1
|
Renz J, Dauda KA, Aga ONL, Diaz-Uriarte R, Löhr IH, Blomberg B, Johnston IG. Evolutionary accumulation modeling in AMR: machine learning to infer and predict evolutionary dynamics of multi-drug resistance. mBio 2025:e0048825. [PMID: 40396716 DOI: 10.1128/mbio.00488-25] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/22/2025] Open
Abstract
Can we understand and predict the evolutionary pathways by which bacteria acquire multi-drug resistance (MDR)? These questions have substantial potential impact in basic biology and in applied approaches to address the global health challenge of antimicrobial resistance (AMR). In this minireview, we discuss how a class of machine-learning approaches called evolutionary accumulation modeling (EvAM) may help reveal these dynamics using genetic and/or phenotypic AMR data sets, without requiring longitudinal sampling. These approaches are well-established in cancer progression and evolutionary biology but currently less used in AMR research. We discuss how EvAM can learn the evolutionary pathways by which drug resistances and other AMR features (for example, mutations driving these resistances) are acquired as pathogens evolve, predict next evolutionary steps, identify influences between AMR features, and explore differences in MDR evolution between regions, demographics, and more. We demonstrate a case study from the literature on MDR evolution in Mycobacterium tuberculosis and discuss the strengths and weaknesses of these approaches, providing links to some approaches for implementation.
Collapse
Affiliation(s)
- Jessica Renz
- Department of Mathematics, University of Bergen, Bergen, Norway
| | - Kazeem A Dauda
- Department of Mathematics, University of Bergen, Bergen, Norway
| | - Olav N L Aga
- Computational Biology Unit, University of Bergen, Bergen, Norway
- Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Ramon Diaz-Uriarte
- Department of Biochemistry, School of Medicine, Universidad Autónoma de Madrid, Madrid, Community of Madrid, Spain
- Instituto de Investigaciones Biomédicas Sols-Morreale (IIBM), CSIC-UAM, Madrid, Community of Madrid, Spain
| | - Iren H Löhr
- Department of Clinical Science, University of Bergen, Bergen, Norway
- Department of Medical Microbiology, Stavanger University Hospital, Stavanger, Norway
| | - Bjørn Blomberg
- Department of Clinical Science, University of Bergen, Bergen, Norway
- Department of Medicine, Haukeland University Hospital, Bergen, Norway
- National Advisory Unit for Tropical Infectious Diseases, Haukeland University Hospital, Bergen, Norway
| | - Iain G Johnston
- Department of Mathematics, University of Bergen, Bergen, Norway
- Computational Biology Unit, University of Bergen, Bergen, Norway
| |
Collapse
|
2
|
Liu C, Wu Y, Zhang Y, Yan Z, Gu D, Zhou H, Dong N, Cai C, Chen G, Zhang R. Effectiveness of antimicrobial agent combinations against carbapenem-producing Klebsiella pneumoniae with KPC variants in China. Front Microbiol 2025; 15:1519319. [PMID: 39911707 PMCID: PMC11794265 DOI: 10.3389/fmicb.2024.1519319] [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: 10/29/2024] [Accepted: 12/30/2024] [Indexed: 02/07/2025] Open
Abstract
Purpose Carbapenem-resistant Klebsiella pneumoniae (CRKP) producing carbapenemases poses a global threat to public health. Antimicrobial agent combinations have been promoted as a potential therapeutic strategy for infections. The most effective antimicrobial combinations against CRKP strains producing different carbapenemases are currently unclear, particularly those producing the KPC variant carbapenemases. This study is aimed to evaluate the effectiveness of various antimicrobial agent combinations against CRKP strains with different carbapenemases. Methods A checkerboard assay involving 24 antimicrobial agent combinations was conducted on 44 strains of carbapenemase-producing CRKP isolated from patients of which 13 CRKP strains carried single KPC variants. The 24 antimicrobial combinations were based on meropenem, polymyxin, tigecycline, ceftazidime/avibactam, respectively. The fractional inhibitory concentration (FIC) indexes were calculated for each combination of antimicrobial agents. Results The distribution of carbapenemases in 44 CRKP strains was as follows: KPC variants (n = 13, 29.5%), KPC-2 (n = 10, 22.7%), metallo-β-lactamases (n = 9, 20.5%), OXA-48-like (n = 12, 27.3%). In the checkerboard assay, the combination of polymyxin and aztreonam exhibited the highest synergistic effect against CRKP strains, with a rate of 95.5% (42/44). This was followed by polymyxin-meropenem at 88.6% (39/44) and polymyxin-levofloxacin at 68.2% (30/44). Additionally, polymyxin-aztreonam combination and polymyxin-meropenem showed the highest sum of synergistic and additive rates of 100.0% against KPC variant-producing CRKP strains. Notably, ceftazidime/avibactam-based combinations exhibited better synergistic effects on KPC variant-producing CRKP strains compared to other CRKP strains with adjusted p value <0.05. Conclusion Our study suggests that the combinations of antimicrobial agent could serve as potential treatment strategies against CRKP infections. Furthermore, the effectiveness of these combinations is influenced by the types of carbapenemases present. Ceftazidime/avibactam-based combinations have showed superior synergistic effects on KPC variant-producing CRKP strains.
Collapse
Affiliation(s)
- Congcong Liu
- Department of Clinical Laboratory, Tianjin First Central Hospital, School of Medicine, Nankai University, Tianjin, China
- Department of Clinical Laboratory, Second Affiliated Hospital of Zhejiang University, School of Medicine, Hangzhou, China
| | - Yuchen Wu
- Department of Clinical Laboratory, Second Affiliated Hospital of Zhejiang University, School of Medicine, Hangzhou, China
| | - Yanyan Zhang
- Department of Clinical Laboratory, Second Affiliated Hospital of Zhejiang University, School of Medicine, Hangzhou, China
| | - Zelin Yan
- Department of Clinical Laboratory, Second Affiliated Hospital of Zhejiang University, School of Medicine, Hangzhou, China
| | - Danxia Gu
- Department of Clinical Laboratory, Second Affiliated Hospital of Zhejiang University, School of Medicine, Hangzhou, China
- Department of Clinical Laboratory, Zhejiang Provincial People's Hospital, Hangzhou, China
| | - Hongwei Zhou
- Department of Clinical Laboratory, Second Affiliated Hospital of Zhejiang University, School of Medicine, Hangzhou, China
| | - Ning Dong
- MOE Key Laboratory of Geriatric Diseases and Immunology, Suzhou Key Laboratory of Pathogen Bioscience and Anti-infective Medicine, School of Biology & Basic Medical Sciences, Suzhou Medical College, Soochow University, Suzhou, China
| | - Chang Cai
- College of Animal Science and Technology, College of Veterinary Medicine, Zhejiang Agricultural and Forestry University, Hangzhou, China
| | - Gongxiang Chen
- Department of Clinical Laboratory, Second Affiliated Hospital of Zhejiang University, School of Medicine, Hangzhou, China
| | - Rong Zhang
- Department of Clinical Laboratory, Second Affiliated Hospital of Zhejiang University, School of Medicine, Hangzhou, China
| |
Collapse
|
3
|
Siedentop B, Kachalov VN, Witzany C, Egger M, Kouyos RD, Bonhoeffer S. The effect of combining antibiotics on resistance: A systematic review and meta-analysis. eLife 2024; 13:RP93740. [PMID: 39704726 PMCID: PMC11661791 DOI: 10.7554/elife.93740] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2024] Open
Abstract
Background Under which conditions antibiotic combination therapy decelerates rather than accelerates resistance evolution is not well understood. We examined the effect of combining antibiotics on within-patient resistance development across various bacterial pathogens and antibiotics. Methods We searched CENTRAL, EMBASE, and PubMed for (quasi)-randomised controlled trials (RCTs) published from database inception to 24 November 2022. Trials comparing antibiotic treatments with different numbers of antibiotics were included. Patients were considered to have acquired resistance if, at the follow-up culture, a resistant bacterium (as defined by the study authors) was detected that had not been present in the baseline culture. We combined results using a random effects model and performed meta-regression and stratified analyses. The trials' risk of bias was assessed with the Cochrane tool. Results 42 trials were eligible and 29, including 5054 patients, qualified for statistical analysis. In most trials, resistance development was not the primary outcome and studies lacked power. The combined odds ratio for the acquisition of resistance comparing the group with the higher number of antibiotics with the comparison group was 1.23 (95% CI 0.68-2.25), with substantial between-study heterogeneity (I2=77%). We identified tentative evidence for potential beneficial or detrimental effects of antibiotic combination therapy for specific pathogens or medical conditions. Conclusions The evidence for combining a higher number of antibiotics compared to fewer from RCTs is scarce and overall compatible with both benefit or harm. Trials powered to detect differences in resistance development or well-designed observational studies are required to clarify the impact of combination therapy on resistance. Funding Support from the Swiss National Science Foundation (grant 310030B_176401 (SB, BS, CW), grant 32FP30-174281 (ME), grant 324730_207957 (RDK)) and from the National Institute of Allergy and Infectious Diseases (NIAID, cooperative agreement AI069924 (ME)) is gratefully acknowledged.
Collapse
Affiliation(s)
- Berit Siedentop
- Institute of Integrative Biology, Department of Environmental Systems Science, ETH ZürichZurichSwitzerland
- Department of Infectious Diseases and Hospital Epidemiology, University Hospital Zurich, University of ZurichZurichSwitzerland
| | - Viacheslav N Kachalov
- Department of Infectious Diseases and Hospital Epidemiology, University Hospital Zurich, University of ZurichZurichSwitzerland
- Institute of Medical Virology, University of ZurichZurichSwitzerland
| | - Christopher Witzany
- Institute of Integrative Biology, Department of Environmental Systems Science, ETH ZürichZurichSwitzerland
| | - Matthias Egger
- Institute of Social and Preventive Medicine (ISPM), University of BernBernSwitzerland
- Population Health Sciences, University of BristolBristolUnited Kingdom
- Centre for Infectious Disease Epidemiology and Research, Faculty of Health Sciences, University of Cape TownCape TownSouth Africa
| | - Roger D Kouyos
- Department of Infectious Diseases and Hospital Epidemiology, University Hospital Zurich, University of ZurichZurichSwitzerland
- Institute of Medical Virology, University of ZurichZurichSwitzerland
| | - Sebastian Bonhoeffer
- Institute of Integrative Biology, Department of Environmental Systems Science, ETH ZürichZurichSwitzerland
| |
Collapse
|
4
|
Min YH, Kim YU, Park MC. In Vitro Synergistic Effect of Lefamulin with Doxycycline, Rifampin, and Quinupristin/Dalfopristin Against Enterococci. Microorganisms 2024; 12:2515. [PMID: 39770718 PMCID: PMC11678898 DOI: 10.3390/microorganisms12122515] [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: 11/05/2024] [Revised: 12/01/2024] [Accepted: 12/04/2024] [Indexed: 01/11/2025] Open
Abstract
The widespread resistance of enterococci to many commonly used antimicrobial agents is a growing concern. Given that the current treatment options for enterococcal infections are limited, the discovery of new therapies, including combination therapies, is necessary. We evaluated double-drug combinations of lefamulin with doxycycline, rifampin, and quinupristin/dalfopristin for in vitro synergy against strains of Enterococcus faecium (E. faecium) and Enterococcus faecalis (E. faecalis) by using checkerboard and time-kill assays. In the checkerboard assay, the synergistic effect of lefamulin with doxycycline and rifampin was observed in 29 (85.3%) and 33 (97.1%) of the 34 different E. faecium strains tested, respectively. These combinations also showed synergistic effects against 17 (94.4%) of the 18 different vancomycin-resistant E. faecium strains. Among the 33 different E. faecalis strains, the combination of lefamulin with doxycycline, quinupristin/dalfopristin, and rifampin displayed synergistic effects in 31 (93.9%), 26 (78.8%), and 20 (60.6%) strains, respectively. No antagonism was observed in any of the combinations. The time-kill assay confirmed the synergistic effects of all these combinations. These synergistic combinations exhibited bacteriostatic activity. Although lefamulin is not currently used to treat enterococcal infections, we suggest that these combinations may serve as alternative drug regimens.
Collapse
Affiliation(s)
- Yu-Hong Min
- College of K-Bio Health, Daegu Haany University, Gyeongsan 38610, Republic of Korea;
| | - Yong-ung Kim
- Department of Pharmaceutical Engineering, Daegu Haany University, Gyeongsan 38610, Republic of Korea;
| | - Min Chul Park
- College of Pharmacy and Inje Institute of Pharmaceutical Sciences and Research, Inje University, Gimhae 50832, Republic of Korea
| |
Collapse
|
5
|
Nazari M, Taheri M, Nouri F, Bahmanzadeh M, Alikhani MY. The antimicrobial and antibiofilm effects of gentamicin, imipenem, and fucoidan combinations against dual-species biofilms of Staphylococcus aureus and Acinetobacter baumannii isolated from diabetic foot ulcers. Ann Clin Microbiol Antimicrob 2024; 23:101. [PMID: 39548455 PMCID: PMC11568526 DOI: 10.1186/s12941-024-00760-w] [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: 08/19/2024] [Accepted: 11/05/2024] [Indexed: 11/18/2024] Open
Abstract
INTRODUCTION Diabetes mellitus is a chronic metabolic disorder characterized by persistent hyperglycemia due to impaired insulin production or utilization, leading to severe health complications. Diabetic foot ulcers (DFUs) represent a major complication, often exacerbated by polymicrobial infections involving Staphylococcus aureus and Acinetobacter baumannii. These pathogens, notorious for their resistance to antibiotics, complicate treatment efforts, especially due to biofilm formation, which enhances bacterial survival and resistance. This study explores the synergistic effects of combining gentamicin, imipenem, and fucoidan, a sulfated polysaccharide with antimicrobial properties, against both planktonic and biofilm forms of S. aureus and A. baumannii. METHODS Isolates of S. aureus and A. baumannii were collected from DFUs and genetically confirmed. Methicillin resistance in S. aureus was identified through disk diffusion and PCR. Biofilm formation, including dual-species biofilms, was analyzed using the microtiter plate method. The antimicrobial efficacy of gentamicin, imipenem, and fucoidan was assessed by determining the minimum inhibitory concentration (MIC), minimum bactericidal concentration (MBC), minimum biofilm inhibitory concentration (MBIC), and minimum biofilm eradication concentration (MBEC). Synergistic interactions were evaluated using the fractional inhibitory concentration index (FICi) and fractional bactericidal concentration index (FBCi). The expression of biofilm-associated genes (icaA in S. aureus and bap in A. baumannii) was analyzed, and the cytotoxicity of fucoidan was assessed. RESULTS The study revealed that 77.4% of S. aureus and all A. baumannii isolates showed multidrug resistance. Among 837 tested conditions for dual-species biofilm formation, 72 resulted in strong biofilm formation and 67 in moderate biofilm formation. The geometric mean MIC values for gentamicin were 12.2 µg/mL for S. aureus, 22.62 µg/mL for A. baumannii, and 5.87 µg/mL for their co-culture; for imipenem, they were 19.84, 9.18, and 3.70 µg/mL, respectively, and for fucoidan, 48.50, 31.20, and 19.65 µg/mL, respectively. The MBC values for gentamicin were 119.42, 128, and 11.75 µg/mL; for imipenem, they were 48.50, 14.92, and 8 µg/mL; and for fucoidan, they were 88.37, 62.62, and 42.48 µg/mL. The MBIC values were 55.71, 119.42, and 18.66 µg/mL for gentamicin; 68.59, 48.50, and 25.39 µg/mL for imipenem; and 153.89, 101.49, and 53.53 µg/mL for fucoidan. The MBEC values were 315.17, 362.03, and 59.25 µg/mL for gentamicin; 207.93, 157.58, and 74.65 µg/mL for imipenem; and 353.55, 189.46, and 99.19 µg/mL for fucoidan. When cultured in planktonic form, the geometric mean FICi and FBCi values indicated additive effects, while co-culture showed FICi values of ≤ 0.5, suggesting a synergistic interaction. Treatment with gentamicin and fucoidan led to significant downregulation of the icaA and bap genes in both single-species and dual-species biofilms of S. aureus and A. baumannii. The reductions in gene expression were more pronounced in dual-species biofilms compared to single-species biofilms. Additionally, treatment with imipenem and fucoidan also resulted in significant downregulation of these genes in both biofilm types. Cytotoxicity assessments indicated that higher concentrations of fucoidan were toxic, yet no harmful effects were noted at the optimal synergistic concentrations used with antibiotics. CONCLUSION In our investigation, we found that combining gentamicin, imipenem, and fucoidan had a synergistic effect on dual-species biofilms of S. aureus and A. baumannii, suggesting potential benefits for treating such infections effectively. This underscores the importance of understanding microbial interactions, antibiotic susceptibility, and biofilm formation in DFUs.
Collapse
Affiliation(s)
- Mohsen Nazari
- Department of Microbiology, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Mohammad Taheri
- Department of Microbiology, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Fatemeh Nouri
- Department of Pharmaceutical Biotechnology, School of Pharmacy, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Maryam Bahmanzadeh
- Department of Anatomical Sciences, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
- Fertility and Infertility Research Center, Avicenna Institute of Clinical Sciences, Avicenna Health Research Institute , Hamadan University of Medical Sciences, Hamadan, Iran
| | - Mohammad Yousef Alikhani
- Department of Microbiology, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran.
- Infectious Disease Research Center, Avicenna Institute of Clinical Sciences, Avicenna Health Research Institute , Hamadan University of Medical Sciences, P.O. Box: 6517838678, Hamadan, Iran.
| |
Collapse
|
6
|
Wang M, Li D, Liu X, Chen C, Frey B, Sui X, Li MH. Global hierarchical meta-analysis to identify the factors for controlling effects of antibiotics on soil microbiota. ENVIRONMENT INTERNATIONAL 2024; 192:109038. [PMID: 39357259 DOI: 10.1016/j.envint.2024.109038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/16/2024] [Revised: 09/16/2024] [Accepted: 09/26/2024] [Indexed: 10/04/2024]
Abstract
It is widely known that antibiotics can affect the structure and function of soil microbial communities, but the specific degree of impact and controlled factors on different indicators remain inconclusive. We conducted a multiple hierarchical mixed effects meta-analysis on 2564 observations that were extracted from 60 publications, to comprehensively assess the impact of antibiotics on soil microbiota. The results showed that antibiotics had significant negative effects on soil microbial biomass, α-diversity and soil enzyme activity. Under neutral initial soil, when soil was derived from agricultural land or had a fine-textured, the negative impacts of antibiotics on soil microbial community were exacerbated. Both single and mixed additions of antibiotics had significant inhibitory effects on soil microbial enzyme activities. The Random Forest model predicted the following key moderators involved in the effects of antibiotics on the soil microbiome, and antibiotics type, soil texture were key moderators on the severity of soil microbial biomass changes. Soil texture, temperature and single or combined application constitute of antibiotics were the main drivers of effects on soil enzyme activities. The reported results can be helpful to assess the ecological risk of antibiotics in a soil environment and provides a scientific basis for the rational of antibiotics use in the soil environment.
Collapse
Affiliation(s)
- Mingyu Wang
- Engineering Research Center of Agricultural Microbiology Technology, Ministry of Education & Heilongjiang Provincial Key Laboratory of Ecological Restoration and Resource Utilization for Cold Region & Key Laboratory of Microbiology, College of Heilongjiang Province & School of Life Sciences, Heilongjiang University, Harbin 150080, PR China
| | - Detian Li
- Griffith School of Environment and Science and the Australian Rivers Institute, Griffith University, Nathan, QLD, Australia
| | - Xiangyu Liu
- Griffith School of Environment and Science and the Australian Rivers Institute, Griffith University, Nathan, QLD, Australia
| | - Chengrong Chen
- Griffith School of Environment and Science and the Australian Rivers Institute, Griffith University, Nathan, QLD, Australia
| | - Beat Frey
- Forest Dynamics, Swiss Federal Institute for Forest, Snow and Landscape Research, Birmensdorf, Switzerland
| | - Xin Sui
- Engineering Research Center of Agricultural Microbiology Technology, Ministry of Education & Heilongjiang Provincial Key Laboratory of Ecological Restoration and Resource Utilization for Cold Region & Key Laboratory of Microbiology, College of Heilongjiang Province & School of Life Sciences, Heilongjiang University, Harbin 150080, PR China.
| | - Mai-He Li
- Forest Dynamics, Swiss Federal Institute for Forest, Snow and Landscape Research, Birmensdorf, Switzerland; Key Laboratory of Geographical Processes and Ecological Security in Changbai Mountains, Ministry of Education, School of Geographical Sciences, Northeast Normal University, Changchun, PR China; School of Life Science, Hebei University, Baoding, PR China.
| |
Collapse
|
7
|
Golden MM, Heppe AC, Zaremba CL, Wuest WM. Metal chelation as an antibacterial strategy for Pseudomonas aeruginosa and Acinetobacter baumannii. RSC Chem Biol 2024; 5:d4cb00175c. [PMID: 39372678 PMCID: PMC11446287 DOI: 10.1039/d4cb00175c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Accepted: 09/23/2024] [Indexed: 10/08/2024] Open
Abstract
It is estimated that by 2050, bacterial infections will cause 1.8 million more deaths than cancer annually, and the current lack of antibiotic drug discovery is only exacerbating the crisis. Two pathogens in particular, Gram-negative bacteria A. baumannii and P. aeruginosa, are of grave concern because of their heightened multi-drug resistance due to a dense, impermeable outer membrane. However, targeting specific cellular processes may prove successful in overcoming bacterial resistance. This review will concentrate on a novel approach to combatting pathogenicity by disarming bacteria through the disruption of metal homeostasis to reduce virulence and enhance antibiotic uptake. The varying levels of success in bringing metallophores to clinical trials, with currently only one FDA-approved siderophore antibiotic to date, will also be detailed.
Collapse
Affiliation(s)
| | - Amelia C Heppe
- Department of Chemistry, Emory University Atlanta GA 30322 USA
| | - Cassandra L Zaremba
- Department of Chemistry and Biochemistry, Denison University Granville OH 43023 USA
| | - William M Wuest
- Department of Chemistry, Emory University Atlanta GA 30322 USA
- Emory Antibiotic Resistance Center, Emory School of Medicine, Emory University Atlanta GA 30322 USA
| |
Collapse
|
8
|
Abichabki N, Gaspar GG, Zacharias LV, Pocente RHC, Lima DAFS, de Freitas NAB, Brancini GTP, Moreira NC, Braga GÚL, Bellissimo-Rodrigues F, Bollela VR, Darini ALC, Andrade LN. In Vitro Synergistic Activity of Rifampicin Combined with Minimal Effective Antibiotic Concentration (MEAC) of Polymyxin B Against Extensively Drug-Resistant, Carbapenem-, and Polymyxin B-Resistant Klebsiella pneumoniae Clinical Isolates. Curr Microbiol 2024; 81:371. [PMID: 39307852 DOI: 10.1007/s00284-024-03897-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2024] [Accepted: 09/11/2024] [Indexed: 10/22/2024]
Abstract
We investigated the in vitro antibacterial activity of the combination rifampicin (RIF) + polymyxin B (PB) against extensively drug-resistant (XDR) Klebsiella pneumoniae isolates. We evaluated clinical isolates co-resistant to PB (non-mcr carriers; eptB, mgrB, pmr operon, and ramA mutations) and to carbapenems (KPC, CTX-M, and SHV producers; including KPC + NDM co-producer), belonging to sequence types (ST) ST16, ST11, ST258, ST340, and ST437. We used the standard broth microdilution method to determine RIF and PB minimum inhibitory concentration (MIC) and the checkerboard assay to evaluate the fractional inhibitory concentration index (FICI) of RIF + PB as well as to investigate the lowest concentrations of RIF and PB that combined (RIF + PB) had antibacterial activity. Time-kill assays were performed to evaluate the synergistic effect of the combination against selected isolates. PB MIC (32-256 µg/mL) and RIF MIC (32-1024 µg/mL) were determined. FICI (<0.5) indicated a synergistic effect for all isolates evaluated for the combination RIF + PB. Our results showed that low concentrations of PB (PB minimal effective antibiotic concentration [MEAC], ≤0.25-1 µg/mL) favor RIF (≤0.03-0.125 µg/mL) to reach the bacterial target and exert antibacterial activity against PB-resistant isolates, and the synergistic effect was also observed in time-kill results. The combination of RIF + PB showed in vitro antibacterial activity against XDR, carbapenem-, and PB-resistant K. pneumoniae and could be further studied as a potential combination therapy, with cost-effectiveness and promising efficacy.
Collapse
Affiliation(s)
- Nathália Abichabki
- School of Pharmaceutical Sciences of Ribeirão Preto (FCFRP), University of São Paulo (USP), Av. Prof. Dr. Zeferino Vaz - Vila Monte Alegre, Ribeirão Preto, SP, 14040-903, Brazil
| | - Gilberto Gambero Gaspar
- Ribeirão Preto Medical School (FMRP), University of São Paulo (USP), Av. Bandeirantes, 3900, Campus da USP - Cidade Universitária, Ribeirão Preto, SP, 14040-900, Brazil
- University Hospital of Ribeirão Preto Medical School (HCFMRP), University of São Paulo (USP), R. Ten. Catão Roxo, 3900 - Vila Monte Alegre, Ribeirão Preto, SP, 14015-010, Brazil
| | - Luísa Vieira Zacharias
- School of Pharmaceutical Sciences of Ribeirão Preto (FCFRP), University of São Paulo (USP), Av. Prof. Dr. Zeferino Vaz - Vila Monte Alegre, Ribeirão Preto, SP, 14040-903, Brazil
| | - Renata Helena Cândido Pocente
- University Hospital of Ribeirão Preto Medical School (HCFMRP), University of São Paulo (USP), R. Ten. Catão Roxo, 3900 - Vila Monte Alegre, Ribeirão Preto, SP, 14015-010, Brazil
| | - Denissani Aparecida Ferrari Santos Lima
- University Hospital of Ribeirão Preto Medical School (HCFMRP), University of São Paulo (USP), R. Ten. Catão Roxo, 3900 - Vila Monte Alegre, Ribeirão Preto, SP, 14015-010, Brazil
| | - Natália Augusta Barbosa de Freitas
- University Hospital of Ribeirão Preto Medical School (HCFMRP), University of São Paulo (USP), R. Ten. Catão Roxo, 3900 - Vila Monte Alegre, Ribeirão Preto, SP, 14015-010, Brazil
| | - Guilherme Thomaz Pereira Brancini
- School of Pharmaceutical Sciences of Ribeirão Preto (FCFRP), University of São Paulo (USP), Av. Prof. Dr. Zeferino Vaz - Vila Monte Alegre, Ribeirão Preto, SP, 14040-903, Brazil
| | - Natália Columbaro Moreira
- School of Pharmaceutical Sciences of Ribeirão Preto (FCFRP), University of São Paulo (USP), Av. Prof. Dr. Zeferino Vaz - Vila Monte Alegre, Ribeirão Preto, SP, 14040-903, Brazil
| | - Gilberto Úbida Leite Braga
- Department of Clinical Analyses, Toxicology and Food Science (DACTB), School of Pharmaceutical Sciences of Ribeirão Preto (FCFRP), University of São Paulo (USP), Av. Prof. Dr. Zeferino Vaz - Vila Monte Alegre, Ribeirão Preto, SP, 14040-903, Brazil
| | - Fernando Bellissimo-Rodrigues
- Ribeirão Preto Medical School (FMRP), University of São Paulo (USP), Av. Bandeirantes, 3900, Campus da USP - Cidade Universitária, Ribeirão Preto, SP, 14040-900, Brazil
| | - Valdes Roberto Bollela
- Ribeirão Preto Medical School (FMRP), University of São Paulo (USP), Av. Bandeirantes, 3900, Campus da USP - Cidade Universitária, Ribeirão Preto, SP, 14040-900, Brazil
- University Hospital of Ribeirão Preto Medical School (HCFMRP), University of São Paulo (USP), R. Ten. Catão Roxo, 3900 - Vila Monte Alegre, Ribeirão Preto, SP, 14015-010, Brazil
| | - Ana Lúcia Costa Darini
- Department of Clinical Analyses, Toxicology and Food Science (DACTB), School of Pharmaceutical Sciences of Ribeirão Preto (FCFRP), University of São Paulo (USP), Av. Prof. Dr. Zeferino Vaz - Vila Monte Alegre, Ribeirão Preto, SP, 14040-903, Brazil
| | - Leonardo Neves Andrade
- Department of Clinical Analyses, Toxicology and Food Science (DACTB), School of Pharmaceutical Sciences of Ribeirão Preto (FCFRP), University of São Paulo (USP), Av. Prof. Dr. Zeferino Vaz - Vila Monte Alegre, Ribeirão Preto, SP, 14040-903, Brazil.
| |
Collapse
|
9
|
Fatsis-Kavalopoulos N, Sánchez-Hevia DL, Andersson DI. Beyond the FIC index: the extended information from fractional inhibitory concentrations (FICs). J Antimicrob Chemother 2024; 79:2394-2396. [PMID: 38997227 PMCID: PMC11368421 DOI: 10.1093/jac/dkae233] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/14/2024] Open
Affiliation(s)
| | - Dione L Sánchez-Hevia
- Department of Medical Biochemistry and Microbiology, Uppsala University, UppsalaSweden
| | - Dan I Andersson
- Department of Medical Biochemistry and Microbiology, Uppsala University, UppsalaSweden
| |
Collapse
|
10
|
So B, Kim J, Jo JK, So H. Recent developments in preventing catheter-related infections based on biofilms: A comprehensive review. BIOMICROFLUIDICS 2024; 18:051506. [PMID: 39397894 PMCID: PMC11470810 DOI: 10.1063/5.0195165] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/31/2023] [Accepted: 09/09/2024] [Indexed: 10/15/2024]
Abstract
Urinary and vascular catheters are among the most commonly used medical devices. However, infections caused by biofilm formation on the surface of catheters are a major cause of healthcare-associated infections. Traditional methods, such as using antimicrobials to prevent such infections, generally have short-term effects, and treatment is challenging owing to the emergence of antimicrobial-resistant bacteria. This review aims to evaluate the limitations of conventional catheter-related infection prevention efficacy, such as currently used antimicrobials, and analyze the efficacy and limitations of potential alternatives to prevent catheter-related infections that have not yet been commercialized, classified by the transition stages of biofilm formation. We intend to provide profound insights into the ideal technologies for preventing catheter-associated tract infections and present perspectives on future directions in this field.
Collapse
Affiliation(s)
- Byeongchan So
- Department of Medical and Digital Engineering, Hanyang University, Seoul 04763, South Korea
| | - Jongwon Kim
- Department of Medical and Digital Engineering, Hanyang University, Seoul 04763, South Korea
| | - Jung Ki Jo
- Department of Urology, College of Medicine, Hanyang University, Seoul 04763, South Korea
| | - Hongyun So
- Institute of Nano Science and Technology, Hanyang University, Seoul 04763, South Korea
| |
Collapse
|
11
|
Bellucci MC, Romani C, Sani M, Volonterio A. Dual Antibiotic Approach: Synthesis and Antibacterial Activity of Antibiotic-Antimicrobial Peptide Conjugates. Antibiotics (Basel) 2024; 13:783. [PMID: 39200083 PMCID: PMC11352213 DOI: 10.3390/antibiotics13080783] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Revised: 08/16/2024] [Accepted: 08/18/2024] [Indexed: 09/01/2024] Open
Abstract
In recent years, bacterial resistance to conventional antibiotics has become a major concern in the medical field. The global misuse of antibiotics in clinics, personal use, and agriculture has accelerated this resistance, making infections increasingly difficult to treat and rendering new antibiotics ineffective more quickly. Finding new antibiotics is challenging due to the complexity of bacterial mechanisms, high costs and low financial incentives for the development of new molecular scaffolds, and stringent regulatory requirements. Additionally, innovation has slowed, with many new antibiotics being modifications of existing drugs rather than entirely new classes. Antimicrobial peptides (AMPs) are a valid alternative to small-molecule antibiotics offering several advantages, including broad-spectrum activity and a lower likelihood of inducing resistance due to their multifaceted mechanisms of action. However, AMPs face challenges such as stability issues in physiological conditions, potential toxicity to human cells, high production costs, and difficulties in large-scale manufacturing. A reliable strategy to overcome the drawbacks associated with the use of small-molecule antibiotics and AMPs is combination therapy, namely the simultaneous co-administration of two or more antibiotics or the synthesis of covalently linked conjugates. This review aims to provide a comprehensive overview of the literature on the development of antibiotic-AMP conjugates, with a particular emphasis on critically analyzing the design and synthetic strategies employed in their creation. In addition to the synthesis, the review will also explore the reported antibacterial activity of these conjugates and, where available, examine any data concerning their cytotoxicity.
Collapse
Affiliation(s)
- Maria Cristina Bellucci
- Department of Food, Environmental, and Nutritional Sciences, Università degli Studi di Milano, Via Celoria 2, 20131 Milano, Italy;
| | - Carola Romani
- Department of Chemistry, Materials, and Chemical Engineering “Giulio Natta”, Politecnico di Milano, Via Mancinelli 7, 20131 Milano, Italy;
| | - Monica Sani
- Consiglio Nazionale delle Ricerche, Istituto di Scienze e Tecnologie Chimica “G. Natta” (SCITEC), Via Mario Bianco 9, 20131 Milano, Italy;
| | - Alessandro Volonterio
- Department of Chemistry, Materials, and Chemical Engineering “Giulio Natta”, Politecnico di Milano, Via Mancinelli 7, 20131 Milano, Italy;
| |
Collapse
|
12
|
Yang A, Song J, Li J, Li Y, Bai S, Zhou C, Wang M, Zhou Y, Wen K, Luo M, Chen P, Liu B, Yang H, Bai Y, Wong WL, Cai Q, Pu H, Qian Y, Hu W, Huang W, Wan M, Zhang C, Feng X. Ligand-Receptor Interaction-Induced Intracellular Phase Separation: A Global Disruption Strategy for Resistance-Free Lethality of Pathogenic Bacteria. J Am Chem Soc 2024; 146:23121-23137. [PMID: 38980064 DOI: 10.1021/jacs.4c04749] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/10/2024]
Abstract
Addressing the global challenge of bacterial resistance demands innovative approaches, among which multitargeting is a widely used strategy. Current strategies of multitargeting, typically achieved through drug combinations or single agents inherently aiming at multiple targets, face challenges such as stringent pharmacokinetic and pharmacodynamic requirements and cytotoxicity concerns. In this report, we propose a bacterial-specific global disruption approach as a vastly expanded multitargeting strategy that effectively disrupts bacterial subcellular organization. This effect is achieved through a pioneering chemical design of ligand-receptor interaction-induced aggregation of small molecules, i.e., DNA-induced aggregation of a diarginine peptidomimetic within bacterial cells. These intracellular aggregates display affinity toward various proteins and thus substantially interfere with essential bacterial functions and rupture bacterial cell membranes in an "inside-out" manner, leading to robust antibacterial activities and suppression of drug resistance. Additionally, biochemical analysis of macromolecule binding affinity, cytoplasmic localization patterns, and bacterial stress responses suggests that this bacterial-specific intracellular aggregation mechanism is fundamentally different from nonselective classic DNA or membrane binding mechanisms. These mechanistic distinctions, along with the peptidomimetic's selective permeation of bacterial membranes, contribute to its favorable biocompatibility and pharmacokinetic properties, enabling its in vivo antimicrobial efficacy in several animal models, including mice-based superficial wound models, subcutaneous abscess models, and septicemia infection models. These results highlight the great promise of ligand-receptor interaction-induced intracellular aggregation in achieving a globally disruptive multitargeting effect, thereby offering potential applications in the treatment of malignant cells, including pathogens, tumor cells, and infected tissues.
Collapse
Affiliation(s)
- Anming Yang
- State Key Laboratory of Chemo/Biosensing and Chemometrics, Hunan Provincial Key Laboratory of Biomacromolecular Chemical Biology, and School of Chemistry and Chemical Engineering, Hunan University, Changsha, Hunan 410082, China
| | - Junfeng Song
- State Key Laboratory of Chemo/Biosensing and Chemometrics, Hunan Provincial Key Laboratory of Biomacromolecular Chemical Biology, and School of Chemistry and Chemical Engineering, Hunan University, Changsha, Hunan 410082, China
| | - Jiaqi Li
- State Key Laboratory of Chemo/Biosensing and Chemometrics, Hunan Provincial Key Laboratory of Biomacromolecular Chemical Biology, and School of Chemistry and Chemical Engineering, Hunan University, Changsha, Hunan 410082, China
| | - Youzhi Li
- State Key Laboratory of Chemo/Biosensing and Chemometrics, Hunan Provincial Key Laboratory of Biomacromolecular Chemical Biology, and School of Chemistry and Chemical Engineering, Hunan University, Changsha, Hunan 410082, China
| | - Silei Bai
- State Key Laboratory of Chemo/Biosensing and Chemometrics, Hunan Provincial Key Laboratory of Biomacromolecular Chemical Biology, and School of Chemistry and Chemical Engineering, Hunan University, Changsha, Hunan 410082, China
| | - Cailing Zhou
- State Key Laboratory of Chemo/Biosensing and Chemometrics, Hunan Provincial Key Laboratory of Biomacromolecular Chemical Biology, and School of Chemistry and Chemical Engineering, Hunan University, Changsha, Hunan 410082, China
| | - Min Wang
- State Key Laboratory of Chemo/Biosensing and Chemometrics, Hunan Provincial Key Laboratory of Biomacromolecular Chemical Biology, and School of Chemistry and Chemical Engineering, Hunan University, Changsha, Hunan 410082, China
| | - Yu Zhou
- State Key Laboratory of Chemo/Biosensing and Chemometrics, Hunan Provincial Key Laboratory of Biomacromolecular Chemical Biology, and School of Chemistry and Chemical Engineering, Hunan University, Changsha, Hunan 410082, China
| | - Kang Wen
- State Key Laboratory of Chemo/Biosensing and Chemometrics, Hunan Provincial Key Laboratory of Biomacromolecular Chemical Biology, and School of Chemistry and Chemical Engineering, Hunan University, Changsha, Hunan 410082, China
| | - Miaomiao Luo
- State Key Laboratory of Chemo/Biosensing and Chemometrics, Hunan Provincial Key Laboratory of Biomacromolecular Chemical Biology, and School of Chemistry and Chemical Engineering, Hunan University, Changsha, Hunan 410082, China
| | - Peiren Chen
- State Key Laboratory of Chemo/Biosensing and Chemometrics, Hunan Provincial Key Laboratory of Biomacromolecular Chemical Biology, and School of Chemistry and Chemical Engineering, Hunan University, Changsha, Hunan 410082, China
| | - Bo Liu
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, No.555 Zuchongzhi Rd, Pudong, Shanghai 201203, China
| | - Huan Yang
- School of Medical Technology, Xuzhou Medical University, Xuzhou 221004, China
| | - Yugang Bai
- State Key Laboratory of Chemo/Biosensing and Chemometrics, Hunan Provincial Key Laboratory of Biomacromolecular Chemical Biology, and School of Chemistry and Chemical Engineering, Hunan University, Changsha, Hunan 410082, China
| | - Wing-Leung Wong
- State Key Laboratory of Chemical Biology and Drug Discovery, Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Kowloon, Hong Kong SAR 999077, China
| | - Qingyun Cai
- State Key Laboratory of Chemo/Biosensing and Chemometrics, Hunan Provincial Key Laboratory of Biomacromolecular Chemical Biology, and School of Chemistry and Chemical Engineering, Hunan University, Changsha, Hunan 410082, China
| | - Huangsheng Pu
- College of Advanced Interdisciplinary Studies & Hunan Provincial Key Laboratory of Novel NanoOptoelectronic Information Materials and Devices, National University of Defense Technology, Changsha, Hunan 410073, China
- Nanhu Laser Laboratory, National University of Defense Technology, Changsha 410073, China
| | - Yu Qian
- State Key Laboratory of Anti-Infective Drug Discovery and Development, Guangdong Provincial Key Laboratory of Chiral Molecule and Drug Discovery, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Wenhao Hu
- State Key Laboratory of Anti-Infective Drug Discovery and Development, Guangdong Provincial Key Laboratory of Chiral Molecule and Drug Discovery, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Wei Huang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, No.555 Zuchongzhi Rd, Pudong, Shanghai 201203, China
| | - Muyang Wan
- College of Biology, Hunan University, Changsha, Hunan 410082, China
| | - Chunhui Zhang
- College of Biology, Hunan University, Changsha, Hunan 410082, China
| | - Xinxin Feng
- State Key Laboratory of Chemo/Biosensing and Chemometrics, Hunan Provincial Key Laboratory of Biomacromolecular Chemical Biology, and School of Chemistry and Chemical Engineering, Hunan University, Changsha, Hunan 410082, China
| |
Collapse
|
13
|
Silva-Rodrigues G, de Castro IM, Borges PHG, Suzukawa HT, de Souza JM, Bartolomeu-Gonçalves G, Pelisson M, Medeiros CIS, Bispo MDLF, de Almeida RSC, Ishida K, Tavares ER, Yamauchi LM, Yamada-Ogatta SF. Geraniol Potentiates the Effect of Fluconazole against Planktonic and Sessile Cells of Azole-Resistant Candida tropicalis: In Vitro and In Vivo Analyses. Pharmaceutics 2024; 16:1053. [PMID: 39204397 PMCID: PMC11360560 DOI: 10.3390/pharmaceutics16081053] [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: 06/27/2024] [Revised: 07/24/2024] [Accepted: 08/03/2024] [Indexed: 09/04/2024] Open
Abstract
Candida tropicalis is regarded as an opportunistic pathogen, causing diseases ranging from superficial infections to life-threatening disseminated infections. The ability of this yeast to form biofilms and develop resistance to antifungals represents a significant therapeutic challenge. Herein, the effect of geraniol (GER), alone and combined with fluconazole (FLZ), was evaluated in the planktonic and sessile cells of azole-resistant C. tropicalis. GER showed a time-dependent fungicidal effect on the planktonic cells, impairing the cell membrane integrity. Additionally, GER inhibited the rhodamine 6G efflux, and the molecular docking analyzes supported the binding affinity of GER to the C. tropicalis Cdr1 protein. GER exhibited a synergism with FLZ against the planktonic and sessile cells, inhibiting the adhesion of the yeast cells and the viability of the 48-h biofilms formed on abiotic surfaces. C. tropicalis biofilms treated with GER, alone or combined with FLZ, displayed morphological and ultrastructural alterations, including a decrease in the stacking layers and the presence of wilted cells. Moreover, neither GER alone nor combined with FLZ caused toxicity, and both treatments prolonged the survival of the Galleria mellonella larvae infected with azole-resistant C. tropicalis. These findings indicate that the combination of GER and FLZ may be a promising strategy to control azole-resistant C. tropicalis infections.
Collapse
Affiliation(s)
- Gislaine Silva-Rodrigues
- Postgraduate Program in Microbiology, Department of Microbiology, State University of Londrina, Londrina 86057-970, Brazil; (G.S.-R.); (I.M.d.C.); (P.H.G.B.); (H.T.S.); (L.M.Y.)
| | - Isabela Madeira de Castro
- Postgraduate Program in Microbiology, Department of Microbiology, State University of Londrina, Londrina 86057-970, Brazil; (G.S.-R.); (I.M.d.C.); (P.H.G.B.); (H.T.S.); (L.M.Y.)
| | - Paulo Henrique Guilherme Borges
- Postgraduate Program in Microbiology, Department of Microbiology, State University of Londrina, Londrina 86057-970, Brazil; (G.S.-R.); (I.M.d.C.); (P.H.G.B.); (H.T.S.); (L.M.Y.)
| | - Helena Tiemi Suzukawa
- Postgraduate Program in Microbiology, Department of Microbiology, State University of Londrina, Londrina 86057-970, Brazil; (G.S.-R.); (I.M.d.C.); (P.H.G.B.); (H.T.S.); (L.M.Y.)
| | - Joyce Marinho de Souza
- Postgraduate Program in Microbiology, Department of Microbiology, State University of Londrina, Londrina 86057-970, Brazil; (G.S.-R.); (I.M.d.C.); (P.H.G.B.); (H.T.S.); (L.M.Y.)
| | - Guilherme Bartolomeu-Gonçalves
- Postgraduate Program in Clinical and Laboratory Pathophysiology, Department of Pathology, Clinical and Toxicological Analysis, State University of Londrina, Londrina 86038-350, Brazil
| | - Marsileni Pelisson
- Postgraduate Program in Clinical and Laboratory Pathophysiology, Department of Pathology, Clinical and Toxicological Analysis, State University of Londrina, Londrina 86038-350, Brazil
| | | | - Marcelle de Lima Ferreira Bispo
- Synthesis of Medicinal Molecules Laboratory, Department of Chemistry, State University of Londrina, Londrina 86057-970, Brazil;
| | - Ricardo Sérgio Couto de Almeida
- Postgraduate Program in Microbiology, Department of Microbiology, State University of Londrina, Londrina 86057-970, Brazil; (G.S.-R.); (I.M.d.C.); (P.H.G.B.); (H.T.S.); (L.M.Y.)
| | - Kelly Ishida
- Laboratory of Antifungal Chemotherapy, Institute of Biomedical Sciences, University of São Paulo, São Paulo 05508-900, Brazil;
| | - Eliandro Reis Tavares
- Department of Medicine, Pontifical Catholic University of Paraná, Campus Londrina, Londrina 86067-000, Brazil;
- Laboratory of Molecular Biology of Microorganisms, Department of Microbiology, State University of Londrina, Londrina 86057-970, Brazil
| | - Lucy Megumi Yamauchi
- Postgraduate Program in Microbiology, Department of Microbiology, State University of Londrina, Londrina 86057-970, Brazil; (G.S.-R.); (I.M.d.C.); (P.H.G.B.); (H.T.S.); (L.M.Y.)
- Laboratory of Molecular Biology of Microorganisms, Department of Microbiology, State University of Londrina, Londrina 86057-970, Brazil
| | - Sueli Fumie Yamada-Ogatta
- Postgraduate Program in Microbiology, Department of Microbiology, State University of Londrina, Londrina 86057-970, Brazil; (G.S.-R.); (I.M.d.C.); (P.H.G.B.); (H.T.S.); (L.M.Y.)
- Postgraduate Program in Clinical and Laboratory Pathophysiology, Department of Pathology, Clinical and Toxicological Analysis, State University of Londrina, Londrina 86038-350, Brazil
- Laboratory of Molecular Biology of Microorganisms, Department of Microbiology, State University of Londrina, Londrina 86057-970, Brazil
| |
Collapse
|
14
|
Ramfol R, van Vuuren S. The interactive effects of medicinal dyes with conventional antimicrobials against skin pathogens. J Appl Microbiol 2024; 135:lxae147. [PMID: 38955371 DOI: 10.1093/jambio/lxae147] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2024] [Revised: 05/21/2024] [Accepted: 07/01/2024] [Indexed: 07/04/2024]
Abstract
AIMS This study aimed to explore potential synergistic effects of medicinal dyes with antimicrobials against pathogens responsible for skin infections. METHODS AND RESULTS Antimicrobial testing was conducted using minimum inhibitory concentrations and minimum bactericidal/fungicidal concentration assays. The fractional inhibitory index (ΣFIC) of combinations was calculated, and isobolograms were constructed on selected combinations. Toxicity studies were conducted using the brine-shrimp lethality assay. Combination (1:1 ratio) studies noted that 26% of dye-antibiotic combinations were synergistic against the Gram-positive strains, 15% against the Gram-negative strains, and 14% against the yeasts. The Mercurochrome: Betadine® combination noted synergy at ratios against all the Staphylococcus aureus strains with ΣFIC values ranging from 0.05 to 0.48. The combination of Gentian violet with Gentamycin noted a 15-fold decrease in toxicity, and a selectivity index of 977.50 against the Escherichia coli (DSM 22314) strain. Time-kill studies were conducted on the combinations with the highest safe selectivity index (SI) value and lowest safe SI value i.e. Gentian violet with Gentamycin and Malachite green with Neomycin. Both combinations demonstrated better antimicrobial activity in comparison to the independent values and the controls. CONCLUSION This study highlights the potential for medicinal dye combinations as a treatment for skin infections.
Collapse
Affiliation(s)
- Rhea Ramfol
- Department of Pharmacy and Pharmacology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg 2193, South Africa
| | - Sandy van Vuuren
- Department of Pharmacy and Pharmacology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg 2193, South Africa
| |
Collapse
|
15
|
Tanveer R, Neale PA, Melvin SD, Leusch FDL. Application of in vitro bioassays to monitor pharmaceuticals in water: A synthesis of chronological analysis, mode of action, and practical insights. CHEMOSPHERE 2024; 359:142255. [PMID: 38729441 DOI: 10.1016/j.chemosphere.2024.142255] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/19/2024] [Revised: 05/02/2024] [Accepted: 05/04/2024] [Indexed: 05/12/2024]
Abstract
Pharmaceutical compounds in wastewater have emerged as a significant concern for the aquatic environment. The use of in vitro bioassays represents a sustainable and cost-effective approach for assessing the potential toxicological risks of these biologically active compounds in wastewater and aligns with ethical considerations in research. It facilitates high-throughput analysis, captures mixture effects, integrates impacts of both known and unknown chemicals, and reduces reliance on animal testing. The core aim of the current review was to explore the practical application of in vitro bioassays in evaluating the environmental impacts of pharmaceuticals in wastewater. This comprehensive review strives to achieve several key objectives. First, it provides a summary categorisation of pharmaceuticals based on their mode of action, providing a structured framework for understanding their ecological significance. Second, a chronological analysis of pharmaceutical research aims to document their prevalence and trends over time, shedding light on evolving environmental challenges. Third, the review critically analyses existing bioassay applications in wastewater, while also examining bioassay coverage of representative compounds within major pharmaceutical classes. Finally, it explores the potential for developing innovative bioassays tailored for water quality monitoring of pharmaceuticals, paving the way for more robust environmental monitoring and risk assessment. Overall, adopting effect-based methods for pharmaceutical monitoring in water holds significant promise. It encompasses a broad spectrum of biological impacts, promotes standardized protocols, and supports a bioassay test battery approach indicative of different endpoints, thereby enhancing the effectiveness of environmental risk assessment.
Collapse
Affiliation(s)
- Rameesha Tanveer
- Australian Rivers Institute, Griffith University, Southport, Qld 4222, Australia.
| | - Peta A Neale
- Australian Rivers Institute, Griffith University, Southport, Qld 4222, Australia.
| | - Steven D Melvin
- Australian Rivers Institute, Griffith University, Southport, Qld 4222, Australia.
| | - Frederic D L Leusch
- Australian Rivers Institute, Griffith University, Southport, Qld 4222, Australia.
| |
Collapse
|
16
|
Liang Y, Zhao H, Li Y, Gao F, Qiu J, Liu Z, Li Q. Joint effects about antibiotics combined using with antibiotics or phytochemicals on Aeromonas hydrophila. MARINE ENVIRONMENTAL RESEARCH 2024; 199:106594. [PMID: 38908112 DOI: 10.1016/j.marenvres.2024.106594] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/08/2024] [Revised: 05/01/2024] [Accepted: 06/04/2024] [Indexed: 06/24/2024]
Abstract
Aeromonas hydrophila is highly prevalent in aquaculture animals and aquaculture environment. Due to the abuse of antibiotics, A. hydrophila can change the antibiotic resistance spectrum directly and affect human health indirectly. The use of combined drugs replacement therapy and the long-term coexistence with drug-resistant bacteria are the reality that human beings have to face in dealing with the problem of antibiotic resistance in the future. This study showed the characteristics and trends through abundant results of combined effects related with the combinations of antibiotic and the combinations of antibiotic and phytochemical on A. hydrophila, and revealed the antagonism probability of combinations of antibiotic and phytochemical is significantly higher than that of the combinations of antibiotic. Meanwhile, the combinations of antibiotic and phytochemical could protect the host cells which also achieved the same effectiveness as combination of antibiotics, and the enrichment pathway was proved to be relatively discrete. In addition, the possible mechanism about the reverse "U" shape of the combined effect curve on wild/antibiotic-resistant bacteria was clarified, and it was confirmed that the antagonism for the combinations of antibiotic and phytochemical might has the significance in inhibiting the evolution of bacterial resistance mutations. This study was aims to provide theoretical basis and some clues for the antibiotic resistance control associated with A. hydrophila.
Collapse
Affiliation(s)
- Yannei Liang
- Key Laboratory of Agro-products Safety & Quality of Ministry of Agriculture, Beijing, PR China; Institute of Quality Standards & Testing Technology for Agro-products, Chinese Academy of Agricultural Sciences, Beijing, PR China
| | - Haiqing Zhao
- Key Laboratory of Agro-products Safety & Quality of Ministry of Agriculture, Beijing, PR China; Institute of Quality Standards & Testing Technology for Agro-products, Chinese Academy of Agricultural Sciences, Beijing, PR China
| | - Yun Li
- Key Laboratory of Agro-products Safety & Quality of Ministry of Agriculture, Beijing, PR China; Institute of Quality Standards & Testing Technology for Agro-products, Chinese Academy of Agricultural Sciences, Beijing, PR China.
| | - Fuqing Gao
- College of Food Science & Nutritional Engineering, China Agricultural University, Beijing, PR China
| | - Jing Qiu
- Key Laboratory of Agro-products Safety & Quality of Ministry of Agriculture, Beijing, PR China; Institute of Quality Standards & Testing Technology for Agro-products, Chinese Academy of Agricultural Sciences, Beijing, PR China
| | - Zhe Liu
- Key Laboratory of Agro-products Safety & Quality of Ministry of Agriculture, Beijing, PR China; Institute of Quality Standards & Testing Technology for Agro-products, Chinese Academy of Agricultural Sciences, Beijing, PR China
| | - Qiongyan Li
- Key Laboratory of Agro-products Safety & Quality of Ministry of Agriculture, Beijing, PR China; Institute of Quality Standards & Testing Technology for Agro-products, Chinese Academy of Agricultural Sciences, Beijing, PR China
| |
Collapse
|
17
|
Siedentop B, Kachalov VN, Witzany C, Egger M, Kouyos RD, Bonhoeffer S. The effect of combining antibiotics on resistance: A systematic review and meta-analysis. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2023.07.10.23292374. [PMID: 37503165 PMCID: PMC10370225 DOI: 10.1101/2023.07.10.23292374] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/29/2023]
Abstract
When and under which conditions antibiotic combination therapy decelerates rather than accelerates resistance evolution is not well understood. We examined the effect of combining antibiotics on within-patient resistance development across various bacterial pathogens and antibiotics. We searched CENTRAL, EMBASE and PubMed for (quasi)-randomised controlled trials (RCTs) published from database inception to November 24th, 2022. Trials comparing antibiotic treatments with different numbers of antibiotics were included. A patient was considered to have acquired resistance if, at the follow-up culture, a resistant bacterium (as defined by the study authors) was detected that had not been present in the baseline culture. We combined results using a random effects model and performed meta-regression and stratified analyses. The trials' risk of bias was assessed with the Cochrane tool. 42 trials were eligible and 29, including 5054 patients, were qualified for statistical analysis. In most trials, resistance development was not the primary outcome and studies lacked power. The combined odds ratio (OR) for the acquisition of resistance comparing the group with the higher number of antibiotics with the comparison group was 1.23 (95% CI 0.68-2.25), with substantial between-study heterogeneity (I 2 =77%). We identified tentative evidence for potential beneficial or detrimental effects of antibiotic combination therapy for specific pathogens or medical conditions. The evidence for combining a higher number of antibiotics compared to fewer from RCTs is scarce and overall, is compatible with both benefit or harm. Trials powered to detect differences in resistance development or well-designed observational studies are required to clarify the impact of combination therapy on resistance.
Collapse
Affiliation(s)
- Berit Siedentop
- Institute of Integrative Biology, Department of Environmental Systems Science, ETH Zürich, Zurich, Switzerland
- Division of Infectious Diseases and Hospital Epidemiology, University Hospital Zürich, University of Zürich, Zürich, Switzerland
| | - Viacheslav N. Kachalov
- Division of Infectious Diseases and Hospital Epidemiology, University Hospital Zürich, University of Zürich, Zürich, Switzerland
- Institute of Medical Virology, University of Zurich, Zurich, Switzerland
| | - Christopher Witzany
- Institute of Integrative Biology, Department of Environmental Systems Science, ETH Zürich, Zurich, Switzerland
| | - Matthias Egger
- Institute of Social and Preventive Medicine (ISPM), University of Bern, Bern, Switzerland
- Population Health Sciences, University of Bristol, Bristol, UK
- Centre for Infectious Disease Epidemiology and Research, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Roger D. Kouyos
- Division of Infectious Diseases and Hospital Epidemiology, University Hospital Zürich, University of Zürich, Zürich, Switzerland
- Institute of Medical Virology, University of Zurich, Zurich, Switzerland
| | - Sebastian Bonhoeffer
- Institute of Integrative Biology, Department of Environmental Systems Science, ETH Zürich, Zurich, Switzerland
| |
Collapse
|
18
|
Wei X, Gao J, Zhou D, Xu C, Chen P, Chen S, Zhang Y, Liu X, Li G, Zhu G, Liu H, Li J, Geng B, Gao L, Cheng Z, Lamont IL, Pletzer D, Jin Y, Jin S, Wu W. Murepavadin promotes the killing efficacies of aminoglycoside antibiotics against Pseudomonas aeruginosa by enhancing membrane potential. Antimicrob Agents Chemother 2024; 68:e0153923. [PMID: 38470195 PMCID: PMC10989017 DOI: 10.1128/aac.01539-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Accepted: 02/19/2024] [Indexed: 03/13/2024] Open
Abstract
Murepavadin is a peptidomimetic that specifically targets the lipopolysaccharide transport protein LptD of Pseudomonas aeruginosa. Here, we found that murepavadin enhances the bactericidal efficacies of tobramycin and amikacin. We further demonstrated that murepavadin enhances bacterial respiration activity and subsequent membrane potential, which promotes intracellular uptake of aminoglycoside antibiotics. In addition, the murepavadin-amikacin combination displayed a synergistic bactericidal effect in a murine pneumonia model.
Collapse
Affiliation(s)
- Xiaoya Wei
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Molecular Microbiology and Technology of the Ministry of Education, Department of Microbiology, College of Life Sciences, Nankai University, Tianjin, China
| | - Jiacong Gao
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Molecular Microbiology and Technology of the Ministry of Education, Department of Microbiology, College of Life Sciences, Nankai University, Tianjin, China
| | - Dandan Zhou
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Molecular Microbiology and Technology of the Ministry of Education, Department of Microbiology, College of Life Sciences, Nankai University, Tianjin, China
| | - Congjuan Xu
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Molecular Microbiology and Technology of the Ministry of Education, Department of Microbiology, College of Life Sciences, Nankai University, Tianjin, China
| | - Ping Chen
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Molecular Microbiology and Technology of the Ministry of Education, Department of Microbiology, College of Life Sciences, Nankai University, Tianjin, China
| | - Shuiping Chen
- Department of Laboratory Medicine, 5th Medical Center of PLA General Hospital, Beijing, China
| | - Yanhong Zhang
- Nankai University Affiliated Hospital (Tianjin Forth Hospital), Tianjin, China
| | - Xuehua Liu
- Tianjin Union Medical Center, Nankai University Affiliated Hospital, Tianjin, China
| | - Guanxian Li
- Tianjin Union Medical Center, Nankai University Affiliated Hospital, Tianjin, China
| | - Guangbo Zhu
- Tianjin Union Medical Center, Nankai University Affiliated Hospital, Tianjin, China
| | - Huimin Liu
- Tianjin Union Medical Center, Nankai University Affiliated Hospital, Tianjin, China
| | - Jinjin Li
- Tianjin Union Medical Center, Nankai University Affiliated Hospital, Tianjin, China
| | - Bin Geng
- Tianjin Union Medical Center, Nankai University Affiliated Hospital, Tianjin, China
| | - Linlin Gao
- Tianjin Union Medical Center, Nankai University Affiliated Hospital, Tianjin, China
| | - Zhihui Cheng
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Molecular Microbiology and Technology of the Ministry of Education, Department of Microbiology, College of Life Sciences, Nankai University, Tianjin, China
| | - Iain L. Lamont
- Department of Biochemistry, University of Otago, Dunedin, New Zealand
| | - Daniel Pletzer
- Department of Microbiology and Immunology, University of Otago, Dunedin, New Zealand
| | - Yongxin Jin
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Molecular Microbiology and Technology of the Ministry of Education, Department of Microbiology, College of Life Sciences, Nankai University, Tianjin, China
| | - Shouguang Jin
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Molecular Microbiology and Technology of the Ministry of Education, Department of Microbiology, College of Life Sciences, Nankai University, Tianjin, China
| | - Weihui Wu
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Molecular Microbiology and Technology of the Ministry of Education, Department of Microbiology, College of Life Sciences, Nankai University, Tianjin, China
| |
Collapse
|
19
|
Imran M, Singh S, Ahmad MN, Malik P, Mukhopadhyay A, Yadav KS, Gupta UD, Mugale MN, Mitra K, Srivastava KK, Chopra S, Mignani S, Apartsin E, Majoral JP, Dasgupta A. Polycationic phosphorous dendrimer potentiates multiple antibiotics against drug-resistant mycobacterial pathogens. Biomed Pharmacother 2024; 173:116289. [PMID: 38452653 DOI: 10.1016/j.biopha.2024.116289] [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: 12/03/2023] [Revised: 02/02/2024] [Accepted: 02/17/2024] [Indexed: 03/09/2024] Open
Abstract
Mycobacterium tuberculosis (Mtb), causative agent of tuberculosis (TB) and non-tubercular mycobacterial (NTM) pathogens such as Mycobacterium abscessus are one of the most critical concerns worldwide due to increased drug-resistance resulting in increased morbidity and mortality. Therefore, focusing on developing novel therapeutics to minimize the treatment period and reducing the burden of drug-resistant Mtb and NTM infections are an urgent and pressing need. In our previous study, we identified anti-mycobacterial activity of orally bioavailable, non-cytotoxic, polycationic phosphorus dendrimer 2G0 against Mtb. In this study, we report ability of 2G0 to potentiate activity of multiple classes of antibiotics against drug-resistant mycobacterial strains. The observed synergy was confirmed using time-kill kinetics and revealed significantly potent activity of the combinations as compared to individual drugs alone. More importantly, no re-growth was observed in any tested combination. The identified combinations were further confirmed in intra-cellular killing assay as well as murine model of NTM infection, where 2G0 potentiated the activity of all tested antibiotics significantly better than individual drugs. Taken together, this nanoparticle with intrinsic antimycobacterial properties has the potential to represents an alternate drug candidate and/or a novel delivery agent for antibiotics of choice for enhancing the treatment of drug-resistant mycobacterial pathogens.
Collapse
Affiliation(s)
- Mohmmad Imran
- Division of Molecular Microbiology & Immunology, CSIR-Central Drug Research Institute, Sitapur Road, Jankipuram Extension, Lucknow, Uttar Pradesh 226031, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Shriya Singh
- Division of Molecular Microbiology & Immunology, CSIR-Central Drug Research Institute, Sitapur Road, Jankipuram Extension, Lucknow, Uttar Pradesh 226031, India
| | - Mohammad Naiyaz Ahmad
- Division of Molecular Microbiology & Immunology, CSIR-Central Drug Research Institute, Sitapur Road, Jankipuram Extension, Lucknow, Uttar Pradesh 226031, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Pradip Malik
- Division of Molecular Microbiology & Immunology, CSIR-Central Drug Research Institute, Sitapur Road, Jankipuram Extension, Lucknow, Uttar Pradesh 226031, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Atri Mukhopadhyay
- Division of Molecular Microbiology & Immunology, CSIR-Central Drug Research Institute, Sitapur Road, Jankipuram Extension, Lucknow, Uttar Pradesh 226031, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Karan Singh Yadav
- Division of Toxicology & Experimental Medicine, CSIR-Central Drug Research Institute, Sitapur Road, Jankipuram Extension, Lucknow, Uttar Pradesh 226031, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Umesh D Gupta
- National JALMA Institute for Leprosy and Other Mycobacterial Diseases, Agra 282001, India
| | - Madhav N Mugale
- Division of Toxicology & Experimental Medicine, CSIR-Central Drug Research Institute, Sitapur Road, Jankipuram Extension, Lucknow, Uttar Pradesh 226031, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Kalyan Mitra
- Electron Microscopy Unit, CSIR-Central Drug Research Institute, Sitapur Road, Lucknow, 226031, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Kishore K Srivastava
- Division of Molecular Microbiology & Immunology, CSIR-Central Drug Research Institute, Sitapur Road, Jankipuram Extension, Lucknow, Uttar Pradesh 226031, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Sidharth Chopra
- Division of Molecular Microbiology & Immunology, CSIR-Central Drug Research Institute, Sitapur Road, Jankipuram Extension, Lucknow, Uttar Pradesh 226031, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Serge Mignani
- Centre d'Etudes et de Recherche sur le Medicament de Normandie (CERMN), Université de Caen Normandie, Caen 14032, France; Centro de Quimica da Madeira, MMRG, Campus da Penteada, Universidade da Madeira, Funchal 9020-105 19, Portugal.
| | - Evgeny Apartsin
- Université de Bordeaux, CNRS, Bordeaux INP, CBMN, UMR 5248, Pessac F-33600, France
| | - Jean-Pierre Majoral
- Laboratoire de Chimie de Coordination du CNRS, 205 Route de Narbonne, BP 44099, 31077 Toulouse Cedex 4, France; LCC-CNRS, Université de Toulouse, CNRS, Toulouse 31400, France.
| | - Arunava Dasgupta
- Division of Molecular Microbiology & Immunology, CSIR-Central Drug Research Institute, Sitapur Road, Jankipuram Extension, Lucknow, Uttar Pradesh 226031, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India.
| |
Collapse
|
20
|
Davis KP, Morales Y, Ende RJ, Peters R, McCabe AL, Mecsas J, Aldridge BB. Critical role of growth medium for detecting drug interactions in Gram-negative bacteria that model in vivo responses. mBio 2024; 15:e0015924. [PMID: 38364199 PMCID: PMC10936441 DOI: 10.1128/mbio.00159-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2024] [Accepted: 01/22/2024] [Indexed: 02/18/2024] Open
Abstract
The rise in infections caused by multidrug-resistant (MDR) bacteria has necessitated a variety of clinical approaches, including the use of antibiotic combinations. Here, we tested the hypothesis that drug-drug interactions vary in different media, and determined which in vitro models best predict drug interactions in the lungs. We systematically studied pair-wise antibiotic interactions in three different media, CAMHB, (a rich lab medium standard for antibiotic susceptibility testing), a urine mimetic medium (UMM), and a minimal medium of M9 salts supplemented with glucose and iron (M9Glu) with three Gram-negative ESKAPE pathogens, Acinetobacter baumannii (Ab), Klebsiella pneumoniae (Kp), and Pseudomonas aeruginosa (Pa). There were pronounced differences in responses to antibiotic combinations between the three bacterial species grown in the same medium. However, within species, PaO1 responded to drug combinations similarly when grown in all three different media, whereas Ab17978 and other Ab clinical isolates responded similarly when grown in CAMHB and M9Glu medium. By contrast, drug interactions in Kp43816, and other Kp clinical isolates poorly correlated across different media. To assess whether any of these media were predictive of antibiotic interactions against Kp in the lungs of mice, we tested three antibiotic combination pairs. In vitro measurements in M9Glu, but not rich medium or UMM, predicted in vivo outcomes. This work demonstrates that antibiotic interactions are highly variable across three Gram-negative pathogens and highlights the importance of growth medium by showing a superior correlation between in vitro interactions in a minimal growth medium and in vivo outcomes. IMPORTANCE Drug-resistant bacterial infections are a growing concern and have only continued to increase during the SARS-CoV-2 pandemic. Though not routinely used for Gram-negative bacteria, drug combinations are sometimes used for serious infections and may become more widely used as the prevalence of extremely drug-resistant organisms increases. To date, reliable methods are not available for identifying beneficial drug combinations for a particular infection. Our study shows variability across strains in how drug interactions are impacted by growth conditions. It also demonstrates that testing drug combinations in tissue-relevant growth conditions for some strains better models what happens during infection and may better inform combination therapy selection.
Collapse
Affiliation(s)
- Kathleen P. Davis
- Department of Molecular Biology and Microbiology, Tufts University School of Medicine, & Stuart B. Levy Center for Integrated Management of Antimicrobial Resistance Boston, Boston, Massachusetts, USA
| | - Yoelkys Morales
- Department of Molecular Biology and Microbiology, Tufts University School of Medicine, & Stuart B. Levy Center for Integrated Management of Antimicrobial Resistance Boston, Boston, Massachusetts, USA
- Graduate School of Biomedical Sciences, Tufts University School of Medicine, Boston, Massachusetts, USA
| | - Rachel J. Ende
- Department of Molecular Biology and Microbiology, Tufts University School of Medicine, & Stuart B. Levy Center for Integrated Management of Antimicrobial Resistance Boston, Boston, Massachusetts, USA
| | - Ryan Peters
- Department of Molecular Biology and Microbiology, Tufts University School of Medicine, & Stuart B. Levy Center for Integrated Management of Antimicrobial Resistance Boston, Boston, Massachusetts, USA
| | - Anne L. McCabe
- Department of Molecular Biology and Microbiology, Tufts University School of Medicine, & Stuart B. Levy Center for Integrated Management of Antimicrobial Resistance Boston, Boston, Massachusetts, USA
- Department of Basic and Clinical Sciences, Albany College of Pharmacy and Health Sciences, Albany, New York, USA
| | - Joan Mecsas
- Department of Molecular Biology and Microbiology, Tufts University School of Medicine, & Stuart B. Levy Center for Integrated Management of Antimicrobial Resistance Boston, Boston, Massachusetts, USA
- Graduate School of Biomedical Sciences, Tufts University School of Medicine, Boston, Massachusetts, USA
| | - Bree B. Aldridge
- Department of Molecular Biology and Microbiology, Tufts University School of Medicine, & Stuart B. Levy Center for Integrated Management of Antimicrobial Resistance Boston, Boston, Massachusetts, USA
- Graduate School of Biomedical Sciences, Tufts University School of Medicine, Boston, Massachusetts, USA
- Department of Biomedical Engineering, Tufts University School of Engineering, Medford, Massachusetts, USA
| |
Collapse
|
21
|
Sadia M, Ahmad I, Aziz S, Khan R, Zahoor M, Ullah R, Ali EA. Carbon-Supported Nanocomposite Synthesis, Characterization, and Application as an Efficient Adsorbent for Ciprofloxacin and Amoxicillin. ACS OMEGA 2024; 9:6815-6827. [PMID: 38371783 PMCID: PMC10870352 DOI: 10.1021/acsomega.3c08161] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Revised: 01/23/2024] [Accepted: 01/26/2024] [Indexed: 02/20/2024]
Abstract
The existence of antibiotics in the environment has recently raised serious concerns about their possible hazards to human health and the water ecosystem. In the current study, an activated carbon-supported nanocomposite, AC-CoFe2O3, was synthesized by a coprecipitation method, characterized, and then applied to adsorb different drugs from water. The synthesized composites were characterized by using energy-dispersive X-ray spectroscopy, Fourier transform infrared spectroscopy, Brunauer-Emmett-Teller plots, and scanning electron microscopy. The adsorption of both Ciprofloxacin (Cipro) and Amoxicillin (Amoxi) antibiotics on the composite followed the pseudo-second-order kinetic model (R2 = 0.9981 and 0.9974 mg g-1 min-1, respectively). Langmuir isotherm was the best-fit model showing 312.17 and 217.76 mg g-1 adsorption capacities for Ciprofloxacin and Amoxicillin, respectively, at 333 K. The negative Gibbs free energy (ΔG°) specified the spontaneity of the method. The positive change in the enthalpy (ΔH) indicated that the adsorption process was assisted by higher temperatures. The different optimized parameters were pH, contact time, adsorbent weight, concentration, and temperature. The maximum adsorption of Cipro was found to be 98.41% at pH 12, while for Amoxi, it was 89.09% at pH 2 at 333 K. The drugs were then successfully determined from natural water samples at optimized conditions using these nanocomposites.
Collapse
Affiliation(s)
- Maria Sadia
- Department of Chemistry, University of Malakand, Lower Dir, Chakdara 18800, Pakistan
| | - Izaz Ahmad
- Department of Chemistry, University of Malakand, Lower Dir, Chakdara 18800, Pakistan
- Department of Chemistry and Chemical Technology, Al-Farabi Kazakh National University, Almaty 050040, Kazakhstan
| | - Shaukat Aziz
- Department of Chemistry, University of Malakand, Lower Dir, Chakdara 18800, Pakistan
| | - Rizwan Khan
- Department of Electrical Engineering, Kwangwoon University Seoul, Seoul 54047, South Korea
| | - Muhammad Zahoor
- Department of Biochemistry, University of Malakand, Chakdara, Lower Dir, Khyber Pakhtunkhwa 18000, Pakistan
| | - Riaz Ullah
- Department of Pharmacognosy, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Essam A Ali
- Department of Pharmaceutical Chemistry, College of Pharmacy King Saud University Riyadh, Riyadh 11451, Saudi Arabia
| |
Collapse
|
22
|
Faure A, Manuse S, Gonin M, Grangeasse C, Jault JM, Orelle C. Daptomycin avoids drug resistance mediated by the BceAB transporter in Streptococcus pneumoniae. Microbiol Spectr 2024; 12:e0363823. [PMID: 38214521 PMCID: PMC10846014 DOI: 10.1128/spectrum.03638-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Accepted: 12/19/2023] [Indexed: 01/13/2024] Open
Abstract
Drug-resistant bacteria are a serious threat to human health as antibiotics are gradually losing their clinical efficacy. Comprehending the mechanism of action of antimicrobials and their resistance mechanisms plays a key role in developing new agents to fight antimicrobial resistance. The lipopeptide daptomycin is an antibiotic that selectively disrupts Gram-positive bacterial membranes, thereby showing slower resistance development than many classical drugs. Consequently, it is often used as a last resort antibiotic to preserve its use as one of the least potent antibiotics at our disposal. The mode of action of daptomycin has been debated but was recently found to involve the formation of a tripartite complex between undecaprenyl precursors of cell wall biosynthesis and the anionic phospholipid phosphatidylglycerol. BceAB-type ABC transporters are known to confer resistance to antimicrobial peptides that sequester some precursors of the peptidoglycan, such as the undecaprenyl pyrophosphate or lipid II. The expression of these transporters is upregulated by dedicated two-component regulatory systems in the presence of antimicrobial peptides that are recognized by the system. Here, we investigated whether daptomycin evades resistance mediated by the BceAB transporter from the bacterial pathogen Streptococcus pneumoniae. Although daptomycin can bind to the transporter, our data showed that the BceAB transporter does not mediate resistance to the drug and its expression is not induced in its presence. These findings show that the pioneering membrane-active daptomycin has the potential to escape the resistance mechanism mediated by BceAB-type transporters and confirm that the development of this class of compounds has promising clinical applications.IMPORTANCEAntibiotic resistance is rising in all parts of the world. New resistance mechanisms are emerging and dangerously spreading, threatening our ability to treat common infectious diseases. Daptomycin is an antimicrobial peptide that is one of the last antibiotics approved for clinical use. Understanding the resistance mechanisms toward last-resort antibiotics such as daptomycin is critical for the success of future antimicrobial therapies. BceAB-type ABC transporters confer resistance to antimicrobial peptides that target precursors of cell-wall synthesis. In this study, we showed that the BceAB transporter from the human pathogen Streptococcus pneumoniae does not confer resistance to daptomycin, suggesting that this drug and other calcium-dependent lipopeptide antibiotics have the potential to evade the action of this type of ABC transporters in other bacterial pathogens.
Collapse
Affiliation(s)
- Agathe Faure
- Molecular Microbiology and Structural Biochemistry (MMSB), UMR 5086 CNRS/University of Lyon, Lyon, France
| | - Sylvie Manuse
- Molecular Microbiology and Structural Biochemistry (MMSB), UMR 5086 CNRS/University of Lyon, Lyon, France
| | - Mathilde Gonin
- Molecular Microbiology and Structural Biochemistry (MMSB), UMR 5086 CNRS/University of Lyon, Lyon, France
| | - Christophe Grangeasse
- Molecular Microbiology and Structural Biochemistry (MMSB), UMR 5086 CNRS/University of Lyon, Lyon, France
| | - Jean-Michel Jault
- Molecular Microbiology and Structural Biochemistry (MMSB), UMR 5086 CNRS/University of Lyon, Lyon, France
| | - Cédric Orelle
- Molecular Microbiology and Structural Biochemistry (MMSB), UMR 5086 CNRS/University of Lyon, Lyon, France
| |
Collapse
|
23
|
Azimzadeh M, Greco G, Farmani A, Pourhajibagher M, Taherkhani A, Alikhani MY, Bahador A. Synergistic effects of nano curcumin mediated photodynamic inactivation and nano-silver@colistin against Pseudomonas aeruginosa biofilms. Photodiagnosis Photodyn Ther 2024; 45:103971. [PMID: 38218569 DOI: 10.1016/j.pdpdt.2024.103971] [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: 11/17/2023] [Revised: 01/10/2024] [Accepted: 01/10/2024] [Indexed: 01/15/2024]
Abstract
BACKGROUND Patients with burn injuries colonized by multidrug-resistant Pseudomonas aeruginosa face increased mortality risk. The efficacy of colistin, a last-resort treatment, is declining as resistance levels rise. P. aeruginosa's robust biofilm exacerbates antibiotic resistance. Photodynamic Inactivation (PDI) shows promise in fighting biofilm. MATERIALS AND METHODS Nano curcumin (nCur) particles were synthesized, and their chemical characteristics were determined using zeta potential (ZP), dynamic light scattering analysis (DLS), energy-dispersive X-ray (EDX) analysis, and fourier transform infrared (FTIR). We conducted an MTT assay to assess the cytotoxicity of nCur-mediated PDI in combination with nanosilver colistin. The fractional biofilm inhibitory concentration (FBIC) of two P. aeruginosa clinical isolates and P. aeruginosa ATCC 27853 during nCur-mediated PDI@AgNPs@CL was determined using a 3-dimensional (3-D) checkerboard assay. To study the effect of nCur-mediated PDI@AgNPs@CL on lasI, lasR, rhlI, rhlR, pelA, and pslA gene expression, Reverse transcription-quantitative polymerase chain reaction (RT-qPCR) was conducted at each isolate's FBIC. The impact of treatments was also investigated using scanning electron microscopy (SEM). RESULTS The ZP and mean DLS values of the nCur were 10.3 mV and 402.6 ± 24.6 nm, respectively. The distinct functional groups of nCur corresponded with the peaks of FTIR absorption. Moreover, the EDX analysis showed the ratios of different metals in nCur. Cell viability percentages of nCur-mediated PDI@AgNPs@CL at FBIC concentrations of clinical isolates Nos. 30, 354, and P. aeruginosa ATCC 27853 were 91.36 %, 83.20 %, and 92.48 %, respectively. nCur-mediated PDI@AgNPs@CL treatment showed synergistic effects in clinical isolates and P. aeruginosa ATCC 27853 in a 3-D checkerboard assay. All six of the investigated genes showed down-regulation after nCur-mediated PDI@AgNPs@CL treatment. The most suppressed gene during nCur-mediated PDI@AgNPs@CL treatment was the rhlR gene (-11.9-fold) of P. aeruginosa ATCC 27853. The SEM micrographs further proved the connecting cement reduction and biofilm mass mitigation following nCur-mediated PDI@AgNPs@CL treatments. CONCLUSIONS The combined effect of nCur-mediated PDI and AgNPs@CL synergistically reduce the formation of biofilm in P. aeruginosa. This may be attributable to the suppression of the genes responsible for regulating the production of biofilms.
Collapse
Affiliation(s)
- Masoud Azimzadeh
- Department of Microbiology, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Grazia Greco
- Department of Veterinary Medicine, University of Bari Aldo Moro, Bari, Italy
| | - Abbas Farmani
- Dental Research Center, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Maryam Pourhajibagher
- Dental Research Center, Dentistry Research Institute, Tehran University of Medical Sciences, Tehran, Iran.
| | - Amir Taherkhani
- Research Center for Molecular Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Mohammad Yousef Alikhani
- Department of Microbiology, Hamadan University of Medical Sciences, Hamadan, Iran; Infectious Disease Research Center, Hamadan University of Medical Sciences, Hamadan, Iran.
| | - Abbas Bahador
- Department of Microbiology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran; Fellowship in Clinical Laboratory Sciences, BioHealth Lab, Tehran, Iran.
| |
Collapse
|
24
|
Kroemer N, Amann LF, Farooq A, Pfaffendorf C, Martens M, Decousser JW, Grégoire N, Nordmann P, Wicha SG. Pharmacokinetic/pharmacodynamic analysis of ceftazidime/avibactam and fosfomycin combinations in an in vitro hollow fiber infection model against multidrug-resistant Escherichia coli. Microbiol Spectr 2024; 12:e0331823. [PMID: 38063387 PMCID: PMC10783110 DOI: 10.1128/spectrum.03318-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Accepted: 11/10/2023] [Indexed: 01/13/2024] Open
Abstract
IMPORTANCE Mechanistic understanding of pharmacodynamic interactions is key for the development of rational antibiotic combination therapies to increase efficacy and suppress the development of resistances. Potent tools to provide those insights into pharmacodynamic drug interactions are semi-mechanistic modeling and simulation techniques. This study uses those techniques to provide a detailed understanding with regard to the direction and strength of the synergy of ceftazidime-avibactam and ceftazidime-fosfomycin in a clinical Escherichia coli isolate expressing extended spectrum beta-lactamase (CTX-M-15 and TEM-4) and carbapenemase (OXA-244) genes. Enhanced killing effects in combination were identified as a driver of the synergy and were translated from static time-kill experiments into the dynamic hollow fiber infection model. These findings in combination with a suppression of the emergence of resistance in combination emphasize a potential clinical benefit with regard to increased efficacy or to allow for dose reductions with maintained effect sizes to avoid toxicity.
Collapse
Affiliation(s)
- Niklas Kroemer
- Institute of Pharmacy, University of Hamburg, Hamburg, Germany
| | - Lisa F. Amann
- Institute of Pharmacy, University of Hamburg, Hamburg, Germany
| | - Aneeq Farooq
- Institute of Pharmacy, University of Hamburg, Hamburg, Germany
| | | | - Miklas Martens
- Institute of Pharmacy, University of Hamburg, Hamburg, Germany
| | - Jean-Winoc Decousser
- Dynamic Team – EA 7380, Faculté de Santé, Université Paris-Est-Créteil Val-De-Marne, Créteil, France
| | - Nicolas Grégoire
- Inserm U1070, Poitiers, France
- UFR de Médecine Pharmacie, Université de Poitiers, Poitiers, France
- Laboratoire de Toxicologie-Pharmacologie, CHU de Poitiers, Poitiers, France
| | - Patrice Nordmann
- Medical and Molecular Microbiology, University of Fribourg, Fribourg, Switzerland
| | | |
Collapse
|
25
|
Zore M, San-Martin-Galindo P, Reigada I, Hanski L, Fallarero A, Yli-Kauhaluoma J, Patel JZ. Design and synthesis of etrasimod derivatives as potent antibacterial agents against Gram-positive bacteria. Eur J Med Chem 2024; 263:115921. [PMID: 37948883 DOI: 10.1016/j.ejmech.2023.115921] [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: 09/14/2023] [Revised: 10/19/2023] [Accepted: 10/27/2023] [Indexed: 11/12/2023]
Abstract
The emergence of multidrug-resistant bacteria along with a declining pipeline of clinically useful antibiotics has led to the urgent need for the development of more effective antibacterial agents. Inspired by our recent report on the antibacterial activity of etrasimod, an immunomodulating drug candidate, we prepared a series of etrasimod derivatives by varying substituents on the phenyl ring, altering the central tricyclic aromatic ring, and modifying the carboxyl group. From this series of compounds, indole derivative 24f was identified as the most potent antibacterial compound, with the minimum inhibitory concentration (MIC) values between 2.5 and 10 μM against various Gram-positive bacteria, including methicillin-resistant Staphylococcus aureus (MRSA), S. epidermidis and enterococci. Moreover, 24f exhibited rapid bactericidal activity against S. aureus, low toxicity and hemolytic activity, and a synergistic effect with gentamicin against S. aureus, MRSA, and Enterococcus faecalis. Furthermore, it was shown that neither etrasimod nor 24f affects S. aureus cell membranes. Importantly, 24f did not induce resistance in S. aureus, representing a significant improvement compared to etrasimod. Finally, the antibacterial activity of etrasimod and 24f against S. aureus and MRSA was confirmed in vivo in a Caenorhabditis elegans infection model. Taken together, our study highlights the value of etrasimod and its derivatives as potential antibacterial candidates for combating infections caused by Gram-positive bacteria.
Collapse
Affiliation(s)
- Matej Zore
- Drug Research Program, Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, Viikinkaari 5 E, FI-00014, Helsinki, Finland
| | - Paola San-Martin-Galindo
- Drug Research Program, Division of Pharmaceutical Biosciences, Faculty of Pharmacy, University of Helsinki, Viikinkaari 5 E, FI-00014, Helsinki, Finland
| | - Inés Reigada
- Drug Research Program, Division of Pharmaceutical Biosciences, Faculty of Pharmacy, University of Helsinki, Viikinkaari 5 E, FI-00014, Helsinki, Finland
| | - Leena Hanski
- Drug Research Program, Division of Pharmaceutical Biosciences, Faculty of Pharmacy, University of Helsinki, Viikinkaari 5 E, FI-00014, Helsinki, Finland
| | - Adyary Fallarero
- Drug Research Program, Division of Pharmaceutical Biosciences, Faculty of Pharmacy, University of Helsinki, Viikinkaari 5 E, FI-00014, Helsinki, Finland
| | - Jari Yli-Kauhaluoma
- Drug Research Program, Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, Viikinkaari 5 E, FI-00014, Helsinki, Finland
| | - Jayendra Z Patel
- Drug Research Program, Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, Viikinkaari 5 E, FI-00014, Helsinki, Finland.
| |
Collapse
|
26
|
Li D, Mo K, Liang B, Huang Y, Tan X, Wang Z, Yang X. The impact of different antibiotic injection regimens on patients with severe infections: A meta-analysis. Int Wound J 2024; 21:e14514. [PMID: 38272804 PMCID: PMC10791546 DOI: 10.1111/iwj.14514] [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: 10/25/2023] [Revised: 11/01/2023] [Accepted: 11/10/2023] [Indexed: 01/27/2024] Open
Abstract
Severe infection is a critical health threat to humans, and antibiotic treatment is one of the main therapeutic approaches. Nevertheless, the efficacy of various antibiotic injection regimens in severe infection patients remains uncertain. This study aimed to comprehensively evaluate the impact of various antibiotic injection strategies on patients with severe infection through a meta-analysis. Relevant research literature was collected by searching databases such as PubMed, Embase, and Cochrane Library. The retrieved literature was screened according to inclusion and exclusion criteria. Relevant data, including study design, sample size, and antibiotic regimens, were extracted from the included studies. The Cochrane Collaboration's Risk of Bias tool was employed to assess the risk of bias in each study. Statistical analysis was performed based on the results of the included studies. A total of 15 articles were included, covering various types of severe infection patients, including pulmonary and abdominal infections. The analysis provided insights into mortality rates, treatment efficacy, adverse reactions (ARs), Acute Physiology and Chronic Health Evaluation (APACHE) scores, among other outcomes. The results indicated that combination therapy was superior to monotherapy in terms of mortality rate, treatment efficacy, and APACHE scores, while the incidence of ARs was lower in the monotherapy group compared to the combination therapy group (p < 0.05). Combination therapy showed better treatment efficacy compared to monotherapy, although it was associated with a higher incidence of ARs.
Collapse
Affiliation(s)
- Da Li
- Intensive Care UnitThe People’s Hospital of Guangxi Zhuang Autonomous RegionNanningChina
| | - Kanglin Mo
- Respiratory EndoscopyThe First Affiliated Hospital of Guangxi Mediacal UniversityNanningChina
| | - Binqi Liang
- Intensive Care UnitThe People’s Hospital of Guangxi Zhuang Autonomous RegionNanningChina
| | | | - Xingling Tan
- Department of General PracticeThe First Affiliated Hospital of Guangxi Mediacal UniversityNanningChina
| | - Zengrui Wang
- Department of General PracticeThe First Affiliated Hospital of Guangxi Mediacal UniversityNanningChina
| | - Xia Yang
- Department of General PracticeThe First Affiliated Hospital of Guangxi Mediacal UniversityNanningChina
| |
Collapse
|
27
|
Igo M, Xu L, Krishna A, Stewart S, Xu L, Li Z, Weaver JL, Stone H, Sacks L, Bensman T, Florian J, Rouse R, Han X. A metagenomic analysis for combination therapy of multiple classes of antibiotics on the prevention of the spread of antibiotic-resistant genes. Gut Microbes 2023; 15:2271150. [PMID: 37908118 PMCID: PMC10621307 DOI: 10.1080/19490976.2023.2271150] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Accepted: 10/11/2023] [Indexed: 11/02/2023] Open
Abstract
Antibiotics used systemically to treat infections may have off-target effects on the gut microbiome, potentially resulting in the emergence of drug-resistant bacteria or selection of pathogenic species. These organisms may present a risk to the host and spread to the environment with a risk of transmission in the community. To investigate the risk of emergent antibiotic resistance in the gut microbiome following systemic treatment with antibiotics, this metagenomic analysis project used next-generation sequencing, a custom-built metagenomics pipeline, and differential abundance analysis to study the effect of antibiotics (ampicillin, ciprofloxacin, and fosfomycin) in monotherapy and different combinations at high and low doses, to determine the effect on resistome and taxonomic composition in the gut of Balb/c mice. The results showed that low-dose monotherapy treatments showed little change in microbiome composition but did show an increase in expression of many antibiotic-resistant genes (ARGs) posttreatment. Dual combination treatments allowed the emergence of some conditionally pathogenic bacteria and some increase in the abundance of ARGs despite a general decrease in microbiota diversity. Triple combination treatment was the most successful in inhibiting emergence of relevant opportunistic pathogens and completely suppressed all ARGs after 72 h of treatment. The relative abundances of mobile genetic elements that can enhance transmission of antibiotic resistance either decreased or remained the same for combination therapy while increasing for low-dose monotherapy. Combination therapy prevented the emergence of ARGs and decreased bacterial diversity, while low-dose monotherapy treatment increased ARGs and did not greatly change bacterial diversity.
Collapse
Affiliation(s)
- Matthew Igo
- Division of Applied Regulatory Science, Office of Clinical Pharmacology, Office of Translational Sciences, Center for Drug Evaluation and Research, U. S. Food and Drug Administration, Silver Spring, MD, USA
| | - Lei Xu
- Division of Applied Regulatory Science, Office of Clinical Pharmacology, Office of Translational Sciences, Center for Drug Evaluation and Research, U. S. Food and Drug Administration, Silver Spring, MD, USA
| | - Ashok Krishna
- Division of Applied Regulatory Science, Office of Clinical Pharmacology, Office of Translational Sciences, Center for Drug Evaluation and Research, U. S. Food and Drug Administration, Silver Spring, MD, USA
| | - Sharron Stewart
- Division of Applied Regulatory Science, Office of Clinical Pharmacology, Office of Translational Sciences, Center for Drug Evaluation and Research, U. S. Food and Drug Administration, Silver Spring, MD, USA
| | - Lin Xu
- Division of Applied Regulatory Science, Office of Clinical Pharmacology, Office of Translational Sciences, Center for Drug Evaluation and Research, U. S. Food and Drug Administration, Silver Spring, MD, USA
| | - Zhihua Li
- Division of Applied Regulatory Science, Office of Clinical Pharmacology, Office of Translational Sciences, Center for Drug Evaluation and Research, U. S. Food and Drug Administration, Silver Spring, MD, USA
| | - James L. Weaver
- Division of Applied Regulatory Science, Office of Clinical Pharmacology, Office of Translational Sciences, Center for Drug Evaluation and Research, U. S. Food and Drug Administration, Silver Spring, MD, USA
| | - Heather Stone
- Office of Medical Policy, Center for Drug Evaluation and Research, U. S. Food and Drug Administration, Silver Spring, MD, USA
| | - Leonard Sacks
- Office of Medical Policy, Center for Drug Evaluation and Research, U. S. Food and Drug Administration, Silver Spring, MD, USA
| | - Timothy Bensman
- Division of Infectious Disease Pharmacology, Office of Clinical Pharmacology, Office of Translational Sciences, Center for Drug Evaluation and Research, U. S. Food and Drug Administration, Silver Spring, MD, USA
| | - Jeffry Florian
- Division of Applied Regulatory Science, Office of Clinical Pharmacology, Office of Translational Sciences, Center for Drug Evaluation and Research, U. S. Food and Drug Administration, Silver Spring, MD, USA
| | - Rodney Rouse
- Division of Applied Regulatory Science, Office of Clinical Pharmacology, Office of Translational Sciences, Center for Drug Evaluation and Research, U. S. Food and Drug Administration, Silver Spring, MD, USA
| | - Xiaomei Han
- Division of Applied Regulatory Science, Office of Clinical Pharmacology, Office of Translational Sciences, Center for Drug Evaluation and Research, U. S. Food and Drug Administration, Silver Spring, MD, USA
| |
Collapse
|
28
|
Khan F, Jeong GJ, Javaid A, Thuy Nguyen Pham D, Tabassum N, Kim YM. Surface adherence and vacuolar internalization of bacterial pathogens to the Candida spp. cells: Mechanism of persistence and propagation. J Adv Res 2023; 53:115-136. [PMID: 36572338 PMCID: PMC10658324 DOI: 10.1016/j.jare.2022.12.013] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Revised: 12/17/2022] [Accepted: 12/21/2022] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND The co-existence of Candida albicans with the bacteria in the host tissues and organs displays interactions at competitive, antagonistic, and synergistic levels. Several pathogenic bacteria take advantage of such types of interaction for their survival and proliferation. The chemical interaction involves the signaling molecules produced by the bacteria or Candida spp., whereas the physical attachment occurs by involving the surface proteins of the bacteria and Candida. In addition, bacterial pathogens have emerged to internalize inside the C. albicans vacuole, which is one of the inherent properties of the endosymbiotic relationship between the bacteria and the eukaryotic host. AIM OF REVIEW The interaction occurring by the involvement of surface protein from diverse bacterial species with Candida species has been discussed in detail in this paper. An in silico molecular docking study was performed between the surface proteins of different bacterial species and Als3P of C. albicans to explain the molecular mechanism involved in the Als3P-dependent interaction. Furthermore, in order to understand the specificity of C. albicans interaction with Als3P, the evolutionary relatedness of several bacterial surface proteins has been investigated. Furthermore, the environmental factors that influence bacterial pathogen internalization into the Candida vacuole have been addressed. Moreover, the review presented future perspectives for disrupting the cross-kingdom interaction and eradicating the endosymbiotic bacterial pathogens. KEY SCIENTIFIC CONCEPTS OF REVIEW With the involvement of cross-kingdom interactions and endosymbiotic relationships, the bacterial pathogens escape from the environmental stresses and the antimicrobial activity of the host immune system. Thus, the study of interactions between Candida and bacterial pathogens is of high clinical significance.
Collapse
Affiliation(s)
- Fazlurrahman Khan
- Marine Integrated Biomedical Technology Center, The National Key Research Institutes in Universities, Pukyong National University, Busan 48513, Republic of Korea; Research Center for Marine Integrated Bionics Technology, Pukyong National University, Busan 48513, Republic of Korea.
| | - Geum-Jae Jeong
- Department of Food Science and Technology, Pukyong National University, Busan 48513, Republic of Korea
| | - Aqib Javaid
- Department of Biotechnology and Bioinformatics, University of Hyderabad, India
| | - Dung Thuy Nguyen Pham
- Institute of Applied Technology and Sustainable Development, Nguyen Tat Thanh University, Ho Chi Minh City 70000, Vietnam
| | - Nazia Tabassum
- Marine Integrated Biomedical Technology Center, The National Key Research Institutes in Universities, Pukyong National University, Busan 48513, Republic of Korea; Research Center for Marine Integrated Bionics Technology, Pukyong National University, Busan 48513, Republic of Korea
| | - Young-Mog Kim
- Marine Integrated Biomedical Technology Center, The National Key Research Institutes in Universities, Pukyong National University, Busan 48513, Republic of Korea; Research Center for Marine Integrated Bionics Technology, Pukyong National University, Busan 48513, Republic of Korea; Department of Food Science and Technology, Pukyong National University, Busan 48513, Republic of Korea.
| |
Collapse
|
29
|
Deusenbery C, Carneiro O, Oberkfell C, Shukla A. Synergy of Antibiotics and Antibiofilm Agents against Methicillin-Resistant Staphylococcus aureus Biofilms. ACS Infect Dis 2023; 9:1949-1963. [PMID: 37646612 DOI: 10.1021/acsinfecdis.3c00239] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/01/2023]
Abstract
Methicillin-resistant Staphylococcus aureus (MRSA) infections are some of the most common antibiotic-resistant infections, often exacerbated by the formation of biofilms. Here, we evaluated six compounds, three common antibiotics used against MRSA and three antibiofilm compounds, in nine combinations to investigate the mechanisms of synergistic eradication of MRSA biofilms. Using metabolic assessment, colony enumeration, confocal fluorescence microscopy, and scanning electron microscopy, we identified two promising combinations of antibiotics with antibiofilm agents against preformed MRSA biofilms. The broad-spectrum protease, proteinase K, and membrane-targeting antibiotic, daptomycin, worked in synergy against MRSA biofilms by manipulating the protein content, increasing access to the cell membrane of biofilm bacteria. We also found that the combination of cationic peptide, IDR-1018, with the cell wall cross-linking inhibitor, vancomycin, exhibited synergy against MRSA biofilms by causing bacterial damage and preventing repair. Our findings identify synergistic combinations of antibiotics and antibiofilm agents, providing insight into mechanisms that may be explored further for the development of effective treatments against MRSA biofilm.
Collapse
Affiliation(s)
- Carly Deusenbery
- School of Engineering, Center for Biomedical Engineering, Brown University, Providence, Rhode Island 02912, United States
| | - Olivia Carneiro
- Therapeutic Sciences Graduate Program, Division of Biology and Medicine, Brown University, Providence, Rhode Island 02912, United States
| | - Carleigh Oberkfell
- School of Engineering, Center for Biomedical Engineering, Brown University, Providence, Rhode Island 02912, United States
| | - Anita Shukla
- School of Engineering, Center for Biomedical Engineering, Brown University, Providence, Rhode Island 02912, United States
| |
Collapse
|
30
|
Jalihal A, Mortazi A, Forson M, Bashiru M, Le T, Oyebade A, Siraj N. Antibiotics Coupled with Photothermal Therapy for the Enhanced Killing of Bacteria. JOURNAL OF BIOCHEMICAL TECHNOLOGY 2023; 14:50-58. [PMID: 38179150 PMCID: PMC10766427 DOI: 10.51847/nplvoycg9u] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/06/2024]
Abstract
In this study, the application of ionic materials as a combination antibiotic drug was investigated. The fluoroquinolone, Norfloxacin, was converted into the ionic form and combined with the cationic dye, IR780+, using an ion-exchange reaction. The resulting ionic combination drug possesses two killing mechanisms in one compound. The antibiotic chemical mechanism along with the photothermal mechanism that was acquired by adding IR780 to the compound led to the development of a combination antibiotic drug. This ionic combination drug consisting of Norfloxacin anion and IR780 cation is easily dispersed in water using sonication waves. The parent compounds and ionic combination drug, dissolved in organic solvent and dispersed in water, were characterized, and the photophysical properties were studied in detail. It was discovered that the aqueous ionic combination drugs exhibited significant changes in absorbance and photoluminescent properties. In aqueous media, the dispersed ionic combination drug exhibited a very broad absorbance with an additional peak around 1000 nm which is advantageous in photothermal. A significant decrease in the quantum yield along with enhanced non-radiative rate constant was observed for the combination drug in the aqueous. The photothermal mechanism is present in both the parent IR780 dye and the ionic combination drug. The ionic combination drug displayed a high light-to-heat conversion efficiency and temperature increase similar to the parent dye. The combination of both killing mechanisms in the ionic combination drug resulted in enhanced antibacterial activity against Escherichia coli as compared to the parent Norfloxacin and IR780-I individually.
Collapse
Affiliation(s)
- Amanda Jalihal
- Department of Chemistry, University of Arkansas at Little Rock, 2801 S. University Ave, Little Rock, AR 72204, USA
| | - Armin Mortazi
- Department of Chemistry, University of Arkansas at Little Rock, 2801 S. University Ave, Little Rock, AR 72204, USA
| | - Mavis Forson
- Department of Chemistry, University of Arkansas at Little Rock, 2801 S. University Ave, Little Rock, AR 72204, USA
| | - Mujeebat Bashiru
- Department of Chemistry, University of Arkansas at Little Rock, 2801 S. University Ave, Little Rock, AR 72204, USA
| | - Thuy Le
- Department of Chemistry, University of Arkansas at Little Rock, 2801 S. University Ave, Little Rock, AR 72204, USA
| | - Adeniyi Oyebade
- Department of Chemistry, University of Arkansas at Little Rock, 2801 S. University Ave, Little Rock, AR 72204, USA
| | - Noureen Siraj
- Department of Chemistry, University of Arkansas at Little Rock, 2801 S. University Ave, Little Rock, AR 72204, USA
| |
Collapse
|
31
|
Yang D, Yu Z, Zheng M, Yang W, Liu Z, Zhou J, Huang L. Artificial intelligence-accelerated high-throughput screening of antibiotic combinations on a microfluidic combinatorial droplet system. LAB ON A CHIP 2023; 23:3961-3977. [PMID: 37605875 DOI: 10.1039/d3lc00647f] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/23/2023]
Abstract
Microfluidic platforms have been employed as an effective tool for drug screening and exhibit the advantages of lower reagent consumption, higher throughput and a higher degree of automation. Despite the great advancement, it remains challenging to screen complex antibiotic combinations in a simple, high-throughput and systematic manner. Meanwhile, the large amounts of datasets generated during the screening process generally outpace the abilities of the conventional manual or semi-automatic data analysis. To address these issues, we propose an artificial intelligence-accelerated high-throughput combinatorial drug evaluation system (AI-HTCDES), which not only allows high-throughput production of antibiotic combinations with varying concentrations, but can also automatically analyze the dynamic growth of bacteria under the action of different antibiotic combinations. Based on this system, several antibiotic combinations displaying an additive effect are discovered, and the dosage regimens of each component in the combinations are determined. This strategy not only provides useful guidance in the clinical use of antibiotic combination therapy and personalized medicine, but also offers a promising tool for the combinatorial screenings of other medicines.
Collapse
Affiliation(s)
- Deyu Yang
- School of Biomedical Engineering, Sun Yat-sen University, Shenzhen 518107, China.
| | - Ziming Yu
- School of Biomedical Engineering, Sun Yat-sen University, Shenzhen 518107, China.
| | - Mengxin Zheng
- School of Biomedical Engineering, Sun Yat-sen University, Shenzhen 518107, China.
| | - Wei Yang
- School of Biomedical Engineering, Sun Yat-sen University, Shenzhen 518107, China.
| | - Zhangcai Liu
- School of Biomedical Engineering, Sun Yat-sen University, Shenzhen 518107, China.
| | - Jianhua Zhou
- School of Biomedical Engineering, Sun Yat-sen University, Shenzhen 518107, China.
- Key Laboratory of Sensing Technology and Biomedical Instruments of Guangdong Province, School of Biomedical Engineering, Sun Yat-sen University, Guangzhou 510275, China
| | - Lu Huang
- School of Biomedical Engineering, Sun Yat-sen University, Shenzhen 518107, China.
- Key Laboratory of Sensing Technology and Biomedical Instruments of Guangdong Province, School of Biomedical Engineering, Sun Yat-sen University, Guangzhou 510275, China
| |
Collapse
|
32
|
Zhao J, Luan Y, Chen Y, Cheng L, Qin Q. Toxicological and transcriptomic-based analysis of monensin and sulfamethazine co-exposure on male SD rats. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2022; 245:114110. [PMID: 36155339 DOI: 10.1016/j.ecoenv.2022.114110] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Revised: 08/31/2022] [Accepted: 09/20/2022] [Indexed: 06/16/2023]
Abstract
Antibiotic residue has become an emerging environmental contaminant, while the toxicological effects and underlying mechanisms caused by the co-exposure to multiple veterinary antibiotics were rarely studied. In this study, male Sprague Dawley rats were exposed to monensin (M) (1, 2, 10 mg/(kg·body weight (BW)) combined with sulfamethazine (S) (60, 120, 600 mg/(kg·BW)) or single drugs for 28 consecutive days. The body weight, hematological and blood biochemical parameters, organ coefficients, and histopathology were analyzed to discover their combined toxicity effect. Transcriptomic analysis was used to reveal the possible mechanisms of their joint toxicity. Compared with the control group, the weight gain rate was significantly reduced in the H-M+S and H-S, and alkaline phosphatase in H-M+S was significantly increased. Furthermore, relative liver and kidneys weight was significantly increased, and the liver of H-M+S showed more severe lesions in histopathological analysis. For H-M+S, H-M and H-S, transcriptomic results showed that 344, 246, and 99 genes were differentially expressed, respectively. The Gene Ontology terms mainly differ in sterol biosynthetic process and steroid hydroxylase activity. The Kyoto Encyclopedia of Genes and Genome pathways showed abnormal retinol metabolism, metabolism of xenobiotics by cytochrome P450, and drug metabolism-cytochrome 450; the common 30 genes were screened from the network of protein-protein interaction. The results showed that mixed contamination of M and S produces hepatotoxicity by interfering with linoleic acid metabolism, retinol metabolism and CYP450 enzyme-dominated drug metabolism. Further analysis showed that Cyp1a2, Cyp2c61, Ugt1a3, and Ugt1a5 might be the key genes. These findings could provide more evidence for investigating the toxic effects and metabolism of mixed antibiotics contamination in mammals.
Collapse
Affiliation(s)
- Junjie Zhao
- College of Veterinary Medicine, China Agricultural University, Beijing 100093, China.
| | - Yehui Luan
- College of Veterinary Medicine, China Agricultural University, Beijing 100093, China.
| | - Yanan Chen
- College of Veterinary Medicine, China Agricultural University, Beijing 100093, China.
| | - Linli Cheng
- College of Veterinary Medicine, China Agricultural University, Beijing 100093, China; National Reference Laboratory on Residue of Veterinary Medicine, Beijing 100093, China; Beijing Key Laboratory of Animal Source Food Safety Testing Technology, Beijing 100093, China.
| | - Qianxi Qin
- College of Veterinary Medicine, China Agricultural University, Beijing 100093, China.
| |
Collapse
|
33
|
Zhong S, Wu X, Zhang D, Du S, Shen J, Xiao L, Zhu Y, Xu Y, Lin Y, Yin L, Rao M, Lu S. Antibiotics in urine from general adults in Shenzhen, China: Demographic-related difference in exposure levels. THE SCIENCE OF THE TOTAL ENVIRONMENT 2022; 843:157070. [PMID: 35780901 DOI: 10.1016/j.scitotenv.2022.157070] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/28/2022] [Revised: 06/24/2022] [Accepted: 06/26/2022] [Indexed: 06/15/2023]
Abstract
Misuse or overuse of antibiotics can have a variety of detrimental microbial effects. However, the body burden of antibiotics in the general population is currently unclear. In this cross-sectional study, we determined four classes of widely-applied antibiotics (3 imidazoles, 2 sulfonamides, 5 quinolones, and 2 chloramphenicols) in urine samples from 1170 adult residents in Shenzhen, China. Antibiotics were detected in 30.8 % of all urine samples with concentrations ranging from <LOD to 3517 μg/mL, among which metronidazole, ofloxacin and florfenicol were predominant. Notably, antibiotics prohibited for human or veterinary use were detected in 21.0 % of samples, indicating that these antibiotics may still be overused in daily life. We found that the presence of antibiotics in urine is associated with being overweight (OR: 1.386, 95 % CI: 1.056-1.819, p = 0.019) and obesity (OR: 1.862, 95 % CI: 1.103-3.146, p = 0.020) in the adult population. Multilinear regression analysis showed that a percent increase of hydroxy metronidazole was related to 9.86 % positive change of body mass index (p = 0.029). Interestingly, we also found total antibiotic concentration higher in the unmarried group (p = 0.006). Besides, consumption of smoked foods was correlated with urinary antibiotic levels (p = 0.001), indicating smoked meat may be a potential exposure source of veterinary antibiotics. These results highlight the need to reduce human exposure to banned antibiotics. Future research could focus on assessing the health risk and other outcomes of antibiotic overuse.
Collapse
Affiliation(s)
- Shihua Zhong
- Shenzhen Institute of Quality and Safety Inspection and Research, Shenzhen, China
| | - Xiaoling Wu
- School of Public Health (Shenzhen), Sun Yat-sen University, Guangzhou, China
| | - Duo Zhang
- School of Public Health (Shenzhen), Sun Yat-sen University, Guangzhou, China
| | - Sijin Du
- School of Public Health (Shenzhen), Sun Yat-sen University, Guangzhou, China
| | - Junchun Shen
- School of Public Health (Shenzhen), Sun Yat-sen University, Guangzhou, China
| | - Lehan Xiao
- School of Public Health (Shenzhen), Sun Yat-sen University, Guangzhou, China
| | - Ying Zhu
- Shenzhen Institute of Quality and Safety Inspection and Research, Shenzhen, China
| | - Yuanyuan Xu
- Shenzhen Institute of Quality and Safety Inspection and Research, Shenzhen, China
| | - Yuli Lin
- School of Public Health (Shenzhen), Sun Yat-sen University, Guangzhou, China
| | - Liuyi Yin
- School of Public Health (Shenzhen), Sun Yat-sen University, Guangzhou, China
| | - Manting Rao
- School of Public Health (Shenzhen), Sun Yat-sen University, Guangzhou, China
| | - Shaoyou Lu
- School of Public Health (Shenzhen), Sun Yat-sen University, Guangzhou, China.
| |
Collapse
|
34
|
Huang F, Cai X, Hou X, Zhang Y, Liu J, Yang L, Liu Y, Liu J. A dynamic covalent polymeric antimicrobial for conquering drug-resistant bacterial infection. EXPLORATION (BEIJING, CHINA) 2022; 2:20210145. [PMID: 37325499 PMCID: PMC10191036 DOI: 10.1002/exp.20210145] [Citation(s) in RCA: 55] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Accepted: 04/27/2022] [Indexed: 06/15/2023]
Abstract
Increasing bacterial drug resistance to antibiotics has posed a major threat to contemporary public health, which resulted in a large number of people suffering from serious infections and ending up dying without any effective therapies every year. Here, a dynamic covalent polymeric antimicrobial, based on phenylboronic acid (PBA)-installed micellar nanocarriers incorporating clinical vancomycin and curcumin, is developed to overcome drug-resistant bacterial infections. The formation of this antimicrobial is facilitated by reversible dynamic covalent interactions between PBA moieties in polymeric micelles and diols in vancomycin, which impart favorable stability in blood circulation and excellent acid-responsiveness in the infection microenvironment. Moreover, the structurally similar aromatic vancomycin and curcumin molecules can afford π-π stacking interaction to realize simultaneous delivery and release of payloads. In comparison with monotherapy, this dynamic covalent polymeric antimicrobial demonstrated more significant eradication of drug-resistant bacteria in vitro and in vivo due to the synergism of the two drugs. Furthermore, the achieved combination therapy shows satisfied biocompatibility without unwanted toxicity. Considering various antibiotics contain diol and aromatic structures, this simple and robust strategy can become a universal platform to combat the ever-threatening drug-resistant infectious diseases.
Collapse
Affiliation(s)
- Fan Huang
- Key Laboratory of Radiopharmacokinetics for Innovative DrugsChinese Academy of Medical Sciences, and Institute of Radiation MedicineChinese Academy of Medical Sciences & Peking Union Medical CollegeTianjinP. R. China
| | - Xiaoyao Cai
- Key Laboratory of Radiopharmacokinetics for Innovative DrugsChinese Academy of Medical Sciences, and Institute of Radiation MedicineChinese Academy of Medical Sciences & Peking Union Medical CollegeTianjinP. R. China
| | - Xiaoxue Hou
- Key Laboratory of Radiopharmacokinetics for Innovative DrugsChinese Academy of Medical Sciences, and Institute of Radiation MedicineChinese Academy of Medical Sciences & Peking Union Medical CollegeTianjinP. R. China
| | - Yumin Zhang
- Key Laboratory of Radiopharmacokinetics for Innovative DrugsChinese Academy of Medical Sciences, and Institute of Radiation MedicineChinese Academy of Medical Sciences & Peking Union Medical CollegeTianjinP. R. China
| | - Jinjian Liu
- Key Laboratory of Radiopharmacokinetics for Innovative DrugsChinese Academy of Medical Sciences, and Institute of Radiation MedicineChinese Academy of Medical Sciences & Peking Union Medical CollegeTianjinP. R. China
| | - Lijun Yang
- Key Laboratory of Radiopharmacokinetics for Innovative DrugsChinese Academy of Medical Sciences, and Institute of Radiation MedicineChinese Academy of Medical Sciences & Peking Union Medical CollegeTianjinP. R. China
| | - Yong Liu
- Engineering Research Center of Clinical Functional Materials and Diagnosis & Treatment Devices of Zhejiang ProvinceWenzhou InstituteUniversity of Chinese Academy of Sciences, Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health)WenzhouZhejiangP. R. China
| | - Jianfeng Liu
- Key Laboratory of Radiopharmacokinetics for Innovative DrugsChinese Academy of Medical Sciences, and Institute of Radiation MedicineChinese Academy of Medical Sciences & Peking Union Medical CollegeTianjinP. R. China
| |
Collapse
|
35
|
Vatankhah M, Khosravi K, Zargar N, Shirvani A, Nekoofar MH, Dianat O. Antibacterial efficacy of antibiotic pastes versus calcium hydroxide intracanal dressing: A systematic review and meta-analysis of ex vivo studies. J Conserv Dent 2022; 25:463-480. [PMID: 36506621 PMCID: PMC9733540 DOI: 10.4103/jcd.jcd_183_22] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2022] [Revised: 05/15/2022] [Accepted: 05/20/2022] [Indexed: 12/15/2022] Open
Abstract
Background Conflicting findings on the potency of antibiotic pastes versus calcium hydroxide (CH) have been evident in the literature. Aims To compare the antibacterial efficacy of single antibiotic paste (SAP), double antibiotic paste (DAP), triple antibiotic paste (TAP), and modified TAP (mTAP) with CH on bacterial biofilms. Methods PubMed, Scopus, and Embase were comprehensively searched until August 23, 2021. The study protocol was registered in the PROSPERO. Ex vivo studies performed on Enterococcus faecalis or polymicrobial biofilms incubated on human/bovine dentin were selected. The quality of the studies was assessed using a customized quality assessment tool. Standardized mean difference (SMD) with a 95% confidence interval (CI) was calculated for the meta-analysis. Meta-regression models were used to identify the sources of heterogeneity and to compare the efficacy of pastes. Results The qualitative and quantitative synthesis included 40 and 23 papers, respectively, out of 1421 search results. TAP (SMD = -3.82; CI, -5.44 to -2.21; P < 0.001) and SAPs (SMD = -2.38; CI, -2.81 to - 1.94; P < 0.001) had significantly higher antibacterial efficacy compared to the CH on E. faecalis biofilm. However, no significant difference was found between the efficacy of DAP (SMD = -2.74; CI, -5.56-0.07; P = 0.06) or mTAP (SMD = -0.28; CI, -0.82-0.26; P = 0.31) and CH. Meta-regression model on E. faecalis showed that SAPs have similar efficacy compared to TAP and significantly better efficacy than DAP. On dual-species (SMD = 0.15; CI, -1.00-1.29; P = 0.80) or multi-species (SMD = 0.23; CI, -0.08-0.55; P = 0.15) biofilms, DAP and CH had similar efficacy. Conclusions Ex vivo evidence showed that antibiotic pastes were either superior or equal to CH. The studied SAPs had considerably higher or similar antibacterial effectiveness compared to DAP, CH, and TAP. Hence, combined antibiotic therapy was not necessarily required for root canal disinfection ex vivo.
Collapse
Affiliation(s)
- Mohammadreza Vatankhah
- Iranian Center for Endodontic Research, Research Institute of Dental Sciences, School of Dentistry, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Kamyar Khosravi
- Iranian Center for Endodontic Research, Research Institute of Dental Sciences, School of Dentistry, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Nazanin Zargar
- Department of Endodontics, School of Dentistry, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Armin Shirvani
- Iranian Center for Endodontic Research, Research Institute of Dental Sciences, School of Dentistry, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | | | - Omid Dianat
- Department of Advanced Oral Sciences and Therapeutics, School of Dentistry, University of Maryland, Baltimore, Maryland, USA
| |
Collapse
|
36
|
Lv J, Liu G, Hao J, Ju Y, Sun B, Sun Y. Computational models, databases and tools for antibiotic combinations. Brief Bioinform 2022; 23:6652783. [PMID: 35915052 DOI: 10.1093/bib/bbac309] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Revised: 07/07/2022] [Accepted: 07/08/2022] [Indexed: 11/13/2022] Open
Abstract
Antibiotic combination is a promising strategy to extend the lifetime of antibiotics and thereby combat antimicrobial resistance. However, screening for new antibiotic combinations is both time-consuming and labor-intensive. In recent years, an increasing number of researchers have used computational models to predict effective antibiotic combinations. In this review, we summarized existing computational models for antibiotic combinations and discussed the limitations and challenges of these models in detail. In addition, we also collected and summarized available data resources and tools for antibiotic combinations. This study aims to help computational biologists design more accurate and interpretable computational models.
Collapse
Affiliation(s)
- Ji Lv
- College of Computer Science and Technology, Jilin University, Changchun, China.,Key Laboratory of Symbolic Computation and Knowledge Engineering of Ministry of Education, Jilin University, Changchun, China
| | - Guixia Liu
- College of Computer Science and Technology, Jilin University, Changchun, China.,Key Laboratory of Symbolic Computation and Knowledge Engineering of Ministry of Education, Jilin University, Changchun, China
| | - Junli Hao
- College of Food Science, Northeast Agricultural University, Harbin, China
| | - Yuan Ju
- Sichuan University Library, Sichuan University, Chengdu, China
| | - Binwen Sun
- Engineering Research Center for New Materials and Precision Treatment Technology of Malignant Tumor Therapy, The Second Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Ying Sun
- Department of Respiratory Medicine, the First Hospital of Jilin University, Changchun, China
| |
Collapse
|
37
|
Rivani E, Endraswari PD, Widodo ADW. Growth kinetics of multiple Acinetobacter baumannii resistotype after meropenem-based antibiotic combination exposure. F1000Res 2022; 11:762. [PMID: 36531260 PMCID: PMC9723411 DOI: 10.12688/f1000research.122221.1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 07/04/2022] [Indexed: 09/10/2024] Open
Abstract
Background: Carbapenems are the treatment of choice for multidrug-resistant (MDR) and extensively drug-resistant (XDR) Acinetobacter baumannii infections, but the emergence of carbapenem-resistant A. baumannii (CRAB) has rendered it ineffective in the vast majority of cases. Combination therapy has grown in popularity over the last decade; this study aims to analyze A.baumannii growth kinetics after exposure to meropenem and ampicillin-sulbactam compared with meropenem and amikacin antibiotic combinations in clinically relevant concentrations. Methods: This experimental laboratory study was conducted on the A.baumannii ATCC 19606 isolate and three clinical isolates that were intermediate or resistant to tested antibiotics. Meropenem and ampicillin-sulbactam, as well as meropenem and amikacin, were tested at four different concentrations against isolates. Turbidity measurements were taken at predetermined time points of 0, 1, 2, 4, 6, 8, and 24 hours following exposure; bacterial concentration was enumerated using the agar plate method, with the results plotted in a time-kill curve. Results: A bactericidal effect was achieved in isolates that were intermediate to ampicillin sulbactam and resistant to meropenem after the administration of meropenem and ampicillin-sulbactam combination with a concentration of 4 µg/ml and 16/8 µg/ml, respectively. The combination of meropenem and ampicillin-sulbactam demonstrated bacteriostatic activity against isolates that were resistant to both antibiotics. Isolates treated with resistant antibiotics showed an increased growth rate compared to the growth control. Conclusion: The combination of meropenem and ampicillin-sulbactam could be a promising combination therapy in treating CRAB infections. The mechanism and degree of antibiotic resistance in the isolates affect the efficacy of antibiotic combinations; further research is needed to corroborate the findings of this study.
Collapse
Affiliation(s)
- Erizka Rivani
- Department of Microbiology, Faculty of Medicine, Sriwijaya University, Palembang, South Sumatera, 30114, Indonesia
- Department of Microbiology, Faculty of Medicine, Airlangga University, Surabaya, East Java, 60115, Indonesia
- Clinical Microbiology Department, Dr. Soetomo General Academic Hospital, Surabaya, East Java, 60286, Indonesia
| | - Pepy Dwi Endraswari
- Department of Microbiology, Faculty of Medicine, Airlangga University, Surabaya, East Java, 60115, Indonesia
- Clinical Microbiology Department, Dr. Soetomo General Academic Hospital, Surabaya, East Java, 60286, Indonesia
| | - Agung Dwi Wahyu Widodo
- Department of Microbiology, Faculty of Medicine, Airlangga University, Surabaya, East Java, 60115, Indonesia
- Clinical Microbiology Department, Dr. Soetomo General Academic Hospital, Surabaya, East Java, 60286, Indonesia
| |
Collapse
|
38
|
Rivani E, Endraswari PD, Widodo ADW. Growth kinetics of multiple Acinetobacter baumannii resistotype after meropenem-based antibiotic combination exposure. F1000Res 2022; 11:762. [PMID: 36531260 PMCID: PMC9723411 DOI: 10.12688/f1000research.122221.2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 10/21/2022] [Indexed: 11/29/2022] Open
Abstract
Background: Carbapenems are the treatment of choice for multidrug-resistant (MDR) and extensively drug-resistant (XDR) Acinetobacter baumannii infections, but the emergence of carbapenem-resistant A. baumannii (CRAB) has rendered it ineffective in the vast majority of cases. Combination therapy has grown in popularity over the last decade; this study aims to analyze A.baumannii growth kinetics after exposure to meropenem and ampicillin-sulbactam compared with meropenem and amikacin antibiotic combinations in clinically relevant concentrations. Methods: This experimental laboratory study was conducted on the A. baumannii ATCC 19606 isolate and three clinical isolates that were intermediate or resistant to tested antibiotics. Meropenem and ampicillin-sulbactam, as well as meropenem and amikacin, were tested at four different concentrations against isolates. Turbidity measurements were taken at predetermined time points of 0, 1, 2, 4, 6, 8, and 24 hours following exposure; bacterial concentration was enumerated using the agar plate method, with the results plotted in a time-kill curve. Results: A bactericidal effect was achieved in isolates that were intermediate to ampicillin-sulbactam and resistant to meropenem after the administration of meropenem and ampicillin-sulbactam combination with a concentration of 4 µg/ml and 16/8 µg/ml, respectively. The combination of meropenem and ampicillin-sulbactam demonstrated bacteriostatic activity against isolates that were resistant to both antibiotics. Isolates treated with resistant antibiotics showed an increased growth rate compared to the growth control. Conclusion: The combination of meropenem and ampicillin-sulbactam could be a promising combination therapy in treating CRAB infections. The mechanism and degree of antibiotic resistance in the isolates affect the efficacy of antibiotic combinations; further research is needed to corroborate the findings of this study.
Collapse
Affiliation(s)
- Erizka Rivani
- Department of Microbiology, Faculty of Medicine, Sriwijaya University, Palembang, South Sumatera, 30114, Indonesia
- Department of Microbiology, Faculty of Medicine, Airlangga University, Surabaya, East Java, 60115, Indonesia
- Clinical Microbiology Department, Dr. Soetomo General Academic Hospital, Surabaya, East Java, 60286, Indonesia
| | - Pepy Dwi Endraswari
- Department of Microbiology, Faculty of Medicine, Airlangga University, Surabaya, East Java, 60115, Indonesia
- Clinical Microbiology Department, Dr. Soetomo General Academic Hospital, Surabaya, East Java, 60286, Indonesia
| | - Agung Dwi Wahyu Widodo
- Department of Microbiology, Faculty of Medicine, Airlangga University, Surabaya, East Java, 60115, Indonesia
- Clinical Microbiology Department, Dr. Soetomo General Academic Hospital, Surabaya, East Java, 60286, Indonesia
| |
Collapse
|
39
|
Ma X, Fu S, Wang Y, Zhao L, Yu W, He Y, Ni W, Gao Z. Proteomics Study of the Synergistic Killing of Tigecycline in Combination With Aminoglycosides Against Carbapenem-Resistant Klebsiella pneumoniae. Front Cell Infect Microbiol 2022; 12:920761. [PMID: 35846751 PMCID: PMC9280366 DOI: 10.3389/fcimb.2022.920761] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Accepted: 05/18/2022] [Indexed: 11/14/2022] Open
Abstract
Co-administration of antibiotics with synergistic effects is one method to combat carbapenem-resistant organisms. Although the synergistic effects of tigecycline combined with aminoglycosides against carbapenem-resistant Klebsiella pneumoniae (CRKP) have been demonstrated in vitro and in animal models, the underlying mechanism remains elusive. Here we used proteomics analysis to assess the short-term bacterial responses to tigecycline and aminoglycosides alone or in combination. Emergence of tigecycline resistance during treatment and the susceptibility of tigecycline-resistant strains to aminoglycosides was further evaluated. The proteomic responses to tigecycline and aminoglycosides were divergent in monotherapy, with proteomic alterations to combination therapy dominated by tigecycline. Adaptive responses to tigecycline were associated with the upregulation of oxidative phosphorylation and translation-related proteins. These responses might confer CRKP hypersensitivity towards aminoglycosides by increasing the drug uptake and binding targets. Meanwhile, tigecycline might perturb adaptive responses to aminoglycosides through inhibition of heat shock response. Tigecycline-resistant strains could be isolated within 24 h exposure even in strains without heteroresistance, and the sensitivity to aminoglycosides significantly increased in resistant strains. Overall, these findings demonstrated that adaption to tigecycline in CRKP was a double-edged sword associated with the synergistic killing in tigecycline–aminoglycoside combination. Evolutionary hypersensitivity can provide novel insight into the mechanisms of antibiotic synergistic effects.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Wentao Ni
- *Correspondence: Wentao Ni, ; Zhancheng Gao,
| | | |
Collapse
|
40
|
Ju YG, Lee HJ, Yim HS, Lee MG, Sohn JW, Yoon YK. In vitro synergistic antimicrobial activity of a combination of meropenem, colistin, tigecycline, rifampin, and ceftolozane/tazobactam against carbapenem-resistant Acinetobacter baumannii. Sci Rep 2022; 12:7541. [PMID: 35534512 PMCID: PMC9085847 DOI: 10.1038/s41598-022-11464-6] [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: 12/14/2021] [Accepted: 04/08/2022] [Indexed: 11/20/2022] Open
Abstract
We investigated the in vitro activity of various antimicrobial combinations against carbapenem-resistant Acinetobacter baumannii (CRAB) isolates. The in vitro activity of six two-drug combinations against CRAB isolates collected from the blood samples of patients with bloodstream infection was evaluated using the checkerboard method and time-kill assay [0.5 ×, 1 ×, and 2 × minimum inhibitory concentration (MIC)] to identify potential synergistic and bactericidal two-drug combinations against CRAB isolates. The effects of meropenem, colistin, tigecycline, rifampin, and ceftolozane/tazobactam combinations were investigated. All 10 CRAB isolates in our study produced the OXA-58-type and OXA-23-type carbapenem-hydrolyzing oxacillinases. The colistin-ceftolozane/tazobactam combination showed synergistic effects in both the time-kill assay (using an antibiotic concentration of 1 × MIC) and the checkerboard method. It also showed bactericidal effects in the time-kill assay. For all 10 CRAB isolates, time-kill curves showed synergistic bactericidal activity of the colistin-ceftolozane/tazobactam combination at 0.5 × MIC. Overall, there was substantial discordance of synergistic activity between the checkerboard microdilution and time-kill assays (with a concordance of 31.7%). Our study demonstrated that two-drug combinations of colistin and ceftolozane/tazobactam could be useful treatment alternatives for CRAB infections. The effects of these antibiotic combinations should be evaluated using in vivo experimental models.
Collapse
|
41
|
Khan F, Tabassum N, Bamunuarachchi NI, Kim YM. Phloroglucinol and Its Derivatives: Antimicrobial Properties toward Microbial Pathogens. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2022; 70:4817-4838. [PMID: 35418233 DOI: 10.1021/acs.jafc.2c00532] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Phloroglucinol (PG) is a natural product isolated from plants, algae, and microorganisms. Aside from that, the number of PG derivatives has expanded due to the discovery of their potential biological roles. Aside from its diverse biological activities, PG and its derivatives have been widely utilized to treat microbial infections caused by bacteria, fungus, and viruses. The rapid emergence of antimicrobial-resistant microbial infections necessitates the chemical synthesis of numerous PG derivatives in order to meet the growing demand for drugs. This review focuses on the use of PG and its derivatives to control microbial infection and the underlying mechanism of action. Furthermore, as future perspectives, some of the various alternative strategies, such as the use of PG and its derivatives in conjugation, nanoformulation, antibiotic combination, and encapsulation, have been thoroughly discussed. This review will enable the researcher to investigate the possible antibacterial properties of PG and its derivatives, either free or in the form of various formulations.
Collapse
Affiliation(s)
- Fazlurrahman Khan
- Research Center for Marine Integrated Bionics Technology, Pukyong National University, Busan 48513, Republic of Korea
| | - Nazia Tabassum
- Industry 4.0 Convergence Bionics Engineering, Pukyong National University, Busan 48513, Republic of Korea
| | | | - Young-Mog Kim
- Research Center for Marine Integrated Bionics Technology, Pukyong National University, Busan 48513, Republic of Korea
- Department of Food Science and Technology, Pukyong National University, Busan 48513, Republic of Korea
| |
Collapse
|
42
|
Abichabki N, Zacharias LV, Moreira NC, Bellissimo-Rodrigues F, Moreira FL, Benzi JRL, Ogasawara TMC, Ferreira JC, Ribeiro CM, Pavan FR, Pereira LRL, Brancini GTP, Braga GÚL, Zuardi AW, Hallak JEC, Crippa JAS, Lanchote VL, Cantón R, Darini ALC, Andrade LN. Potential cannabidiol (CBD) repurposing as antibacterial and promising therapy of CBD plus polymyxin B (PB) against PB-resistant gram-negative bacilli. Sci Rep 2022; 12:6454. [PMID: 35440801 PMCID: PMC9018834 DOI: 10.1038/s41598-022-10393-8] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Accepted: 03/04/2022] [Indexed: 01/24/2023] Open
Abstract
This study aimed to assess the ultrapure cannabidiol (CBD) antibacterial activity and to investigate the antibacterial activity of the combination CBD + polymyxin B (PB) against Gram-negative (GN) bacteria, including PB-resistant Gram-negative bacilli (GNB). We used the standard broth microdilution method, checkerboard assay, and time-kill assay. CBD exhibited antibacterial activity against Gram-positive bacteria, lipooligosaccharide (LOS)-expressing GN diplococcus (GND) (Neisseria gonorrhoeae, Neisseria meningitidis, Moraxella catarrhalis), and Mycobacterium tuberculosis, but not against GNB. For most of the GNB studied, our results showed that low concentrations of PB (≤ 2 µg/mL) allow CBD (≤ 4 µg/mL) to exert antibacterial activity against GNB (e.g., Klebsiella pneumoniae, Escherichia coli, Acinetobacter baumannii), including PB-resistant GNB. CBD + PB also showed additive and/or synergistic effect against LOS-expressing GND. Time-kill assays results showed that the combination CBD + PB leads to a greater reduction in the number of colony forming units per milliliter compared to CBD and PB alone, at the same concentration used in combination, and the combination CBD + PB was synergistic for all four PB-resistant K. pneumoniae isolates evaluated. Our results show that CBD has translational potential and should be further explored as a repurposed antibacterial agent in clinical trials. The antibacterial efficacy of the combination CBD + PB against multidrug-resistant and extensively drug-resistant GNB, especially PB-resistant K. pneumoniae, is particularly promising.
Collapse
Affiliation(s)
- Nathália Abichabki
- Department of Clinical Analyses, Toxicology and Food Science (DACTB), School of Pharmaceutical Sciences of Ribeirao Preto (FCFRP), University of São Paulo (USP), Av. do Café, s/nº, Campus Universitário, Ribeirão Preto, SP, 14040-903, Brazil
| | - Luísa V Zacharias
- Department of Clinical Analyses, Toxicology and Food Science (DACTB), School of Pharmaceutical Sciences of Ribeirao Preto (FCFRP), University of São Paulo (USP), Av. do Café, s/nº, Campus Universitário, Ribeirão Preto, SP, 14040-903, Brazil
| | - Natália C Moreira
- Department of Clinical Analyses, Toxicology and Food Science (DACTB), School of Pharmaceutical Sciences of Ribeirao Preto (FCFRP), University of São Paulo (USP), Av. do Café, s/nº, Campus Universitário, Ribeirão Preto, SP, 14040-903, Brazil
| | - Fernando Bellissimo-Rodrigues
- Department of Social Medicine, Ribeirão Preto Medical School (FMRP), University of São Paulo (USP), Ribeirão Preto, SP, Brazil
| | - Fernanda L Moreira
- Department of Clinical Analyses, Toxicology and Food Science (DACTB), School of Pharmaceutical Sciences of Ribeirao Preto (FCFRP), University of São Paulo (USP), Av. do Café, s/nº, Campus Universitário, Ribeirão Preto, SP, 14040-903, Brazil
| | - Jhohann R L Benzi
- Department of Clinical Analyses, Toxicology and Food Science (DACTB), School of Pharmaceutical Sciences of Ribeirao Preto (FCFRP), University of São Paulo (USP), Av. do Café, s/nº, Campus Universitário, Ribeirão Preto, SP, 14040-903, Brazil
| | - Tânia M C Ogasawara
- Department of Clinical Analyses, Toxicology and Food Science (DACTB), School of Pharmaceutical Sciences of Ribeirao Preto (FCFRP), University of São Paulo (USP), Av. do Café, s/nº, Campus Universitário, Ribeirão Preto, SP, 14040-903, Brazil
| | - Joseane C Ferreira
- Department of Clinical Analyses, Toxicology and Food Science (DACTB), School of Pharmaceutical Sciences of Ribeirao Preto (FCFRP), University of São Paulo (USP), Av. do Café, s/nº, Campus Universitário, Ribeirão Preto, SP, 14040-903, Brazil
| | - Camila M Ribeiro
- Department of Biological Sciences, School of Pharmaceutical Sciences (FCF), São Paulo State University (UNESP), Araraquara, SP, Brazil
| | - Fernando R Pavan
- Department of Biological Sciences, School of Pharmaceutical Sciences (FCF), São Paulo State University (UNESP), Araraquara, SP, Brazil
| | - Leonardo R L Pereira
- Department of Pharmaceutical Sciences (DCF), School of Pharmaceutical Sciences of Ribeirão Preto (FCFRP), University of São Paulo (USP), Ribeirão Preto, SP, Brazil
| | - Guilherme T P Brancini
- Department of Clinical Analyses, Toxicology and Food Science (DACTB), School of Pharmaceutical Sciences of Ribeirao Preto (FCFRP), University of São Paulo (USP), Av. do Café, s/nº, Campus Universitário, Ribeirão Preto, SP, 14040-903, Brazil
| | - Gilberto Ú L Braga
- Department of Clinical Analyses, Toxicology and Food Science (DACTB), School of Pharmaceutical Sciences of Ribeirao Preto (FCFRP), University of São Paulo (USP), Av. do Café, s/nº, Campus Universitário, Ribeirão Preto, SP, 14040-903, Brazil
| | - Antonio W Zuardi
- Department of Neurosciences and Behavioral Sciences, Ribeirão Preto Medical School (FMRP), University of São Paulo (USP), Ribeirão Preto, SP, Brazil
- National Institute of Science and Technology for Translational Medicine (INCT-TM), Conselho Nacional de Desenvolvimento Científico e Tecnológico (CNPq), Brasília, DF, Brazil
| | - Jaime E C Hallak
- Department of Neurosciences and Behavioral Sciences, Ribeirão Preto Medical School (FMRP), University of São Paulo (USP), Ribeirão Preto, SP, Brazil
- National Institute of Science and Technology for Translational Medicine (INCT-TM), Conselho Nacional de Desenvolvimento Científico e Tecnológico (CNPq), Brasília, DF, Brazil
| | - José A S Crippa
- Department of Neurosciences and Behavioral Sciences, Ribeirão Preto Medical School (FMRP), University of São Paulo (USP), Ribeirão Preto, SP, Brazil
- National Institute of Science and Technology for Translational Medicine (INCT-TM), Conselho Nacional de Desenvolvimento Científico e Tecnológico (CNPq), Brasília, DF, Brazil
| | - Vera L Lanchote
- Department of Clinical Analyses, Toxicology and Food Science (DACTB), School of Pharmaceutical Sciences of Ribeirao Preto (FCFRP), University of São Paulo (USP), Av. do Café, s/nº, Campus Universitário, Ribeirão Preto, SP, 14040-903, Brazil
| | - Rafael Cantón
- Hospital Universitario Ramón y Cajal and Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Madrid, Spain
| | - Ana Lúcia C Darini
- Department of Clinical Analyses, Toxicology and Food Science (DACTB), School of Pharmaceutical Sciences of Ribeirao Preto (FCFRP), University of São Paulo (USP), Av. do Café, s/nº, Campus Universitário, Ribeirão Preto, SP, 14040-903, Brazil
| | - Leonardo N Andrade
- Department of Clinical Analyses, Toxicology and Food Science (DACTB), School of Pharmaceutical Sciences of Ribeirao Preto (FCFRP), University of São Paulo (USP), Av. do Café, s/nº, Campus Universitário, Ribeirão Preto, SP, 14040-903, Brazil.
| |
Collapse
|
43
|
Cantrell JM, Chung CH, Chandrasekaran S. Machine learning to design antimicrobial combination therapies: promises and pitfalls. Drug Discov Today 2022; 27:1639-1651. [DOI: 10.1016/j.drudis.2022.04.006] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Revised: 01/20/2022] [Accepted: 04/04/2022] [Indexed: 01/13/2023]
|
44
|
Li H, Maimaitiming M, Zhou Y, Li H, Wang P, Liu Y, Schäberle TF, Liu Z, Wang CY. Discovery of Marine Natural Products as Promising Antibiotics against Pseudomonas aeruginosa. Mar Drugs 2022; 20:192. [PMID: 35323491 PMCID: PMC8954164 DOI: 10.3390/md20030192] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Revised: 03/02/2022] [Accepted: 03/03/2022] [Indexed: 12/29/2022] Open
Abstract
Pseudomonas aeruginosa, one of the most intractable Gram-negative bacteria, has become a public health threat due to its outer polysaccharide layer, efflux transporter system, and high level of biofilm formation, all of which contribute to multi-drug resistance. Even though it is a pathogen of the highest concern, the status of the antibiotic development pipeline is unsatisfactory. In this review, we summarize marine natural products (MNPs) isolated from marine plants, animals, and microorganisms which possess unique structures and promising antibiotic activities against P. aeruginosa. In the last decade, nearly 80 such MNPs, ranging from polyketides to alkaloids, peptides, and terpenoids, have been discovered. Representative compounds exhibited impressive in vitro anti-P. aeruginosa activities with MIC values in the single-digit nanomolar range and in vivo efficacy in infectious mouse models. For some of the compounds, the preliminary structure-activity-relationship (SAR) and anti-bacterial mechanisms of selected compounds were introduced. Compounds that can disrupt biofilm formation or membrane integrity displayed potent inhibition of multi-resistant clinical P. aeruginosa isolates and could be considered as lead compounds for future development. Challenges on how to translate hits into useful candidates for clinical development are also proposed and discussed.
Collapse
Affiliation(s)
- Haoran Li
- Institute of Evolution & Marine Biodiversity, School of Medicine and Pharmacy, College of Food Science and Engineering, Ocean University of China, Qingdao 266003, China
- Laboratory for Marine Drugs and Bioproducts, Qingdao National Laboratory for Marine Science and Technology, Qingdao 266237, China
| | - Mireguli Maimaitiming
- Institute of Evolution & Marine Biodiversity, School of Medicine and Pharmacy, College of Food Science and Engineering, Ocean University of China, Qingdao 266003, China
- Laboratory for Marine Drugs and Bioproducts, Qingdao National Laboratory for Marine Science and Technology, Qingdao 266237, China
| | - Yue Zhou
- Institute of Evolution & Marine Biodiversity, School of Medicine and Pharmacy, College of Food Science and Engineering, Ocean University of China, Qingdao 266003, China
- Laboratory for Marine Drugs and Bioproducts, Qingdao National Laboratory for Marine Science and Technology, Qingdao 266237, China
| | - Huaxuan Li
- Institute of Evolution & Marine Biodiversity, School of Medicine and Pharmacy, College of Food Science and Engineering, Ocean University of China, Qingdao 266003, China
- Laboratory for Marine Drugs and Bioproducts, Qingdao National Laboratory for Marine Science and Technology, Qingdao 266237, China
| | - Pingyuan Wang
- Institute of Evolution & Marine Biodiversity, School of Medicine and Pharmacy, College of Food Science and Engineering, Ocean University of China, Qingdao 266003, China
- Laboratory for Marine Drugs and Bioproducts, Qingdao National Laboratory for Marine Science and Technology, Qingdao 266237, China
| | - Yang Liu
- Institute for Insect Biotechnology, Justus-Liebig-University Giessen, Ohlebergsweg 12, 35392 Giessen, Germany
| | - Till F Schäberle
- Institute for Insect Biotechnology, Justus-Liebig-University Giessen, Ohlebergsweg 12, 35392 Giessen, Germany
- Branch for Bioresources, Fraunhofer Institute for Molecular Biology and Applied Ecology (IME), 35392 Giessen, Germany
- Partner Site Giessen-Marburg-Langen, German Center for Infection Research (DZIF), 35392 Giessen, Germany
| | - Zhiqing Liu
- Institute of Evolution & Marine Biodiversity, School of Medicine and Pharmacy, College of Food Science and Engineering, Ocean University of China, Qingdao 266003, China
- Laboratory for Marine Drugs and Bioproducts, Qingdao National Laboratory for Marine Science and Technology, Qingdao 266237, China
| | - Chang-Yun Wang
- Institute of Evolution & Marine Biodiversity, School of Medicine and Pharmacy, College of Food Science and Engineering, Ocean University of China, Qingdao 266003, China
- Laboratory for Marine Drugs and Bioproducts, Qingdao National Laboratory for Marine Science and Technology, Qingdao 266237, China
| |
Collapse
|
45
|
Davis K, Greenstein T, Viau Colindres R, Aldridge BB. Leveraging laboratory and clinical studies to design effective antibiotic combination therapy. Curr Opin Microbiol 2021; 64:68-75. [PMID: 34628295 PMCID: PMC8671129 DOI: 10.1016/j.mib.2021.09.006] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Revised: 07/28/2021] [Accepted: 09/16/2021] [Indexed: 01/21/2023]
Abstract
Interest in antibiotic combination therapy is increasing due to antimicrobial resistance and a slowing antibiotic pipeline. However, aside from specific indications, combination therapy in the clinic is often not administered systematically; instead, it is used at the physician's discretion as a bet-hedging mechanism to increase the chances of appropriately targeting a pathogen(s) with an unknown antibiotic resistance profile. Some recent clinical trials have been unable to demonstrate superior efficacy of combination therapy over monotherapy. Other trials have shown a benefit of combination therapy in defined circumstances consistent with recent studies indicating that factors including species, strain, resistance profile, and microenvironment affect drug combination efficacy and drug interactions. In this review, we discuss how a careful study design that takes these factors into account, along with the different drug interaction and potency metrics for assessing combination performance, may provide the necessary insight to understand the best clinical use-cases for combination therapy.
Collapse
Affiliation(s)
- Kathleen Davis
- Department of Molecular Biology & Microbiology, Tufts University School of Medicine, United States; Stuart B. Levy Center for Integrated Management of Antimicrobial Resistance, United States
| | - Talia Greenstein
- Stuart B. Levy Center for Integrated Management of Antimicrobial Resistance, United States; Graduate School of Biomedical Sciences, Tufts University School of Medicine, United States
| | - Roberto Viau Colindres
- Stuart B. Levy Center for Integrated Management of Antimicrobial Resistance, United States; Department of Geographic Medicine and Infectious Diseases, Tufts Medical Center, United States
| | - Bree B Aldridge
- Department of Molecular Biology & Microbiology, Tufts University School of Medicine, United States; Stuart B. Levy Center for Integrated Management of Antimicrobial Resistance, United States; Graduate School of Biomedical Sciences, Tufts University School of Medicine, United States
| |
Collapse
|
46
|
Feizi S, Cooksley CM, Nepal R, Psaltis AJ, Wormald PJ, Vreugde S. Silver nanoparticles as a bioadjuvant of antibiotics against biofilm-mediated infections with methicillin-resistant Staphylococcus aureus and Pseudomonas aeruginosa in chronic rhinosinusitis patients. Pathology 2021; 54:453-459. [PMID: 34844745 DOI: 10.1016/j.pathol.2021.08.014] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Revised: 08/11/2021] [Accepted: 08/24/2021] [Indexed: 11/26/2022]
Abstract
Infectious diseases caused by antibiotic-resistant bacteria in planktonic and biofilm form are difficult to treat with conventional antibiotics. Silver nanoparticles (Ag NPs) can be used as alternatives to antibiotics and can alter the susceptibility of bacteria to antibiotics. Here, the antibacterial properties of 16 different antibiotics and Ag NPs, alone and in combination, were tested against clinical isolates of Pseudomonas aeruginosa (n=3), Staphylococcus aureus (n=3) and methicillin-resistant Staphylococcus aureus (MRSA) (n=2) isolated from chronic rhinosinusitis (CRS) patients. The microdilution method and resazurin assay were used to determine the minimum inhibitory concentration and minimum biofilm eradication concentration for planktonic and biofilm forms, respectively. Results showed that Ag NPs and gentamicin combinations had synergistic antibacterial activity against P. aeruginosa planktonic and biofilm forms and MRSA biofilms. Furthermore, additive effects against biofilms were seen for combinations of Ag NPs with tobramycin or ciprofloxacin against P. aeruginosa; with mupirocin against MRSA; and with augmentin, doxycycline, azithromycin and clindamycin against S. aureus. Moreover, additive effects against planktonic forms were observed for combinations of Ag NPs with tobramycin, ciprofloxacin, imipenem, ceftazidime and aztreonam against P. aeruginosa; with gentamicin or linezolid against MRSA; and with doxycycline or clindamycin against S. aureus. In conclusion, Ag NP-antibiotic combinations can result in enhanced antimicrobial action against P. aeruginosa, MRSA and S. aureus clinical isolates in planktonic and biofilm forms and can be used in the context of CRS with confirmed infection.
Collapse
Affiliation(s)
- Sholeh Feizi
- Department of Surgery-Otolaryngology Head and Neck Surgery, Basil Hetzel Institute for Translational Health Research, Central Adelaide Local Health Network, Woodville South, SA, Australia; The University of Adelaide, Adelaide, SA, Australia
| | - Clare M Cooksley
- Department of Surgery-Otolaryngology Head and Neck Surgery, Basil Hetzel Institute for Translational Health Research, Central Adelaide Local Health Network, Woodville South, SA, Australia; The University of Adelaide, Adelaide, SA, Australia
| | - Roshan Nepal
- Department of Surgery-Otolaryngology Head and Neck Surgery, Basil Hetzel Institute for Translational Health Research, Central Adelaide Local Health Network, Woodville South, SA, Australia; The University of Adelaide, Adelaide, SA, Australia
| | - Alkis James Psaltis
- Department of Surgery-Otolaryngology Head and Neck Surgery, Basil Hetzel Institute for Translational Health Research, Central Adelaide Local Health Network, Woodville South, SA, Australia; The University of Adelaide, Adelaide, SA, Australia
| | - Peter-John Wormald
- Department of Surgery-Otolaryngology Head and Neck Surgery, Basil Hetzel Institute for Translational Health Research, Central Adelaide Local Health Network, Woodville South, SA, Australia; The University of Adelaide, Adelaide, SA, Australia
| | - Sarah Vreugde
- Department of Surgery-Otolaryngology Head and Neck Surgery, Basil Hetzel Institute for Translational Health Research, Central Adelaide Local Health Network, Woodville South, SA, Australia; The University of Adelaide, Adelaide, SA, Australia.
| |
Collapse
|
47
|
Lin L, Chi J, Yan Y, Luo R, Feng X, Zheng Y, Xian D, Li X, Quan G, Liu D, Wu C, Lu C, Pan X. Membrane-disruptive peptides/peptidomimetics-based therapeutics: Promising systems to combat bacteria and cancer in the drug-resistant era. Acta Pharm Sin B 2021; 11:2609-2644. [PMID: 34589385 PMCID: PMC8463292 DOI: 10.1016/j.apsb.2021.07.014] [Citation(s) in RCA: 85] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Revised: 07/08/2021] [Accepted: 07/12/2021] [Indexed: 02/05/2023] Open
Abstract
Membrane-disruptive peptides/peptidomimetics (MDPs) are antimicrobials or anticarcinogens that present a general killing mechanism through the physical disruption of cell membranes, in contrast to conventional chemotherapeutic drugs, which act on precise targets such as DNA or specific enzymes. Owing to their rapid action, broad-spectrum activity, and mechanisms of action that potentially hinder the development of resistance, MDPs have been increasingly considered as future therapeutics in the drug-resistant era. Recently, growing experimental evidence has demonstrated that MDPs can also be utilized as adjuvants to enhance the therapeutic effects of other agents. In this review, we evaluate the literature around the broad-spectrum antimicrobial properties and anticancer activity of MDPs, and summarize the current development and mechanisms of MDPs alone or in combination with other agents. Notably, this review highlights recent advances in the design of various MDP-based drug delivery systems that can improve the therapeutic effect of MDPs, minimize side effects, and promote the co-delivery of multiple chemotherapeutics, for more efficient antimicrobial and anticancer therapy.
Collapse
Affiliation(s)
- Liming Lin
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
- College of Pharmacy, Jinan University, Guangzhou 511443, China
| | - Jiaying Chi
- College of Pharmacy, Jinan University, Guangzhou 511443, China
| | - Yilang Yan
- College of Pharmacy, Jinan University, Guangzhou 511443, China
| | - Rui Luo
- College of Pharmacy, Jinan University, Guangzhou 511443, China
| | - Xiaoqian Feng
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
- College of Pharmacy, Jinan University, Guangzhou 511443, China
| | - Yuwei Zheng
- College of Pharmacy, Jinan University, Guangzhou 511443, China
| | - Dongyi Xian
- College of Pharmacy, Jinan University, Guangzhou 511443, China
| | - Xin Li
- The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou 510630, China
| | - Guilan Quan
- College of Pharmacy, Jinan University, Guangzhou 511443, China
| | - Daojun Liu
- Shantou University Medical College, Shantou 515041, China
| | - Chuanbin Wu
- College of Pharmacy, Jinan University, Guangzhou 511443, China
| | - Chao Lu
- College of Pharmacy, Jinan University, Guangzhou 511443, China
| | - Xin Pan
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| |
Collapse
|
48
|
Mazurek P, Yuusuf NA, Silau H, Mordhorst H, Pamp SJ, Brook MA, Skov AL. Simultaneous delivery of several antimicrobial drugs from multi‐compartment glycerol‐silicone membranes. J Appl Polym Sci 2021. [DOI: 10.1002/app.50780] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
- Piotr Mazurek
- Danish Polymer Centre, Department of Chemical Engineering Technical University of Denmark Kongens Lyngby Denmark
| | - Nuura A. Yuusuf
- Research Group for Genomic Epidemiology National Food Institute, Technical University of Denmark Kongens Lyngby Denmark
| | - Harald Silau
- Danish Polymer Centre, Department of Chemical Engineering Technical University of Denmark Kongens Lyngby Denmark
| | - Hanne Mordhorst
- Research Group for Genomic Epidemiology National Food Institute, Technical University of Denmark Kongens Lyngby Denmark
| | - Sünje J. Pamp
- Research Group for Genomic Epidemiology National Food Institute, Technical University of Denmark Kongens Lyngby Denmark
| | - Michael A. Brook
- Department of Chemistry and Chemical Biology McMaster University Hamilton Ontario Canada
| | - Anne L. Skov
- Danish Polymer Centre, Department of Chemical Engineering Technical University of Denmark Kongens Lyngby Denmark
- Glysious, R&D Kongens Lyngby Denmark
| |
Collapse
|
49
|
Mechanistic insights into synergy between nalidixic acid and tetracycline against clinical isolates of Acinetobacter baumannii and Escherichia coli. Commun Biol 2021; 4:542. [PMID: 33972678 PMCID: PMC8110569 DOI: 10.1038/s42003-021-02074-5] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Accepted: 04/01/2021] [Indexed: 02/03/2023] Open
Abstract
The increasing prevalence of antimicrobial resistance has become a global health problem. Acinetobacter baumannii is an important nosocomial pathogen due to its capacity to persist in the hospital environment. It has a high mortality rate and few treatment options. Antibiotic combinations can help to fight multi-drug resistant (MDR) bacterial infections, but they are rarely used in the clinics and mostly unexplored. The interaction between bacteriostatic and bactericidal antibiotics are mostly reported as antagonism based on the results obtained in the susceptible model laboratory strain Escherichia coli. However, in the present study, we report a synergistic interaction between nalidixic acid and tetracycline against clinical multi-drug resistant A. baumannii and E. coli. Here we provide mechanistic insight into this dichotomy. The synergistic combination was studied by checkerboard assay and time-kill curve analysis. We also elucidate the mechanism behind this synergy using several techniques such as fluorescence spectroscopy, flow cytometry, fluorescence microscopy, morphometric analysis, and real-time polymerase chain reaction. Nalidixic acid and tetracycline combination displayed synergy against most of the MDR clinical isolates of A. baumannii and E. coli but not against susceptible isolates. Finally, we demonstrate that this combination is also effective in vivo in an A. baumannii/Caenorhabditis elegans infection model (p < 0.001).
Collapse
|
50
|
Deusenbery C, Wang Y, Shukla A. Recent Innovations in Bacterial Infection Detection and Treatment. ACS Infect Dis 2021; 7:695-720. [PMID: 33733747 DOI: 10.1021/acsinfecdis.0c00890] [Citation(s) in RCA: 112] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Bacterial infections are a major threat to human health, exacerbated by increasing antibiotic resistance. These infections can result in tremendous morbidity and mortality, emphasizing the need to identify and treat pathogenic bacteria quickly and effectively. Recent developments in detection methods have focused on electrochemical, optical, and mass-based biosensors. Advances in these systems include implementing multifunctional materials, microfluidic sampling, and portable data-processing to improve sensitivity, specificity, and ease of operation. Concurrently, advances in antibacterial treatment have largely focused on targeted and responsive delivery for both antibiotics and antibiotic alternatives. Antibiotic alternatives described here include repurposed drugs, antimicrobial peptides and polymers, nucleic acids, small molecules, living systems, and bacteriophages. Finally, closed-loop therapies are combining advances in the fields of both detection and treatment. This review provides a comprehensive summary of the current trends in detection and treatment systems for bacterial infections.
Collapse
Affiliation(s)
- Carly Deusenbery
- School of Engineering, Center for Biomedical Engineering, Institute for Molecular and Nanoscale Innovation, Brown University, Providence, Rhode Island 02912, United States
| | - Yingying Wang
- Department of Chemistry, Brown University, Providence, Rhode Island 02912, United States
| | - Anita Shukla
- School of Engineering, Center for Biomedical Engineering, Institute for Molecular and Nanoscale Innovation, Brown University, Providence, Rhode Island 02912, United States
| |
Collapse
|