1
|
Priyanka, Sharma M, Vaid B, Bharti R, Raut S, Jolly RS, Khatri N. Comprehensive safety and toxicity analysis of 2,2'-Bipyridine derivatives in combating MRSA biofilm formation and persistence. Front Cell Infect Microbiol 2025; 15:1493679. [PMID: 39925377 PMCID: PMC11802822 DOI: 10.3389/fcimb.2025.1493679] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2024] [Accepted: 01/02/2025] [Indexed: 02/11/2025] Open
Abstract
Introduction Methicillin-resistant Staphylococcus aureus (MRSA) infections have become arduous to treat due to their capacity to form biofilms, develop persistence, and exhibit significant antimicrobial resistance. These factors contribute to the complexity of managing MRSA infections and highlight the urgent need for innovative treatment strategies. Objectives This endeavor aims to evaluate the safety of 2,2'-Bipyridine (2,2'-Bipy) derivatives and their antimicrobial, anti-biofilm, and anti-persister activities in treating MRSA Infections. Methods Six derivatives were screened for their ADMET properties and tested for minimum inhibitory concentrations against various bacterial strains using agar well diffusion and broth dilution. Safety studies were conducted through hemolysis tests, cell viability assays, and in vivo acute oral toxicity examinations. Bactericidal mechanisms and biofilm disruption effects were analyzed using crystal violet staining and confocal microscopy assays. The murine thigh infection model was also used to investigate the in vivo efficacy. Results All derivatives exhibited favorable physicochemical profiles and ADMET properties and are predicted to be safe based on their drug-like properties. in vitro studies demonstrated that derivatives are non-toxic to 3T3 L1, and in vivo studies confirmed their safety in mice at a dose of 300 mg/kg and their non-hemolytic nature against rabbit red blood cells. All compounds showed potent antibacterial activity against the tested bacteria, including the resistant MRSA strain 831. They inhibited biofilm formation and eradicated biofilms in a dose-dependent manner against MTCC 737 and MRSA 831, and they effectively eliminated MRSA persister cells, outperforming the reference antibiotic vancomycin. These derivatives were found to depolarize the mitochondrial membrane and accumulate intracellular reactive oxygen species. These derivatives significantly reduced the bacterial load in the murine thigh infection model. Conclusion The study concluded that 2,2'-Bipy derivatives possess significant antimicrobial activity, are non-toxic, and are effective in inhibiting biofilm formation and killing persister cells.
Collapse
Affiliation(s)
- Priyanka
- IMTECH Centre for Animal Resources & Experimentation (iCARE), Council of Scientific and Industrial Research (CSIR)-Institute of Microbial Technology, Chandigarh, India
- Academy of Scientific and Innovative Research, Ghaziabad, India
| | - Mohini Sharma
- IMTECH Centre for Animal Resources & Experimentation (iCARE), Council of Scientific and Industrial Research (CSIR)-Institute of Microbial Technology, Chandigarh, India
| | - Bhavna Vaid
- IMTECH Centre for Animal Resources & Experimentation (iCARE), Council of Scientific and Industrial Research (CSIR)-Institute of Microbial Technology, Chandigarh, India
- PG Department of Chemistry, Sri Guru Tegh Bahadur (SGTB) Khalsa College, Sri Anandpur Sahib, Punjab, India
| | - Ram Bharti
- IMTECH Centre for Animal Resources & Experimentation (iCARE), Council of Scientific and Industrial Research (CSIR)-Institute of Microbial Technology, Chandigarh, India
- Academy of Scientific and Innovative Research, Ghaziabad, India
| | - Sachin Raut
- IMTECH Centre for Animal Resources & Experimentation (iCARE), Council of Scientific and Industrial Research (CSIR)-Institute of Microbial Technology, Chandigarh, India
- Academy of Scientific and Innovative Research, Ghaziabad, India
| | - R. S. Jolly
- Council of Scientific and Industrial Research (CSIR)-Institute of Microbial Technology, Chandigarh, India
| | - Neeraj Khatri
- IMTECH Centre for Animal Resources & Experimentation (iCARE), Council of Scientific and Industrial Research (CSIR)-Institute of Microbial Technology, Chandigarh, India
- Academy of Scientific and Innovative Research, Ghaziabad, India
| |
Collapse
|
2
|
Ge R, Zhao H, Tang Q, Chandarajoti K, Bai H, Wang X, Zhang K, Ye W, Han X, Wang C, Zhou W. A novel α-mangostin derivative synergistic to antibiotics against MRSA with unique mechanisms. Microbiol Spectr 2024; 12:e0163124. [PMID: 39508612 PMCID: PMC11619392 DOI: 10.1128/spectrum.01631-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2024] [Accepted: 10/08/2024] [Indexed: 11/15/2024] Open
Abstract
Methicillin-resistant Staphylococcus aureus (MRSA) remains a leading cause of hospital-acquired infections, often linked to complicated treatments, increased mortality risk, and significant cost burdens. Several antibacterial agents have been developed to address MRSA resistance. In this study, potential agents to combat MRSA resistance were explored, with the antibacterial activity of synthesized α-mangostin (α-MG) derivatives being evaluated alongside investigations into their cellular mechanisms against MRSA2. α-MG-4, featuring an allyl group at C3 of the lead compound α-MG, restored the sensitivity of MRSA2 to penicillin, enrofloxacin, and gentamicin, while also demonstrating improved safety profiles. Although α-MG-4 alone was ineffective against MRSA2, it exhibited an optimal synergistic ratio in vitro when combined with these antibiotics. This significant synergistic antibacterial effect was further confirmed in vivo using a mouse skin abscess model. Additionally, the synergistic mechanisms revealed that α-MG-4 was associated with changes in membrane permeability and inhibition of the MepA and NorA genes, which encode the efflux pumps of MRSA2. α-MG-4 also inhibited PBP2a expression, potentially by occupying a crucial binding site in a dose-dependent manner.IMPORTANCEMethicillin-resistant Staphylococcus aureus (MRSA)'s resistance to multiple antibiotics poses significant health and safety concerns. A novel α-mangostin (α-MG) derivative, α-MG-4, was first identified as a xanthone-based PBP2a inhibitor that reverses MRSA2 resistance to penicillin. The synergistic antibacterial effects of α-MG-4 were linked to increased cell membrane permeability and the inhibition of genes involved in efflux pump function.
Collapse
Affiliation(s)
- Rile Ge
- School of Pharmaceutical Sciences, South-Central Minzu University, Wuhan, China
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai, China
| | - Haiyan Zhao
- School of Pharmaceutical Sciences, South-Central Minzu University, Wuhan, China
| | - Qun Tang
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai, China
| | - Kasemsiri Chandarajoti
- Department of Pharmaceutical Chemistry, Faculty of Pharmaceutical Sciences, Prince of Songkla University, Hat Yai, Songkhla, Thailand
- Drug Delivery System Excellence Center, Faculty of Pharmaceutical Sciences, Prince of Songkla University, Hat-Yai, Songkhla, Thailand
| | - Han Bai
- Department of Clinical Pharmacy, College of Pharmacy, Guilin Medical University, Guilin, Guangxi, China
| | - Xiaoyang Wang
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai, China
- Key laboratory of Veterinary Chemical Drugs and Pharmaceutics, Ministry of Agriculture and Rural Affairs, Shanghai Research Institute, Chinese Academy of Agricultural Sciences, Shanghai, China
| | - Keyu Zhang
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai, China
- Key laboratory of Veterinary Chemical Drugs and Pharmaceutics, Ministry of Agriculture and Rural Affairs, Shanghai Research Institute, Chinese Academy of Agricultural Sciences, Shanghai, China
| | - Wenchong Ye
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai, China
- Key laboratory of Veterinary Chemical Drugs and Pharmaceutics, Ministry of Agriculture and Rural Affairs, Shanghai Research Institute, Chinese Academy of Agricultural Sciences, Shanghai, China
| | - Xiangan Han
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai, China
| | - Chunmei Wang
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai, China
- Key laboratory of Veterinary Chemical Drugs and Pharmaceutics, Ministry of Agriculture and Rural Affairs, Shanghai Research Institute, Chinese Academy of Agricultural Sciences, Shanghai, China
| | - Wen Zhou
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai, China
- Key laboratory of Veterinary Chemical Drugs and Pharmaceutics, Ministry of Agriculture and Rural Affairs, Shanghai Research Institute, Chinese Academy of Agricultural Sciences, Shanghai, China
| |
Collapse
|
3
|
Bugyna L, Bilská K, Boháč P, Pribus M, Bujdák J, Bujdáková H. Anti-Biofilm Effect of Hybrid Nanocomposite Functionalized with Erythrosine B on Staphylococcus aureus Due to Photodynamic Inactivation. Molecules 2024; 29:3917. [PMID: 39202995 PMCID: PMC11357139 DOI: 10.3390/molecules29163917] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Revised: 08/05/2024] [Accepted: 08/13/2024] [Indexed: 09/03/2024] Open
Abstract
Resistant biofilms formed by Staphylococcus aureus on medical devices pose a constant medical threat. A promising alternative to tackle this problem is photodynamic inactivation (PDI). This study focuses on a polyurethane (PU) material with an antimicrobial surface consisting of a composite based on silicate, polycation, and erythrosine B (EryB). The composite was characterized using X-ray diffraction and spectroscopy methods. Anti-biofilm effectiveness was determined after PDI by calculation of CFU mL-1. The liquid PU precursors penetrated a thin silicate film resulting in effective binding of the PU/silicate composite and the PU bulk phases. The incorporation of EryB into the composite matrix did not significantly alter the spectral properties or photoactivity of the dye. A green LED lamp and laser were used for PDI, while irradiation was performed for different periods. Preliminary experiments with EryB solutions on planktonic cells and biofilms optimized the conditions for PDI on the nanocomposite materials. Significant eradication of S. aureus biofilm on the composite surface was achieved by irradiation with an LED lamp and laser for 1.5 h and 10 min, respectively, resulting in a 10,000-fold reduction in biofilm growth. These results demonstrate potential for the development of antimicrobial polymer surfaces for modification of medical materials and devices.
Collapse
Affiliation(s)
- Larysa Bugyna
- Department of Microbiology and Virology, Faculty of Natural Sciences, Comenius University in Bratislava, Ilkovičova 6, 842 15 Bratislava, Slovakia; (L.B.); (K.B.)
| | - Katarína Bilská
- Department of Microbiology and Virology, Faculty of Natural Sciences, Comenius University in Bratislava, Ilkovičova 6, 842 15 Bratislava, Slovakia; (L.B.); (K.B.)
| | - Peter Boháč
- Institute of Inorganic Chemistry, Slovak Academy of Sciences, Dúbravská Cesta 9, 845 36 Bratislava, Slovakia; (P.B.); (M.P.); (J.B.)
| | - Marek Pribus
- Institute of Inorganic Chemistry, Slovak Academy of Sciences, Dúbravská Cesta 9, 845 36 Bratislava, Slovakia; (P.B.); (M.P.); (J.B.)
| | - Juraj Bujdák
- Institute of Inorganic Chemistry, Slovak Academy of Sciences, Dúbravská Cesta 9, 845 36 Bratislava, Slovakia; (P.B.); (M.P.); (J.B.)
- Department of Physical and Theoretical Chemistry, Faculty of Natural Sciences, Comenius University in Bratislava, Ilkovičova 6, 842 15 Bratislava, Slovakia
| | - Helena Bujdáková
- Department of Microbiology and Virology, Faculty of Natural Sciences, Comenius University in Bratislava, Ilkovičova 6, 842 15 Bratislava, Slovakia; (L.B.); (K.B.)
| |
Collapse
|
4
|
Zhu L, Yang X, Fu X, Yang P, Lin X, Wang F, Shen Z, Wang J, Sun F, Qiu Z. Pheromone cCF10 inhibits the antibiotic persistence of Enterococcus faecalis by modulating energy metabolism. Front Microbiol 2024; 15:1408701. [PMID: 39040910 PMCID: PMC11260814 DOI: 10.3389/fmicb.2024.1408701] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Accepted: 06/24/2024] [Indexed: 07/24/2024] Open
Abstract
Introduction Bacterial resistance presents a major challenge to both the ecological environment and human well-being, with persistence playing a key role. Multiple studies were recently undertaken to examine the factors influencing the formation of persisters and the underlying process, with a primary focus on Gram-negative bacteria and Staphylococcus aureus (Gram-positive bacteria). Enterococcus faecalis (E. faecalis) is capable of causing a variety of infectious diseases, but there have been few studies of E. faecalis persisters. Previous studies have shown that the sex pheromone cCF10 secreted by E. faecalis induces conjugative plasmid transfer. However, whether the pheromone cCF10 regulates the persistence of E. faecalis has not been investigated. Methods As a result, we investigated the effect and potential molecular mechanism of pheromone cCF10 in regulating the formation of persisters in E. faecalis OG1RF using a persistent bacteria model. Results and discussion The metabolically active E. faecalis OG1RF reached a persistence state and temporarily tolerated lethal antibiotic concentrations after 8 h of levofloxacin hydrochloride (20 mg/mL) exposure, exhibiting a persistence rate of 0.109 %. During the growth of E. faecalis OG1RF, biofilm formation was a critical factor contributing to antibiotic persistence, whereas 10 ng/mL cCF10 blocked persister cell formation. Notably, cCF10 mediated the antibiotic persistence of E. faecalis OG1RF via regulating metabolic activity rather than suppressing biofilm formation. The addition of cCF10 stimulated the Opp system and entered bacterial cells, inhibiting (p)ppGpp accumulation, thus maintaining the metabolically active state of bacteria and reducing persister cell generation. These findings offer valuable insights into the formation, as well as the control mechanism of E. faecalis persisters.
Collapse
Affiliation(s)
- Li Zhu
- School of Environmental and Chemical Engineering, Xi’an Polytechnic University, Xi’an, China
- Key Laboratory of Risk Assessment and Control for Environment and Food Safety, Tianjin Institute of Environmental and Operational Medicine, Tianjin, China
| | - Xiaobo Yang
- Key Laboratory of Risk Assessment and Control for Environment and Food Safety, Tianjin Institute of Environmental and Operational Medicine, Tianjin, China
| | - Xinyue Fu
- Key Laboratory of Risk Assessment and Control for Environment and Food Safety, Tianjin Institute of Environmental and Operational Medicine, Tianjin, China
- College of Oceanography and Ecological Science, Shanghai Ocean University, Shanghai, China
| | - Panpan Yang
- Key Laboratory of Risk Assessment and Control for Environment and Food Safety, Tianjin Institute of Environmental and Operational Medicine, Tianjin, China
- School of Public Health, North China University of Science and Technology, Tangshan, China
| | - Xiaoli Lin
- Key Laboratory of Risk Assessment and Control for Environment and Food Safety, Tianjin Institute of Environmental and Operational Medicine, Tianjin, China
- Key Laboratory of Karst Geological Resources and Environment, Guizhou University, Guizhou, China
| | - Feng Wang
- Key Laboratory of Risk Assessment and Control for Environment and Food Safety, Tianjin Institute of Environmental and Operational Medicine, Tianjin, China
- College of Oceanography and Ecological Science, Shanghai Ocean University, Shanghai, China
| | - Zhiqiang Shen
- Key Laboratory of Risk Assessment and Control for Environment and Food Safety, Tianjin Institute of Environmental and Operational Medicine, Tianjin, China
| | - Jingfeng Wang
- Key Laboratory of Risk Assessment and Control for Environment and Food Safety, Tianjin Institute of Environmental and Operational Medicine, Tianjin, China
| | - Feilong Sun
- School of Environmental and Chemical Engineering, Xi’an Polytechnic University, Xi’an, China
| | - Zhigang Qiu
- Key Laboratory of Risk Assessment and Control for Environment and Food Safety, Tianjin Institute of Environmental and Operational Medicine, Tianjin, China
| |
Collapse
|
5
|
Tang Z, Feng J, Challa M, Rowthu SR, Xiong S, Zou C, Li J, Verma CS, Peng H, He X, Huang C, He Y. Discovery of novel Thymol-TPP antibiotics that eradicate MRSA persisters. Eur J Med Chem 2024; 270:116381. [PMID: 38604097 DOI: 10.1016/j.ejmech.2024.116381] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2024] [Revised: 03/22/2024] [Accepted: 04/01/2024] [Indexed: 04/13/2024]
Abstract
The high prevalence of methicillin-resistant Staphylococcus aureus (MRSA) strains and the formation of non-growing, dormant "persisters" subsets help bacteria evade antibiotic treatment and enhance bacterial resistance, which poses a serious threat to human life and health. It is urgent to discover novel antibacterial therapies effective against MRSA persisters. Thymol is a common nutraceutical with weak antibacterial and antitumor activities. A series of Thymol triphenylphosphine (TPP) conjugates (TPP-Thy3) was designed and synthesized. These compounds showed significantly improved inhibitory activity against Gram-positive bacteria compared with Thymol. Among them, Thy3d displayed a low probability of resistance selection and showed excellent biocompatibility. Interestingly, Thy3d elicited a rapid killing effect of MRSA persisters (99.999%) at high concentration. Fluorescence experiments, electron microscopy, molecular dynamics simulation and bilayer experiment confirmed that Thy3d conjugates exerted potent antimicrobial activity by disrupting the integrity of the membrane of bacterial even the persister. Furthermore, Thy3d exhibited considerable efficacy in a mouse model of subcutaneous murine MRSA infection. In summary, TPP-Thy3 conjugates are a series of novel antibacterial agents and could serve as a new therapeutic strategy for combating antibiotic resistance.
Collapse
Affiliation(s)
- Ziyi Tang
- Chongqing Institute of Green and Intelligent Technology, Chinese Academy of Sciences, Chongqing, 400714, China
| | - Jizhou Feng
- Chongqing Key Laboratory of Natural Product Synthesis and Drug Research, School of Pharmaceutical Sciences, Chongqing University, Chongqing, 401331, China
| | - Mahesh Challa
- Chongqing Key Laboratory of Natural Product Synthesis and Drug Research, School of Pharmaceutical Sciences, Chongqing University, Chongqing, 401331, China
| | - Sankara Rao Rowthu
- Chongqing Key Laboratory of Natural Product Synthesis and Drug Research, School of Pharmaceutical Sciences, Chongqing University, Chongqing, 401331, China
| | - Shuxin Xiong
- Chongqing Key Laboratory of Natural Product Synthesis and Drug Research, School of Pharmaceutical Sciences, Chongqing University, Chongqing, 401331, China
| | - Cheng Zou
- Chongqing Key Laboratory of Natural Product Synthesis and Drug Research, School of Pharmaceutical Sciences, Chongqing University, Chongqing, 401331, China
| | - Jianguo Li
- Singapore Eye Research Institute, Singapore, 169856, Singapore; Bioinformatics Institute, A*STAR, 30 Biopolis Street, Matrix, 138671, Singapore
| | - Chandra Shekhar Verma
- Bioinformatics Institute, A*STAR, 30 Biopolis Street, Matrix, 138671, Singapore; Department of Biological Sciences, National University of Singapore, 117543, Singapore; School of Biological Sciences, Nanyang Technological University, 637551, Singapore
| | - Haibo Peng
- Chongqing Academy of Science and Technology, Chongqing, 401123, China
| | - Xiaoli He
- Chongqing Institute of Green and Intelligent Technology, Chinese Academy of Sciences, Chongqing, 400714, China
| | - Chao Huang
- Chongqing Key Laboratory of Natural Product Synthesis and Drug Research, School of Pharmaceutical Sciences, Chongqing University, Chongqing, 401331, China.
| | - Yun He
- Chongqing Key Laboratory of Natural Product Synthesis and Drug Research, School of Pharmaceutical Sciences, Chongqing University, Chongqing, 401331, China; BayRay Innovation Center, Shenzhen Bay Laboratory, Shenzhen, 518132, China.
| |
Collapse
|
6
|
Huang L, Zhu L, Yan J, Lin Y, Ding D, He L, Li Y, Ying Y, Shen L, Jiang Y, Cai H, Jiang T. Genomic characterization and outbreak investigations of methicillin-resistant Staphylococcus aureus in a county-level hospital in China. Front Microbiol 2024; 15:1387855. [PMID: 38638904 PMCID: PMC11025083 DOI: 10.3389/fmicb.2024.1387855] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2024] [Accepted: 03/11/2024] [Indexed: 04/20/2024] Open
Abstract
Methicillin-resistant Staphylococcus aureus (MRSA) is a common pathogen contributing to healthcare-associated infections, which can result in multiple sites infections. The epidemiological characteristics of MRSA exhibit variability among distinct regions and healthcare facilities. The aim of this study was to investigate the molecular epidemiology and nosocomial outbreak characteristics of MRSA in a county-level hospital in China. A total of 130 non-repetitive MRSA strains were collected from December 2020 to November 2021. Whole-genome sequencing (WGS) was performed to identify antimicrobial resistance and virulence factors. Phylogenetic analysis was conducted to ascertain genetic diversity and phylogenetic relationships. Independent transmission scenarios were determined by the phylogeny derived from single nucleotide polymorphisms (SNPs) within the core genome. All the MRSA isolates were collected from the intensive care unit (30.00%, 39/130), the department of otorhinolaryngology (10.00%, 13/130) and the department of burn unit (9.23%, 12/130). The clinical samples mainly included phlegm (53.85%, 70/130), purulent fluid (24.62%, 32/130), and secretions (8.46%, 11/130). The resistance rates to erythromycin, clindamycin and ciprofloxacin were 75.38, 40.00, and 39.23%, respectively. All the isolates belonged to 11 clonal complexes (CCs), with the major prevalent types were CC5, CC59, and CC398, accounting for 30.00% (39/130), 29.23% (38/130), and 16.92% (22/130), respectively. Twenty sequence types (STs) were identified, and ST59 (25.38%, 33/130) was the dominant lineage, followed by ST5 (23.84%, 31/130) and ST398 (16.92%, 22/130). Three different SCCmec types were investigated, most of isolates were type IV (33.85%, 44/130), followed by type II (27.69%, 36/130) and type III (0.77%, 1/130). The common clonal structures included CC5-ST5-t2460-SCCmec IIa, CC59-ST59-t437-SCCmec IV and CC398-ST398-t034-SCCmec (-), with rates of 16.92% (22/130), 14.62% (19/130), and 13.84% (18/130), respectively. Only 12 panton-valentine leucocidin (PVL) positive strains were identified. Two independent clonal outbreaks were detected, one consisting of 22 PVL-negative strains belongs to CC5-ST5-t2460-SCCmec IIa and the other consisting of 8 PVL-negative strains belongs to CC5-ST5-t311-SCCmec IIa. Overall, our study indicated that the CC5 lineage emerged as the predominant epidemic clone of MRSA, responsible for nosocomial outbreaks and transmission within a county-level hospital in China, highlighting the necessity to strengthen infection control measures for MRSA in such healthcare facilities.
Collapse
Affiliation(s)
- Linyao Huang
- Department of Clinical Laboratory, The First People's Hospital of Wenling, Affiliated Wenling Hospital, Wenzhou Medical University, Wenling, China
| | - Liangrong Zhu
- Department of Pharmacy, Wenling Hospital of Traditional Chinese Medicine, Affiliated Wenling Traditional Chinese Medicine Hospital, Zhejiang Chinese Medical University, Wenling, China
| | - Jianxin Yan
- Department of Clinical Laboratory, The First People's Hospital of Wenling, Affiliated Wenling Hospital, Wenzhou Medical University, Wenling, China
| | - Yajing Lin
- Department of Clinical Laboratory, The First People's Hospital of Wenling, Affiliated Wenling Hospital, Wenzhou Medical University, Wenling, China
| | - Ding Ding
- Department of Clinical Laboratory, The First People's Hospital of Wenling, Affiliated Wenling Hospital, Wenzhou Medical University, Wenling, China
| | - Long He
- Department of Clinical Laboratory, The First People's Hospital of Wenling, Affiliated Wenling Hospital, Wenzhou Medical University, Wenling, China
| | - Yexuzi Li
- Department of Critical Care Medicine, The First People's Hospital of Wenling, Affiliated Wenling Hospital, Wenzhou Medical University, Wenling, China
| | - Yi Ying
- Department of Traditional Chinese Medicine, The Affiliated Xianju’s Hospital, Hangzhou Medical College, Xianju, China
| | - Lijiong Shen
- Department of Clinical Laboratory, The First People's Hospital of Wenling, Affiliated Wenling Hospital, Wenzhou Medical University, Wenling, China
| | - Yuhan Jiang
- School of Public Health, Hangzhou Medical College, Hangzhou, China
| | - Haijun Cai
- Burn Unit, The First People's Hospital of Wenling, Affiliated Wenling Hospital, Wenzhou Medical University, Wenling, China
| | - Tian Jiang
- Department of Clinical Laboratory, The First People's Hospital of Wenling, Affiliated Wenling Hospital, Wenzhou Medical University, Wenling, China
| |
Collapse
|
7
|
Pashizeh F, Mansouri A, Bazzazan S, Abdihaji M, Khaleghian M, Bazzazan S, Rezei N, Eskandari A, Mashayekhi F, Heydari M, Tavakkoli Yaraki M. Bioresponsive gingerol-loaded alginate-coated niosomal nanoparticles for targeting intracellular bacteria and cancer cells. Int J Biol Macromol 2024; 258:128957. [PMID: 38154726 DOI: 10.1016/j.ijbiomac.2023.128957] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Revised: 12/14/2023] [Accepted: 12/19/2023] [Indexed: 12/30/2023]
Abstract
Targeting and treating intracellular pathogen infections has been long-standing challenge, particularly in light of the escalating prevalence of antimicrobial resistance. Herein, an optimum formulation of alginate (AL)-coated niosome-based carriers for delivery of herbal extract Gingerol (Gin) was developed to treat intracellular pathogen infections and cancer cells. We used Gin-Nio@AL as a model drug to assess its efficacy against Gram-negative/positive bacteria and breast cancer cell lines. Our investigation affirmed its heightened antibacterial and anticancer properties. The antibacterial activity of Gin-Nio@AL against intracellular Staphylococcus aureus (S. aureus) and pseudomonas aeruginosa (P. aeruginosa) was also tested. In the current study, the niosome nanoparticles containing herbal extract Gingerol were optimized regarding lipid content and Surfactant per Cholesterol molar ratio. The developed formulation provided potential advantages, such as smooth globular surface morphology, small diameter (240.68 nm), pH-sensitive sustained release, and high entrapment efficiency (94.85 %). The release rate of Gin from AL-coated niosomes (Gin-Nio@AL) in physiological and acidic pH is lower than uncoated nanoparticles (Gin-Nio). Besides, the release rate of Gin from niosomal formulations increased in acidic pH. The Gin-Nio@AL demonstrated good antimicrobial activity against S. aureus and P. aeruginosa, and compared to Gin-Nio, the MIC values decreased to 7.82 ± 0.00 and 1.95 ± 0.00 μg/mL, respectively. In addition, the time-kill assay results showed that the developed formulation significantly reduced the number of bacteria in both strains compared to other tested groups. The microtiter data and scanning electron microscope micrography showed that Gin-Nio@AL has a more significant inhibitory effect on biofilm formation than Gin-Nio and Gin. The cell cytotoxicity evaluation showed that Gin-Nio@AL reduced the survival rate of MDA-MB-231 cancer cells to 52.4 % and 45.2 % after 48 h and 72 h, respectively. The elimination of intracellular pathogens was investigated through a breast cancer cell infection in an in vitro model. Gin-Nio@AL exhibited an enhanced and sustained intracellular antibacterial activity against pathogens-infected breast cancer cells compared to other tested formulations. Overall, Gin-Nio@AL enables the triggered release and targeting of intra-extra cellular bacteria and cancer cells and provides a novel and promising candidate for treating intracellular pathogen infections and cancer cells.
Collapse
Affiliation(s)
- Fatemeh Pashizeh
- Department of Immunology, School of Medicine, Shahid Sadoughi University of Medical Science Yazd, Iran
| | - Afsoun Mansouri
- School of Pharmacy and Pharmaceutical Sciences, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Saina Bazzazan
- Department of Community Medicine, Mashhad Branch, Islamic Azad University, Mashhad, Iran
| | - Mohammadreza Abdihaji
- Department of Biology, The Center for Genomics and Bioinformatics, Indiana University, Bloomington, IN, USA
| | | | - Saba Bazzazan
- Department of Community Medicine, Mashhad Branch, Islamic Azad University, Mashhad, Iran
| | - Niloufar Rezei
- School of Chemical Engineering, College of Engineering, University of Tehran, Tehran, Iran
| | - Alireza Eskandari
- CTERC, NRITLD, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Farzaneh Mashayekhi
- Rasoul Akram Hospital, Iran University of Medical Sciences (IUMS), Tehran, Iran
| | - Maryam Heydari
- Department of Cell and Molecular Biology, Faculty of Biological Sciences, Kharazmi University, Tehran, Iran
| | - Mohammad Tavakkoli Yaraki
- School of Natural Sciences, Faculty of Science and Engineering, Macquarie University, Sydney, NSW 2109, Australia.
| |
Collapse
|
8
|
Hamad M, Al-Marzooq F, Srinivasulu V, Sulaiman A, Menon V, Ramadan WS, El-Awady R, Al-Tel TH. Antimicrobial activity of nature-inspired molecules against multidrug-resistant bacteria. Front Microbiol 2024; 14:1336856. [PMID: 38318129 PMCID: PMC10838778 DOI: 10.3389/fmicb.2023.1336856] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2023] [Accepted: 12/27/2023] [Indexed: 02/07/2024] Open
Abstract
Multidrug-resistant bacterial infections present a serious challenge to global health. In addition to the spread of antibiotic resistance, some bacteria can form persister cells which are tolerant to most antibiotics and can lead to treatment failure or relapse. In the present work, we report the discovery of a new class of small molecules with potent antimicrobial activity against Gram-positive bacteria and moderate activity against Gram-negative drug-resistant bacterial pathogens. The lead compound SIMR 2404 had a minimal inhibitory concentration (MIC) of 2 μg/mL against methicillin-resistant Staphylococcus aureus (MRSA) and vancomycin-intermediate Staphylococcus aureus (VISA). The MIC values against Gram-negative bacteria such as Escherichia coli and Actinobacteria baumannii were between 8-32 μg/mL. Time-kill experiments show that compound SIMR 2404 can rapidly kill tested bacteria. Compound SIMR 2404 was also found to rapidly kill MRSA persisters which display high levels of tolerance to conventional antibiotics. In antibiotic evolution experiments, MRSA quickly developed resistance to ciprofloxacin but failed to develop resistance to compound SIMR 2404 even after 24 serial passages. Compound SIMR 2404 was not toxic to normal human fibroblast at a concentration of 4 μg/mL which is twice the MIC concentration against MRSA. However, at a concentration of 8 μg/mL or higher, it showed cytotoxic activity indicating that it is not ideal as a candidate against Gram-negative bacteria. The acceptable toxicity profile and rapid antibacterial activity against MRSA highlight the potential of these molecules for further studies as anti-MRSA agents.
Collapse
Affiliation(s)
- Mohamad Hamad
- Sharjah Institute for Medical Research, University of Sharjah, Sharjah, United Arab Emirates
- College of Health Sciences, University of Sharjah, Sharjah, United Arab Emirates
| | - Farah Al-Marzooq
- College of Medicine and Health Sciences, UAE University, Al Ain, United Arab Emirates
| | - Vunnam Srinivasulu
- Sharjah Institute for Medical Research, University of Sharjah, Sharjah, United Arab Emirates
| | - Ashna Sulaiman
- Sharjah Institute for Medical Research, University of Sharjah, Sharjah, United Arab Emirates
| | - Varsha Menon
- Sharjah Institute for Medical Research, University of Sharjah, Sharjah, United Arab Emirates
| | - Wafaa S. Ramadan
- Sharjah Institute for Medical Research, University of Sharjah, Sharjah, United Arab Emirates
| | - Raafat El-Awady
- Sharjah Institute for Medical Research, University of Sharjah, Sharjah, United Arab Emirates
- College of Pharmacy, University of Sharjah, Sharjah, United Arab Emirates
| | - Taleb H. Al-Tel
- Sharjah Institute for Medical Research, University of Sharjah, Sharjah, United Arab Emirates
- College of Pharmacy, University of Sharjah, Sharjah, United Arab Emirates
| |
Collapse
|
9
|
Nazli A, Tao W, You H, He X, He Y. Treatment of MRSA Infection: Where are We? Curr Med Chem 2024; 31:4425-4460. [PMID: 38310393 DOI: 10.2174/0109298673249381231130111352] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2023] [Revised: 07/10/2023] [Accepted: 10/10/2023] [Indexed: 02/05/2024]
Abstract
Staphylococcus aureus is a leading cause of septicemia, endocarditis, pneumonia, skin and soft tissue infections, bone and joint infections, and hospital-acquired infections. In particular, methicillin-resistant Staphylococcus aureus (MRSA) is associated with high morbidity and mortality, and continues to be a major public health problem. The emergence of multidrug-resistant MRSA strains along with the wide consumption of antibiotics has made anti-MRSA treatment a huge challenge. Novel treatment strategies (e.g., novel antimicrobials and new administrations) against MRSA are urgently needed. In the past decade, pharmaceutical companies have invested more in the research and development (R&D) of new antimicrobials and strategies, spurred by favorable policies. All research articles were collected from authentic online databases, including Google Scholar, PubMed, Scopus, and Web of Science, by using different combinations of keywords, including 'anti-MRSA', 'antibiotic', 'antimicrobial', 'clinical trial', 'clinical phase', clinical studies', and 'pipeline'. The information extracted from articles was compared to information provided on the drug manufacturer's website and Clinical Trials.gov (https://clinicaltrials.gov/) to confirm the latest development phase of anti-MRSA agents. The present review focuses on the current development status of new anti-MRSA strategies concerning chemistry, pharmacological target(s), indications, route of administration, efficacy and safety, pharmacokinetics, and pharmacodynamics, and aims to discuss the challenges and opportunities in developing drugs for anti-MRSA infections.
Collapse
Affiliation(s)
- Adila Nazli
- Chongqing Key Laboratory of Natural Product Synthesis and Drug Research, School of Pharmaceutical Sciences, Chongqing University, Chongqing, 401331, China
| | - Wenlan Tao
- Chongqing School, University of Chinese Academy of Sciences (UCAS Chongqing), Chongqing, 400714, China
| | - Hengyao You
- Chongqing Key Laboratory of Natural Product Synthesis and Drug Research, School of Pharmaceutical Sciences, Chongqing University, Chongqing, 401331, China
| | - Xiaoli He
- Chongqing School, University of Chinese Academy of Sciences (UCAS Chongqing), Chongqing, 400714, China
| | - Yun He
- Chongqing Key Laboratory of Natural Product Synthesis and Drug Research, School of Pharmaceutical Sciences, Chongqing University, Chongqing, 401331, China
| |
Collapse
|
10
|
Mette M, Connolly W, Vishwanath N, Allu S, Whitaker C, Stone BK, Antoci V, Born CT, Garcia DR, Garcia D. Silver Carboxylate as an Antibiotic-Independent Antimicrobial: A Review of Current Formulations, in vitro Efficacy, and Clinical Relevance. MEDICAL RESEARCH ARCHIVES 2022; 10:10.18103/mra.v10i12.3388. [PMID: 36874620 PMCID: PMC9979826 DOI: 10.18103/mra.v10i12.3388] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
The increasing prevalence of multi-drug resistant pathogens has led to a renewed focus on the use of silver as an antibiotic-independent antimicrobial. Unfortunately, the use of many silver formulations may be limited by an uncontrolled release of silver with the potential for significant cytotoxic effects. Silver carboxylate (AgCar) has emerged as an alternative formulation of silver with the potential to mitigate these concerns while still displaying significant bactericidal activity. This article reviews the efficacy of silver carboxylate formulations as a promising novel antibiotic-independent antimicrobial. This study was conducted through a search of five electronic databases (PubMed, Embase, MEDLINE, Cochrane Library, and Web of Science) for relevant studies up to September 2022. Searches were conducted for types of "silver carboxylate" formulations. Sources were compiled based on title and abstract and screened for inclusion based on relevance and study design. A review of the antimicrobial activity and cytotoxicity of silver carboxylate was compiled based on this search. Current body of data suggests that silver carboxylate shows promise as an emerging antibiotic-independent antimicrobial, with significant bactericidal effects while minimizing cytotoxicity. Silver carboxylate addresses several of the limitations of more primitive formulations, including controlled dosing and fewer negative effects on eukaryotic cell lines. These factors are concentration-dependent and largely rely on the vehicle system used to deliver it. Although several silver carboxylate-based formulations like titanium dioxide/polydimethylsiloxane (TiO2/PDMS) matrix-eluting AgCar have shown promising results in vitro, and could potentially be utilized independently or in conjunction with current and future antimicrobial therapies, there is a need for further in vivo studies to validate their overall safety and efficacy profile.
Collapse
Affiliation(s)
- Makena Mette
- Warren Alpert Medical School of Brown University, Providence, RI.,Weiss Center for Orthopaedic Trauma Research, Rhode Island Hospital, Providence, RI
| | - William Connolly
- Warren Alpert Medical School of Brown University, Providence, RI.,Weiss Center for Orthopaedic Trauma Research, Rhode Island Hospital, Providence, RI
| | - Neel Vishwanath
- Warren Alpert Medical School of Brown University, Providence, RI.,Weiss Center for Orthopaedic Trauma Research, Rhode Island Hospital, Providence, RI
| | - Sai Allu
- Warren Alpert Medical School of Brown University, Providence, RI.,Weiss Center for Orthopaedic Trauma Research, Rhode Island Hospital, Providence, RI
| | - Colin Whitaker
- Warren Alpert Medical School of Brown University, Providence, RI.,Weiss Center for Orthopaedic Trauma Research, Rhode Island Hospital, Providence, RI
| | - Benjamin K Stone
- Warren Alpert Medical School of Brown University, Providence, RI.,Weiss Center for Orthopaedic Trauma Research, Rhode Island Hospital, Providence, RI
| | - Valentin Antoci
- Warren Alpert Medical School of Brown University, Providence, RI.,Weiss Center for Orthopaedic Trauma Research, Rhode Island Hospital, Providence, RI.,Department of Orthopaedic Surgery, Warren Alpert Medical School of Brown University, Providence, RI
| | - Christopher T Born
- Warren Alpert Medical School of Brown University, Providence, RI.,Weiss Center for Orthopaedic Trauma Research, Rhode Island Hospital, Providence, RI.,Department of Orthopaedic Surgery, Warren Alpert Medical School of Brown University, Providence, RI
| | - Dioscaris R Garcia
- Warren Alpert Medical School of Brown University, Providence, RI.,Weiss Center for Orthopaedic Trauma Research, Rhode Island Hospital, Providence, RI.,Department of Orthopaedic Surgery, Warren Alpert Medical School of Brown University, Providence, RI
| | | |
Collapse
|
11
|
Kamble E, Sanghvi P, Pardesi K. Synergistic effect of antibiotic combinations on Staphylococcus aureus biofilms and their persister cell populations. Biofilm 2022; 4:100068. [PMID: 35198967 PMCID: PMC8844754 DOI: 10.1016/j.bioflm.2022.100068] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2021] [Revised: 01/17/2022] [Accepted: 01/18/2022] [Indexed: 11/01/2022] Open
Abstract
Treatment of staphylococcal infections is difficult due to multidrug resistance with their persister forms posing an added threat of recalcitrant infections. Antibiotic combinations are widely studied as an alternative strategy to combat them; therefore, they merit further investigation into their effect on the number of persister cells. In the present study, the fractional inhibitory concentrations of antibiotic combinations ciprofloxacin-daptomycin, ciprofloxacin-vancomycin, daptomycin-tobramycin, and tobramycin-vancomycin (checkerboard assay) were determined against two previously studied clinical (S48 and J6) and one standard (NCIM 5021) isolate of Staphylococcus aureus. They showed synergistic effects with a 2 to 256-fold reduction in MICs. All combinations also resulted in inhibition and disruption of biofilms in a concentration-dependent manner. All antibiotic combinations, except ciprofloxacin-daptomycin, showed total biofilm inhibition at 100X MICs. Similarly, antibiotic combination at 100X MIC showed 77–97% disruption of preformed biofilms. Time-kill assays performed at a 100X MIC combination against stationary-phase cells showed a two to six log10 reduction in CFU followed by a plateau indicating the presence of persisters. Significant differences were observed in persister cell fraction remaining after treatment with antibiotic combinations compared to monotherapies (p < 0.05) and therefore merit further investigation in clinical use for treatment against persisters.
Collapse
|
12
|
Mahjabeen F, Saha U, Mostafa MN, Siddique F, Ahsan E, Fathma S, Tasnim A, Rahman T, Faruq R, Sakibuzzaman M, Dilnaz F, Ashraf A. An Update on Treatment Options for Methicillin-Resistant Staphylococcus aureus (MRSA) Bacteremia: A Systematic Review. Cureus 2022; 14:e31486. [DOI: 10.7759/cureus.31486] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/13/2022] [Indexed: 11/16/2022] Open
|
13
|
Mohiuddin SG, Ghosh S, Kavousi P, Orman MA. Proton Motive Force Inhibitors Are Detrimental to Methicillin-Resistant Staphylococcus aureus Strains. Microbiol Spectr 2022; 10:e0202422. [PMID: 35943153 PMCID: PMC9430991 DOI: 10.1128/spectrum.02024-22] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Accepted: 07/08/2022] [Indexed: 11/20/2022] Open
Abstract
Methicillin-resistant Staphylococcus aureus (MRSA) strains are tolerant of conventional antibiotics, making them extremely dangerous. Previous studies have shown the effectiveness of proton motive force (PMF) inhibitors at killing bacterial cells; however, whether these agents can launch a new treatment strategy to eliminate antibiotic-tolerant cells mandates further investigation. Here, using known PMF inhibitors and two different MRSA isolates, we showed that the bactericidal potency of PMF inhibitors seemed to correlate with their ability to disrupt PMF and permeabilize cell membranes. By screening a small chemical library to verify this correlation, we identified a subset of chemicals (including nordihydroguaiaretic acid, gossypol, trifluoperazine, and amitriptyline) that strongly disrupted PMF in MRSA cells by dissipating either the transmembrane electric potential (ΔΨ) or the proton gradient (ΔpH). These drugs robustly permeabilized cell membranes and reduced MRSA cell levels below the limit of detection. Overall, our study further highlights the importance of cellular PMF as a target for designing new bactericidal therapeutics for pathogens. IMPORTANCE Methicillin-resistant Staphylococcus aureus (MRSA) emerged as a major hypervirulent pathogen that causes severe health care-acquired infections. These pathogens can be multidrug-tolerant cells, which can facilitate the recurrence of chronic infections and the emergence of diverse antibiotic-resistant mutants. In this study, we aimed to investigate whether proton motive force (PMF) inhibitors can launch a new treatment strategy to eliminate MRSA cells. Our in-depth analysis showed that PMF inhibitors that strongly dissipate either the transmembrane electric potential or the proton gradient can robustly permeabilize cell membranes and reduce MRSA cell levels below the limit of detection.
Collapse
Affiliation(s)
- Sayed Golam Mohiuddin
- Department of Chemical and Biomolecular Engineering, University of Houston, Houston, Texas, USA
| | - Sreyashi Ghosh
- Department of Chemical and Biomolecular Engineering, University of Houston, Houston, Texas, USA
| | - Pouria Kavousi
- Department of Chemical and Biomolecular Engineering, University of Houston, Houston, Texas, USA
| | - Mehmet A. Orman
- Department of Chemical and Biomolecular Engineering, University of Houston, Houston, Texas, USA
| |
Collapse
|
14
|
Kim SM, Zou G, Kim H, Kang M, Ahn S, Heo HY, Kim JS, Lim KM, Ausubel FM, Mylonakis E, Gao H, Kim W. Antimicrobial activity of the membrane-active compound nTZDpa is enhanced at low pH. Biomed Pharmacother 2022; 150:112977. [PMID: 35447554 DOI: 10.1016/j.biopha.2022.112977] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Revised: 04/03/2022] [Accepted: 04/12/2022] [Indexed: 11/30/2022] Open
Abstract
The opportunistic human pathogen Staphylococcus aureus can evade antibiotics by acquiring antibiotic resistance genes or by entering into a non-growing dormant state. Moreover, the particular circumstances of a specific infection site, such as acidity or anaerobicity, often weaken antibiotic potency. Decreased bacterial susceptibility combined with diminished antibiotic potency is responsible for high failure rates when treating S. aureus infections. Here, we report that the membrane-active antimicrobial agent nTZDpa does not only exhibit enhanced antibiotic activity against multidrug-resistant Gram-positive pathogens in acidic pH, but also retains antimicrobial potency under anaerobic conditions. This agent completely eradicated highly antibiotic-tolerant cells and biofilms formed by methicillin-resistant S. aureus at pH 5.5 at concentrations at which it was not potent at pH 7.4. Furthermore, nTZDpa was more potent at synergistically potentiating gentamicin killing against antibiotic-tolerant MRSA cells at low pH than at high pH. All-atom molecular dynamics simulations combined with membrane-permeabilization assays revealed that the neutral form of nTZDpa, which contains carboxylic acid, is more effective than the deprotonated form at penetrating the bacterial membrane and plays an essential role in membrane activity. An acidic pH increases the proportion of the neutrally charged nTZDpa, which results in antimicrobial enhancement. Our results provide key insights into rational design of pH-sensitive membrane-active antimicrobials and antibiotic adjuvants that are effective in an infection environment. These findings demonstrate that nTZDpa is a promising lead compound for developing new therapeutics against hard-to-cure infections caused by drug-resistant and -tolerant S. aureus.
Collapse
Affiliation(s)
- Soo Min Kim
- College of Pharmacy, Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul 03760, Republic of Korea
| | - Guijin Zou
- Institute of High Performance Computing, A⁎STAR, Singapore 138632, Singapore
| | - Hyerim Kim
- College of Pharmacy, Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul 03760, Republic of Korea
| | - Minjeong Kang
- College of Pharmacy, Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul 03760, Republic of Korea
| | - Soyeon Ahn
- College of Pharmacy, Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul 03760, Republic of Korea
| | - Hee Young Heo
- College of Pharmacy, Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul 03760, Republic of Korea
| | - Jae-Seok Kim
- Department of Laboratory Medicine, Kangdong Sacred Heart Hospital, Hallym University College of Medicine, Seoul 05355, Republic of Korea
| | - Kyung-Min Lim
- College of Pharmacy, Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul 03760, Republic of Korea
| | - Frederick M Ausubel
- Department of Molecular Biology, Massachusetts General Hospital, Boston, MA 02114, USA; Department of Genetics, Harvard Medical School, Boston, MA 02115, USA
| | - Eleftherios Mylonakis
- Division of Infectious Diseases, Rhode Island Hospital, Warren Alpert Medical School of Brown University, Providence, RI 02903, USA
| | - Huajian Gao
- Institute of High Performance Computing, A⁎STAR, Singapore 138632, Singapore; School of Mechanical and Aerospace Engineering, College of Engineering, Nanyang Technological University, Singapore 639789, Singapore
| | - Wooseong Kim
- College of Pharmacy, Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul 03760, Republic of Korea.
| |
Collapse
|
15
|
Leucyl-tRNA Synthetase Inhibitor, D-Norvaline, in Combination with Oxacillin, Is Effective against Methicillin-Resistant Staphylococcus aureus. Antibiotics (Basel) 2022; 11:antibiotics11050683. [PMID: 35625327 PMCID: PMC9137938 DOI: 10.3390/antibiotics11050683] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Revised: 05/11/2022] [Accepted: 05/16/2022] [Indexed: 11/17/2022] Open
Abstract
Methicillin-resistant Staphylococcus aureus (MRSA) is a pathogenic bacterium that causes severe diseases in humans. For decades, MRSA has acquired substantial resistance against conventional antibiotics through regulatory adaptation, thereby posing a challenge for treating MRSA infection. One of the emerging strategies to combat MRSA is the combinatory use of antibacterial agents. Based on the dramatic change in phospholipid fatty acid (PLFA) composition of MRSA in previous results, this study investigated branched-chain amino acid derivatives (precursors of fatty acid synthesis of cell membrane) and discovered the antimicrobial potency of D-norvaline. The compound, which can act synergistically with oxacillin, is among the three leucine-tRNA synthetase inhibitors with high potency to inhibit MRSA cell growth and biofilm formation. PLFA analysis and membrane properties revealed that D-norvaline decreased the overall amount of PLFA, increasing the fluidity and decreasing the hydrophobicity of the bacterial cell membrane. Additionally, we observed genetic differences to explore the response to D-norvaline. Furthermore, deletion mutants and clinically isolated MRSA strains were treated with D-norvaline. The study revealed that D-norvaline, with low concentrations of oxacillin, was effective in killing several MRSA strains. In summary, our findings provide a new combination of aminoacyl-tRNA synthetase inhibitor D-norvaline and oxacillin, which is effective against MRSA.
Collapse
|
16
|
Hamad M, Al-Marzooq F, Srinivasulu V, Omar HA, Sulaiman A, Zaher DM, Orive G, Al-Tel TH. Antibacterial Activity of Small Molecules Which Eradicate Methicillin-Resistant Staphylococcus aureus Persisters. Front Microbiol 2022; 13:823394. [PMID: 35178043 PMCID: PMC8846302 DOI: 10.3389/fmicb.2022.823394] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2021] [Accepted: 01/11/2022] [Indexed: 11/13/2022] Open
Abstract
The serious challenge posed by multidrug-resistant bacterial infections with concomitant treatment failure and high mortality rates presents an urgent threat to the global health. We herein report the discovery of a new class of potent antimicrobial compounds that are highly effective against Gram-positive bacteria, including methicillin-resistant Staphylococcus aureus (MRSA). The compounds were efficiently synthesized in one-pot employing a cascade of Groebke-Blackburn-Bienaymé and aza-Michael addition reactions. Phenotypic screening of the pilot library against various bacterial species including methicillin-sensitive and MRSA strains, has identified potent chemotypes with minimal inhibitory concentrations (MIC) of 3.125-6.25 μg/ml. The most potent compounds were fast-acting at eradicating exponentially growing MRSA, with killing achieved after 30 min of exposure to the compounds. They were also able to kill MRSA persister cells which are tolerant to most available medications. Microscopic analysis using fluorescence microscope and atomic force microscope indicate that these compounds lead to disruption of bacterial cell envelopes. Most notably, bacterial resistance toward these compounds was not observed after 20 serial passages in stark contrast to the significant resistance developed rapidly upon exposure to a clinically relevant antibiotic. Furthermore, the compounds did not induce significant hemolysis to human red blood cells. In vivo safety studies revealed a high safety profile of these motifs. These small molecules hold a promise for further studies and development as new antibacterial agents against MRSA infections.
Collapse
Affiliation(s)
- Mohamad Hamad
- Sharjah Institute for Medical Research, University of Sharjah, Sharjah, United Arab Emirates.,College of Health Sciences, University of Sharjah, Sharjah, United Arab Emirates
| | - Farah Al-Marzooq
- Sharjah Institute for Medical Research, University of Sharjah, Sharjah, United Arab Emirates.,College of Medicine and Health Sciences, UAE University, Al Ain, United Arab Emirates
| | - Vunnam Srinivasulu
- Sharjah Institute for Medical Research, University of Sharjah, Sharjah, United Arab Emirates
| | - Hany A Omar
- Sharjah Institute for Medical Research, University of Sharjah, Sharjah, United Arab Emirates.,College of Pharmacy, University of Sharjah, Sharjah, United Arab Emirates
| | - Ashna Sulaiman
- Sharjah Institute for Medical Research, University of Sharjah, Sharjah, United Arab Emirates
| | - Dana M Zaher
- Sharjah Institute for Medical Research, University of Sharjah, Sharjah, United Arab Emirates
| | - Gorka Orive
- NanoBioCel Group, Laboratory of Pharmaceutics, School of Pharmacy, University of the Basque Country (UPV/EHU), Bilbao, Spain
| | - Taleb H Al-Tel
- Sharjah Institute for Medical Research, University of Sharjah, Sharjah, United Arab Emirates.,College of Pharmacy, University of Sharjah, Sharjah, United Arab Emirates
| |
Collapse
|
17
|
Yu JH, Xu XF, Hou W, Meng Y, Huang MY, Lin J, Chen WM. Synthetic cajaninstilbene acid derivatives eradicate methicillin-resistant Staphylococcus aureus persisters and biofilms. Eur J Med Chem 2021; 224:113691. [PMID: 34274830 DOI: 10.1016/j.ejmech.2021.113691] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2021] [Revised: 07/05/2021] [Accepted: 07/05/2021] [Indexed: 11/30/2022]
Abstract
The Staphylococcus aureus can switch to a transient genotype-invariant dormancy, known as a persister, to survive treatment with high doses of antibiotics. This transient persister is an important reason underlying its resistance. There is an urgent need to find new antibacterial agents capable of eradicating methicillin-resistant S. aureus (MRSA) persisters. In this study, 37 new derivatives of cajaninstilbene acid (CSA) were designed and synthesized, and their biological activity against MRSA persisters was evaluated. Most of the newly synthesized derivatives exhibit more potent antimicrobial properties against S. aureus and MRSA than CSA itself, and 23 of the 37 derivatives show a tendency to eradicate MRSA persisters. A representative compound (A6) was demonstrated to target bacterial cell membranes. It eradicated the adherent biofilm of MRSA in a concentration dependent manner, and showed a synergistic antibacterial effect with piperacilin. In a model mouse abscess caused by MRSA persisters, A6 effectively reduced the bacterial load in vivo. These results indicate that A6 is a potential candidate for treatment of MRSA persister infections.
Collapse
Affiliation(s)
- Jia-Hui Yu
- College of Pharmacy, Jinan University, Guangzhou, 510632, PR China
| | - Xiao-Fang Xu
- College of Pharmacy, Jinan University, Guangzhou, 510632, PR China
| | - Wen Hou
- College of Pharmacy, Jinan University, Guangzhou, 510632, PR China
| | - Ying Meng
- College of Pharmacy, Jinan University, Guangzhou, 510632, PR China
| | - Mei-Yan Huang
- College of Pharmacy, Jinan University, Guangzhou, 510632, PR China
| | - Jing Lin
- College of Pharmacy, Jinan University, Guangzhou, 510632, PR China.
| | - Wei-Min Chen
- College of Pharmacy, Jinan University, Guangzhou, 510632, PR China.
| |
Collapse
|
18
|
da Cunha BR, Zoio P, Fonseca LP, Calado CRC. Technologies for High-Throughput Identification of Antibiotic Mechanism of Action. Antibiotics (Basel) 2021; 10:565. [PMID: 34065815 PMCID: PMC8151116 DOI: 10.3390/antibiotics10050565] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2021] [Revised: 05/05/2021] [Accepted: 05/10/2021] [Indexed: 01/23/2023] Open
Abstract
There are two main strategies for antibiotic discovery: target-based and phenotypic screening. The latter has been much more successful in delivering first-in-class antibiotics, despite the major bottleneck of delayed Mechanism-of-Action (MOA) identification. Although finding new antimicrobial compounds is a very challenging task, identifying their MOA has proven equally challenging. MOA identification is important because it is a great facilitator of lead optimization and improves the chances of commercialization. Moreover, the ability to rapidly detect MOA could enable a shift from an activity-based discovery paradigm towards a mechanism-based approach. This would allow to probe the grey chemical matter, an underexplored source of structural novelty. In this study we review techniques with throughput suitable to screen large libraries and sufficient sensitivity to distinguish MOA. In particular, the techniques used in chemical genetics (e.g., based on overexpression and knockout/knockdown collections), promoter-reporter libraries, transcriptomics (e.g., using microarrays and RNA sequencing), proteomics (e.g., either gel-based or gel-free techniques), metabolomics (e.g., resourcing to nuclear magnetic resonance or mass spectrometry techniques), bacterial cytological profiling, and vibrational spectroscopy (e.g., Fourier-transform infrared or Raman scattering spectroscopy) were discussed. Ultimately, new and reinvigorated phenotypic assays bring renewed hope in the discovery of a new generation of antibiotics.
Collapse
Affiliation(s)
- Bernardo Ribeiro da Cunha
- Institute for Bioengineering and Biosciences (iBB), Instituto Superior Técnico (IST), Universidade de Lisboa (UL), Av. Rovisco Pais, 1049-001 Lisboa, Portugal; (B.R.d.C.); (P.Z.); (L.P.F.)
| | - Paulo Zoio
- Institute for Bioengineering and Biosciences (iBB), Instituto Superior Técnico (IST), Universidade de Lisboa (UL), Av. Rovisco Pais, 1049-001 Lisboa, Portugal; (B.R.d.C.); (P.Z.); (L.P.F.)
- CIMOSM—Centro de Investigação em Modelação e Optimização de Sistemas Multifuncionais, ISEL—Instituto Superior de Engenharia de Lisboa, Instituto Politécnico de Lisboa, R. Conselheiro Emídio Navarro 1, 1959-007 Lisboa, Portugal
| | - Luís P. Fonseca
- Institute for Bioengineering and Biosciences (iBB), Instituto Superior Técnico (IST), Universidade de Lisboa (UL), Av. Rovisco Pais, 1049-001 Lisboa, Portugal; (B.R.d.C.); (P.Z.); (L.P.F.)
| | - Cecília R. C. Calado
- CIMOSM—Centro de Investigação em Modelação e Optimização de Sistemas Multifuncionais, ISEL—Instituto Superior de Engenharia de Lisboa, Instituto Politécnico de Lisboa, R. Conselheiro Emídio Navarro 1, 1959-007 Lisboa, Portugal
| |
Collapse
|
19
|
Devlin H, Fulaz S, Hiebner DW, O’Gara JP, Casey E. Enzyme-Functionalized Mesoporous Silica Nanoparticles to Target Staphylococcus aureus and Disperse Biofilms. Int J Nanomedicine 2021; 16:1929-1942. [PMID: 33727807 PMCID: PMC7954034 DOI: 10.2147/ijn.s293190] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Accepted: 02/09/2021] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND Staphylococcus aureus biofilms pose a unique challenge in healthcare due to their tolerance to a wide range of antimicrobial agents. The high cost and lengthy timeline to develop novel therapeutic agents have pushed researchers to investigate the use of nanomaterials to deliver antibiofilm agents and target biofilm infections more efficiently. Previous studies have concentrated on improving the efficacy of antibiotics by deploying nanoparticles as nanocarriers. However, the dispersal of the extracellular polymeric substance (EPS) matrix in biofilm-associated infections is also critical to the development of novel nanoparticle-based therapies. METHODS This study evaluated the efficacy of enzyme-functionalized mesoporous silica nanoparticles (MSNs) against methicillin-resistant S. aureus (MRSA) and methicillin-sensitive S. aureus (MSSA) biofilms. MSNs were functionalized with the enzyme lysostaphin, which causes cell lysis of S. aureus bacteria. This was combined with two other enzyme functionalized MSNs, serrapeptase and DNase I which will degrade protein and eDNA in the EPS matrix, to enhance eradication of the biofilm. Cell viability after treatment with enzyme-functionalized MSNs was assessed using a MTT assay and CLSM, while crystal violet staining was used to assess EPS removal. RESULTS The efficacy of all three enzymes against S. aureus cells and biofilms was significantly improved when they were immobilized onto MSNs. Treatment efficacy was further enhanced when the three enzymes were used in combination against both MRSA and MSSA. Regardless of biofilm maturity (24 or 48 h), near-complete dispersal and killing of MRSA biofilms were observed after treatment with the enzyme-functionalized MSNs. Disruption of mature MSSA biofilms with a polysaccharide EPS was less efficient, but cell viability was significantly reduced. CONCLUSION The combination of these three enzymes and their functionalization onto nanoparticles might extend the therapeutic options for the treatment of S. aureus infections, particularly those with a biofilm component.
Collapse
Affiliation(s)
- Henry Devlin
- UCD School of Chemical and Bioprocess Engineering, University College Dublin, Dublin, Ireland
| | - Stephanie Fulaz
- UCD School of Chemical and Bioprocess Engineering, University College Dublin, Dublin, Ireland
| | - Dishon Wayne Hiebner
- UCD School of Chemical and Bioprocess Engineering, University College Dublin, Dublin, Ireland
| | - James P O’Gara
- Department of Microbiology, School of Natural Sciences, National University of Ireland, Galway, Ireland
| | - Eoin Casey
- UCD School of Chemical and Bioprocess Engineering, University College Dublin, Dublin, Ireland
| |
Collapse
|
20
|
Dadi NCT, Dohál M, Medvecká V, Bujdák J, Koči K, Zahoranová A, Bujdáková H. Physico-Chemical Characterization and Antimicrobial Properties of Hybrid Film Based on Saponite and Phloxine B. Molecules 2021; 26:E325. [PMID: 33435210 PMCID: PMC7827291 DOI: 10.3390/molecules26020325] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Revised: 12/25/2020] [Accepted: 12/29/2020] [Indexed: 11/23/2022] Open
Abstract
This research was aimed at the preparation of a hybrid film based on a layered silicate saponite (Sap) with the immobilized photosensitizer phloxine B (PhB). Sap was selected because of its high cation exchange capacity, ability to exfoliate into nanolayers, and to modify different surfaces. The X-ray diffraction of the films confirmed the intercalation of both the surfactant and PhB molecules in the Sap film. The photosensitizer retained its photoactivity in the hybrid films, as shown by fluorescence spectra measurements. The water contact angles and the measurement of surface free energy demonstrated the hydrophilic nature of the hybrid films. Antimicrobial effectiveness, assessed by the photodynamic inactivation on hybrid films, was tested against a standard strain and against methicillin-resistant bacteria of Staphylococcus aureus (MRSA). One group of samples was irradiated (green LED light; 2.5 h) and compared to nonirradiated ones. S. aureus strains manifested a reduction in growth from 1-log10 to over 3-log10 compared to the control samples with Sap only, and defects in S. aureus cells were proven by scanning electron microscopy. The results proved the optimal photo-physical properties and anti-MRSA potential of this newly designed hybrid system that reflects recent progress in the modification of surfaces for various medical applications.
Collapse
Affiliation(s)
- Nitin Chandra teja Dadi
- Department of Microbiology and Virology, Faculty of Natural Sciences, Comenius University in Bratislava, 842 15 Bratislava, Slovakia; (N.C.t.D.); (M.D.); (K.K.)
| | - Matúš Dohál
- Department of Microbiology and Virology, Faculty of Natural Sciences, Comenius University in Bratislava, 842 15 Bratislava, Slovakia; (N.C.t.D.); (M.D.); (K.K.)
| | - Veronika Medvecká
- Department of Experimental Physics, Faculty of Mathematics, Physics and Informatics, Comenius University in Bratislava, 842 48 Bratislava, Slovakia; (V.M.); (A.Z.)
| | - Juraj Bujdák
- Department of Physical and Theoretical Chemistry, Faculty of Natural Sciences, Comenius University in Bratislava, 842 15 Bratislava, Slovakia;
- Institute of Inorganic Chemistry of SAS, 845 36 Bratislava, Slovakia
| | - Kamila Koči
- Department of Microbiology and Virology, Faculty of Natural Sciences, Comenius University in Bratislava, 842 15 Bratislava, Slovakia; (N.C.t.D.); (M.D.); (K.K.)
| | - Anna Zahoranová
- Department of Experimental Physics, Faculty of Mathematics, Physics and Informatics, Comenius University in Bratislava, 842 48 Bratislava, Slovakia; (V.M.); (A.Z.)
| | - Helena Bujdáková
- Department of Microbiology and Virology, Faculty of Natural Sciences, Comenius University in Bratislava, 842 15 Bratislava, Slovakia; (N.C.t.D.); (M.D.); (K.K.)
| |
Collapse
|
21
|
Heinzinger LR, Johnson A, Wurster JI, Nilson R, Penumutchu S, Belenky P. Oxygen and Metabolism: Digesting Determinants of Antibiotic Susceptibility in the Gut. iScience 2020; 23:101875. [PMID: 33354661 PMCID: PMC7744946 DOI: 10.1016/j.isci.2020.101875] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Microbial metabolism is a major determinant of antibiotic susceptibility. Environmental conditions that modify metabolism, notably oxygen availability and redox potential, can directly fine-tune susceptibility to antibiotics. Despite this, relatively few studies have discussed these modifications within the gastrointestinal tract and their implication on in vivo drug activity and the off-target effects of antibiotics in the gut. In this review, we discuss the environmental and biogeographical complexity of the gastrointestinal tract in regard to oxygen availability and redox potential, addressing how the heterogeneity of gut microhabitats may modify antibiotic activity in vivo. We contextualize the current literature surrounding oxygen availability and antibiotic efficacy and discuss empirical treatments. We end by discussing predicted patterns of antibiotic activity in prominent microbiome taxa, given gut heterogeneity, oxygen availability, and polymicrobial interactions. We also propose additional work required to fully elucidate the role of oxygen metabolism on antibiotic susceptibility in the context of the gut.
Collapse
Affiliation(s)
- Lauren R. Heinzinger
- Department of Microbiology and Immunology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, The State University of New York, Buffalo, NY 14214, USA
| | - Angus Johnson
- Department of Biological Science, Binghamton University, Binghamton, NY 13902, USA
| | - Jenna I. Wurster
- Department of Molecular Microbiology and Immunology, Brown University, Providence, RI 02912, USA
| | - Rachael Nilson
- Department of Molecular Microbiology and Immunology, Brown University, Providence, RI 02912, USA
| | - Swathi Penumutchu
- Department of Molecular Microbiology and Immunology, Brown University, Providence, RI 02912, USA
| | - Peter Belenky
- Department of Molecular Microbiology and Immunology, Brown University, Providence, RI 02912, USA
| |
Collapse
|
22
|
Xu J, Yao H, Wang S, Li H, Hou X. Mangiferin Inhibits Apoptosis and Autophagy Induced by Staphylococcus aureus in RAW264.7 Cells. J Inflamm Res 2020; 13:847-857. [PMID: 33177860 PMCID: PMC7650040 DOI: 10.2147/jir.s280091] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Accepted: 10/13/2020] [Indexed: 12/22/2022] Open
Abstract
Purpose Staphylococcus aureus (S. aureus) is an important bacterial pathogen, which creates infective inflammation to human being and animals. Mangiferin (MG) is one of the natural flavonoids with anti-inflammatory, anti-bacterial, and anti-oxidative properties. However, the anti-apoptosis and anti-autophagy of MG are unknown. Hence, this study was aimed to research the inhibition of MG on S. aureus-induced apoptosis and autophagy in RAW264.7 cells. Methods The RAW264.7 cells were pretreated with MG, or pretreated with SP600125 or anisomycin synchronously, and then infected with S. aureus (MOI=100:1). The viability and proliferation status of RAW264.7 cells were detected by MTT and EdU assay. The relative expression of TNF-α, IL-6 and IL-10 protein was tested with ELISA. The levels of Bax, Bcl-2, caspase-3, c-Jun N-terminal kinase (JNK), extracellular-regulated protein kinase (ERK), p38, LC3, Beclin-1, p62, phosphorylated JNK, phosphorylated p38 and phosphorylated ERK in cells were detected by Western blotting. The apoptosis rate of RAW264.7 cells was analyzed by flow cytometric assay. Results The study showed that MG significantly attenuated RAW264.7 cells apoptosis and autophagy caused by S. aureus. MG alleviated S. aureus-induced apoptosis by down-regulating the protein level of active caspase-3 and Bax and up-regulating the level of Bcl-2. MG also inhibited S. aureus-induced autophagy via decreasing the protein level of LC3-II/LC3-I and Beclin-1 or increasing the protein expression of p62. This protective role was dependent on the up-regulation of JNK signal pathway, which was confirmed by using JNK agonist and inhibitor. Conclusion Our results demonstrated that MG might protect RAW264.7 cells from S. aureus-induced apoptosis and autophagy via inhibiting JNK/Bax-dependent signal pathway. Therefore, MG may be a potential agent against pathological cell damage induced by S. aureus infection.
Collapse
Affiliation(s)
- Jun Xu
- Beijing Key Laboratory of Traditional Chinese Veterinary Medicine, National Demonstration Center for Experimental Animal Education, Department of Veterinary Medicine, Beijing University of Agriculture, Beijing, People's Republic of China
| | - Hua Yao
- Beijing Key Laboratory of Traditional Chinese Veterinary Medicine, National Demonstration Center for Experimental Animal Education, Department of Veterinary Medicine, Beijing University of Agriculture, Beijing, People's Republic of China
| | - Shichen Wang
- Beijing Key Laboratory of Traditional Chinese Veterinary Medicine, National Demonstration Center for Experimental Animal Education, Department of Veterinary Medicine, Beijing University of Agriculture, Beijing, People's Republic of China
| | - Huanrong Li
- Beijing Key Laboratory of Traditional Chinese Veterinary Medicine, National Demonstration Center for Experimental Animal Education, Department of Veterinary Medicine, Beijing University of Agriculture, Beijing, People's Republic of China
| | - Xiaolin Hou
- Beijing Key Laboratory of Traditional Chinese Veterinary Medicine, National Demonstration Center for Experimental Animal Education, Department of Veterinary Medicine, Beijing University of Agriculture, Beijing, People's Republic of China
| |
Collapse
|
23
|
Khan F, Pham DTN, Tabassum N, Oloketuyi SF, Kim YM. Treatment strategies targeting persister cell formation in bacterial pathogens. Crit Rev Microbiol 2020; 46:665-688. [DOI: 10.1080/1040841x.2020.1822278] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Affiliation(s)
- Fazlurrahman Khan
- Institute of Food Science, Pukyong National University, Busan, Korea
| | - Dung Thuy Nguyen Pham
- Department of Food Science and Technology, Pukyong National University, Busan, Korea
| | - Nazia Tabassum
- Industrial Convergence Bionix Engineering, Pukyong National University, Busan, Korea
| | | | - Young-Mog Kim
- Institute of Food Science, Pukyong National University, Busan, Korea
- Department of Food Science and Technology, Pukyong National University, Busan, Korea
| |
Collapse
|
24
|
Lacoma A, Usón L, Mendoza G, Sebastián V, Garcia-Garcia E, Muriel-Moreno B, Domínguez J, Arruebo M, Prat C. Novel intracellular antibiotic delivery system against Staphylococcus aureus: cloxacillin-loaded poly(d,l-lactide-co-glycolide) acid nanoparticles. Nanomedicine (Lond) 2020; 15:1189-1203. [DOI: 10.2217/nnm-2019-0371] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Aim: First, to compare in vitro minimum inhibitory concentrations (MIC) of free cloxacillin and cloxacillin-containing nanoparticles (NP) against methicillin-susceptible (MSSA) and resistant Staphylococcus aureus (MRSA) and second, to assess NP antimicrobial activity against intracellular S. aureus. Methods: Poly(d,l-lactide-co-glycolide) acid (PLGA)-NP were loaded with cloxacillin and physico-chemically characterized. MICs were determined for reference strains Newman-(MSSA) and USA300-(MRSA). Murine alveolar macrophages were infected, and bacterial intracellular survival was assessed after incubating with free-cloxacillin or PLGA-cloxacillin-NP. Results & conclusion: For both isolates, MICs for antibiotic-loaded-NP were lower than those obtained with free cloxacillin, indicating that the drug encapsulation improves antimicrobial activity. A sustained antibiotic release was demonstrated when using the PLGA-cloxacillin-NP. When considering the lowest concentrations, the use of drug-loaded NP enabled a higher reduction of intracellular bacterial load.
Collapse
Affiliation(s)
- Alicia Lacoma
- Microbiology Department, Hospital Universitari Germans Trias i Pujol, Fundació Institut d’Investigació en Ciències de la Salut GermansTrias i Pujol, Universitat Autònoma de Barcelona, Badalona, Spain
- CIBER Enfermedades Respiratorias (CIBERES), Instituto de Salud Carlos III, Spain
| | - Laura Usón
- Institute of Nanoscience of Aragon (INA), Department of Chemical Engineering & Environmental Technologies, University of Zaragoza & Instituto de Ciencia de Materiales de Aragón (ICMA), Universidad de Zaragoza-CSIC, 50018, Zaragoza, Spain
- CIBER Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Instituto de Salud Carlos III, Spain
| | - Gracia Mendoza
- Institute of Nanoscience of Aragon (INA), Department of Chemical Engineering & Environmental Technologies, University of Zaragoza & Instituto de Ciencia de Materiales de Aragón (ICMA), Universidad de Zaragoza-CSIC, 50018, Zaragoza, Spain
- CIBER Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Instituto de Salud Carlos III, Spain
| | - Victor Sebastián
- Institute of Nanoscience of Aragon (INA), Department of Chemical Engineering & Environmental Technologies, University of Zaragoza & Instituto de Ciencia de Materiales de Aragón (ICMA), Universidad de Zaragoza-CSIC, 50018, Zaragoza, Spain
- CIBER Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Instituto de Salud Carlos III, Spain
| | - Esther Garcia-Garcia
- Microbiology Department, Hospital Universitari Germans Trias i Pujol, Fundació Institut d’Investigació en Ciències de la Salut GermansTrias i Pujol, Universitat Autònoma de Barcelona, Badalona, Spain
| | - Beatriz Muriel-Moreno
- Microbiology Department, Hospital Universitari Germans Trias i Pujol, Fundació Institut d’Investigació en Ciències de la Salut GermansTrias i Pujol, Universitat Autònoma de Barcelona, Badalona, Spain
| | - Jose Domínguez
- Microbiology Department, Hospital Universitari Germans Trias i Pujol, Fundació Institut d’Investigació en Ciències de la Salut GermansTrias i Pujol, Universitat Autònoma de Barcelona, Badalona, Spain
- CIBER Enfermedades Respiratorias (CIBERES), Instituto de Salud Carlos III, Spain
| | - Manuel Arruebo
- Institute of Nanoscience of Aragon (INA), Department of Chemical Engineering & Environmental Technologies, University of Zaragoza & Instituto de Ciencia de Materiales de Aragón (ICMA), Universidad de Zaragoza-CSIC, 50018, Zaragoza, Spain
- CIBER Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Instituto de Salud Carlos III, Spain
| | - Cristina Prat
- Microbiology Department, Hospital Universitari Germans Trias i Pujol, Fundació Institut d’Investigació en Ciències de la Salut GermansTrias i Pujol, Universitat Autònoma de Barcelona, Badalona, Spain
- CIBER Enfermedades Respiratorias (CIBERES), Instituto de Salud Carlos III, Spain
- Julius Centre for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| |
Collapse
|
25
|
Abstract
Aim To demonstrate that myrrh oil preferentially kills nongrowing bacteria and causes no resistance development. Method Growth inhibition was determined on regular plates or plates without nutrients, which were later overlaid with soft agar containing nutrients to continue growth. Killing experiments were done in broth and in buffer without nutrients. Results Bacterial cells were inhibited preferentially in the absence of nutrients or when growth was halted by a bacteriostatic antibiotic. After five passages in myrrh oil, surviving colonies showed no resistance to the antibiotic. Conclusion Myrrh oil has the potential to be a commercially viable antibiotic that kills persister cells and causes no resistance development. This is a rare example of an antibiotic that can preferentially kill nongrowing bacteria.
Collapse
|
26
|
Lu K, Hou W, Xu XF, Chen Q, Li Z, Lin J, Chen WM. Biological evaluation and chemoproteomics reveal potential antibacterial targets of a cajaninstilbene-acid analogue. Eur J Med Chem 2020; 188:112026. [DOI: 10.1016/j.ejmech.2019.112026] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2019] [Revised: 12/29/2019] [Accepted: 12/29/2019] [Indexed: 12/17/2022]
|
27
|
She P, Li S, Zhou L, Luo Z, Liao J, Xu L, Zeng X, Chen T, Liu Y, Wu Y. Insights into idarubicin antimicrobial activity against methicillin-resistant Staphylococcus aureus. Virulence 2020; 11:636-651. [PMID: 32423280 PMCID: PMC7549941 DOI: 10.1080/21505594.2020.1770493] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2019] [Revised: 02/02/2020] [Accepted: 02/14/2020] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND MRSA is a major concern in community settings and in health care. The emergence of biofilms and persister cells substantially increases its antimicrobial resistance. It is very urgent to develop new antimicrobials to solve this problem. OBJECTIVE Idarubicin was profiled to assess its antimicrobial effects in vitro and in vivo, and the underlying mechanisms. METHODS We investigated the antimicrobial effects of idarubicin against MRSA by time-kill analysis. The antibiofilm efficacy of idarubicin was assessed by crystal violet and XTT staining, followed by laser confocal microscopy observation. The mechanisms underlying the antimicrobial effects were studied by transmission electron microscopy, all-atom molecular dynamic simulations, SYTOX staining, surface plasma resonance, and DNA gyrase inhibition assay. Further, we addressed the antimicrobial efficacy in wound and subcutaneous abscess infection in vivo. RESULTS Idarubicin kills MRSA cells by disrupting the lipid bilayers and interrupting the DNA topoisomerase IIA subunits, and idarubicin shows synergistic antimicrobial effects with fosfomycin. Through synergy with a single dose treatment fosfomycin and the addition of the cell protector amifostine, the cytotoxicity and cardiotoxicity of idarubicin were significantly reduced without affecting its antimicrobial effects. Idarubicin alone or in combination with fosfomycin exhibited considerable efficacy in a subcutaneous abscess mouse model of MRSA infection. In addition, idarubicin also showed a low probability of causing resistance and good postantibiotic effects. CONCLUSIONS Idarubicin and its analogs have the potential to become a new class of antimicrobials for the treatment of MRSA-related infections.
Collapse
Affiliation(s)
- Pengfei She
- Department of Clinical Laboratory, The Third Xiangya Hospital of Central South University, Changsha, R.P. China
| | - Shijia Li
- Department of Clinical Laboratory, The Third Xiangya Hospital of Central South University, Changsha, R.P. China
| | - Linying Zhou
- Department of Clinical Laboratory, The Third Xiangya Hospital of Central South University, Changsha, R.P. China
| | - Zhen Luo
- Department of Clinical Laboratory, The Third Xiangya Hospital of Central South University, Changsha, R.P. China
| | - Jinfeng Liao
- Department of Clinical Laboratory, The Third Xiangya Hospital of Central South University, Changsha, R.P. China
| | - Lanlan Xu
- Department of Clinical Laboratory, The Third Xiangya Hospital of Central South University, Changsha, R.P. China
| | - Xianghai Zeng
- Department of Clinical Laboratory, The Third Xiangya Hospital of Central South University, Changsha, R.P. China
| | - Ti Chen
- Department of Clinical Laboratory, The Third Xiangya Hospital of Central South University, Changsha, R.P. China
| | - Yaqian Liu
- Department of Clinical Laboratory, The Third Xiangya Hospital of Central South University, Changsha, R.P. China
| | - Yong Wu
- Department of Clinical Laboratory, The Third Xiangya Hospital of Central South University, Changsha, R.P. China
| |
Collapse
|
28
|
Synthesis and structure-activity relationships of novel abietane diterpenoids with activity against Staphylococcus aureus. Future Med Chem 2019; 11:3109-3124. [PMID: 31838897 DOI: 10.4155/fmc-2019-0192] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Aim: To find alternative compounds against methicillin-resistant Staphylococcus aureus (MRSA) and methicillin-susceptible S. aureus (MSSA), novel derivatives from dehydroabietic acid were synthesized. Methods & results: Compound 12 was the most effective against 15 MRSA and 11 MSSA with minimum inhibitory concentration values ranging from 3.9 to 15.6 μg/ml. Although less active than 12, compound 11, followed by 25 and 13, also exhibited anti-staphylococcal activity. Additional studies showed that compound 12 is devoid of toxic effect on non-target cells. A structure-activity relationship study revealed that an oxime at C-13 together with a hydroxyl at C-12 could play a key role in the activity. Conclusion: These structures, in particular compound 12, could arise as templates for the development of agents against MRSA and MSSA.
Collapse
|
29
|
Wang X, Chen J, Wang W, Jaunarajs A, Wang X. Tryptoline-based benzothiazoles re-sensitize MRSA to β-lactam antibiotics. Bioorg Med Chem 2019; 27:115095. [PMID: 31521461 PMCID: PMC6779328 DOI: 10.1016/j.bmc.2019.115095] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2019] [Revised: 09/04/2019] [Accepted: 09/06/2019] [Indexed: 01/03/2023]
Abstract
Resistance-modifying agents (RMAs) offer a promising solution to combat bacterial antibiotic resistance. Here we report the discovery and structure-activity relationships of a new class of RMAs with a novel tryptoline-based benzothiazole scaffold. Our most potent compound in this series (4ad) re-sensitizes multiple MRSA strains to cephalosporins at low concentrations (2 μg/mL) and has low mammalian cytotoxicity with a half growth inhibitory concentration (GI50) > 100 μg/mL in human cervical carcinoma (HeLa) cells. In addition, the same core scaffold with different substitutions also gives good antibacterial activity against MRSA.
Collapse
Affiliation(s)
- Xinfeng Wang
- Department of Chemistry, University of Colorado, Boulder, CO 80309, United States
| | - Jinsen Chen
- Department of Chemistry, University of Colorado, Boulder, CO 80309, United States
| | - Wei Wang
- Department of Chemistry, University of Colorado, Boulder, CO 80309, United States
| | - Anna Jaunarajs
- Department of Chemistry, University of Colorado, Boulder, CO 80309, United States
| | - Xiang Wang
- Department of Chemistry, University of Colorado, Boulder, CO 80309, United States.
| |
Collapse
|
30
|
Antimicrobial Activity of Exebacase (Lysin CF-301) against the Most Common Causes of Infective Endocarditis. Antimicrob Agents Chemother 2019; 63:AAC.01078-19. [PMID: 31332073 DOI: 10.1128/aac.01078-19] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2019] [Accepted: 07/16/2019] [Indexed: 12/22/2022] Open
Abstract
Exebacase, a recombinantly produced lysin (cell wall hydrolase), and comparator antibiotics were tested by the broth microdilution method against strain sets of Staphylococcus and Streptococcus spp., which are the most common causes of infective endocarditis in humans. Exebacase was active against all Staphylococcus spp. tested, including S. aureus and coagulase-negative staphylococci (MIC50/90, 0.5/1 μg/ml). Activity against Streptococcus spp. was variable, with S. pyogenes, S. agalactiae, and S. dysgalactiae (MIC50/90, 1/2 μg/ml) among the most susceptible.
Collapse
|
31
|
A Phase 1, Randomized, Single-Ascending-Dose Study To Investigate the Safety, Tolerability, and Pharmacokinetics of DSTA4637S, an Anti- Staphylococcus aureus Thiomab Antibody-Antibiotic Conjugate, in Healthy Volunteers. Antimicrob Agents Chemother 2019; 63:AAC.02588-18. [PMID: 30910894 DOI: 10.1128/aac.02588-18] [Citation(s) in RCA: 69] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2018] [Accepted: 03/17/2019] [Indexed: 01/10/2023] Open
Abstract
Staphylococcus aureus causes serious bacterial infections with high morbidity and mortality, necessitating the discovery of new antibiotics. DSTA4637S is a novel antibody-antibiotic conjugate designed to target intracellular S. aureus that is not adequately eliminated by current standard-of-care antibiotics. DSTA4637S is composed of an anti-S. aureus Thiomab human immunoglobulin G1 (IgG1) monoclonal antibody linked to a novel rifamycin-class antibiotic (4-dimethylaminopiperidino-hydroxybenzoxazino rifamycin [dmDNA31]) via a protease-cleavable linker. Phagocytic cells ingest DSTA4637S-bound S. aureus, and intracellular cathepsins cleave the linker, releasing dmDNA31and killing intracellular S. aureus This first-in-human, randomized, double-blind, placebo-controlled, single-ascending-dose phase 1 trial analyzed the safety, pharmacokinetics, and immunogenicity of DSTA4637S in healthy volunteers. Thirty healthy male and female volunteers, 18-65 years old, were randomized into five cohorts receiving single intravenous (i.v.) doses of 5, 15, 50, 100, and 150 mg/kg of DSTA4637S or placebo (4 active:2 placebo). Subjects were followed for 85 days after dosing. No subject withdrew from the study, and no serious or severe adverse events occurred. One moderate infusion-related reaction (150 mg/kg DSTA4637S) occurred. No clinically meaningful or dose-related changes in laboratory parameters or vital signs occurred. Pharmacokinetics of plasma DSTA4637S conjugate and serum DSTA4637S total antibody were dose proportional. Systemic exposure of unconjugated dmDNA31 was low. No DSTA4637S-induced anti-drug antibody responses were observed. DSTA4637S was generally safe and well tolerated as a single i.v. dose in healthy volunteers. DSTA4637S has a favorable safety and pharmacokinetic profile that supports future development as a novel therapeutic for S. aureus infections. (This study has been registered at ClinicalTrials.gov under identifier NCT02596399.).
Collapse
|