1
|
Lubrano P, Smollich F, Schramm T, Lorenz E, Alvarado A, Eigenmann SC, Stadelmann A, Thavapalan S, Waffenschmidt N, Glatter T, Hoffmann N, Müller J, Peter S, Drescher K, Link H. Metabolic mutations reduce antibiotic susceptibility of E. coli by pathway-specific bottlenecks. Mol Syst Biol 2025; 21:274-293. [PMID: 39748127 PMCID: PMC11876631 DOI: 10.1038/s44320-024-00084-z] [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: 07/02/2024] [Revised: 12/09/2024] [Accepted: 12/12/2024] [Indexed: 01/04/2025] Open
Abstract
Metabolic variation across pathogenic bacterial strains can impact their susceptibility to antibiotics and promote the evolution of antimicrobial resistance (AMR). However, little is known about how metabolic mutations influence metabolism and which pathways contribute to antibiotic susceptibility. Here, we measured the antibiotic susceptibility of 15,120 Escherichia coli mutants, each with a single amino acid change in one of 346 essential proteins. Across all mutants, we observed modest increases of the minimal inhibitory concentration (twofold to tenfold) without any cases of major resistance. Most mutants that showed reduced susceptibility to either of the two tested antibiotics carried mutations in metabolic genes. The effect of metabolic mutations on antibiotic susceptibility was antibiotic- and pathway-specific: mutations that reduced susceptibility against the β-lactam antibiotic carbenicillin converged on purine nucleotide biosynthesis, those against the aminoglycoside gentamicin converged on the respiratory chain. In addition, metabolic mutations conferred tolerance to carbenicillin by reducing growth rates. These results, along with evidence that metabolic bottlenecks are common among clinical E. coli isolates, highlight the contribution of metabolic mutations for AMR.
Collapse
Affiliation(s)
- Paul Lubrano
- Interfaculty Institute of Microbiology and Infection Medicine, University of Tübingen, Auf der Morgenstelle 24, 72076, Tübingen, Germany
- Cluster of Excellence "Controlling Microbes to Fight Infections", University of Tübingen, 72076, Tübingen, Germany
- M3 Research Center, Otfried-Müller-Straße 37, University of Tübingen, 72076, Tübingen, Germany
| | - Fabian Smollich
- Interfaculty Institute of Microbiology and Infection Medicine, University of Tübingen, Auf der Morgenstelle 24, 72076, Tübingen, Germany
- Cluster of Excellence "Controlling Microbes to Fight Infections", University of Tübingen, 72076, Tübingen, Germany
- M3 Research Center, Otfried-Müller-Straße 37, University of Tübingen, 72076, Tübingen, Germany
| | - Thorben Schramm
- Interfaculty Institute of Microbiology and Infection Medicine, University of Tübingen, Auf der Morgenstelle 24, 72076, Tübingen, Germany
- Institute of Molecular Systems Biology, ETH Zurich, Otto-Stern-Weg 3, 8093, Zürich, Switzerland
| | - Elisabeth Lorenz
- Interfaculty Institute of Microbiology and Infection Medicine, University of Tübingen, Auf der Morgenstelle 24, 72076, Tübingen, Germany
| | - Alejandra Alvarado
- Interfaculty Institute of Microbiology and Infection Medicine, University of Tübingen, Auf der Morgenstelle 24, 72076, Tübingen, Germany
- Cluster of Excellence "Controlling Microbes to Fight Infections", University of Tübingen, 72076, Tübingen, Germany
| | | | - Amelie Stadelmann
- Interfaculty Institute of Microbiology and Infection Medicine, University of Tübingen, Auf der Morgenstelle 24, 72076, Tübingen, Germany
- Cluster of Excellence "Controlling Microbes to Fight Infections", University of Tübingen, 72076, Tübingen, Germany
- M3 Research Center, Otfried-Müller-Straße 37, University of Tübingen, 72076, Tübingen, Germany
| | - Sevvalli Thavapalan
- Interfaculty Institute of Microbiology and Infection Medicine, University of Tübingen, Auf der Morgenstelle 24, 72076, Tübingen, Germany
- Cluster of Excellence "Controlling Microbes to Fight Infections", University of Tübingen, 72076, Tübingen, Germany
- M3 Research Center, Otfried-Müller-Straße 37, University of Tübingen, 72076, Tübingen, Germany
| | - Nils Waffenschmidt
- Interfaculty Institute of Microbiology and Infection Medicine, University of Tübingen, Auf der Morgenstelle 24, 72076, Tübingen, Germany
| | - Timo Glatter
- Max Planck Institute for Terrestrial Microbiology, Karl-von-Frisch-Straße 10, 35043, Marburg, Germany
| | - Nadine Hoffmann
- Cluster of Excellence "Controlling Microbes to Fight Infections", University of Tübingen, 72076, Tübingen, Germany
- Institute of Medical Microbiology and Hygiene, University of Tübingen, Elfriede-Aulhorn-Str. 6, 72076, Tübingen, Germany
| | - Jennifer Müller
- Institute of Medical Microbiology and Hygiene, University of Tübingen, Elfriede-Aulhorn-Str. 6, 72076, Tübingen, Germany
- NGS Competence Center Tübingen (NCCT), 72076, Tübingen, Germany
| | - Silke Peter
- Cluster of Excellence "Controlling Microbes to Fight Infections", University of Tübingen, 72076, Tübingen, Germany
- Institute of Medical Microbiology and Hygiene, University of Tübingen, Elfriede-Aulhorn-Str. 6, 72076, Tübingen, Germany
- NGS Competence Center Tübingen (NCCT), 72076, Tübingen, Germany
| | - Knut Drescher
- Biozentrum, University of Basel, Spitalstrasse 41, 4056, Basel, Switzerland
| | - Hannes Link
- Interfaculty Institute of Microbiology and Infection Medicine, University of Tübingen, Auf der Morgenstelle 24, 72076, Tübingen, Germany.
- Cluster of Excellence "Controlling Microbes to Fight Infections", University of Tübingen, 72076, Tübingen, Germany.
- M3 Research Center, Otfried-Müller-Straße 37, University of Tübingen, 72076, Tübingen, Germany.
| |
Collapse
|
2
|
Campey A, Łapińska U, Chait R, Tsaneva-Atanasova K, Pagliara S. Antibiotic resistant bacteria survive treatment by doubling while shrinking. mBio 2024; 15:e0237524. [PMID: 39565111 PMCID: PMC11633386 DOI: 10.1128/mbio.02375-24] [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: 08/02/2024] [Accepted: 10/22/2024] [Indexed: 11/21/2024] Open
Abstract
Many antibiotics that are used in healthcare, farming, and aquaculture end up in environments with different spatial structures that might promote heterogeneity in the emergence of antibiotic resistance. However, the experimental evolution of microbes at sub-inhibitory concentrations of antibiotics has been mainly carried out at the population level which does not allow capturing single-cell responses to antibiotics. Here, we investigate and compare the emergence of resistance to ciprofloxacin in Escherichia coli in well-mixed and structured environments using experimental evolution, genomics, and microfluidics-based time-lapse microscopy. We discover that resistance to ciprofloxacin and cross-resistance to other antibiotics is stronger in the well-mixed environment due to the emergence of target mutations, whereas efflux regulator mutations emerge in the structured environment. The latter mutants also harbor sub-populations of persisters that survive high concentrations of ciprofloxacin that inhibit bacterial growth at the population level. In contrast, genetically resistant bacteria that display target mutations also survive high concentrations of ciprofloxacin that inhibit their growth via population-level antibiotic tolerance. These resistant and tolerant bacteria keep doubling while shrinking in size in the presence of ciprofloxacin and regain their original size after antibiotic removal, which constitutes a newly discovered phenotypic response. This new knowledge sheds light on the diversity of strategies employed by bacteria to survive antibiotics and poses a stepping stone for understanding the link between mutations at the population level and phenotypic single-cell responses. IMPORTANCE The evolution of antimicrobial resistance poses a pressing challenge to global health with an estimated 5 million deaths associated with antimicrobial resistance every year globally. Here, we investigate the diversity of strategies employed by bacteria to survive antibiotics. We discovered that bacteria evolve genetic resistance to antibiotics while simultaneously displaying tolerance to very high doses of antibiotics by doubling while shrinking in size.
Collapse
Affiliation(s)
- Adrian Campey
- Living Systems Institute and Biosciences, University of Exeter, Exeter, Devon, United Kingdom
| | - Urszula Łapińska
- Living Systems Institute and Biosciences, University of Exeter, Exeter, Devon, United Kingdom
| | - Remy Chait
- Living Systems Institute and Biosciences, University of Exeter, Exeter, Devon, United Kingdom
| | - Krasimira Tsaneva-Atanasova
- Living Systems Institute and Biosciences, University of Exeter, Exeter, Devon, United Kingdom
- EPSRC Hub for Quantitative Modelling in Healthcare, University of Exeter, Exeter, Devon, United Kingdom
| | - Stefano Pagliara
- Living Systems Institute and Biosciences, University of Exeter, Exeter, Devon, United Kingdom
| |
Collapse
|
3
|
Ma Z, Hou B, Liao A, Tan Y, Tan C, Jiang Y. Light-Activable Inhibitor Overcomes Antimicrobial Resistance and Regulates Antibacterial Activity. J Med Chem 2024; 67:20455-20466. [PMID: 39540544 DOI: 10.1021/acs.jmedchem.4c01923] [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: 11/16/2024]
Abstract
Overuse of antibiotics and the widespread environmental accumulation of antibiotics drive the evolution and spread of antimicrobial resistance, posing a significant global health threat by reducing the effectiveness of available treatments and increasing the risk of untreatable infections. We designed and synthesized PhoPS, a novel photocaged β-lactamase inhibitor, which incorporates the pharmacophore of sulbactam caged with a photoresponsive moiety of o-nitrobiphenyl derivative. Experimental results demonstrate its rapid photoactivation, good stability in solution, and light-activated β-lactamase inhibition in vitro. PhoPS displays synergy with a cephalosporin antibiotic cefoperazone against both susceptible and resistant strains of Escherichia coli and biofilm formation. Additionally, PhoPS treatment demonstrates the potential to suppress the development of resistance in E. coli. These findings suggest that PhoPS offers a promising approach for restoring the efficacy of existing antibiotics and mitigating the emergence of AMR.
Collapse
Affiliation(s)
- Zhuang Ma
- The State Key Laboratory of Chemical Oncogenomics, Institute of Biopharmaceutical and Health Engineering, Shenzhen International Graduate School, Tsinghua University, Shenzhen 518055, P. R. China
| | - Boxuan Hou
- The State Key Laboratory of Chemical Oncogenomics, Institute of Biopharmaceutical and Health Engineering, Shenzhen International Graduate School, Tsinghua University, Shenzhen 518055, P. R. China
| | - Anhui Liao
- The State Key Laboratory of Chemical Oncogenomics, Institute of Biopharmaceutical and Health Engineering, Shenzhen International Graduate School, Tsinghua University, Shenzhen 518055, P. R. China
| | - Ying Tan
- The State Key Laboratory of Chemical Oncogenomics, Institute of Biopharmaceutical and Health Engineering, Shenzhen International Graduate School, Tsinghua University, Shenzhen 518055, P. R. China
- Open FIESTA, Shenzhen International Graduate School, Tsinghua University, Shenzhen 518055, P. R. China
| | - Chunyan Tan
- The State Key Laboratory of Chemical Oncogenomics, Institute of Biopharmaceutical and Health Engineering, Shenzhen International Graduate School, Tsinghua University, Shenzhen 518055, P. R. China
- Open FIESTA, Shenzhen International Graduate School, Tsinghua University, Shenzhen 518055, P. R. China
| | - Yuyang Jiang
- School of Pharmaceutical Sciences, Tsinghua University, Beijing 100084, P. R. China
| |
Collapse
|
4
|
Berinson B, Davies E, Torpner J, Flinkfeldt L, Fernberg J, Åman A, Bergqvist J, Öhrn H, Ångström J, Johansson C, Jäder K, Andersson H, Ghaderi E, Rolf M, Sundqvist M, Rohde H, Fernandez-Zafra T, Malmberg C. A multicenter evaluation of a novel microfluidic rapid AST assay for Gram-negative bloodstream infections. J Clin Microbiol 2024; 62:e0045824. [PMID: 39324811 PMCID: PMC11481479 DOI: 10.1128/jcm.00458-24] [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: 03/25/2024] [Accepted: 09/07/2024] [Indexed: 09/27/2024] Open
Abstract
Common phenotypic methods for antimicrobial susceptibility testing (AST) of bacteria are slow, labor intensive, and display considerable technical variability. The QuickMIC system provides rapid AST using a microfluidic linear gradient. Here, we evaluate the performance of QuickMIC at four different laboratories with regard to speed, precision, accuracy, and reproducibility in comparison to broth microdilution (BMD). Spiked (n = 411) and clinical blood cultures (n = 148) were tested with the QuickMIC Gram-negative panel and compared with BMD for the 12 on-panel antibiotics, and 10 defined strains were run at each site to measure reproducibility. Logistic and linear regression analysis was applied to explore factors affecting assay performance. The overall essential agreement and categorical agreement between QuickMIC and BMD were 95.6% and 96.0%, respectively. Very major error, major error, and minor error rates were 1.0%, 0.6%, and 2.4%, respectively. Inter-laboratory reproducibility between the sites was high at 98.9% using the acceptable standard of ±1 twofold dilution. The mean in-instrument analysis time overall was 3 h 13 min (SD: 29 min). Regression analysis indicated that QuickMIC is robust with regard to initial inoculum and delay time after blood culture positivity. We conclude that QuickMIC can be used to rapidly measure MIC directly from blood cultures in clinical settings with high reproducibility, precision, and accuracy. The microfluidics-generated linear gradient ensures high reproducibility between laboratories, thus allowing a high level of trust in MIC values from single testing, at the cost of reduced measurement range compared to BMD. IMPORTANCE Increasing antimicrobial resistance underscores the need for new diagnostic solutions to guide therapy, but traditional antimicrobial susceptibility testing (AST) is often inadequate in time-critical diseases such as sepsis. This work presents a novel and rapid AST system with a rapid turnaround of results, which may help reduce the time to guided therapy, possibly allowing early de-escalation of broad-spectrum empirical therapy as well as rapid adjustments to treatments when coverage is lacking.
Collapse
Affiliation(s)
- Benjamin Berinson
- Institute of Medical Microbiology, Virology and Hygiene, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | | | | | | | | | | | | | | | | | | | - Klara Jäder
- Department of Clinical Microbiology, Uppsala University Hospital, Uppsala, Sweden
| | - Helena Andersson
- Department of Clinical Microbiology, Uppsala University Hospital, Uppsala, Sweden
| | - Ehsan Ghaderi
- Department of Clinical Microbiology, Uppsala University Hospital, Uppsala, Sweden
| | - Maria Rolf
- Department of Laboratory Medicine, Clinical Microbiology, Örebro University Hospital, Örebro, Sweden
| | - Martin Sundqvist
- Department of Laboratory Medicine, Clinical Microbiology, Faculty of Medicine and Health, Örebro University, Örebro, Sweden
| | - Holger Rohde
- Institute of Medical Microbiology, Virology and Hygiene, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | | | - Christer Malmberg
- Gradientech AB, Uppsala, Sweden
- Department of Medical Sciences, Uppsala University, Uppsala, Sweden
| |
Collapse
|
5
|
Oliveira M, Antunes W, Mota S, Madureira-Carvalho Á, Dinis-Oliveira RJ, Dias da Silva D. An Overview of the Recent Advances in Antimicrobial Resistance. Microorganisms 2024; 12:1920. [PMID: 39338594 PMCID: PMC11434382 DOI: 10.3390/microorganisms12091920] [Citation(s) in RCA: 20] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2024] [Revised: 09/15/2024] [Accepted: 09/18/2024] [Indexed: 09/30/2024] Open
Abstract
Antimicrobial resistance (AMR), frequently considered a major global public health threat, requires a comprehensive understanding of its emergence, mechanisms, advances, and implications. AMR's epidemiological landscape is characterized by its widespread prevalence and constantly evolving patterns, with multidrug-resistant organisms (MDROs) creating new challenges every day. The most common mechanisms underlying AMR (i.e., genetic mutations, horizontal gene transfer, and selective pressure) contribute to the emergence and dissemination of new resistant strains. Therefore, mitigation strategies (e.g., antibiotic stewardship programs-ASPs-and infection prevention and control strategies-IPCs) emphasize the importance of responsible antimicrobial use and surveillance. A One Health approach (i.e., the interconnectedness of human, animal, and environmental health) highlights the necessity for interdisciplinary collaboration and holistic strategies in combating AMR. Advancements in novel therapeutics (e.g., alternative antimicrobial agents and vaccines) offer promising avenues in addressing AMR challenges. Policy interventions at the international and national levels also promote ASPs aiming to regulate antimicrobial use. Despite all of the observed progress, AMR remains a pressing concern, demanding sustained efforts to address emerging threats and promote antimicrobial sustainability. Future research must prioritize innovative approaches and address the complex socioecological dynamics underlying AMR. This manuscript is a comprehensive resource for researchers, policymakers, and healthcare professionals seeking to navigate the complex AMR landscape and develop effective strategies for its mitigation.
Collapse
Affiliation(s)
- Manuela Oliveira
- Associate Laboratory i4HB—Institute for Health and Bioeconomy, University Institute of Health Sciences—CESPU, Avenida Central de Gandra 1317, 4585-116 Gandra, Portugal; (Á.M.-C.); (D.D.d.S.)
- UCIBIO—Research Unit on Applied Molecular Biosciences, Translational Toxicology Research Laboratory, University Institute of Health Sciences (1H-TOXRUN, IUCS-CESPU), Avenida Central de Gandra 1317, 4585-116 Gandra, Portugal
| | - Wilson Antunes
- Instituto Universitário Militar, CINAMIL, Unidade Militar Laboratorial de Defesa Biológica e Química, Avenida Doutor Alfredo Bensaúde, 4 piso, do LNM, 1849-012 Lisbon, Portugal
| | - Salete Mota
- ULSEDV—Unidade Local De Saúde De Entre Douro Vouga, Unidade de Santa Maria da Feira e Hospital S. Sebastião, Rua Dr. Cândido Pinho, 4520-211 Santa Maria da Feira, Portugal
| | - Áurea Madureira-Carvalho
- Associate Laboratory i4HB—Institute for Health and Bioeconomy, University Institute of Health Sciences—CESPU, Avenida Central de Gandra 1317, 4585-116 Gandra, Portugal; (Á.M.-C.); (D.D.d.S.)
- UCIBIO—Applied Molecular Biosciences Unit, Forensics and Biomedical Sciences Research Laboratory, University Institute of Health Sciences (1H-TOXRUN, IUCS-CESPU), Avenida Central de Gandra 1317, 4585-116 Gandra, Portugal
- Department of Public Health and Forensic Sciences and Medical Education, Faculty of Medicine, University of Porto, Alameda Prof. Hernâni Monteiro, 4200-319 Porto, Portugal
| | - Ricardo Jorge Dinis-Oliveira
- Associate Laboratory i4HB—Institute for Health and Bioeconomy, University Institute of Health Sciences—CESPU, Avenida Central de Gandra 1317, 4585-116 Gandra, Portugal; (Á.M.-C.); (D.D.d.S.)
- UCIBIO—Research Unit on Applied Molecular Biosciences, Translational Toxicology Research Laboratory, University Institute of Health Sciences (1H-TOXRUN, IUCS-CESPU), Avenida Central de Gandra 1317, 4585-116 Gandra, Portugal
- Department of Public Health and Forensic Sciences and Medical Education, Faculty of Medicine, University of Porto, Alameda Prof. Hernâni Monteiro, 4200-319 Porto, Portugal
- FOREN—Forensic Science Experts, Avenida Dr. Mário Moutinho 33-A, 1400-136 Lisbon, Portugal
| | - Diana Dias da Silva
- Associate Laboratory i4HB—Institute for Health and Bioeconomy, University Institute of Health Sciences—CESPU, Avenida Central de Gandra 1317, 4585-116 Gandra, Portugal; (Á.M.-C.); (D.D.d.S.)
- UCIBIO—Applied Molecular Biosciences Unit, Forensics and Biomedical Sciences Research Laboratory, University Institute of Health Sciences (1H-TOXRUN, IUCS-CESPU), Avenida Central de Gandra 1317, 4585-116 Gandra, Portugal
- REQUIMTE/LAQV, ESS, Polytechnic of Porto, Rua Dr. António Bernardino de Almeida, 4200-072 Porto, Portugal
- Associate Laboratory i4HB—Institute for Health and Bioeconomy, University of Porto, Rua de Jorge Viterbo Ferreira 228, 4050-313 Porto, Portugal
- UCIBIO—Applied Molecular Biosciences Unit, Laboratory of Toxicology, Faculty of Pharmacy, University of Porto, Rua de Jorge Viterbo Ferreira 228, 4050-313 Porto, Portugal
| |
Collapse
|
6
|
Fang Z, Tu S, Huang J. A bibliometric analysis of One Health approach in research on antimicrobial resistance. SCIENCE IN ONE HEALTH 2024; 3:100077. [PMID: 39350920 PMCID: PMC11440798 DOI: 10.1016/j.soh.2024.100077] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Accepted: 09/01/2024] [Indexed: 10/04/2024]
Abstract
Background Antimicrobial resistance (AMR) is a global public health threat that requires actions through One Health intervention. This study aims to trace the historical development of One Health research on AMR to provide evidence supporting future research and actions. Methods A bibliometric analysis is conducted with One Health articles in the field of antimicrobial resistance (AMR-OH articles) retrieved from the Web of Science Core Collection (WoSCC). AMR-OH articles refer to articles in the field of AMR that simultaneously involve elements from human health and at least one other domain, including animals, environment, or plants. Three research periods were identified based on the development of global actions in combating AMR. Descriptive analysis of publications, keyword cluster analysis, annual trending topic analysis, and co-authorship analysis were conducted using R software, V OSV iewer, and Pajek. Results The results indicated that the percentage of AMR-OH articles among all AMR articles increased from 5.21% in 1990 to 20.01% in 2023. Key topics in the current AMR-OH articles included the mechanism of AMR, AMR epidemiology, and public health control strategies. Epidemiological research initially focused on human and animal health and then shifted to environmental factors in the third period. Research at the molecular level focused on the mechanisms of AMR transmission in various domains, along with the dynamics and diversity of antibiotic resistance genes (ARGs). The co-authorship analysis suggested a significant increase in cooperation among low- and middle-income countries in the third period. Conclusion The scope of epidemiological research on AMR has expanded by including human, animal, and environmental areas. Moreover, genetic and molecular level research represents the forefront of this field, offering innovative tools to combat AMR in the future. This study suggests further research to translate existing findings into practical implementation of the One Health strategy, and to support globally consistent action in combating AMR.
Collapse
Affiliation(s)
- Zheyi Fang
- Shanghai Institute of Infectious Disease and Biosecurity, School of Public Health, Fudan University, Shanghai 200032, China
- Global Health Institutes, Fudan University, Shanghai 200032, China
| | - Shiyi Tu
- Shanghai Institute of Infectious Disease and Biosecurity, School of Public Health, Fudan University, Shanghai 200032, China
- Global Health Institutes, Fudan University, Shanghai 200032, China
| | - Jiayan Huang
- Shanghai Institute of Infectious Disease and Biosecurity, School of Public Health, Fudan University, Shanghai 200032, China
- Global Health Institutes, Fudan University, Shanghai 200032, China
| |
Collapse
|
7
|
Alotaibi G. Prevalence, pandemic, preventions and policies to overcome antimicrobial resistance. Saudi J Biol Sci 2024; 31:104032. [PMID: 38854892 PMCID: PMC11157277 DOI: 10.1016/j.sjbs.2024.104032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2024] [Revised: 05/22/2024] [Accepted: 05/25/2024] [Indexed: 06/11/2024] Open
Abstract
Antimicrobial resistance (AMR) is a growing concern in Asia, and it is essential to understand the prevalence, pandemic, prevention, and policies to overcome it. According to the World Health Organization (WHO), AMR is one of the main causes of death; in 2019, it was linked to 4.95 million fatalities and caused about 1.27 million deaths. A core package of actions has been provided by WHO to help countries prioritize their needs when creating, carrying out, and overseeing national action plans on antimicrobial resistance. Using a people-cantered approach to AMR, the interventions address the needs and obstacles that individuals and patients encounter when trying to obtain healthcare. The people-cantered core package of AMR treatments seeks to improve public and policymakers; awareness and comprehension of AMR by changing the narrative of AMR to emphasize the needs of people and systemic impairments. Additionally, it backs a more comprehensive and programmatic national response to AMR, which emphasizes the value of fair and inexpensive access to high-quality healthcare services for the avoidance, identification, and management of drug-resistant diseases. The report signals increasing resistance to antibiotics in bacterial infections in humans and the need for better data. In conclusion, the prevalence of AMR in Asia is a significant public health concern, and it is crucial to implement policies and interventions to overcome it.
Collapse
Affiliation(s)
- Ghallab Alotaibi
- Department of Pharmacology, College of Pharmacy, Shaqra University, Riyadh 11961, Saudi Arabia
| |
Collapse
|
8
|
Nantongo M, Nguyen DC, Bethel CR, Taracila MA, Li Q, Dousa KM, Shin E, Kurz SG, Nguyen L, Kreiswirth BN, Boom WH, Plummer MS, Bonomo RA. Durlobactam, a Diazabicyclooctane β-Lactamase Inhibitor, Inhibits BlaC and Peptidoglycan Transpeptidases of Mycobacterium tuberculosis. ACS Infect Dis 2024; 10:1767-1779. [PMID: 38619138 DOI: 10.1021/acsinfecdis.4c00119] [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: 04/16/2024]
Abstract
Peptidoglycan synthesis is an underutilized drug target in Mycobacterium tuberculosis (Mtb). Diazabicyclooctanes (DBOs) are a class of broad-spectrum β-lactamase inhibitors that also inhibit certain peptidoglycan transpeptidases that are important in mycobacterial cell wall synthesis. We evaluated the DBO durlobactam as an inhibitor of BlaC, the Mtb β-lactamase, and multiple Mtb peptidoglycan transpeptidases (PonA1, LdtMt1, LdtMt2, LdtMt3, and LdtMt5). Timed electrospray ionization mass spectrometry (ESI-MS) captured acyl-enzyme complexes with BlaC and all transpeptidases except LdtMt5. Inhibition kinetics demonstrated durlobactam was a potent and efficient DBO inhibitor of BlaC (KI app 9.2 ± 0.9 μM, k2/K 5600 ± 560 M-1 s-1) and similar to clavulanate (KI app 3.3 ± 0.6 μM, k2/K 8400 ± 840 M-1 s-1); however, durlobactam had a lower turnover number (tn = kcat/kinact) than clavulanate (1 and 8, respectively). KI app values with durlobactam and clavulanate were similar for peptidoglycan transpeptidases, but ESI-MS captured durlobactam complexes at more time points. Molecular docking and simulation demonstrated several productive interactions of durlobactam in the active sites of BlaC, PonA1, and LdtMt2. Antibiotic susceptibility testing was conducted on 11 Mtb isolates with amoxicillin, ceftriaxone, meropenem, imipenem, clavulanate, and durlobactam. Durlobactam had a minimum inhibitory concentration (MIC) range of 0.5-16 μg/mL, similar to the ranges for meropenem (1-32 μg/mL) and imipenem (0.5-64 μg/mL). In β-lactam + durlobactam combinations (1:1 mass/volume), MICs were lowered 4- to 64-fold for all isolates except one with meropenem-durlobactam. This work supports further exploration of novel β-lactamase inhibitors that target BlaC and Mtb peptidoglycan transpeptidases.
Collapse
Affiliation(s)
- Mary Nantongo
- Department of Molecular Biology and Microbiology, Case Western Reserve University (CWRU), Cleveland, Ohio 44106, United States
- Research Service, Louis Stokes Veterans Affairs Medical Center, Cleveland, Ohio 44106, United States
| | - David C Nguyen
- Division of Infectious Diseases, Department of Pediatrics and Division of Infectious Diseases, and Department of Internal Medicine, Rush University Medical Center, Chicago, Illinois 60612, United States
| | - Christopher R Bethel
- Research Service, Louis Stokes Veterans Affairs Medical Center, Cleveland, Ohio 44106, United States
| | - Magdalena A Taracila
- Research Service, Louis Stokes Veterans Affairs Medical Center, Cleveland, Ohio 44106, United States
- Department of Medicine, Case Western Reserve University (CWRU), Cleveland, Ohio 44106, United States
| | - Qing Li
- Department of Medicine, Case Western Reserve University (CWRU), Cleveland, Ohio 44106, United States
| | - Khalid M Dousa
- Research Service, Louis Stokes Veterans Affairs Medical Center, Cleveland, Ohio 44106, United States
- Department of Medicine, Case Western Reserve University (CWRU), Cleveland, Ohio 44106, United States
- Medical Service, Veterans Affairs Northeast Ohio Healthcare System (VANEOHS), Cleveland, Ohio 44106, United States
| | - Eunjeong Shin
- Research Service, Louis Stokes Veterans Affairs Medical Center, Cleveland, Ohio 44106, United States
- Department of Medicine, Case Western Reserve University (CWRU), Cleveland, Ohio 44106, United States
| | - Sebastian G Kurz
- Department of Internal Medicine VIII, Medical Oncology and Pneumology, University of Tübingen, 72076 Tübingen, Germany
| | - Liem Nguyen
- Department of Molecular Biology and Microbiology, Case Western Reserve University (CWRU), Cleveland, Ohio 44106, United States
| | - Barry N Kreiswirth
- Center for Discovery and Innovation, Hackensack, New Jersey 07110, United States
| | - W Henry Boom
- Department of Molecular Biology and Microbiology, Case Western Reserve University (CWRU), Cleveland, Ohio 44106, United States
- Department of Medicine, Case Western Reserve University (CWRU), Cleveland, Ohio 44106, United States
| | - Mark S Plummer
- Biopharmaworks, Groton, Connecticut 06340, United States
| | - Robert A Bonomo
- Department of Molecular Biology and Microbiology, Case Western Reserve University (CWRU), Cleveland, Ohio 44106, United States
- Department of Medicine, Case Western Reserve University (CWRU), Cleveland, Ohio 44106, United States
- Medical Service, Veterans Affairs Northeast Ohio Healthcare System (VANEOHS), Cleveland, Ohio 44106, United States
- CWRU-Cleveland VAMC Center for Antibiotic Resistance and Epidemiology (Case VA CARES), Cleveland, Ohio 44106, United States
- Departments of Biochemistry, Pharmacology, and Proteomics and Bioinformatics, CWRU, Cleveland, Ohio 44106, United States
- Cleveland Geriatrics Research Education and Clinical Center (GRECC), VANEOHS, Cleveland, Ohio 44106, United States
| |
Collapse
|
9
|
Ahmad N, Bukhari SNA, Hussain MA, Ejaz H, Munir MU, Amjad MW. Nanoparticles incorporated hydrogels for delivery of antimicrobial agents: developments and trends. RSC Adv 2024; 14:13535-13564. [PMID: 38665493 PMCID: PMC11043667 DOI: 10.1039/d4ra00631c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Accepted: 04/01/2024] [Indexed: 04/28/2024] Open
Abstract
The prevention and treatment of microbial infections is an imminent global public health concern due to the poor antimicrobial performance of the existing antimicrobial regime and rapidly emerging antibiotic resistance in pathogenic microbes. In order to overcome these problems and effectively control bacterial infections, various new treatment modalities have been identified. To attempt this, various micro- and macro-molecular antimicrobial agents that function by microbial membrane disruption have been developed with improved antimicrobial activity and lesser resistance. Antimicrobial nanoparticle-hydrogels systems comprising antimicrobial agents (antibiotics, biological extracts, and antimicrobial peptides) loaded nanoparticles or antimicrobial nanoparticles (metal or metal oxide) constitute an important class of biomaterials for the prevention and treatment of infections. Hydrogels that incorporate nanoparticles can offer an effective strategy for delivering antimicrobial agents (or nanoparticles) in a controlled, sustained, and targeted manner. In this review, we have described an overview of recent advancements in nanoparticle-hydrogel hybrid systems for antimicrobial agent delivery. Firstly, we have provided an overview of the nanoparticle hydrogel system and discussed various advantages of these systems in biomedical and pharmaceutical applications. Thereafter, different hybrid hydrogel systems encapsulating antibacterial metal/metal oxide nanoparticles, polymeric nanoparticles, antibiotics, biological extracts, and antimicrobial peptides for controlling infections have been reviewed in detail. Finally, the challenges and future prospects of nanoparticle-hydrogel systems have been discussed.
Collapse
Affiliation(s)
- Naveed Ahmad
- Department of Pharmaceutics, College of Pharmacy, Jouf University Sakaka 72388 Aljouf Saudi Arabia
| | - Syed Nasir Abbas Bukhari
- Department of Pharmaceutical Chemistry, College of Pharmacy, Jouf University Sakaka 72388 Aljouf Saudi Arabia
| | - Muhammad Ajaz Hussain
- Centre for Organic Chemistry, School of Chemistry, University of the Punjab Lahore 54590 Pakistan
| | - Hasan Ejaz
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Jouf University Sakaka 72388 Aljouf Saudi Arabia
| | - Muhammad Usman Munir
- Australian Institute for Bioengineering & Nanotechnology, The University of Queensland Brisbane Queens-land 4072 Australia
| | - Muhammad Wahab Amjad
- 6 Center for Ultrasound Molecular Imaging and Therapeutics, School of Medicine, University of Pittsburgh 15213 Pittsburgh Pennsylvania USA
| |
Collapse
|
10
|
Brunskill I, Somanader DS, Perrin M, Barkema HW, Hillier S, Hindmarch S, Topp E, Weese JS, Wright GD, Morris AM. Seeing the whole elephant: designing 'one health' governance to fight antimicrobial resistance. Clin Microbiol Infect 2024; 30:419-422. [PMID: 37802304 DOI: 10.1016/j.cmi.2023.09.021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Revised: 09/13/2023] [Accepted: 09/27/2023] [Indexed: 10/08/2023]
Affiliation(s)
| | - Deborah S Somanader
- Antimicrobial Stewardship Program, Sinai Health-University Health Network, Toronto, Ontario, Canada
| | | | - Herman W Barkema
- One Health at UCalgary, University of Calgary, Calgary, Alberta, Canada
| | - Sean Hillier
- Faculty of Health, York University, Toronto, Ontario, Canada
| | - Suzanne Hindmarch
- Department of Political Science, University of New Brunswick, Fredericton, New Brunswick, Canada
| | - Ed Topp
- National Research Institute for Agriculture, Food and the Environment, University of Burgundy, Dijon, France
| | | | - Gerard D Wright
- Michael G. DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, Ontario, Canada
| | - Andrew M Morris
- Antimicrobial Stewardship Program, Sinai Health-University Health Network, Toronto, Ontario, Canada.
| |
Collapse
|
11
|
Bhardwaj M, Kour D, Rai G, Bhattacharya S, Manhas D, Vij B, Kumar A, Mukherjee D, Ahmed Z, Gandhi SG, Nandi U. EIDD-1931 Treatment Tweaks CYP3A4 and CYP2C8 in Arthritic Rats to Expedite Drug Interaction: Implication in Oral Therapy of Molnupiravir. ACS OMEGA 2024; 9:13982-13993. [PMID: 38559969 PMCID: PMC10976394 DOI: 10.1021/acsomega.3c09287] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Revised: 01/26/2024] [Accepted: 02/08/2024] [Indexed: 04/04/2024]
Abstract
EIDD-1931 is the active form of molnupiravir, an orally effective drug approved by the United States Food and Drug Administration (USFDA) against COVID-19. Pharmacokinetic alteration can cause untoward drug interaction (drug-drug/disease-drug), but hardly any information is known about this recently approved drug. Therefore, we first investigated the impact of the arthritis state on the oral pharmacokinetics of EIDD-1931 using a widely accepted complete Freund's adjuvant (CFA)-induced rat model of rheumatoid arthritis (RA) after ascertaining the disease occurrence by paw swelling measurement and X-ray examination. Comparative oral pharmacokinetic assessment of EIDD-1931 (normal state vs arthritis state) showed that overall plasma exposure was augmented (1.7-fold) with reduced clearance (0.54-fold), suggesting its likelihood of dose adjustment in arthritis conditions. In order to elucidate the effect of EIDD-1931 treatment at a therapeutic regime (normal state vs arthritis state) on USFDA-recommended panel of cytochrome P450 (CYP) enzymes (CYP1A2, CYP2B6, CYP2C8, CYP2C9, CYP2C19, CYP2D6, and CYP3A4) for drug interaction using the same disease model, we monitored protein and mRNA expressions (rat homologs) in liver tissue by western blotting (WB) and real time-polymerase chain reaction (RT-PCR), respectively. Results reveal that EIDD-1931 treatment could strongly influence CYP3A4 and CYP2C8 among experimental proteins/mRNAs. Although CYP2C8 regulation upon EIDD-1931 treatment resembles similar behavior under the arthritis state, results dictate a potentially reverse phenomenon for CYP3A4. Moreover, the lack of any CYP inhibitory effect by EIDD-1931 in human/rat liver microsomes (HLM/RLM) helps to ascertain EIDD-1931 treatment-mediated disease-drug interaction and the possibility of drug-drug interaction with disease-modifying antirheumatic drugs (DMARDs) upon coadministration. As elevated proinflammatory cytokine levels are prevalent in RA and nuclear factor-kappa B (NF-kB) and nuclear receptors control CYP expressions, further studies should focus on understanding the regulation of affected CYPs to subside unexpected drug interaction.
Collapse
Affiliation(s)
- Mahir Bhardwaj
- Pharmacology
Division, CSIR-Indian Institute of Integrative
Medicine, Jammu 180001, India
- Academy
of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Dilpreet Kour
- Pharmacology
Division, CSIR-Indian Institute of Integrative
Medicine, Jammu 180001, India
- Academy
of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Garima Rai
- Infectious
Diseases Division, CSIR-Indian Institute
of Integrative Medicine, Jammu 180001, India
| | - Srija Bhattacharya
- Natural
Product and Medicinal Chemistry Division, CSIR-Indian Institute of Integrative Medicine, Jammu 180001, India
| | - Diksha Manhas
- Pharmacology
Division, CSIR-Indian Institute of Integrative
Medicine, Jammu 180001, India
- Academy
of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Bhavna Vij
- Pharmacology
Division, CSIR-Indian Institute of Integrative
Medicine, Jammu 180001, India
| | - Ajay Kumar
- Pharmacology
Division, CSIR-Indian Institute of Integrative
Medicine, Jammu 180001, India
- Academy
of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Debaraj Mukherjee
- Academy
of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
- Natural
Product and Medicinal Chemistry Division, CSIR-Indian Institute of Integrative Medicine, Jammu 180001, India
- Department
of Chemical Sciences, Bose institute, Kolkata 700091, India
| | - Zabeer Ahmed
- Pharmacology
Division, CSIR-Indian Institute of Integrative
Medicine, Jammu 180001, India
- Academy
of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Sumit G. Gandhi
- Academy
of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
- Infectious
Diseases Division, CSIR-Indian Institute
of Integrative Medicine, Jammu 180001, India
| | - Utpal Nandi
- Pharmacology
Division, CSIR-Indian Institute of Integrative
Medicine, Jammu 180001, India
- Academy
of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| |
Collapse
|
12
|
Cotter E, Pultar F, Riniker S, Altmann KH. Experimental and Theoretical Studies on the Reactions of Aliphatic Imines with Isocyanates. Chemistry 2024; 30:e202304272. [PMID: 38226702 DOI: 10.1002/chem.202304272] [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: 12/21/2023] [Revised: 01/14/2024] [Accepted: 01/15/2024] [Indexed: 01/17/2024]
Abstract
In the context of a project aiming at the replacement of the 3-substituted β-lactam ring in classical β-lactam antibiotics by an N(3)-acyl-1,3-diazetidinone moiety, we have investigated the reaction of isocyanates with imines derived from allyl glycinate and differently substituted propionaldehydes. Imines of aromatic aldehydes with anilines have been reported to react with acyl isocyanates to give 1,3-diazetidinones or 2,3-dihydro-4H-1,3,5-oxadiazin-4-ones, via [2+2] or [4+2] cycloaddition, respectively. However, neither of these products was formed with imines derived from allyl glycinate and 2-(mono)methyl propionaldehydes. α,α-Dimethylation of the imine enabled the [4+2] cycloaddition pathway, but the desired 1,3-diazetidinone products were not observed. Surprisingly, the imines obtained from thioesters of 2,2-dimethyl 3-oxo propionic acid reacted with aryl isocyanates or with benzyl isocyanate to give 5,5-dimethyl-2,4-dioxo-6-(aryl-/alkylthio)tetrahydropyrimidines, via thiol displacement and re-addition to a putative six-membered iminium intermediate. These experimental results obtained for the reactions could be rationalized by DFT calculations. In addition, we have shown that N(3)-acyl-1,3-diazetidinone and 2,3-dihydro-4H-1,3,5-oxadiazin-4-one products can be distinguished based on experimental IR data in combination with theoretical reference spectra employing the IR spectra alignment (IRSA) algorithm. This discrimination was not possible by means of 1 H, 13 C, or 15 N NMR spectroscopy.
Collapse
Affiliation(s)
- Etienne Cotter
- Department of Chemistry and Applied Biosciences, Institute of Pharmaceutical Sciences, ETH Zürich, Vladimir-Prelog-Weg 4, 8093, Zürich, Switzerland
| | - Felix Pultar
- ETH Zürich, Department of Chemistry and Applied Biosciences, Institute of Molecular Physical Science, 8093, Zürich, Switzerland
| | - Sereina Riniker
- ETH Zürich, Department of Chemistry and Applied Biosciences, Institute of Molecular Physical Science, 8093, Zürich, Switzerland
| | - Karl-Heinz Altmann
- Department of Chemistry and Applied Biosciences, Institute of Pharmaceutical Sciences, ETH Zürich, Vladimir-Prelog-Weg 4, 8093, Zürich, Switzerland
| |
Collapse
|
13
|
Chang SC, Kao CY, Lin LC, Hidrosollo JH, Lu JJ. Lugdunin production and activity in Staphylococcus lugdunensis isolates are associated with its genotypes. Microbiol Spectr 2023; 11:e0129823. [PMID: 37732790 PMCID: PMC10580833 DOI: 10.1128/spectrum.01298-23] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Accepted: 07/20/2023] [Indexed: 09/22/2023] Open
Abstract
Lugdunin produced by Staphylococcus lugdunensis has been shown to have broad inhibitory activity against Gram-positive bacteria; however, lugdunin activity among S. lugdunensis isolates and its association with different agr, SCCmec, and sequence types remain unclear. We used matrix-assisted laser desorption ionization-time-of-flight mass spectrometry to identify S. lugdunensis and collected 202 S. lugdunensis samples for further assays. Agar spot tests were performed to characterize S. lugdunensis lugdunin production and activity. Multilocus sequence typing, SCCmec, and agr genotyping were performed on S. lugdunensis. In all, 91 Staphylococcus aureus strains with varying vancomycin susceptibilities were used to examine lugdunin activity in S. lugdunensis. In total, 48 S. lugdunensis strains (23.8%) were found to be oxacillin-resistant S. lugdunensis (ORSL), whereas 154 (76.2%) were classified as oxacillin-sensitive S. lugdunensis (OSSL). Moreover, 16 (33.3%) ORSL and 35 (22.7%) OSSL strains showed antibacterial activity against S. aureus. Our data showed that most lugdunin-producing ORSL strains (14/48, 29.2%) were of ST3-SCCmec V-agr II genotypes, whereas most lugdunin-producing OSSL strains (15/154, 9.7%) were of ST3-agr II, followed by ST1-agr I (10/154, 6.5%). Our data also revealed that lugdunin exhibited weak inhibitory activity against the VISA ST239 isolate. In addition, we observed that ST239 VSSA was more resistant to lugdunin than ST5, ST59, and ST45 VSSA. Taken together, our data pioneered the epidemiology of lugdunin production in S. lugdunensis isolates and revealed its association with genotypes. However, further molecular and bioinformatics investigations are needed to elucidate the regulatory mechanisms of lugdunin production and activity. IMPORTANCE Lugdunin is active against both methicillin-resistant Staphylococcus aureus and vancomycin-resistant enterococci by dissipating their membrane potential. However, the association of lugdunin activity with the genotypes of Staphylococcus lugdunensis has not been addressed. Here, we show the high prevalence of lugdunin-producing strains among ST1 (83.3%), ST2 (66.7%), and ST3 (53.3%) S. lugdunensis. Moreover, we identified the antibacterial activity of lugdunin-producing strains against VISA and hVISA. These results shed light on the potential application of lugdunin for the treatment of drug-resistant pathogens.
Collapse
Affiliation(s)
- Shih-Cheng Chang
- Department of Laboratory Medicine, Linkou Chang Gung Memorial Hospital, Taoyuan, Taiwan
- Department of Medical Biotechnology and Laboratory Science, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Cheng-Yen Kao
- Institute of Microbiology and Immunology, College of Life Sciences, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Lee-Chung Lin
- Department of Laboratory Medicine, Linkou Chang Gung Memorial Hospital, Taoyuan, Taiwan
| | - Jazon Harl Hidrosollo
- Institute of Microbiology and Immunology, College of Life Sciences, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Jang-Jih Lu
- Department of Laboratory Medicine, Linkou Chang Gung Memorial Hospital, Taoyuan, Taiwan
- Department of Medical Biotechnology and Laboratory Science, College of Medicine, Chang Gung University, Taoyuan, Taiwan
- Department of Medicine, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| |
Collapse
|
14
|
Zahra NUA, Vagiona AC, Uddin R, Andrade-Navarro MA. Selection of Multi-Drug Targets against Drug-Resistant Mycobacterium tuberculosis XDR1219 Using the Hyperbolic Mapping of the Protein Interaction Network. Int J Mol Sci 2023; 24:14050. [PMID: 37762354 PMCID: PMC10530867 DOI: 10.3390/ijms241814050] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Revised: 09/06/2023] [Accepted: 09/12/2023] [Indexed: 09/29/2023] Open
Abstract
Tuberculosis remains the leading cause of death from a single pathogen. On the other hand, antimicrobial resistance (AMR) makes it increasingly difficult to deal with this disease. We present the hyperbolic embedding of the Mycobacterium tuberculosis protein interaction network (mtbPIN) of resistant strain (MTB XDR1219) to determine the biological relevance of its latent geometry. In this hypermap, proteins with similar interacting partners occupy close positions. An analysis of the hypermap of available drug targets (DTs) and their direct and intermediate interactors was used to identify potentially useful drug combinations and drug targets. We identify rpsA and rpsL as close DTs targeted by different drugs (pyrazinamide and aminoglycosides, respectively) and propose that the combination of these drugs could have a synergistic effect. We also used the hypermap to explain the effects of drugs that affect multiple DTs, for example, forcing the bacteria to deal with multiple stresses like ethambutol, which affects the synthesis of both arabinogalactan and lipoarabinomannan. Our strategy uncovers novel potential DTs, such as dprE1 and dnaK proteins, which interact with two close DT pairs: arabinosyltransferases (embC and embB), Ser/Thr protein kinase (pknB) and RNA polymerase (rpoB), respectively. Our approach provides mechanistic explanations for existing drugs and suggests new DTs. This strategy can also be applied to the study of other resistant strains.
Collapse
Affiliation(s)
- Noor ul Ain Zahra
- Lab 103 PCMD ext., Dr. Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi 75270, Pakistan;
- Institute of Organismic and Molecular Evolution, Faculty of Biology, Johannes Gutenberg University, Hans-Dieter-Hüsch-Weg 15, 55128 Mainz, Germany;
| | - Aimilia-Christina Vagiona
- Institute of Organismic and Molecular Evolution, Faculty of Biology, Johannes Gutenberg University, Hans-Dieter-Hüsch-Weg 15, 55128 Mainz, Germany;
| | - Reaz Uddin
- Lab 103 PCMD ext., Dr. Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi 75270, Pakistan;
| | - Miguel A. Andrade-Navarro
- Institute of Organismic and Molecular Evolution, Faculty of Biology, Johannes Gutenberg University, Hans-Dieter-Hüsch-Weg 15, 55128 Mainz, Germany;
| |
Collapse
|
15
|
Liu DY, Phillips L, Wilson DM, Fulton KM, Twine SM, Wong A, Linington RG. Collateral sensitivity profiling in drug-resistant Escherichia coli identifies natural products suppressing cephalosporin resistance. Nat Commun 2023; 14:1976. [PMID: 37031190 PMCID: PMC10082850 DOI: 10.1038/s41467-023-37624-4] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Accepted: 03/22/2023] [Indexed: 04/10/2023] Open
Abstract
The rapid emergence of antimicrobial resistance presents serious health challenges to the management of infectious diseases, a problem that is further exacerbated by slowing rates of antimicrobial drug discovery in recent years. The phenomenon of collateral sensitivity (CS), whereby resistance to one drug is accompanied by increased sensitivity to another, provides new opportunities to address both these challenges. Here, we present a high-throughput screening platform termed Collateral Sensitivity Profiling (CSP) to map the difference in bioactivity of large chemical libraries across 29 drug-resistant strains of E. coli. CSP screening of 80 commercial antimicrobials demonstrated multiple CS interactions. Further screening of a 6195-member natural product library revealed extensive CS relationships in nature. In particular, we report the isolation of known and new analogues of borrelidin A with potent CS activities against cephalosporin-resistant strains. Co-dosing ceftazidime with borrelidin A slows broader cephalosporin resistance with no recognizable resistance to borrelidin A itself.
Collapse
Affiliation(s)
- Dennis Y Liu
- Department of Chemistry, Simon Fraser University, 8888 University Dr., V5A 1S6, Burnaby, BC, Canada
| | - Laura Phillips
- Department of Biology, Carleton University, 1125 Colonel By Dr., K1S 5B6, Ottawa, ON, Canada
| | - Darryl M Wilson
- Department of Chemistry, Simon Fraser University, 8888 University Dr., V5A 1S6, Burnaby, BC, Canada
| | - Kelly M Fulton
- Human Health Therapeutics Research Center, National Research Council Canada, 100 Sussex Dr., K1N 5A2, Ottawa, ON, Canada
| | - Susan M Twine
- Department of Biology, Carleton University, 1125 Colonel By Dr., K1S 5B6, Ottawa, ON, Canada
- Human Health Therapeutics Research Center, National Research Council Canada, 100 Sussex Dr., K1N 5A2, Ottawa, ON, Canada
| | - Alex Wong
- Department of Biology, Carleton University, 1125 Colonel By Dr., K1S 5B6, Ottawa, ON, Canada
- Institute for Advancing Health Through Agriculture, Texas A&M AgriLife, 1500 Research Parkway, 77845, College Station, TX, USA
| | - Roger G Linington
- Department of Chemistry, Simon Fraser University, 8888 University Dr., V5A 1S6, Burnaby, BC, Canada.
| |
Collapse
|
16
|
Lau WYV, Taylor PK, Brinkman FSL, Lee AHY. Pathogen-associated gene discovery workflows for novel antivirulence therapeutic development. EBioMedicine 2023; 88:104429. [PMID: 36628845 PMCID: PMC9843249 DOI: 10.1016/j.ebiom.2022.104429] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 11/23/2022] [Accepted: 12/15/2022] [Indexed: 01/11/2023] Open
Abstract
Novel therapeutics to manage bacterial infections are urgently needed as the impact and prevalence of antimicrobial resistance (AMR) grows. Antivirulence therapeutics are an alternative approach to antibiotics that aim to attenuate virulence rather than target bacterial essential functions, while minimizing microbiota perturbation and the risk of AMR development. Beyond known virulence factors, pathogen-associated genes (PAGs; genes found only in pathogens to date) may play an important role in virulence or host association. Many identified PAGs encode uncharacterized hypothetical proteins and represent an untapped wealth of novel drug targets. Here, we review current advances in antivirulence drug research and development, including PAG identification, and provide a comprehensive workflow from the discovery of antivirulence drug targets to drug discovery. We highlight the importance of integrating bioinformatic/genomic-based methods for novel virulence factor discovery, coupled with experimental characterization, into existing drug screening platforms to develop novel and effective antivirulence drugs.
Collapse
Affiliation(s)
- Wing Yin Venus Lau
- Department of Molecular Biology and Biochemistry, Simon Fraser University, Burnaby, BC, Canada
| | - Patrick K Taylor
- Department of Molecular Biology and Biochemistry, Simon Fraser University, Burnaby, BC, Canada
| | - Fiona S L Brinkman
- Department of Molecular Biology and Biochemistry, Simon Fraser University, Burnaby, BC, Canada.
| | - Amy H Y Lee
- Department of Molecular Biology and Biochemistry, Simon Fraser University, Burnaby, BC, Canada.
| |
Collapse
|
17
|
Albuquerque PC, Zicker F, Fonseca BP. Advancing drug repurposing research: Trends, collaborative networks, innovation and knowledge leaders. Drug Discov Today 2022; 27:103396. [DOI: 10.1016/j.drudis.2022.103396] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Revised: 07/08/2022] [Accepted: 10/06/2022] [Indexed: 11/03/2022]
|
18
|
Monteiro KLC, Silva ON, Dos Santos Nascimento IJ, Mendonça Júnior FJB, Aquino PGV, da Silva-Júnior EF, de Aquino TM. Medicinal Chemistry of Inhibitors Targeting Resistant Bacteria. Curr Top Med Chem 2022; 22:1983-2028. [PMID: 35319372 DOI: 10.2174/1568026622666220321124452] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2021] [Revised: 02/01/2022] [Accepted: 02/13/2022] [Indexed: 12/15/2022]
Abstract
The discovery of antibiotics was a revolutionary feat that provided countless health benefits. The identification of penicillin by Alexander Fleming initiated the era of antibiotics, represented by constant discoveries that enabled effective treatments for the different classes of diseases caused by bacteria. However, the indiscriminate use of these drugs allowed the emergence of resistance mechanisms of these microorganisms against the available drugs. In addition, the constant discoveries in the 20th century generated a shortage of new molecules, worrying health agencies and professionals about the appearance of multidrug-resistant strains against available drugs. In this context, the advances of recent years in molecular biology and microbiology have allowed new perspectives in drug design and development, using the findings related to the mechanisms of bacterial resistance to generate new drugs that are not affected by such mechanisms and supply new molecules to be used to treat resistant bacterial infections. Besides, a promising strategy against bacterial resistance is the combination of drugs through adjuvants, providing new expectations in designing new antibiotics and new antimicrobial therapies. Thus, this manuscript will address the main mechanisms of bacterial resistance under the understanding of medicinal chemistry, showing the main active compounds against efflux mechanisms, and also the application of the use of drug delivery systems, and finally, the main potential natural products as adjuvants or with promising activity against resistant strains.
Collapse
Affiliation(s)
- Kadja Luana Chagas Monteiro
- Research Group on Therapeutic Strategies - GPET, Laboratory of Synthesis and Research in Medicinal Chemistry - LSPMED, Institute of Chemistry and Biotechnology, Federal University of Alagoas, 57072-970, Maceió, Alagoas, Brazil
| | - Osmar Nascimento Silva
- Faculty of Pharmacy, University Center of Anápolis, Unievangélica, 75083-515, Anápolis, Goiás, Brazil
| | - Igor José Dos Santos Nascimento
- Research Group on Therapeutic Strategies - GPET, Laboratory of Synthesis and Research in Medicinal Chemistry - LSPMED, Institute of Chemistry and Biotechnology, Federal University of Alagoas, 57072-970, Maceió, Alagoas, Brazil
| | | | | | - Edeildo Ferreira da Silva-Júnior
- Laboratory of Medicinal Chemistry, Institute of Pharmaceutical Sciences, Federal University of Alagoas, 57072-970, Maceió, Alagoas, Brazil
| | - Thiago Mendonça de Aquino
- Research Group on Therapeutic Strategies - GPET, Laboratory of Synthesis and Research in Medicinal Chemistry - LSPMED, Institute of Chemistry and Biotechnology, Federal University of Alagoas, 57072-970, Maceió, Alagoas, Brazil
| |
Collapse
|
19
|
Frossard TM, Trapp N, Altmann KH. Studies Towards the Total Synthesis of Amycolamicin: A Chiral Auxiliary‐Based Diels‐Alder Approach towards the Decalin Core. European J Org Chem 2022. [DOI: 10.1002/ejoc.202200761] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Affiliation(s)
- Thomas M. Frossard
- ETH Zürich: Eidgenossische Technische Hochschule Zurich Chemistry and Applied Biosciences SWITZERLAND
| | - Nils Trapp
- ETH Zürich: Eidgenossische Technische Hochschule Zurich Chemistry and Applied Biosciences SWITZERLAND
| | - Karl-Heinz Altmann
- ETH Zurich Deptm. of Chemistry and Applied Biosciences Vladimir-Prelog-Weg 1- 5/10 8093 Zurich SWITZERLAND
| |
Collapse
|
20
|
Butler MS, Gigante V, Sati H, Paulin S, Al-Sulaiman L, Rex JH, Fernandes P, Arias CA, Paul M, Thwaites GE, Czaplewski L, Alm RA, Lienhardt C, Spigelman M, Silver LL, Ohmagari N, Kozlov R, Harbarth S, Beyer P. Analysis of the Clinical Pipeline of Treatments for Drug-Resistant Bacterial Infections: Despite Progress, More Action Is Needed. Antimicrob Agents Chemother 2022; 66:e0199121. [PMID: 35007139 PMCID: PMC8923189 DOI: 10.1128/aac.01991-21] [Citation(s) in RCA: 156] [Impact Index Per Article: 52.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
There is an urgent global need for new strategies and drugs to control and treat multidrug-resistant bacterial infections. In 2017, the World Health Organization (WHO) released a list of 12 antibiotic-resistant priority pathogens and began to critically analyze the antibacterial clinical pipeline. This review analyzes "traditional" and "nontraditional" antibacterial agents and modulators in clinical development current on 30 June 2021 with activity against the WHO priority pathogens mycobacteria and Clostridioides difficile. Since 2017, 12 new antibacterial drugs have been approved globally, but only vaborbactam belongs to a new antibacterial class. Also innovative is the cephalosporin derivative cefiderocol, which incorporates an iron-chelating siderophore that facilitates Gram-negative bacteria cell entry. Overall, there were 76 antibacterial agents in clinical development (45 traditional and 31 nontraditional), with 28 in phase 1, 32 in phase 2, 12 in phase 3, and 4 under regulatory evaluation. Forty-one out of 76 (54%) targeted WHO priority pathogens, 16 (21%) were against mycobacteria, 15 (20%) were against C. difficile, and 4 (5%) were nontraditional agents with broad-spectrum effects. Nineteen of the 76 antibacterial agents have new pharmacophores, and 4 of these have new modes of actions not previously exploited by marketed antibacterial drugs. Despite there being 76 antibacterial clinical candidates, this analysis indicated that there were still relatively few clinically differentiated antibacterial agents in late-stage clinical development, especially against critical-priority pathogens. We believe that future antibacterial research and development (R&D) should focus on the development of innovative and clinically differentiated candidates that have clear and feasible progression pathways to the market.
Collapse
Affiliation(s)
- Mark S. Butler
- MSBChem Consulting, Brisbane, Queensland, Australia
- Institute for Molecular Bioscience, University of Queensland, Brisbane, Queensland, Australia
| | | | - Hatim Sati
- Antimicrobial Resistance Division, WHO, Geneva, Switzerland
| | - Sarah Paulin
- Antimicrobial Resistance Division, WHO, Geneva, Switzerland
| | | | - John H. Rex
- F2G Limited, Eccles, Manchester, United Kingdom
- McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Prabhavathi Fernandes
- Scientific Advisory Committee, GARDP, Geneva, Switzerland
- The National Biodefense Science Board, U.S. Department of Health and Human Services, Washington, DC, USA
| | - Cesar A. Arias
- Center for Infectious Diseases Research, Houston Methodist Research Institute, Houston, Texas, USA
- Center for Infectious Diseases, UTHealth School of Public Health, Houston, Texas, USA
| | - Mical Paul
- Infectious Diseases Institute, Rambam Health Care Campus, Haifa, Israel
- The Ruth and Bruce Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, Israel
| | - Guy E. Thwaites
- Oxford University Clinical Research Unit, Ho Chi Minh City, Vietnam
- Centre for Tropical Medicine and Global Health, Nuffield Department of Clinical Medicine, Oxford University, Oxford, United Kingdom
| | - Lloyd Czaplewski
- Chemical Biology Ventures Ltd., Abingdon, Oxfordshire, United Kingdom
| | | | - Christian Lienhardt
- Université de Montpellier, INSERM, Institut de Recherche pour le Développement, Montpellier, France
| | | | | | - Norio Ohmagari
- National Center for Global Health and Medicine, Tokyo, Japan
| | - Roman Kozlov
- Institute of Antimicrobial Chemotherapy, Smolensk State Medical University, Smolensk, Russia
| | - Stephan Harbarth
- National Center for Infection Prevention, Swissnoso, Bern, Switzerland
- Infection Control Programme, Geneva University Hospitals and Faculty of Medicine, WHO Collaborating Center for Patient Safety, Geneva, Switzerland
| | - Peter Beyer
- Antimicrobial Resistance Division, WHO, Geneva, Switzerland
| |
Collapse
|
21
|
Haindongo EH, Funtua B, Singu B, Hedimbi M, Kalemeera F, Hamman J, Vainio O, Hakanen AJ, Vuopio J. Antimicrobial resistance among bacteria isolated from urinary tract infections in females in Namibia, 2016-2017. Antimicrob Resist Infect Control 2022; 11:33. [PMID: 35151360 PMCID: PMC8840701 DOI: 10.1186/s13756-022-01066-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Accepted: 01/21/2022] [Indexed: 01/08/2023] Open
Abstract
Background The emergence of antimicrobial resistance (AMR) among bacterial pathogens demands a local understanding of the epidemiological situation. This information is needed both for clinical treatment decision-making purposes as well as for the revision of current care guidelines. Clinical AMR data from Namibia is sparse, whilst urinary tract infections remain not only widespread but they disproportionally affect females. This paper aims to describe the national antimicrobial resistance situation of major bacterial uropathogens in females within the 14 Namibian regions. Method Retrospective countrywide information on clinical urine cultures performed in females in Namibia in 2016–2017 was obtained from the national public health laboratory, Namibia Institute of Pathology (NIP). The data set included both microbiological findings as well as antimicrobial susceptibility test (AST) results. The AST was done as per the Clinical and Laboratory Standards Institute (CLSI) guidelines. Resistance to 3rd generation cephalosporins was indicative of Extended Spectrum-ß-lactamase (ESBL) production. Data analysis was done with WHONET using expert interpretation rules. Results In total, 22,259 urinary cultures were performed, of which 13,673 (61.4%) were culture positive. Gram-negative bacterial species accounted for 72.6% of the findings. The most common pathogens identified were Escherichia coli, Klebsiella pneumoniae and Proteus mirabilis. Most of these were from young females, with a median age ranging from 28 to 32 years for the various pathogens. Resistance to ampicillin was 77.7% in E. coli and 84.9% in K. pneumoniae. In E. coli, resistance to 1st line empiric therapy antibiotic, nitrofurantoin, was below 13%, except for one region that showed 59.2% resistance. Resistance to third generation cephalosporin (3GC) was used as a proxy for ESBL production. By year 2017, 3GC resistance was 22%, 31.4% and 8.3% for E. coli, K. pneumoniae and P. mirabilis, respectively. Conclusion We report high resistance to ampicillin, quinolones and sulfamethoxazole-trimethoprim amongst E. coli. Resistance rates to third-generation cephalosporins was also concerningly high at 22%. Resistance to carbapenems was low. However, superiority of nitrofurantoin was found, which provides rational support for the usefulness of nitrofurantoin as an empiric therapy regimen for the treatment of urinary tract infections in this setting. Supplementary Information The online version contains supplementary material available at 10.1186/s13756-022-01066-2.
Collapse
Affiliation(s)
- Erastus H Haindongo
- School of Medicine, Faculty of Health Sciences, University of Namibia, Windhoek, Namibia. .,Institute of Biomedicine, University of Turku, Turku, Finland.
| | - Binta Funtua
- School of Pharmacy, Faculty of Health Sciences, University of Namibia, Windhoek, Namibia
| | - Boni Singu
- School of Pharmacy, Faculty of Health Sciences, University of Namibia, Windhoek, Namibia
| | - Marius Hedimbi
- School of Medicine, Faculty of Health Sciences, University of Namibia, Windhoek, Namibia
| | - Francis Kalemeera
- School of Pharmacy, Faculty of Health Sciences, University of Namibia, Windhoek, Namibia
| | - Jana Hamman
- Namibia Institute of Pathology, Windhoek, Namibia
| | - Olli Vainio
- Faculty of Medicine, University of Turku, Turku, Finland
| | - Antti J Hakanen
- Institute of Biomedicine, University of Turku, Turku, Finland.,Department of Clinical Microbiology, Laboratory Division, Turku University Hospital, Turku, Finland
| | - Jaana Vuopio
- Institute of Biomedicine, University of Turku, Turku, Finland.,Department of Clinical Microbiology, Laboratory Division, Turku University Hospital, Turku, Finland
| |
Collapse
|
22
|
Bridi H, Pustay AP, Bordignon SADL, Picoli SU, von Poser GL, Ferraz ADBF. Antimicrobial activity of dimeric acylphloroglucinols isolated from southern Brazilian Hypericum species against to resistant bacterial. Nat Prod Res 2022; 36:6448-6452. [PMID: 35142580 DOI: 10.1080/14786419.2022.2038596] [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: 10/19/2022]
Abstract
The hexanic extracts of Hypericum austrobrasiliense, H. caprifoliatum, H. denudatum, H. pedersenii and H. polyanthemum, and three isolated dimeric acylphloroglucinols (uliginosin B, japonicine A and hyperbrasilol B) were assayed for their antimicrobial activity against some Gram-positive and Gram-negative bacteria (including resistant strains). These extracts were assayed using the disc diffusion test, and the results indicated that the tested species did non exhibit activity on the Gram-negative strains. Subsequently, the minimum inhibitory concentration (MIC) and the minimum bactericidal concentration (MBC) were measured using the broth dilution technique adopted to macrodillution. The most susceptible strains were the MRSA and the S. aureus MLSb. Regarding these pathogens, the better MIC values were obtained with the extracts from H. austrobrasiliense, H. caprifoliatum and H. pedersenii. The acylphloroglucinols uliginosin B and hyperbrasilol B presented the lowest MIC values against Enterococcus faecalis, Staphylococcus aureus, MRSA and S. aureus MLSb resistance.
Collapse
Affiliation(s)
- Henrique Bridi
- Programa de Pós Graduação em Ciências Farmacêuticas, Faculdade de Farmácia, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, RS, Brazil
| | - Ana Paula Pustay
- Instituto de Ciências da Saúde, Universidade Feevale (FEEVALE), Novo Hamburgo, RS, Brazil
| | | | - Simone Ulrich Picoli
- Instituto de Ciências da Saúde, Universidade Feevale (FEEVALE), Novo Hamburgo, RS, Brazil
| | - Gilsane Lino von Poser
- Programa de Pós Graduação em Ciências Farmacêuticas, Faculdade de Farmácia, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, RS, Brazil
| | - Alexandre de Barros Falcão Ferraz
- Programa de Pós Graduação em Biologia Celular e Molecular Aplicada as Ciências Farmacêuticas (PPGBioSaúde), Universidade Luterana do Brasil (ULBRA), Canoas, RS, Brazil
| |
Collapse
|
23
|
Singh S, Numan A, Cinti S. Point-of-Care for Evaluating Antimicrobial Resistance through the Adoption of Functional Materials. Anal Chem 2022; 94:26-40. [PMID: 34802244 PMCID: PMC8756393 DOI: 10.1021/acs.analchem.1c03856] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Affiliation(s)
- Sima Singh
- IES
Institute of Pharmacy, IES University Campus, Kalkheda, Ratibad Main Road, Bhopal 462044, Madhya Pradesh, India
| | - Arshid Numan
- Graphene
& Advanced 2D Materials Research Group (GAMRG), School of Engineering
and Technology, Sunway University, 5, Jalan University, Bandar Sunway, 47500 Petaling
Jaya, Selangor, Malaysia
| | - Stefano Cinti
- Department
of Pharmacy, University of Naples “Federico
II”, Via D. Montesano 49, 80131 Naples, Italy
- BAT
Center−Interuniversity Center for Studies on Bioinspired Agro-Environmental
Technology, University of Napoli Federico
II, 80055 Naples, Italy
| |
Collapse
|
24
|
Barreca D, Trombetta D, Smeriglio A, Mandalari G, Romeo O, Felice MR, Gattuso G, Nabavi SM. Food flavonols: Nutraceuticals with complex health benefits and functionalities. Trends Food Sci Technol 2021. [DOI: 10.1016/j.tifs.2021.03.030] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
25
|
Li S, Wei R, Lin Y, Feng Z, Zhang Z, Wang Z, Chen Y, Ma J, Yan Y, Sun J, Sun T, Chen Z, Li S, Wang H. A Preliminary Study of Antibiotic Resistance Genes in Domestic Honey Produced in China. Foodborne Pathog Dis 2021; 18:859-866. [PMID: 34415782 DOI: 10.1089/fpd.2020.2877] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Antibiotic resistance genes (ARGs) are emerging contaminants that pose a health risk to humans worldwide. Little information on ARGs in bee honey is available. This study profiles ARGs in bee honey samples produced in China, the biggest producer in the world. Of 317 known ARGs encoding resistance to 8 classes of antibiotics, 212 were found in collected honey samples by a real-time quantitative polymerase chain reaction approach. Occurrence frequencies of genes providing resistance to FCA (fluoroquinolone, quinolone, florfenicol, chloramphenicol, and amphenicol) and aminoglycosides were 21.0% and 18.5%, respectively. Frequencies of genes encoding efflux pumps were 42.5% and those of destructase genes 36.6%, indicating that these two mechanisms were predominant for resistance. Nine plasmid-mediated quinolone resistance genes were detected. Of the nine transposase genes known to be involved in antibiotic resistance, eight were found in the samples examined, with tnpA-4, tnpA-5, and tnpA-6 being more abundant. The abundance of the transposase genes was associated with genes conferring resistance to tetracyclines (r = 0.648, p < 0.01), macrolide-lincosamide-streptogramin B (r = 0.642, p < 0.01), FCA (r = 0.517, p < 0.01), and aminoglycosides (r = 0.401, 0.01 < p < 0.05). This is the first study on the abundance and diversity of ARGs in Chinese bee honey products. These findings suggest that bee honey may be a significant source of ARGs that might pose threat to public health. Further research is required to collect more samples in diverse geographic regions in China to make a more comprehensive judgment of ARG in bee honey.
Collapse
Affiliation(s)
- Sisi Li
- Shanghai Key Laboratory of Veterinary Biotechnology, Key Laboratory of Urban Agriculture, Ministry of Agriculture and Rural Areas, School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai, China
| | - Renjie Wei
- Shanghai Key Laboratory of Veterinary Biotechnology, Key Laboratory of Urban Agriculture, Ministry of Agriculture and Rural Areas, School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai, China
| | - Yingzheng Lin
- Technical Center for Animal, Plant, and Food Inspection and Quarantine of Shanghai Customs, Shanghai, China
| | - Zhu Feng
- Shanghai Key Laboratory of Veterinary Biotechnology, Key Laboratory of Urban Agriculture, Ministry of Agriculture and Rural Areas, School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai, China
| | - Zhenyang Zhang
- Shanghai Key Laboratory of Veterinary Biotechnology, Key Laboratory of Urban Agriculture, Ministry of Agriculture and Rural Areas, School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai, China
| | - Zhaofei Wang
- Shanghai Key Laboratory of Veterinary Biotechnology, Key Laboratory of Urban Agriculture, Ministry of Agriculture and Rural Areas, School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai, China
| | - Yuqiang Chen
- Shanghai Key Laboratory of Veterinary Biotechnology, Key Laboratory of Urban Agriculture, Ministry of Agriculture and Rural Areas, School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai, China
| | - Jingjiao Ma
- Shanghai Key Laboratory of Veterinary Biotechnology, Key Laboratory of Urban Agriculture, Ministry of Agriculture and Rural Areas, School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai, China
| | - Yaxian Yan
- Shanghai Key Laboratory of Veterinary Biotechnology, Key Laboratory of Urban Agriculture, Ministry of Agriculture and Rural Areas, School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai, China
| | - Jianhe Sun
- Shanghai Key Laboratory of Veterinary Biotechnology, Key Laboratory of Urban Agriculture, Ministry of Agriculture and Rural Areas, School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai, China
| | - Tao Sun
- Shanghai Key Laboratory of Veterinary Biotechnology, Key Laboratory of Urban Agriculture, Ministry of Agriculture and Rural Areas, School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai, China
| | - Zhifei Chen
- Technical Center for Animal, Plant, and Food Inspection and Quarantine of Shanghai Customs, Shanghai, China
| | - Shuqing Li
- Technical Center for Animal, Plant, and Food Inspection and Quarantine of Shanghai Customs, Shanghai, China
| | - Hengan Wang
- Shanghai Key Laboratory of Veterinary Biotechnology, Key Laboratory of Urban Agriculture, Ministry of Agriculture and Rural Areas, School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
26
|
Sun H, Yu Y, Zhang Y, Li J, Cheng Y, Huang S, Wang W, Zhang X. Glycosylated Nanotherapeutics with β-Lactamase Reversible Competitive Inhibitory Activity Reinvigorates Antibiotics against Gram-Negative Bacteria. Biomacromolecules 2021; 22:2834-2849. [PMID: 34164980 DOI: 10.1021/acs.biomac.1c00231] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Antibiotics are currently first-line therapy for bacterial infections. However, the curative effect of antibiotic remedies is limited due to increasingly prevalent bacterial resistance. The strategy to reverse intrinsic acquired drug resistance presents a promising option for reinvigorating antibiotic therapy. Here, we developed a β-lactamase-inhibiting macromolecule composed of benzoxaborole and dextran for precise transport of β-lactam antibiotics to strains overexpressing β-lactamase. Benzoxaborole-derived nanotherapeutics enabled specific recognition and rapid internalization, and the nanotherapeutics with a high affinity toward bacteria distinctly inhibited the catalytic activity of bacterially secreted β-lactamase by a reversible competitive mechanism. Thus, the system entrapping cefoxitin harbored a significantly enhanced ability to kill drug-resistant Escherichia coli compared to the ability of the drug by specifically overcoming the membrane barrier and acquired resistance mechanism of β-lactamase overproduction. The reversible competitive nanotherapeutics exhibited a robust therapeutic efficacy in rat wounds infected with drug-resistant bacteria; the efficacy was due to efficient bacterial elimination and collateral benzoxaborole-dependent amelioration of the inflammatory response. The above results offered insights into the facile design of precise macromolecular adjuvants to exclusively reverse the acquired bacterial resistance mechanism and increase the utility of antibiotic therapies against antibiotic-resistant bacterial infections.
Collapse
Affiliation(s)
- Haonan Sun
- Key Laboratory of Functional Polymer Materials of Ministry Education, Institute of Polymer Chemistry, College of Chemistry, Nankai University, Tianjin 300071, China
| | - Yunjian Yu
- Key Laboratory of Functional Polymer Materials of Ministry Education, Institute of Polymer Chemistry, College of Chemistry, Nankai University, Tianjin 300071, China
| | - Yufei Zhang
- Key Laboratory of Functional Polymer Materials of Ministry Education, Institute of Polymer Chemistry, College of Chemistry, Nankai University, Tianjin 300071, China
| | - Jie Li
- Key Laboratory of Functional Polymer Materials of Ministry Education, Institute of Polymer Chemistry, College of Chemistry, Nankai University, Tianjin 300071, China
| | - Yijie Cheng
- Key Laboratory of Functional Polymer Materials of Ministry Education, Institute of Polymer Chemistry, College of Chemistry, Nankai University, Tianjin 300071, China
| | - Siyuan Huang
- Key Laboratory of Functional Polymer Materials of Ministry Education, Institute of Polymer Chemistry, College of Chemistry, Nankai University, Tianjin 300071, China
| | - Wenbo Wang
- Key Laboratory of Functional Polymer Materials of Ministry Education, Institute of Polymer Chemistry, College of Chemistry, Nankai University, Tianjin 300071, China
| | - Xinge Zhang
- Key Laboratory of Functional Polymer Materials of Ministry Education, Institute of Polymer Chemistry, College of Chemistry, Nankai University, Tianjin 300071, China
| |
Collapse
|
27
|
The Design of Alapropoginine, a Novel Conjugated Ultrashort Antimicrobial Peptide with Potent Synergistic Antimicrobial Activity in Combination with Conventional Antibiotics. Antibiotics (Basel) 2021; 10:antibiotics10060712. [PMID: 34199154 PMCID: PMC8231522 DOI: 10.3390/antibiotics10060712] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Revised: 06/10/2021] [Accepted: 06/11/2021] [Indexed: 12/01/2022] Open
Abstract
(1) Background: Antimicrobial resistance represents an urgent health dilemma facing the global human population. The development of novel antimicrobial agents is needed to face the rising number of resistant bacteria. Ultrashort antimicrobial peptides (USAMPs) are considered promising antimicrobial agents that meet the required criteria of novel antimicrobial drug development. (2) Methods: Alapropoginine was rationally designed by incorporating arginine (R), biphenylalanine (B), and naproxen to create an ultrashort hexapeptide. The antimicrobial activity of alapropoginine was evaluated against different strains of bacteria. The hemolytic activity of alapropoginine was also investigated against human erythrocytes. Finally, synergistic studies with antibiotics were performed using the checkerboard technique and the determination of the fractional inhibitory index. (3) Results: Alapropoginine displayed potent antimicrobial activities against reference and multi-drug-resistant bacteria with MIC values of as low as 28.6 µg/mL against methicillin-resistant S. aureus. Alapropoginine caused negligible toxicity toward human red blood cells. Moreover, the synergistic studies showed improved activities for the combined conventional antibiotics with a huge reduction in their antimicrobial concentrations. (4) Conclusions: The present study indicates that alapropoginine exhibits promising antimicrobial activity against reference and resistant strains of bacteria with negligible hemolytic activity. Additionally, the peptide displays synergistic or additive effects when combined with several antibiotics.
Collapse
|
28
|
Gomes NGM, Madureira-Carvalho Á, Dias-da-Silva D, Valentão P, Andrade PB. Biosynthetic versatility of marine-derived fungi on the delivery of novel antibacterial agents against priority pathogens. Biomed Pharmacother 2021; 140:111756. [PMID: 34051618 DOI: 10.1016/j.biopha.2021.111756] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Revised: 05/13/2021] [Accepted: 05/19/2021] [Indexed: 11/24/2022] Open
Abstract
Despite the increasing number of novel marine natural products being reported from fungi in the last three decades, to date only the broad-spectrum cephalosporin C can be tracked back as marine fungal-derived drug. Cephalosporins were isolated in the early 1940s from a strain of Acremonium chrysogenum obtained in a sample collected in sewage water in the Sardinian coast, preliminary findings allowing the discovery of cephalosporin C. Since then, bioprospection of marine fungi has been enabling the identification of several metabolites with antibacterial effects, many of which proving to be active against multi-drug resistant strains, available data suggesting also that some might fuel the pharmaceutical firepower towards some of the bacterial pathogens classified as a priority by the World Health Organization. Considering the success of their terrestrial counterparts on the discovery and development of several antibiotics that are nowadays used in the clinical setting, marine fungi obviously come into mind as producers of new prototypes to counteract antibiotic-resistant bacteria that are no longer responding to available treatments. We mainly aim to provide a snapshot on those metabolites that are likely to proceed to advanced preclinical development, not only based on their antibacterial potency, but also considering their targets and modes of action, and activity against priority pathogens.
Collapse
Affiliation(s)
- Nelson G M Gomes
- REQUIMTE/LAQV, Laboratório de Farmacognosia, Departamento de Química, Faculdade de Farmácia, Universidade do Porto, R. Jorge Viterbo Ferreira, nº 228, 4050-313 Porto, Portugal.
| | - Áurea Madureira-Carvalho
- REQUIMTE/LAQV, Laboratório de Farmacognosia, Departamento de Química, Faculdade de Farmácia, Universidade do Porto, R. Jorge Viterbo Ferreira, nº 228, 4050-313 Porto, Portugal; IINFACTS-Institute of Research and Advanced Training in Health Sciences and Technologies, Department of Sciences, University Institute of Health Sciences (IUCS), CESPU, CRL, Gandra, Portugal.
| | - Diana Dias-da-Silva
- IINFACTS-Institute of Research and Advanced Training in Health Sciences and Technologies, Department of Sciences, University Institute of Health Sciences (IUCS), CESPU, CRL, Gandra, Portugal; UCIBIO, REQUIMTE, Laboratory of Toxicology, Faculty of Pharmacy, University of Porto, R. Jorge Viterbo Ferreira, nº 228, 4050-313 Porto, Portugal.
| | - Patrícia Valentão
- REQUIMTE/LAQV, Laboratório de Farmacognosia, Departamento de Química, Faculdade de Farmácia, Universidade do Porto, R. Jorge Viterbo Ferreira, nº 228, 4050-313 Porto, Portugal.
| | - Paula B Andrade
- REQUIMTE/LAQV, Laboratório de Farmacognosia, Departamento de Química, Faculdade de Farmácia, Universidade do Porto, R. Jorge Viterbo Ferreira, nº 228, 4050-313 Porto, Portugal.
| |
Collapse
|
29
|
Shchelik IS, Tomio A, Gademann K. Design, Synthesis, and Biological Evaluation of Light-Activated Antibiotics. ACS Infect Dis 2021; 7:681-692. [PMID: 33656844 DOI: 10.1021/acsinfecdis.1c00015] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
The spatial and temporal control of bioactivity of small molecules by light (photopharmacology) constitutes a promising approach for study of biological processes and ultimately for the treatment of diseases. In this study, we investigated two different "caged" antibiotic classes that can undergo remote activation with UV-light at λ = 365 nm, via the conjugation of deactivating and photocleavable units through a short synthetic sequence. The two widely used antibiotics vancomycin and cephalosporin were thus enhanced in their performance by rendering them photoresponsive and thereby suppressing undesired off-site activity. The antimicrobial activity against Bacillus subtilis ATCC 6633, Staphylococcus aureus ATCC 29213, S. aureus ATCC 43300 (MRSA), Escherichia coli ATCC 25922, and Pseudomonas aeruginosa ATCC 27853 could be spatiotemporally controlled with light. Both molecular series displayed a good activity window. The vancomycin derivative displayed excellent values against Gram-positive strains after uncaging, and the next-generation caged cephalosporin derivative achieved good and broad activity against both Gram-positive and Gram-negative strains after photorelease.
Collapse
Affiliation(s)
- Inga S. Shchelik
- Department of Chemistry, University of Zurich, Winterthurerstrasse 190, 8057 Zurich, Switzerland
| | - Andrea Tomio
- Department of Chemistry, University of Zurich, Winterthurerstrasse 190, 8057 Zurich, Switzerland
| | - Karl Gademann
- Department of Chemistry, University of Zurich, Winterthurerstrasse 190, 8057 Zurich, Switzerland
| |
Collapse
|
30
|
Pseudomonas aeruginosa elastase (LasB) as a therapeutic target. Drug Discov Today 2021; 26:2108-2123. [PMID: 33676022 DOI: 10.1016/j.drudis.2021.02.026] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Revised: 02/03/2021] [Accepted: 02/17/2021] [Indexed: 02/08/2023]
Abstract
Why is P. aeruginosa LasB elastase an attractive target for antivirulence therapy and what is the state-of-the art in LasB inhibitor design and development?
Collapse
|