical use for decades outside the United States. We found that minimum inhibitory concentrations of fusidic acid ranged from 0.31 to 4 µg/mL among 17 M. genitalium strains including laboratory-passaged and low-passage clinical isolates. Time-kill data indicate that bactericidal killing occurs when M. genitalium is exposed to ≥10 µg/mL for 48 h, comparing favorably to serum concentrations obtained from typical loading dose regimens. Resistance to fusidic acid was associated with mutations in fusA consistent with the known mechanism of action in which fusidic acid inhibits protein synthesis by binding to elongation factor G. Interestingly, no mutants resistant to >10 µg/mL fusidic acid were obtained and a resistant strain containing a F435Y mutation in FusA was impaired for growth in vitro. These data suggest that fusidic acid may be a promising option for the treatment of M. genitalium infections.
生殖支原体是男性尿道炎和女性宫颈炎、阴道炎和盆腔炎的常见病因,其抗菌药耐药性在生殖支原体中极为常见。由于支原体对许多抗生素类别具有内在和获得性抗药性,因此生殖器支原体感染的治疗非常困难。我们开展了一项计划,从真菌天然产物中找出对 M. genitalium 有活性的新型抗菌素。Ramularia coccinea 的提取物中含有一种具有强效活性的分子,这种分子后来被鉴定为夫西地酸,夫西地酸是一种夫西地烷类抗生素,在美国以外的国家已在临床上使用了几十年。我们发现,在 17 种 M. genitalium 菌株(包括实验室通过的和低通过率的临床分离株)中,夫西地酸的最小抑菌浓度介于 0.31 至 4 µg/mL 之间。杀菌时间数据表明,当生殖器畸形芽孢杆菌暴露在≥10 µg/mL 的浓度下 48 小时,就会发生杀菌作用,这与典型的负荷剂量疗法所获得的血清浓度相比较,效果更佳。对夫西地酸的耐药性与 FusA 中的突变有关,这与已知的作用机制一致,即夫西地酸通过与延伸因子 G 结合抑制蛋白质合成。这些数据表明,夫西地酸可能是治疗生殖器疱疹病毒感染的一种很有前景的选择。
{"title":"The fungal natural product fusidic acid demonstrates potent activity against <i>Mycoplasma genitalium</i>.","authors":"Gwendolyn E Wood, Jin Woo Lee, Thilini Peramuna, Karen L Wendt, Caroline M Kim, Laarni Kendra T Aguila, Claire L Calderon, Robert H Cichewicz","doi":"10.1128/aac.01006-24","DOIUrl":"10.1128/aac.01006-24","url":null,"abstract":"<p><p>Antimicrobial resistance is extremely common in <i>Mycoplasma genitalium</i>, a frequent cause of urethritis in men and cervicitis, vaginitis, and pelvic inflammatory disease in women. Treatment of <i>M. genitalium</i> infections is difficult due to intrinsic and acquired resistance to many antibiotic classes. We undertook a program to identify novel antimicrobials with activity against <i>M. genitalium</i> from fungal natural products. Extracts of <i>Ramularia coccinea</i> contained a molecule with potent activity that was subsequently identified as fusidic acid, a fusidane-type antibiotic that has been in clinical use for decades outside the United States. We found that minimum inhibitory concentrations of fusidic acid ranged from 0.31 to 4 µg/mL among 17 <i>M</i>. <i>genitalium</i> strains including laboratory-passaged and low-passage clinical isolates. Time-kill data indicate that bactericidal killing occurs when <i>M. genitalium</i> is exposed to ≥10 µg/mL for 48 h, comparing favorably to serum concentrations obtained from typical loading dose regimens. Resistance to fusidic acid was associated with mutations in <i>fusA</i> consistent with the known mechanism of action in which fusidic acid inhibits protein synthesis by binding to elongation factor G. Interestingly, no mutants resistant to >10 µg/mL fusidic acid were obtained and a resistant strain containing a F435Y mutation in FusA was impaired for growth <i>in vitro</i>. These data suggest that fusidic acid may be a promising option for the treatment of <i>M. genitalium</i> infections.</p>","PeriodicalId":8152,"journal":{"name":"Antimicrobial Agents and Chemotherapy","volume":" ","pages":"e0100624"},"PeriodicalIF":4.1,"publicationDate":"2024-10-08","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11459954/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142103774","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-10-08Epub Date: 2024-09-05DOI: 10.1128/aac.00753-24
Vidhu Singh, Hemraj Nandanwar
Gram-negative bacteria (GNB) pose a major global public health challenge as they exhibit a remarkable level of resistance to antibiotics. One of the factors responsible for promoting resistance against a wide range of antibiotics is the outer membrane (OM) of Gram-negative bacteria. The OM acts as a barrier that prevents the entry of numerous antibiotics by reducing their influx (due to membrane impermeability) and enhancing their efflux (with the help of efflux pumps). Our study focuses on analyzing the effect of IMT-P8, a cell-penetrating peptide (CPP), to enhance the influx of various Gram-positive specific antibiotics in multi-drug resistant Gram-negative pathogens. In the mechanistic experiments, IMT-P8 permeabilizes the OM at the same concentrations at which it enhances the activity of various antibiotics against GNB. Cytoplasmic membrane permeabilization was also observed at these concentrations, indicating that IMT-P8 acts on both the outer and cytoplasmic membranes. IMT-P8 interferes with the intrinsic resistance mechanism of GNB and has the potential to make Gram-positive specific antibiotics effective against GNB. IMT-P8 extends the post-antibiotic effect and in combination with antibiotics shows anti-persister activity. The IMT-P8/fusidic acid combination is effective in eliminating intracellular pathogens. IMT-P8 with negligible toxicity displayed good efficacy in murine lung and thigh infection models. Based on these findings, IMT-P8 is a potential antibiotic adjuvant to treat Gram-negative bacterial infections that pose a health hazard.
{"title":"IMT-P8 potentiates Gram-positive specific antibiotics in intrinsically resistant Gram-negative bacteria.","authors":"Vidhu Singh, Hemraj Nandanwar","doi":"10.1128/aac.00753-24","DOIUrl":"10.1128/aac.00753-24","url":null,"abstract":"<p><p>Gram-negative bacteria (GNB) pose a major global public health challenge as they exhibit a remarkable level of resistance to antibiotics. One of the factors responsible for promoting resistance against a wide range of antibiotics is the outer membrane (OM) of Gram-negative bacteria. The OM acts as a barrier that prevents the entry of numerous antibiotics by reducing their influx (due to membrane impermeability) and enhancing their efflux (with the help of efflux pumps). Our study focuses on analyzing the effect of IMT-P8, a cell-penetrating peptide (CPP), to enhance the influx of various Gram-positive specific antibiotics in multi-drug resistant Gram-negative pathogens. In the mechanistic experiments, IMT-P8 permeabilizes the OM at the same concentrations at which it enhances the activity of various antibiotics against GNB. Cytoplasmic membrane permeabilization was also observed at these concentrations, indicating that IMT-P8 acts on both the outer and cytoplasmic membranes. IMT-P8 interferes with the intrinsic resistance mechanism of GNB and has the potential to make Gram-positive specific antibiotics effective against GNB. IMT-P8 extends the post-antibiotic effect and in combination with antibiotics shows anti-persister activity. The IMT-P8/fusidic acid combination is effective in eliminating intracellular pathogens. IMT-P8 with negligible toxicity displayed good efficacy in murine lung and thigh infection models. Based on these findings, IMT-P8 is a potential antibiotic adjuvant to treat Gram-negative bacterial infections that pose a health hazard.</p>","PeriodicalId":8152,"journal":{"name":"Antimicrobial Agents and Chemotherapy","volume":" ","pages":"e0075324"},"PeriodicalIF":4.1,"publicationDate":"2024-10-08","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11459922/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142131586","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Limited treatment options and multidrug-resistant (MDR) Klebsiella pneumoniae present a significant therapeutic challenge, underscoring the need for novel approaches. Drug repurposing is a promising tool for augmenting the activity of many antibiotics. This study aimed to identify novel synergistic drug combinations against K. pneumoniae based on drug repurposing. We used the clinically isolated GN 172867 MDR strain of K. pneumoniae to determine the reversal resistance activity of zidovudine (AZT). The combined effects of AZT and various antibiotics, including nitrofurantoin (NIT) and omadacycline (OMC), were examined using the checkerboard method, growth curves, and crystal violet assays to assess biofilms. An in vitro combination activity testing was carried out in 12 isolates of K. pneumoniae. In vivo murine urinary tract and lung infection models were used to evaluate the therapeutic effects of AZT + NIT and AZT + OMC, respectively. The fractional inhibitory concentration index and growth curve demonstrated that AZT synergized with NIT or OMC against K. pneumoniae strains. In addition, AZT + NIT inhibited biofilm formation and cleared mature biofilms. In vivo, compared with untreated GN 172867-infected mice, AZT + NIT and AZT + OMC treatment decreased colony counts in multiple tissues (P < 0.05) and pathological scores in the bladder and kidneys (P < 0.05) and increased the survival rate by 60% (P < 0.05). This study evaluated the combination of AZT and antibiotics to treat drug-resistant K. pneumoniae infections and found novel drug combinations for the treatment of acute urinary tract infections. These findings suggest that AZT may exert significant anti-resistance activity.
有限的治疗方案和耐多药(MDR)肺炎克雷伯氏菌给治疗带来了巨大挑战,突出了对新方法的需求。药物再利用是增强多种抗生素活性的有效手段。本研究的目的是在药物再利用的基础上确定针对肺炎克雷伯菌的新型协同药物组合。我们利用临床分离的肺炎克雷伯菌 GN 172867 MDR 菌株来确定齐多夫定(AZT)的逆转抗药性活性。我们使用棋盘格法、生长曲线以及评估生物膜的结晶紫测定法,检验了 AZT 与硝基呋喃妥因(NIT)和奥美拉唑霉素(OMC)等多种抗生素的联合作用。在 12 个肺炎双球菌分离物中进行了体外组合活性测试。体内小鼠尿道和肺部感染模型分别用于评估 AZT + NIT 和 AZT + OMC 的治疗效果。部分抑制浓度指数和生长曲线表明,AZT 与 NIT 或 OMC 对肺炎克雷伯菌株有协同作用。此外,AZT + NIT 还能抑制生物膜的形成并清除成熟的生物膜。在体内,与未经治疗的 GN 172867 感染小鼠相比,AZT + NIT 和 AZT + OMC 治疗降低了多个组织中的菌落计数(P < 0.05)以及膀胱和肾脏的病理评分(P < 0.05),并提高了 60% 的存活率(P < 0.05)。这项研究评估了 AZT 与抗生素联合治疗耐药肺炎双球菌感染的效果,并发现了治疗急性尿路感染的新型药物组合。这些研究结果表明,AZT 可发挥显著的抗耐药性活性。
{"title":"Zidovudine in synergistic combination with nitrofurantoin or omadacycline: <i>in vitro</i> and in murine urinary tract or lung infection evaluation against multidrug-resistant <i>Klebsiella pneumoniae</i>.","authors":"Ping Tian, Qing-Qing Li, Ming-Juan Guo, Yun-Zhu Zhu, Rong-Qing Zhu, Ya-Qin Guo, Yi Yang, Yan-Yan Liu, Liang Yu, Ya-Sheng Li, Jia-Bin Li","doi":"10.1128/aac.00344-24","DOIUrl":"10.1128/aac.00344-24","url":null,"abstract":"<p><p>Limited treatment options and multidrug-resistant (MDR) <i>Klebsiella pneumoniae</i> present a significant therapeutic challenge, underscoring the need for novel approaches. Drug repurposing is a promising tool for augmenting the activity of many antibiotics. This study aimed to identify novel synergistic drug combinations against <i>K. pneumoniae</i> based on drug repurposing. We used the clinically isolated GN 172867 MDR strain of <i>K. pneumoniae</i> to determine the reversal resistance activity of zidovudine (AZT). The combined effects of AZT and various antibiotics, including nitrofurantoin (NIT) and omadacycline (OMC), were examined using the checkerboard method, growth curves, and crystal violet assays to assess biofilms. An <i>in vitro</i> combination activity testing was carried out in 12 isolates of <i>K. pneumoniae. In vivo</i> murine urinary tract and lung infection models were used to evaluate the therapeutic effects of AZT + NIT and AZT + OMC, respectively. The fractional inhibitory concentration index and growth curve demonstrated that AZT synergized with NIT or OMC against <i>K. pneumoniae</i> strains. In addition, AZT + NIT inhibited biofilm formation and cleared mature biofilms. <i>In vivo</i>, compared with untreated GN 172867-infected mice, AZT + NIT and AZT + OMC treatment decreased colony counts in multiple tissues (<i>P</i> < 0.05) and pathological scores in the bladder and kidneys (<i>P</i> < 0.05) and increased the survival rate by 60% (<i>P</i> < 0.05). This study evaluated the combination of AZT and antibiotics to treat drug-resistant <i>K. pneumoniae</i> infections and found novel drug combinations for the treatment of acute urinary tract infections. These findings suggest that AZT may exert significant anti-resistance activity.</p>","PeriodicalId":8152,"journal":{"name":"Antimicrobial Agents and Chemotherapy","volume":" ","pages":"e0034424"},"PeriodicalIF":4.1,"publicationDate":"2024-10-08","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11459972/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142078880","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
The escalating prevalence of antibiotic-resistant bacterial infections necessitates urgent alternative therapeutic strategies. Phage therapy, which employs bacteriophages to specifically target pathogenic bacteria, emerges as a promising solution. This review examines the efficacy of phage therapy in zebrafish models, both embryos and adults, which are proven and reliable for simulating human infectious diseases. We synthesize findings from recent studies that utilized these models to assess phage treatments against various bacterial pathogens, including Enterococcus faecalis, Pseudomonas aeruginosa, Mycobacterium abscessus, Staphylococcus aureus, Klebsiella pneumoniae, Acinetobacter baumannii, and Escherichia coli. Methods of phage administration, such as circulation injection and bath immersion, are detailed alongside evaluations of survival rates and bacterial load reductions. Notably, combination therapies of phages with antibiotics show enhanced efficacy, as evidenced by improved survival rates and synergistic effects in reducing bacterial loads. We also discuss the transition from zebrafish embryos to adult models, emphasizing the increased complexity of immune responses. This review highlights the valuable contribution of the zebrafish model to advancing phage therapy research, particularly in the face of rising antibiotic resistance and the urgent need for alternative treatments.
{"title":"Zebrafish as an effective model for evaluating phage therapy in bacterial infections: a promising strategy against human pathogens.","authors":"Lucile Plumet, Denis Costechareyre, Jean-Philippe Lavigne, Karima Kissa, Virginie Molle","doi":"10.1128/aac.00829-24","DOIUrl":"10.1128/aac.00829-24","url":null,"abstract":"<p><p>The escalating prevalence of antibiotic-resistant bacterial infections necessitates urgent alternative therapeutic strategies. Phage therapy, which employs bacteriophages to specifically target pathogenic bacteria, emerges as a promising solution. This review examines the efficacy of phage therapy in zebrafish models, both embryos and adults, which are proven and reliable for simulating human infectious diseases. We synthesize findings from recent studies that utilized these models to assess phage treatments against various bacterial pathogens, including <i>Enterococcus faecalis</i>, <i>Pseudomonas aeruginosa</i>, <i>Mycobacterium abscessus</i>, <i>Staphylococcus aureus</i>, <i>Klebsiella pneumoniae</i>, <i>Acinetobacter baumannii</i>, and <i>Escherichia coli</i>. Methods of phage administration, such as circulation injection and bath immersion, are detailed alongside evaluations of survival rates and bacterial load reductions. Notably, combination therapies of phages with antibiotics show enhanced efficacy, as evidenced by improved survival rates and synergistic effects in reducing bacterial loads. We also discuss the transition from zebrafish embryos to adult models, emphasizing the increased complexity of immune responses. This review highlights the valuable contribution of the zebrafish model to advancing phage therapy research, particularly in the face of rising antibiotic resistance and the urgent need for alternative treatments.</p>","PeriodicalId":8152,"journal":{"name":"Antimicrobial Agents and Chemotherapy","volume":" ","pages":"e0082924"},"PeriodicalIF":4.1,"publicationDate":"2024-10-08","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11460995/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142153035","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}