1
|
Joseph J, Boby S, Mooyottu S, Muyyarikkandy MS. Antibiotic potentiators as a promising strategy for combating antibiotic resistance. NPJ ANTIMICROBIALS AND RESISTANCE 2025; 3:53. [PMID: 40481260 PMCID: PMC12144181 DOI: 10.1038/s44259-025-00112-4] [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: 01/06/2025] [Accepted: 05/01/2025] [Indexed: 06/11/2025]
Abstract
Antimicrobial resistance (AMR) poses a critical global health challenge. It arises from pathogens' resistance to antibiotics due to misuse, overuse, and insufficient regulation. As new antibiotics emerge slowly, antibiotic potentiators can enhance existing treatments against resistant strains. Challenges such as toxicity and regulatory barriers necessitate further studies to optimize these agents. This review examines the mechanisms, sources, and recent advancements in antibiotic potentiation while highlighting its potential to combat AMR.
Collapse
Affiliation(s)
- Jiddu Joseph
- Department of Animal and Food Sciences, University of Delaware, Newark, DE, 19716, USA
| | - Sanya Boby
- Department of Animal and Food Sciences, University of Delaware, Newark, DE, 19716, USA
| | | | | |
Collapse
|
2
|
Khan RT, Sharma V, Khan SS, Rasool S. Prevention and potential remedies for antibiotic resistance: current research and future prospects. Front Microbiol 2024; 15:1455759. [PMID: 39421555 PMCID: PMC11484029 DOI: 10.3389/fmicb.2024.1455759] [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: 06/27/2024] [Accepted: 09/13/2024] [Indexed: 10/19/2024] Open
Abstract
The increasing threat of antibiotic resistance and shrinking treatment options for infections have pushed mankind into a difficult position. The looming threat of the return of the pre-antibiotic era has caused a sense of urgency to protect and conserve the potency of antibiotic therapy. One of the perverse effects of antibiotic resistance is the dissemination of its causative agents from non-clinically important strains to clinically important strains and vice versa. The popular saying "Prevention is better than cure" is appropriate for tackling antibiotic resistance. On the one hand, new and effective antibiotics are required; on the other hand, better measures for the use of antibiotics, along with increased awareness in the general public related to antibiotic use, are essential. Awareness, especially of appropriate antibiotic use, antibiotic resistance, its dissemination, and potential threats, can help greatly in controlling the use and abuse of antibiotics, and the containment of antibiotic resistance. Antibiotic drugs' effectiveness can be enhanced by producing novel antibiotic analogs or adding adjuvants to current antibiotics. Combinatorial therapy of antibiotics has proven successful in treating multidrug-resistant (MDR) bacterial infections. This review aims to highlight the current global situation of antibiotic resistance and discuss the methods used to monitor, prevent, inhibit, or reverse bacterial resistance mechanisms in the fight against antibiotic resistance.
Collapse
Affiliation(s)
| | | | | | - Shafaq Rasool
- Molecular Biology Lab, School of Biotechnology, Shri Mata Vaishno Devi University, Katra, India
| |
Collapse
|
3
|
Otun SO, Graca R, Achilonu I. Combating Aminoglycoside Resistance: From Structural and Functional Characterisation to Therapeutic Challenges with RKAAT. Curr Protein Pept Sci 2024; 25:454-468. [PMID: 38314602 DOI: 10.2174/0113892037278814231226104509] [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: 09/18/2023] [Revised: 12/04/2023] [Accepted: 12/07/2023] [Indexed: 02/06/2024]
Abstract
A comprehensive knowledge of aminoglycoside-modifying enzymes (AMEs) and their role in bacterial resistance mechanisms is urgently required due to the rising incidence of antibiotic resistance, particularly in Klebsiella pneumoniae infections. This study explores the essential features of AMEs, including their structural and functional properties, the processes by which they contribute to antibiotic resistance, and the therapeutic importance of aminoglycosides. The study primarily examines the Recombinant Klebsiella pneumoniae Aminoglycoside Adenylyl Transferase (RKAAT), particularly emphasizing its biophysical characteristics and the sorts of resistance it imparts. Furthermore, this study examines the challenges presented by RKAAT-mediated resistance, an evaluation of treatment methods and constraints, and options for controlling infection. The analysis provides a prospective outlook on strategies to address and reduce antibiotic resistance. This extensive investigation seeks to provide vital insights into the continuing fight against bacterial resistance, directing future research efforts and medicinal approaches.
Collapse
Affiliation(s)
- Sarah Oluwatobi Otun
- Department of Molecular and Cell Biology, Protein Structure-function Unit, University of Witwatersrand, Johannesburg, South Africa
| | - Richard Graca
- Department of Molecular and Cell Biology, Protein Structure-function Unit, University of Witwatersrand, Johannesburg, South Africa
| | - Ikechukwu Achilonu
- Department of Molecular and Cell Biology, Protein Structure-function Unit, University of Witwatersrand, Johannesburg, South Africa
| |
Collapse
|
4
|
Xiao G, Li J, Sun Z. The Combination of Antibiotic and Non-Antibiotic Compounds Improves Antibiotic Efficacy against Multidrug-Resistant Bacteria. Int J Mol Sci 2023; 24:15493. [PMID: 37895172 PMCID: PMC10607837 DOI: 10.3390/ijms242015493] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Revised: 10/19/2023] [Accepted: 10/21/2023] [Indexed: 10/29/2023] Open
Abstract
Bacterial antibiotic resistance, especially the emergence of multidrug-resistant (MDR) strains, urgently requires the development of effective treatment strategies. It is always of interest to delve into the mechanisms of resistance to current antibiotics and target them to promote the efficacy of existing antibiotics. In recent years, non-antibiotic compounds have played an important auxiliary role in improving the efficacy of antibiotics and promoting the treatment of drug-resistant bacteria. The combination of non-antibiotic compounds with antibiotics is considered a promising strategy against MDR bacteria. In this review, we first briefly summarize the main resistance mechanisms of current antibiotics. In addition, we propose several strategies to enhance antibiotic action based on resistance mechanisms. Then, the research progress of non-antibiotic compounds that can promote antibiotic-resistant bacteria through different mechanisms in recent years is also summarized. Finally, the development prospects and challenges of these non-antibiotic compounds in combination with antibiotics are discussed.
Collapse
Affiliation(s)
| | | | - Zhiliang Sun
- College of Veterinary Medicine, Hunan Agricultural University, Changsha 410128, China; (G.X.); (J.L.)
| |
Collapse
|
5
|
Magaña AJ, Sklenicka J, Pinilla C, Giulianotti M, Chapagain P, Santos R, Ramirez MS, Tolmasky ME. Restoring susceptibility to aminoglycosides: identifying small molecule inhibitors of enzymatic inactivation. RSC Med Chem 2023; 14:1591-1602. [PMID: 37731693 PMCID: PMC10507813 DOI: 10.1039/d3md00226h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Accepted: 07/21/2023] [Indexed: 09/22/2023] Open
Abstract
Growing resistance to antimicrobial medicines is a critical health problem that must be urgently addressed. Adding to the increasing number of patients that succumb to infections, there are other consequences to the rise in resistance like the compromise of several medical procedures and dental work that are heavily dependent on infection prevention. Since their introduction in the clinics, aminoglycoside antibiotics have been a critical component of the armamentarium to treat infections. Still, the increase in resistance and their side effects led to a decline in their utilization. However, numerous current factors, like the urgent need for antimicrobials and their favorable properties, led to renewed interest in these drugs. While efforts to design new classes of aminoglycosides refractory to resistance mechanisms and with fewer toxic effects are starting to yield new promising molecules, extending the useful life of those already in use is essential. For this, numerous research projects are underway to counter resistance from different angles, like inhibition of expression or activity of resistance components. This review focuses on selected examples of one aspect of this quest, the design or identification of small molecule inhibitors of resistance caused by enzymatic modification of the aminoglycoside. These compounds could be developed as aminoglycoside adjuvants to overcome resistant infections.
Collapse
Affiliation(s)
- Angel J Magaña
- Center for Applied Biotechnology Studies, Department of Biological Science, College of Natural Sciences and Mathematics, California State University Fullerton Fullerton CA 92831 USA
| | - Jan Sklenicka
- Center for Applied Biotechnology Studies, Department of Biological Science, College of Natural Sciences and Mathematics, California State University Fullerton Fullerton CA 92831 USA
| | - Clemencia Pinilla
- Center for Translational Science, Florida International University Port St. Lucie FL 34987 USA
| | - Marc Giulianotti
- Center for Translational Science, Florida International University Port St. Lucie FL 34987 USA
| | - Prem Chapagain
- Department of Physics, Florida International University Miami FL 33199 USA
- Biomolecular Sciences Institute, Florida International University Miami FL 33199 USA
| | - Radleigh Santos
- Department of Mathematics, Nova Southeastern University Fort Lauderdale FL 33314 USA
| | - Maria Soledad Ramirez
- Center for Applied Biotechnology Studies, Department of Biological Science, College of Natural Sciences and Mathematics, California State University Fullerton Fullerton CA 92831 USA
| | - Marcelo E Tolmasky
- Center for Applied Biotechnology Studies, Department of Biological Science, College of Natural Sciences and Mathematics, California State University Fullerton Fullerton CA 92831 USA
| |
Collapse
|
6
|
Darby EM, Trampari E, Siasat P, Gaya MS, Alav I, Webber MA, Blair JMA. Molecular mechanisms of antibiotic resistance revisited. Nat Rev Microbiol 2023; 21:280-295. [PMID: 36411397 DOI: 10.1038/s41579-022-00820-y] [Citation(s) in RCA: 506] [Impact Index Per Article: 253.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/17/2022] [Indexed: 11/22/2022]
Abstract
Antibiotic resistance is a global health emergency, with resistance detected to all antibiotics currently in clinical use and only a few novel drugs in the pipeline. Understanding the molecular mechanisms that bacteria use to resist the action of antimicrobials is critical to recognize global patterns of resistance and to improve the use of current drugs, as well as for the design of new drugs less susceptible to resistance development and novel strategies to combat resistance. In this Review, we explore recent advances in understanding how resistance genes contribute to the biology of the host, new structural details of relevant molecular events underpinning resistance, the identification of new resistance gene families and the interactions between different resistance mechanisms. Finally, we discuss how we can use this information to develop the next generation of antimicrobial therapies.
Collapse
Affiliation(s)
- Elizabeth M Darby
- College of Medical and Dental Sciences, Institute of Microbiology and Infection, University of Birmingham, Birmingham, UK
| | | | - Pauline Siasat
- College of Medical and Dental Sciences, Institute of Microbiology and Infection, University of Birmingham, Birmingham, UK
| | | | - Ilyas Alav
- College of Medical and Dental Sciences, Institute of Microbiology and Infection, University of Birmingham, Birmingham, UK
| | - Mark A Webber
- Quadram Institute Bioscience, Norwich Research Park, Norwich, UK.
- Medical School, University of East Anglia, Norwich Research Park, Norwich, UK.
| | - Jessica M A Blair
- College of Medical and Dental Sciences, Institute of Microbiology and Infection, University of Birmingham, Birmingham, UK.
| |
Collapse
|
7
|
Dhanda G, Acharya Y, Haldar J. Antibiotic Adjuvants: A Versatile Approach to Combat Antibiotic Resistance. ACS OMEGA 2023; 8:10757-10783. [PMID: 37008128 PMCID: PMC10061514 DOI: 10.1021/acsomega.3c00312] [Citation(s) in RCA: 70] [Impact Index Per Article: 35.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Accepted: 02/21/2023] [Indexed: 06/13/2023]
Abstract
The problem of antibiotic resistance is on the rise, with multidrug-resistant strains emerging even to the last resort antibiotics. The drug discovery process is often stalled by stringent cut-offs required for effective drug design. In such a scenario, it is prudent to delve into the varying mechanisms of resistance to existing antibiotics and target them to improve antibiotic efficacy. Nonantibiotic compounds called antibiotic adjuvants which target bacterial resistance can be used in combination with obsolete drugs for an improved therapeutic regime. The field of "antibiotic adjuvants" has gained significant traction in recent years where mechanisms other than β-lactamase inhibition have been explored. This review discusses the multitude of acquired and inherent resistance mechanisms employed by bacteria to resist antibiotic action. The major focus of this review is how to target these resistance mechanisms by the use of antibiotic adjuvants. Different types of direct acting and indirect resistance breakers are discussed including enzyme inhibitors, efflux pump inhibitors, inhibitors of teichoic acid synthesis, and other cellular processes. The multifaceted class of membrane-targeting compounds with poly pharmacological effects and the potential of host immune-modulating compounds have also been reviewed. We conclude with providing insights about the existing challenges preventing clinical translation of different classes of adjuvants, especially membrane-perturbing compounds, and a framework about the possible directions which can be pursued to fill this gap. Antibiotic-adjuvant combinatorial therapy indeed has immense potential to be used as an upcoming orthogonal strategy to conventional antibiotic discovery.
Collapse
Affiliation(s)
- Geetika Dhanda
- Antimicrobial
Research Laboratory, New Chemistry Unit and School of Advanced
Materials, Jawaharlal Nehru Centre for Advanced
Scientific Research (JNCASR), Jakkur, Bengaluru 560064, Karnataka, India
| | - Yash Acharya
- Antimicrobial
Research Laboratory, New Chemistry Unit and School of Advanced
Materials, Jawaharlal Nehru Centre for Advanced
Scientific Research (JNCASR), Jakkur, Bengaluru 560064, Karnataka, India
| | - Jayanta Haldar
- Antimicrobial
Research Laboratory, New Chemistry Unit and School of Advanced
Materials, Jawaharlal Nehru Centre for Advanced
Scientific Research (JNCASR), Jakkur, Bengaluru 560064, Karnataka, India
| |
Collapse
|
8
|
Greve JM, Cowan JA. Activity and Synergy of Cu-ATCUN Antimicrobial Peptides. Int J Mol Sci 2022; 23:ijms232214151. [PMID: 36430622 PMCID: PMC9692552 DOI: 10.3390/ijms232214151] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Revised: 11/08/2022] [Accepted: 11/11/2022] [Indexed: 11/19/2022] Open
Abstract
Antibiotic resistance demands innovative strategies and therapies. The pairs of antimicrobial peptides tested in this work show broad-spectrum synergy and are capable of interacting with diverse bacterial membranes. In most cases, the ATCUN motif enhanced the activity of peptides tested in combination. Our studies also show CP10A to be a multifaceted peptide, displaying both cell membrane and intracellular activity and acting as a chameleon, improving the activity of other peptides as needed. The results of the synergy experiments demonstrate the importance of varied modes of action and how these changes can affect the ability to combat pathogens, while also illustrating the value of the metal-binding domain in enhancing the activity of antimicrobial peptides in combination.
Collapse
|
9
|
Escobar‐Salom M, Torrens G, Jordana‐Lluch E, Oliver A, Juan C. Mammals' humoral immune proteins and peptides targeting the bacterial envelope: from natural protection to therapeutic applications against multidrug‐resistant
Gram
‐negatives. Biol Rev Camb Philos Soc 2022; 97:1005-1037. [PMID: 35043558 PMCID: PMC9304279 DOI: 10.1111/brv.12830] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Revised: 12/12/2021] [Accepted: 12/15/2021] [Indexed: 12/11/2022]
Abstract
Mammalian innate immunity employs several humoral ‘weapons’ that target the bacterial envelope. The threats posed by the multidrug‐resistant ‘ESKAPE’ Gram‐negative pathogens (Klebsiella pneumoniae, Acinetobacter baumannii, Pseudomonas aeruginosa, and Enterobacter spp.) are forcing researchers to explore new therapeutic options, including the use of these immune elements. Here we review bacterial envelope‐targeting (peptidoglycan and/or membrane‐targeting) proteins/peptides of the mammalian immune system that are most likely to have therapeutic applications. Firstly we discuss their general features and protective activity against ESKAPE Gram‐negatives in the host. We then gather, integrate, and discuss recent research on experimental therapeutics harnessing their bactericidal power, based on their exogenous administration and also on the discovery of bacterial and/or host targets that improve the performance of this endogenous immunity, as a novel therapeutic concept. We identify weak points and knowledge gaps in current research in this field and suggest areas for future work to obtain successful envelope‐targeting therapeutic options to tackle the challenge of antimicrobial resistance.
Collapse
Affiliation(s)
- María Escobar‐Salom
- Department of Microbiology University Hospital Son Espases‐Health Research Institute of the Balearic Islands (IdISBa) Carretera de Valldemossa 79 Palma Balearic Islands 07010 Spain
| | - Gabriel Torrens
- Department of Microbiology University Hospital Son Espases‐Health Research Institute of the Balearic Islands (IdISBa) Carretera de Valldemossa 79 Palma Balearic Islands 07010 Spain
| | - Elena Jordana‐Lluch
- Department of Microbiology University Hospital Son Espases‐Health Research Institute of the Balearic Islands (IdISBa) Carretera de Valldemossa 79 Palma Balearic Islands 07010 Spain
| | - Antonio Oliver
- Department of Microbiology University Hospital Son Espases‐Health Research Institute of the Balearic Islands (IdISBa) Carretera de Valldemossa 79 Palma Balearic Islands 07010 Spain
| | - Carlos Juan
- Department of Microbiology University Hospital Son Espases‐Health Research Institute of the Balearic Islands (IdISBa) Carretera de Valldemossa 79 Palma Balearic Islands 07010 Spain
| |
Collapse
|
10
|
Zhu Y, Hao W, Wang X, Ouyang J, Deng X, Yu H, Wang Y. Antimicrobial peptides, conventional antibiotics, and their synergistic utility for the treatment of drug-resistant infections. Med Res Rev 2022; 42:1377-1422. [PMID: 34984699 DOI: 10.1002/med.21879] [Citation(s) in RCA: 129] [Impact Index Per Article: 43.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2021] [Revised: 12/09/2021] [Accepted: 12/23/2021] [Indexed: 12/13/2022]
Abstract
Antimicrobial peptides (AMPs), also known as host defense peptides (HDPs), are important effector immune defense molecules in multicellular organisms. AMPs exert their antimicrobial activities through several mechanisms; thus far, induction of drug resistance through AMPs has been regarded as unlikely. Therefore, they have great potential as new generation antimicrobial agents. To date, more than 30 AMP-related drugs are in the clinical trial phase. In recent years, studies show that some AMPs and conventional antibiotics have synergistic effects. The combined use of AMPs and antibiotics can kill drug-resistant pathogens, prevent drug resistance, and significantly improve the therapeutic effects of antibiotics. In this review, we discuss the progress in synergistic studies on AMPs and conventional antibiotics. An overview of the current understanding of the functional scope of AMPs, ongoing clinical trials, and challenges in the development processes are also presented.
Collapse
Affiliation(s)
- Yiyun Zhu
- Department of Biopharmaceutical Sciences, College of Pharmaceutical Sciences, Soochow University, Suzhou, Jiangsu, China
| | - Weijing Hao
- Department of Biopharmaceutical Sciences, College of Pharmaceutical Sciences, Soochow University, Suzhou, Jiangsu, China
| | - Xia Wang
- Department of Biopharmaceutical Sciences, College of Pharmaceutical Sciences, Soochow University, Suzhou, Jiangsu, China
| | - Jianhong Ouyang
- Department of Biopharmaceutical Sciences, College of Pharmaceutical Sciences, Soochow University, Suzhou, Jiangsu, China
| | - Xinyi Deng
- Department of Biopharmaceutical Sciences, College of Pharmaceutical Sciences, Soochow University, Suzhou, Jiangsu, China
| | - Haining Yu
- Department of Bioscience and Biotechnology, Dalian University of Technology, Dalian, Liaoning, China
| | - Yipeng Wang
- Department of Biopharmaceutical Sciences, College of Pharmaceutical Sciences, Soochow University, Suzhou, Jiangsu, China
| |
Collapse
|
11
|
Porto WF. Virtual screening of peptides with high affinity for SARS-CoV-2 main protease. Comput Biol Med 2021; 133:104363. [PMID: 33862305 PMCID: PMC8018786 DOI: 10.1016/j.compbiomed.2021.104363] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2021] [Revised: 03/23/2021] [Accepted: 03/24/2021] [Indexed: 12/26/2022]
Abstract
The current pandemic of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has caused more than 2,000,000 deaths worldwide. Currently, vaccine development and drug repurposing have been the main strategies to find a COVID-19 treatment. However, the development of new drugs could be the solution if the main strategies fail. Here, a virtual screening of pentapeptides was applied in order to identify peptides with high affinity to SARS-CoV-2 main protease (Mpro). Over 70,000 peptides were screened employing a genetic algorithm that uses a docking score as the fitness function. The algorithm was coupled with a RESTful API to persist data and avoid redundancy. The docking exhaustiveness was adapted to the number of peptides in each virtual screening step, where the higher the number of peptides, the lower the docking exhaustiveness. Two potential peptides were selected (HHYWH and HYWWT), which have higher affinity to Mpro than to human proteases. Albeit preliminary, the data presented here provide some basis for the rational design of peptide-based drugs to treat COVID-19.
Collapse
|
12
|
Thompson Z, Greve JM, Cowan JA. Enhanced Synergism and Mechanism of Action Studies of Synthetic Antimicrobial Metallopeptides. ChemMedChem 2021; 16:2112-2120. [PMID: 33825350 DOI: 10.1002/cmdc.202100063] [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: 01/25/2021] [Indexed: 11/08/2022]
Abstract
Antimicrobial peptides (AMPs) are found throughout most kingdoms of life, are an important part of host immunity, and have been shown to act synergistically in various organisms to ameliorate bacterial infections. Herein, we report the synergistic behavior observed between two AMPs, Sub5 and CP10A, against E. coli. In addition, enhanced synergistic activity against E. coli and MRSA 43300 for two derivatives of Sub5, extended with the amino-terminal copper and nickel (ATCUN) binding motif, is observed when dosed together with CP10A, while displaying little cytotoxicity towards human dermal fibroblasts. All three combinations of peptides co-localized within bacterial cells as evidenced by fluorescence confocal microscopy. Investigations into the mechanism of synergy shows that all peptides indirectly damage DNA within cells, while only the ATCUN derivatives can oxidize phospholipids. Combinations of peptides were also shown to upregulate the concentration of reactive oxygen species within both E. coli and MRSA 43300. These results suggest that the production of reactive oxygen species is an important aspect mechanistically and further highlights the potential of these metallopeptides to aid in the treatment of antibiotic-resistant infections.
Collapse
Affiliation(s)
- Zechariah Thompson
- Chemistry and Biochemistry, The Ohio State University, Evans Laboratory of Chemistry 100, West 18th Avenue, Columbus, Ohio, 43210, USA
| | - Jenna M Greve
- Chemistry and Biochemistry, The Ohio State University, Evans Laboratory of Chemistry 100, West 18th Avenue, Columbus, Ohio, 43210, USA
| | - James Allan Cowan
- Chemistry and Biochemistry, The Ohio State University, Evans Laboratory of Chemistry 100, West 18th Avenue, Columbus, Ohio, 43210, USA
| |
Collapse
|
13
|
Hobson C, Chan AN, Wright GD. The Antibiotic Resistome: A Guide for the Discovery of Natural Products as Antimicrobial Agents. Chem Rev 2021; 121:3464-3494. [PMID: 33606500 DOI: 10.1021/acs.chemrev.0c01214] [Citation(s) in RCA: 105] [Impact Index Per Article: 26.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
The use of life-saving antibiotics has long been plagued by the ability of pathogenic bacteria to acquire and develop an array of antibiotic resistance mechanisms. The sum of these resistance mechanisms, the antibiotic resistome, is a formidable threat to antibiotic discovery, development, and use. The study and understanding of the molecular mechanisms in the resistome provide the basis for traditional approaches to combat resistance, including semisynthetic modification of naturally occurring antibiotic scaffolds, the development of adjuvant therapies that overcome resistance mechanisms, and the total synthesis of new antibiotics and their analogues. Using two major classes of antibiotics, the aminoglycosides and tetracyclines as case studies, we review the success and limitations of these strategies when used to combat the many forms of resistance that have emerged toward natural product-based antibiotics specifically. Furthermore, we discuss the use of the resistome as a guide for the genomics-driven discovery of novel antimicrobials, which are essential to combat the growing number of emerging pathogens that are resistant to even the newest approved therapies.
Collapse
Affiliation(s)
- Christian Hobson
- Department of Biochemistry and Biomedical Sciences, Michael G. DeGroote Institute for Infectious Disease Research, McMaster University, 1280 Main Street West, Hamilton, Ontario L8S 4L8, Canada
| | - Andrew N Chan
- Department of Biochemistry and Biomedical Sciences, Michael G. DeGroote Institute for Infectious Disease Research, McMaster University, 1280 Main Street West, Hamilton, Ontario L8S 4L8, Canada
| | - Gerard D Wright
- Department of Biochemistry and Biomedical Sciences, Michael G. DeGroote Institute for Infectious Disease Research, McMaster University, 1280 Main Street West, Hamilton, Ontario L8S 4L8, Canada
| |
Collapse
|
14
|
Jouybari MA, Ahanjan M, Mirzaei B, Goli HR. Role of aminoglycoside-modifying enzymes and 16S rRNA methylase (ArmA) in resistance of Acinetobacter baumannii clinical isolates against aminoglycosides. Rev Soc Bras Med Trop 2021; 54:e05992020. [PMID: 33533819 PMCID: PMC7849326 DOI: 10.1590/0037-8682-0599-2020] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Accepted: 11/12/2020] [Indexed: 11/22/2022] Open
Abstract
INTRODUCTION: This study aimed to determine the role of genes encoding aminoglycoside-modifying enzymes (AMEs) and 16S rRNA methylase (ArmA) in Acinetobacter baumannii clinical isolates. METHODS: We collected 100 clinical isolates of A. baumannii and identified and confirmed them using microbiological tests and assessment of the OXA-51 gene. Antibiotic susceptibility testing was carried out using disk agar diffusion and micro-broth dilution methods. The presence of AME genes and ArmA was detected by PCR and multiplex PCR. RESULTS: The most and least effective antibiotics in this study were netilmicin and ciprofloxacin with 68% and 100% resistance rates, respectively. According to the minimum inhibitory concentration test, 94% of the isolates were resistant to gentamicin, tobramycin, and streptomycin, while the highest susceptibility (20%) was observed against netilmicin. The proportion of strains harboring the aminoglycoside resistance genes was as follows: APH(3′)-VIa (aphA6) (77%), ANT(2”)-Ia (aadB) (73%), ANT(3”)-Ia (aadA1) (33%), AAC(6′)-Ib (aacA4) (33%), ArmA (22%), and AAC(3)-IIa (aacC2) (19%). Among the 22 gene profiles detected in this study, the most prevalent profiles included APH(3′)-VIa + ANT(2”)-Ia (39 isolates, 100% of which were kanamycin-resistant), and AAC(3)-IIa + AAC(6′)-Ib + ANT(3”)-Ia + APH(3′)-VIa + ANT(2”)-Ia (14 isolates, all of which were resistant to gentamicin, kanamycin, and streptomycin). CONCLUSIONS: High minimum inhibitory concentration of aminoglycosides in isolates with the simultaneous presence of AME- and ArmA-encoding genes indicated the importance of these genes in resistance to aminoglycosides. However, control of their spread could be effective in the treatment of infections caused by A. baumannii.
Collapse
Affiliation(s)
- Maryam Asadi Jouybari
- Mazandaran University of Medical Sciences, Faculty of Medicine, Molecular and Cell Biology Research Centre, Sari, Iran.,Mazandaran University of Medical Sciences, Faculty of Medicine, Department of Medical Microbiology and Virology, Sari, Iran
| | - Mohammad Ahanjan
- Mazandaran University of Medical Sciences, Faculty of Medicine, Molecular and Cell Biology Research Centre, Sari, Iran.,Mazandaran University of Medical Sciences, Faculty of Medicine, Department of Medical Microbiology and Virology, Sari, Iran
| | - Bahman Mirzaei
- Zanjan University of Medical Sciences, School of Medicine, Department of Medical Microbiology and Virology, Zanjan, Iran
| | - Hamid Reza Goli
- Mazandaran University of Medical Sciences, Faculty of Medicine, Molecular and Cell Biology Research Centre, Sari, Iran.,Mazandaran University of Medical Sciences, Faculty of Medicine, Department of Medical Microbiology and Virology, Sari, Iran
| |
Collapse
|
15
|
Smirnova MP, Kolodkin NI, Kolobov AA, Afonin VG, Afonina IV, Stefanenko LI, Shpen' VM, Shamova OV, Kolobov AA. Indolicidin analogs with broad-spectrum antimicrobial activity and low hemolytic activity. Peptides 2020; 132:170356. [PMID: 32593681 DOI: 10.1016/j.peptides.2020.170356] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Revised: 06/23/2020] [Accepted: 06/24/2020] [Indexed: 12/19/2022]
Abstract
To create a broad-spectrum peptide biocide, we synthesized 45 analogs of antimicrobial peptide indolicidin (H-Ile-Leu-Pro-Trp-Lys-Trp-Pro-Trp-Trp-Pro-Trp-Arg-Arg-NH2). Among them the peptides H-Ile-Leu-Pro-(2-Me)Phe-Lys-(2-Me)Phe-Pro-(2-Me)Phe-(2-Me)Phe-Pro-(2-Me)Phe-Arg-Arg-NH2 and HN2-(CH2)10-Ile-Leu-Pro-D-Phe-Lys-D-Phe-Pro-D-Phe-D-Phe-Pro-D-Phe-Arg-Arg-NH2 have the broadest spectrum of antimicrobial activity and the lowest hemolytic activity. They are active against all 11 tested strains of Gram-positive bacteria, Gram-negative bacteria and fungi with MIC50 from 0.9 to 6.1 μg/ml (0.5 to 3.2 μM), being up to 3 times more active than indolicidin, and are at least 1.8 times less hemolytically active than indolicidin (reached the detection limit). These peptides are patented and could be used for further drug development as antimicrobials.
Collapse
Affiliation(s)
- M P Smirnova
- Institute of Highly Pure Biopreparations, Saint-Petersburg, Russia; Verta Ltd., Saint-Petersburg, Russia
| | - N I Kolodkin
- Institute of Highly Pure Biopreparations, Saint-Petersburg, Russia; Verta Ltd., Saint-Petersburg, Russia
| | - A A Kolobov
- Institute of Highly Pure Biopreparations, Saint-Petersburg, Russia; Verta Ltd., Saint-Petersburg, Russia
| | - V G Afonin
- Institute of Highly Pure Biopreparations, Saint-Petersburg, Russia; Verta Ltd., Saint-Petersburg, Russia
| | - I V Afonina
- Institute of Highly Pure Biopreparations, Saint-Petersburg, Russia; Verta Ltd., Saint-Petersburg, Russia
| | - L I Stefanenko
- Institute of Highly Pure Biopreparations, Saint-Petersburg, Russia; Verta Ltd., Saint-Petersburg, Russia
| | - V M Shpen'
- Institute of Highly Pure Biopreparations, Saint-Petersburg, Russia; Verta Ltd., Saint-Petersburg, Russia
| | - O V Shamova
- Institute of Experimental Medicine, Saint-Petersburg, Russia
| | - A A Kolobov
- Institute of Highly Pure Biopreparations, Saint-Petersburg, Russia; Verta Ltd., Saint-Petersburg, Russia.
| |
Collapse
|
16
|
Laws M, Shaaban A, Rahman KM. Antibiotic resistance breakers: current approaches and future directions. FEMS Microbiol Rev 2020; 43:490-516. [PMID: 31150547 PMCID: PMC6736374 DOI: 10.1093/femsre/fuz014] [Citation(s) in RCA: 177] [Impact Index Per Article: 35.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2019] [Accepted: 05/30/2019] [Indexed: 12/15/2022] Open
Abstract
Infections of antibiotic-resistant pathogens pose an ever-increasing threat to mankind. The investigation of novel approaches for tackling the antimicrobial resistance crisis must be part of any global response to this problem if an untimely reversion to the pre-penicillin era of medicine is to be avoided. One such promising avenue of research involves so-called antibiotic resistance breakers (ARBs), capable of re-sensitising resistant bacteria to antibiotics. Although some ARBs have previously been employed in the clinical setting, such as the β-lactam inhibitors, we posit that the broader field of ARB research can yet yield a greater diversity of more effective therapeutic agents than have been previously achieved. This review introduces the area of ARB research, summarises the current state of ARB development with emphasis on the various major classes of ARBs currently being investigated and their modes of action, and offers a perspective on the future direction of the field.
Collapse
Affiliation(s)
- Mark Laws
- Institute of Pharmaceutical Sciences, School of Cancer and Pharmaceutical Sciences, King's College London, Franklin-Wilkins Building, 150 Stamford Street, London, SE1 9NH
| | - Ali Shaaban
- Institute of Pharmaceutical Sciences, School of Cancer and Pharmaceutical Sciences, King's College London, Franklin-Wilkins Building, 150 Stamford Street, London, SE1 9NH
| | - Khondaker Miraz Rahman
- Institute of Pharmaceutical Sciences, School of Cancer and Pharmaceutical Sciences, King's College London, Franklin-Wilkins Building, 150 Stamford Street, London, SE1 9NH
| |
Collapse
|
17
|
Ruden S, Rieder A, Chis Ster I, Schwartz T, Mikut R, Hilpert K. Synergy Pattern of Short Cationic Antimicrobial Peptides Against Multidrug-Resistant Pseudomonas aeruginosa. Front Microbiol 2019; 10:2740. [PMID: 31849888 PMCID: PMC6901909 DOI: 10.3389/fmicb.2019.02740] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2019] [Accepted: 11/11/2019] [Indexed: 12/18/2022] Open
Abstract
With the rise of various multidrug-resistant (MDR) pathogenic bacteria, worldwide health care is under pressure to respond. Conventional antibiotics are failing and the development of novel classes and alternative strategies is a major priority. Antimicrobial peptides (AMPs) cannot only kill MDR bacteria, but also can be used synergistically with conventional antibiotics. We selected 30 short AMPs from different origins and measured their synergy in combination with polymyxin B, piperacillin, ceftazidime, cefepime, meropenem, imipenem, tetracycline, erythromycin, kanamycin, tobramycin, amikacin, gentamycin, and ciprofloxacin. In total, 403 unique combinations were tested against an MDR Pseudomonas aeruginosa isolate (PA910). As a measure of the synergistic effects, fractional inhibitory concentrations (FICs) were determined using microdilution assays with FICs ranges between 0.25 and 2. A high number of combinations between peptides and polymyxin B, erythromycin, and tetracycline were found to be synergistic. Novel variants of indolicidin also showed a high frequency in synergist interaction. Single amino acid substitutions within the peptides can have a very strong effect on the ability to synergize, making it possible to optimize future drugs toward synergistic interaction.
Collapse
Affiliation(s)
- Serge Ruden
- Institute of Biological Interfaces, Karlsruhe Institute of Technology, Karlsruhe, Germany.,Institute of Functional Interfaces, Karlsruhe Institute of Technology, Karlsruhe, Germany
| | - Annika Rieder
- Institute of Functional Interfaces, Karlsruhe Institute of Technology, Karlsruhe, Germany
| | - Irina Chis Ster
- Institute of Infection and Immunity, St George's, University of London, London, United Kingdom
| | - Thomas Schwartz
- Institute of Functional Interfaces, Karlsruhe Institute of Technology, Karlsruhe, Germany
| | - Ralf Mikut
- Institute for Automation and Applied Informatics, Karlsruhe Institute of Technology, Karlsruhe, Germany
| | - Kai Hilpert
- Institute of Functional Interfaces, Karlsruhe Institute of Technology, Karlsruhe, Germany.,Institute of Infection and Immunity, St George's, University of London, London, United Kingdom.,Institute of Microstructure Technology, Karlsruhe Institute of Technology, Karlsruhe, Germany
| |
Collapse
|
18
|
Verma A, Nayek A, Kumar A, Singh R, Salotra P. Elucidation of role of an acetyltransferase like protein in paromomycin resistance in Leishmania donovani using in silico and in vitro approaches. J Biomol Struct Dyn 2019; 38:4449-4460. [PMID: 31625467 DOI: 10.1080/07391102.2019.1682674] [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: 10/25/2022]
Abstract
Paromomycin, an aminoglycoside antibiotic, is an effective treatment for VL (visceral leishmaniasis) in India. The modification of aminoglycoside antibiotics by enzymes such as aminoglycoside acetyltransferases is the predominant mechanism of resistance to antibiotics in bacterial system. In the present study, we identified and characterized LdATLP (an acetyltransferase-like protein) and elucidated its role in paromomycin resistance in Leishmania donovani. Gene encoding LdATLP was consistently up-regulated (>2fold) in three distinct paromomycin resistant in comparison with sensitive parasites, although the gene sequence was identical in the two. In silico analysis revealed that LdATLP consisted of conserved GNAT (GCN5-related N-Acetyltransferase) domain which is characteristic of aminoglycoside N-acetyltransferases. Evolutionary relationship among LdATLP of Leishmania and aminoglycoside acetyltransferases of bacteria was established by phylogenetic analysis. The 3D structure of LdATLP, predicted by ab-initio modeling, constituted 6 α-helices and 6 β-sheets. A few residues, such as R175, R177, E196, R197, V198, V200, K202, R205, C206, D208, G210, R211, R215, A234, S237, S238, K239, D240, F241 and Y242 of GNAT domain were predicted to be present at active site. Molecular docking of LdATLP with paromomycin or indolicidin (broad spectrum inhibitor of aminoglycoside modifying enzymes), followed by molecular dynamics simulation of docked complex suggested that both paromomycin and indolicidin bind to LdATLP with comparable free energy of binding. In vitro studies revealed that in the presence of indolicidin, paromomycin resistant parasites exhibited reversion of phenotype into sensitive parasites with marked increase in paromomycin susceptibility, suggesting the role of LdATLP in paromomycin resistance.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Aditya Verma
- ICMR-National Institute of Pathology, Safdarjung Hospital Campus, New Delhi, India.,Faculty of Health and Biological Sciences, Symbiosis International (Deemed University), Pune, India
| | - Arnab Nayek
- ICMR-National Institute of Pathology, Safdarjung Hospital Campus, New Delhi, India
| | - Amit Kumar
- ICMR Computational Genomics Center, Division of ISRM, ICMR, New Delhi, India
| | - Ruchi Singh
- ICMR-National Institute of Pathology, Safdarjung Hospital Campus, New Delhi, India
| | - Poonam Salotra
- ICMR-National Institute of Pathology, Safdarjung Hospital Campus, New Delhi, India
| |
Collapse
|
19
|
Abstract
Many antibiotics available in the clinic today directly inhibit bacterial translation. Despite the past success of such drugs, their efficacy is diminishing with the spread of antibiotic resistance. Through the use of ribosomal modifications, ribosomal protection proteins, translation elongation factors and mistranslation, many pathogens are able to establish resistance to common therapeutics. However, current efforts in drug discovery are focused on overcoming these obstacles through the modification or discovery of new treatment options. Here, we provide an overview for common mechanisms of resistance to translation-targeting drugs and summarize several important breakthroughs in recent drug development.
Collapse
Affiliation(s)
- Anne Witzky
- 1 Department of Molecular Genetics, Ohio State University , Columbus, OH 43210 , USA.,2 Center for RNA Biology, Ohio State University , Columbus, OH 43210 , USA
| | - Rodney Tollerson
- 2 Center for RNA Biology, Ohio State University , Columbus, OH 43210 , USA.,3 Department of Microbiology, Ohio State University , Columbus, OH 43210 , USA
| | - Michael Ibba
- 2 Center for RNA Biology, Ohio State University , Columbus, OH 43210 , USA.,3 Department of Microbiology, Ohio State University , Columbus, OH 43210 , USA
| |
Collapse
|
20
|
Drug combinations: a strategy to extend the life of antibiotics in the 21st century. Nat Rev Microbiol 2019; 17:141-155. [PMID: 30683887 DOI: 10.1038/s41579-018-0141-x] [Citation(s) in RCA: 530] [Impact Index Per Article: 88.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2018] [Accepted: 11/22/2018] [Indexed: 01/03/2023]
Abstract
Antimicrobial resistance threatens a resurgence of life-threatening bacterial infections and the potential demise of many aspects of modern medicine. Despite intensive drug discovery efforts, no new classes of antibiotics have been developed into new medicines for decades, in large part owing to the stringent chemical, biological and pharmacological requisites for effective antibiotic drugs. Combinations of antibiotics and of antibiotics with non-antibiotic activity-enhancing compounds offer a productive strategy to address the widespread emergence of antibiotic-resistant strains. In this Review, we outline a theoretical and practical framework for the development of effective antibiotic combinations.
Collapse
|
21
|
Wang J, Dou X, Song J, Lyu Y, Zhu X, Xu L, Li W, Shan A. Antimicrobial peptides: Promising alternatives in the post feeding antibiotic era. Med Res Rev 2018; 39:831-859. [PMID: 30353555 DOI: 10.1002/med.21542] [Citation(s) in RCA: 359] [Impact Index Per Article: 51.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2018] [Revised: 09/11/2018] [Accepted: 09/12/2018] [Indexed: 12/15/2022]
Abstract
Antimicrobial peptides (AMPs), critical components of the innate immune system, are widely distributed throughout the animal and plant kingdoms. They can protect against a broad array of infection-causing agents, such as bacteria, fungi, parasites, viruses, and tumor cells, and also exhibit immunomodulatory activity. AMPs exert antimicrobial activities primarily through mechanisms involving membrane disruption, so they have a lower likelihood of inducing drug resistance. Extensive studies on the structure-activity relationship have revealed that net charge, hydrophobicity, and amphipathicity are the most important physicochemical and structural determinants endowing AMPs with antimicrobial potency and cell selectivity. This review summarizes the recent advances in AMPs development with respect to characteristics, structure-activity relationships, functions, antimicrobial mechanisms, expression regulation, and applications in food, medicine, and animals.
Collapse
Affiliation(s)
- Jiajun Wang
- Institute of Animal Nutrition, Department of Animal Nutrition, Northeast Agricultural University, Harbin, China
| | - Xiujing Dou
- Institute of Animal Nutrition, Department of Animal Nutrition, Northeast Agricultural University, Harbin, China
| | - Jing Song
- Institute of Animal Nutrition, Department of Animal Nutrition, Northeast Agricultural University, Harbin, China
| | - Yinfeng Lyu
- Institute of Animal Nutrition, Department of Animal Nutrition, Northeast Agricultural University, Harbin, China
| | - Xin Zhu
- Institute of Animal Nutrition, Department of Animal Nutrition, Northeast Agricultural University, Harbin, China
| | - Lin Xu
- Institute of Animal Nutrition, Department of Animal Nutrition, Northeast Agricultural University, Harbin, China
| | - Weizhong Li
- Institute of Animal Nutrition, Department of Animal Nutrition, Northeast Agricultural University, Harbin, China
| | - Anshan Shan
- Institute of Animal Nutrition, Department of Animal Nutrition, Northeast Agricultural University, Harbin, China
| |
Collapse
|
22
|
Parulekar RS, Sonawane KD. Insights into the antibiotic resistance and inhibition mechanism of aminoglycoside phosphotransferase from
Bacillus cereus
: In silico and in vitro perspective. J Cell Biochem 2018; 119:9444-9461. [DOI: 10.1002/jcb.27261] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2018] [Accepted: 06/22/2018] [Indexed: 01/13/2023]
Affiliation(s)
| | - Kailas Dashrath Sonawane
- Department of Microbiology Shivaji University Kolhapur Maharashtra India
- Structural Bioinformatics Unit, Department of Biochemistry Shivaji University Kolhapur Maharashtra India
| |
Collapse
|
23
|
Zárate SG, Claure MLDLC, Benito-Arenas R, Revuelta J, Santana AG, Bastida A. Overcoming Aminoglycoside Enzymatic Resistance: Design of Novel Antibiotics and Inhibitors. Molecules 2018; 23:molecules23020284. [PMID: 29385736 PMCID: PMC6017855 DOI: 10.3390/molecules23020284] [Citation(s) in RCA: 65] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2017] [Revised: 01/12/2018] [Accepted: 01/26/2018] [Indexed: 11/17/2022] Open
Abstract
Resistance to aminoglycoside antibiotics has had a profound impact on clinical practice. Despite their powerful bactericidal activity, aminoglycosides were one of the first groups of antibiotics to meet the challenge of resistance. The most prevalent source of clinically relevant resistance against these therapeutics is conferred by the enzymatic modification of the antibiotic. Therefore, a deeper knowledge of the aminoglycoside-modifying enzymes and their interactions with the antibiotics and solvent is of paramount importance in order to facilitate the design of more effective and potent inhibitors and/or novel semisynthetic aminoglycosides that are not susceptible to modifying enzymes.
Collapse
Affiliation(s)
- Sandra G. Zárate
- Facultad de Tecnología-Carrera de Ingeniería Química, Universidad Mayor Real y Pontificia de San Francisco Xavier de Chuquisaca, Regimiento Campos 180, Casilla 60-B, Sucre, Bolivia;
| | - M. Luisa De la Cruz Claure
- Facultad de Ciencias Químico Farmacéuticas y Bioquímicas, Universidad Mayor Real y Pontificia de San Francisco Xavier de Chuquisaca, Dalence 51, Casilla 497, Sucre, Bolivia;
| | - Raúl Benito-Arenas
- Departmento de Química Bio-Orgánica, Instituto de Química Orgánica General (CSIC), Juan de la Cierva 3, 28006 Madrid, Spain; (R.B.-A.); (J.R.)
| | - Julia Revuelta
- Departmento de Química Bio-Orgánica, Instituto de Química Orgánica General (CSIC), Juan de la Cierva 3, 28006 Madrid, Spain; (R.B.-A.); (J.R.)
| | - Andrés G. Santana
- Departmento de Química Bio-Orgánica, Instituto de Química Orgánica General (CSIC), Juan de la Cierva 3, 28006 Madrid, Spain; (R.B.-A.); (J.R.)
- Correspondence: (A.G.S.); (A.B.); Tel: +34-915-612-800 (A.B.)
| | - Agatha Bastida
- Departmento de Química Bio-Orgánica, Instituto de Química Orgánica General (CSIC), Juan de la Cierva 3, 28006 Madrid, Spain; (R.B.-A.); (J.R.)
- Correspondence: (A.G.S.); (A.B.); Tel: +34-915-612-800 (A.B.)
| |
Collapse
|
24
|
Alexander JL, Yu Z, Cowan JA. Amino Terminal Copper and Nickel Binding Motif Derivatives of Ovispirin-3 Display Increased Antimicrobial Activity via Lipid Oxidation. J Med Chem 2017; 60:10047-10055. [PMID: 29172482 DOI: 10.1021/acs.jmedchem.7b01117] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Affiliation(s)
- Jessica L Alexander
- Evans Laboratory of Chemistry, The Ohio State University, 100 West 18th Avenue, Columbus, Ohio 43210, United States
| | - Zhen Yu
- Evans Laboratory of Chemistry, The Ohio State University, 100 West 18th Avenue, Columbus, Ohio 43210, United States
| | - J. A Cowan
- Evans Laboratory of Chemistry, The Ohio State University, 100 West 18th Avenue, Columbus, Ohio 43210, United States
| |
Collapse
|
25
|
Deshayes S, Xian W, Schmidt NW, Kordbacheh S, Lieng J, Wang J, Zarmer S, Germain SS, Voyen L, Thulin J, Wong GCL, Kasko AM. Designing Hybrid Antibiotic Peptide Conjugates To Cross Bacterial Membranes. Bioconjug Chem 2017; 28:793-804. [DOI: 10.1021/acs.bioconjchem.6b00725] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Affiliation(s)
| | | | - Nathan W. Schmidt
- Department
of Pharmaceutical Chemistry, University of California, San Francisco, 555 Mission Bay Boulevard South, San Francisco, California 94158, United States
| | | | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Wright GD. Antibiotic Adjuvants: Rescuing Antibiotics from Resistance. Trends Microbiol 2016; 24:862-871. [DOI: 10.1016/j.tim.2016.06.009] [Citation(s) in RCA: 388] [Impact Index Per Article: 43.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2016] [Revised: 06/20/2016] [Accepted: 06/27/2016] [Indexed: 10/21/2022]
|
27
|
Power BH, Smith N, Downer B, Alisaraie L. Insight into the mechanism of chemical modification of antibacterial agents by antibiotic resistance enzymeO-phosphotransferase-IIIA. Chem Biol Drug Des 2016; 89:84-97. [DOI: 10.1111/cbdd.12835] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2016] [Revised: 07/22/2016] [Accepted: 07/25/2016] [Indexed: 11/30/2022]
Affiliation(s)
- Blake Hollett Power
- School of Pharmacy; Memorial University of Newfoundland; St. John's Newfoundland Canada
| | - Nathan Smith
- School of Pharmacy; Memorial University of Newfoundland; St. John's Newfoundland Canada
| | - Brandon Downer
- School of Pharmacy; Memorial University of Newfoundland; St. John's Newfoundland Canada
| | - Laleh Alisaraie
- School of Pharmacy; Memorial University of Newfoundland; St. John's Newfoundland Canada
- Department of Chemistry; Memorial University of Newfoundland; St. John's Newfoundland Canada
| |
Collapse
|
28
|
Willby MJ, Green KD, Gajadeera CS, Hou C, Tsodikov OV, Posey JE, Garneau-Tsodikova S. Potent Inhibitors of Acetyltransferase Eis Overcome Kanamycin Resistance in Mycobacterium tuberculosis. ACS Chem Biol 2016; 11:1639-46. [PMID: 27010218 DOI: 10.1021/acschembio.6b00110] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
A major cause of tuberculosis (TB) resistance to the aminoglycoside kanamycin (KAN) is the Mycobacterium tuberculosis (Mtb) acetyltransferase Eis. Upregulation of this enzyme is responsible for inactivation of KAN through acetylation of its amino groups. A 123 000-compound high-throughput screen (HTS) yielded several small-molecule Eis inhibitors that share an isothiazole S,S-dioxide heterocyclic core. These were investigated for their structure-activity relationships. Crystal structures of Eis in complex with two potent inhibitors show that these molecules are bound in the conformationally adaptable aminoglycoside binding site of the enzyme, thereby obstructing binding of KAN for acetylation. Importantly, we demonstrate that several Eis inhibitors, when used in combination with KAN against resistant Mtb, efficiently overcome KAN resistance. This approach paves the way toward development of novel combination therapies against aminoglycoside-resistant TB.
Collapse
Affiliation(s)
- Melisa J. Willby
- Division of Tuberculosis
Elimination, National Center for HIV/AIDS, Viral Hepatitis, STD, and
TB Prevention, Centers for Disease Control and Prevention, Atlanta, Georgia 30329, United States
| | - Keith D. Green
- Department
of Pharmaceutical Sciences, University of Kentucky, Lexington, Kentucky 40536-0596, United States
| | - Chathurada S. Gajadeera
- Department
of Pharmaceutical Sciences, University of Kentucky, Lexington, Kentucky 40536-0596, United States
| | - Caixia Hou
- Department
of Pharmaceutical Sciences, University of Kentucky, Lexington, Kentucky 40536-0596, United States
| | - Oleg V. Tsodikov
- Department
of Pharmaceutical Sciences, University of Kentucky, Lexington, Kentucky 40536-0596, United States
| | - James E. Posey
- Division of Tuberculosis
Elimination, National Center for HIV/AIDS, Viral Hepatitis, STD, and
TB Prevention, Centers for Disease Control and Prevention, Atlanta, Georgia 30329, United States
| | - Sylvie Garneau-Tsodikova
- Department
of Pharmaceutical Sciences, University of Kentucky, Lexington, Kentucky 40536-0596, United States
| |
Collapse
|
29
|
Liu X, Cao R, Wang S, Jia J, Fei H. Amphipathicity Determines Different Cytotoxic Mechanisms of Lysine- or Arginine-Rich Cationic Hydrophobic Peptides in Cancer Cells. J Med Chem 2016; 59:5238-47. [DOI: 10.1021/acs.jmedchem.5b02016] [Citation(s) in RCA: 67] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Affiliation(s)
- Xiaoli Liu
- CAS Key Laboratory of Nano-Bio
Interface, Division of Nanobiomedicine, Suzhou Institute of Nano-Tech
and Nano-Bionics, Chinese Academy of Sciences, 398 Ruoshui Road, Suzhou Industrial Park, Suzhou, Jiangsu 215123, P. R. China
- School of Life Science, Shanghai University, 99 Shangda Road, Baoshan District, Shanghai 200444, P. R. China
| | - Rui Cao
- CAS Key Laboratory of Nano-Bio
Interface, Division of Nanobiomedicine, Suzhou Institute of Nano-Tech
and Nano-Bionics, Chinese Academy of Sciences, 398 Ruoshui Road, Suzhou Industrial Park, Suzhou, Jiangsu 215123, P. R. China
| | - Sha Wang
- CAS Key Laboratory of Nano-Bio
Interface, Division of Nanobiomedicine, Suzhou Institute of Nano-Tech
and Nano-Bionics, Chinese Academy of Sciences, 398 Ruoshui Road, Suzhou Industrial Park, Suzhou, Jiangsu 215123, P. R. China
- School of Pharmacy, Xi’an Jiaotong University, Xi’an, Shaanxi 710061, P. R. China
| | - Junli Jia
- CAS Key Laboratory of Nano-Bio
Interface, Division of Nanobiomedicine, Suzhou Institute of Nano-Tech
and Nano-Bionics, Chinese Academy of Sciences, 398 Ruoshui Road, Suzhou Industrial Park, Suzhou, Jiangsu 215123, P. R. China
| | - Hao Fei
- CAS Key Laboratory of Nano-Bio
Interface, Division of Nanobiomedicine, Suzhou Institute of Nano-Tech
and Nano-Bionics, Chinese Academy of Sciences, 398 Ruoshui Road, Suzhou Industrial Park, Suzhou, Jiangsu 215123, P. R. China
| |
Collapse
|
30
|
Chiem K, Jani S, Fuentes B, Lin DL, Rasche ME, Tolmasky ME. Identification of an Inhibitor of the Aminoglycoside 6'- N-Acetyltransferase type Ib [AAC(6')-Ib] by Glide Molecular Docking. MEDCHEMCOMM 2016; 7:184-189. [PMID: 26973774 PMCID: PMC4784703 DOI: 10.1039/c5md00316d] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
The aminoglycoside 6'-N-acetyltransferase type Ib, AAC(6')-Ib, confers resistance to clinically relevant aminoglycosides and is the most widely distributed enzyme among AAC(6')-I-producing Gram-negative pathogens. An alternative to counter the action of this enzyme is the development of inhibitors. Glide is a computational strategy for rapidly docking ligands to protein sites and estimating their binding affinities. We docked a collection of 280,000 compounds from 7 sub-libraries of the Chembridge library as ligands to the aminoglycoside binding site of AAC(6')-Ib. We identified a compound, 1-[3-(2-aminoethyl)benzyl]-3-(piperidin-1-ylmethyl)pyrrolidin-3-ol (compound 1), that inhibited the acetylation of aminoglycosides in vitro with IC50 values of 39.7 and 34.9 µM when the aminoglycoside substrates assayed were kanamycin A or amikacin, respectively. The growth of an amikacin-resistant Acinetobacter baumannii clinical strain was inhibited in the presence of a combination of amikacin and compound 1.
Collapse
Affiliation(s)
- Kevin Chiem
- Center for Applied Biotechnology Studies, Department of Biological Science, College of Natural Sciences and Mathematics, California State University Fullerton, Fullerton, CA 92834-6850, United States
| | - Saumya Jani
- Center for Applied Biotechnology Studies, Department of Biological Science, College of Natural Sciences and Mathematics, California State University Fullerton, Fullerton, CA 92834-6850, United States
| | - Brooke Fuentes
- Center for Applied Biotechnology Studies, Department of Biological Science, College of Natural Sciences and Mathematics, California State University Fullerton, Fullerton, CA 92834-6850, United States
| | - David L. Lin
- Center for Applied Biotechnology Studies, Department of Biological Science, College of Natural Sciences and Mathematics, California State University Fullerton, Fullerton, CA 92834-6850, United States
| | - Madeline E. Rasche
- Center for Applied Biotechnology Studies, Department of Chemistry and Biochemistry, College of Natural Sciences and Mathematics, California State University Fullerton, Fullerton, CA 92834-6850, United States
| | - Marcelo E. Tolmasky
- Center for Applied Biotechnology Studies, Department of Biological Science, College of Natural Sciences and Mathematics, California State University Fullerton, Fullerton, CA 92834-6850, United States
| |
Collapse
|
31
|
Sur A, Pradhan B, Banerjee A, Aich P. Immune activation efficacy of indolicidin is enhanced upon conjugation with carbon nanotubes and gold nanoparticles. PLoS One 2015; 10:e0123905. [PMID: 25876153 PMCID: PMC4398554 DOI: 10.1371/journal.pone.0123905] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2014] [Accepted: 03/02/2015] [Indexed: 12/28/2022] Open
Abstract
Antibiotic resistance is concern of today's world. Search for alternative molecules, for treatment and immune stimulation, remains at the forefront. One such group of biomolecules with promise, along the line of immune stimulation or therapy, is host defense peptide (HDP). These molecules, however, are required at a higher dose to be effective which leads to high cost. To alleviate such problems, an aid can be used to achieve similar efficacy but at a smaller effective dose of the immune stimulant. We hypothesised that by conjugating HDPs with carbon nanotubes and/or gold nanoparticles, it would be possible to stimulate a protective immune response in host system at a lower dosage of HDP. In this report, we characterized, using biophysical methodologies, conjugation of Indolicidin, as a representative of HDP. We further established efficacy of peptide-nanomaterial conjugates in activating innate immunity and protecting against pathogen infection in vitro at a significantly small dose.
Collapse
Affiliation(s)
- Abhinav Sur
- School of Biological Sciences, National Institute of Science Education and Research, Bhubaneswar, India
| | - Biswaranjan Pradhan
- School of Biological Sciences, National Institute of Science Education and Research, Bhubaneswar, India
| | - Arka Banerjee
- School of Biological Sciences, National Institute of Science Education and Research, Bhubaneswar, India
| | - Palok Aich
- School of Biological Sciences, National Institute of Science Education and Research, Bhubaneswar, India
- * E-mail:
| |
Collapse
|
32
|
Hirsch DR, Cox G, D'Erasmo MP, Shakya T, Meck C, Mohd N, Wright GD, Murelli RP. Inhibition of the ANT(2")-Ia resistance enzyme and rescue of aminoglycoside antibiotic activity by synthetic α-hydroxytropolones. Bioorg Med Chem Lett 2014; 24:4943-7. [PMID: 25283553 PMCID: PMC4798002 DOI: 10.1016/j.bmcl.2014.09.037] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2014] [Revised: 09/09/2014] [Accepted: 09/11/2014] [Indexed: 11/17/2022]
Abstract
Aminoglycoside-2"-O-nucleotidyltransferase ANT(2")-Ia is an aminoglycoside resistance enzyme prevalent among Gram-negative bacteria, and is one of the most common determinants of enzyme-dependant aminoglycoside-resistance. The following report outlines the use of our recently described oxidopyrylium cycloaddition/ring-opening strategy in the synthesis and profiling of a library of synthetic α-hydroxytropolones against ANT(2")-Ia. In addition, we show that two of these synthetic constructs are capable of rescuing gentamicin activity against ANT-(2")-Ia-expressing bacteria.
Collapse
Affiliation(s)
- Danielle R Hirsch
- Department of Chemistry, Brooklyn College, The City University of New York, 2900 Bedford Avenue, Brooklyn, NY 11210, United States; Department of Chemistry, The Graduate Center, The City University of New York, 365 Fifth Avenue, New York, NY 10016, United States
| | - Georgina Cox
- M. G. DeGroote Institute for Infectious Disease Research, Department of Biochemistry and Biomedical Sciences, McMaster University, 1280 Main Street West, Hamilton, ON L8N 4K1, Canada
| | - Michael P D'Erasmo
- Department of Chemistry, Brooklyn College, The City University of New York, 2900 Bedford Avenue, Brooklyn, NY 11210, United States; Department of Chemistry, The Graduate Center, The City University of New York, 365 Fifth Avenue, New York, NY 10016, United States
| | - Tushar Shakya
- M. G. DeGroote Institute for Infectious Disease Research, Department of Biochemistry and Biomedical Sciences, McMaster University, 1280 Main Street West, Hamilton, ON L8N 4K1, Canada
| | - Christine Meck
- Department of Chemistry, Brooklyn College, The City University of New York, 2900 Bedford Avenue, Brooklyn, NY 11210, United States; Department of Chemistry, The Graduate Center, The City University of New York, 365 Fifth Avenue, New York, NY 10016, United States
| | - Noushad Mohd
- Department of Chemistry, Brooklyn College, The City University of New York, 2900 Bedford Avenue, Brooklyn, NY 11210, United States
| | - Gerard D Wright
- M. G. DeGroote Institute for Infectious Disease Research, Department of Biochemistry and Biomedical Sciences, McMaster University, 1280 Main Street West, Hamilton, ON L8N 4K1, Canada
| | - Ryan P Murelli
- Department of Chemistry, Brooklyn College, The City University of New York, 2900 Bedford Avenue, Brooklyn, NY 11210, United States; Department of Chemistry, The Graduate Center, The City University of New York, 365 Fifth Avenue, New York, NY 10016, United States.
| |
Collapse
|
33
|
Strategies to overcome the action of aminoglycoside-modifying enzymes for treating resistant bacterial infections. Future Med Chem 2014; 5:1285-309. [PMID: 23859208 DOI: 10.4155/fmc.13.80] [Citation(s) in RCA: 93] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Shortly after the discovery of the first antibiotics, bacterial resistance began to emerge. Many mechanisms give rise to resistance; the most prevalent mechanism of resistance to the aminoglycoside (AG) family of antibiotics is the action of aminoglycoside-modifying enzymes (AMEs). Since the identification of these modifying enzymes, many efforts have been put forth to prevent their damaging alterations of AGs. These diverse strategies are discussed within this review, including: creating new AGs that are unaffected by AMEs; developing inhibitors of AMEs to be co-delivered with AGs; or regulating AME expression. Modern high-throughput methods as well as drug combinations and repurposing are highlighted as recent drug-discovery efforts towards fighting the increasing antibiotic resistance crisis.
Collapse
|
34
|
Lin DL, Tran T, Adams C, Alam JY, Herron SR, Tolmasky ME. Inhibitors of the aminoglycoside 6'-N-acetyltransferase type Ib [AAC(6')-Ib] identified by in silico molecular docking. Bioorg Med Chem Lett 2013; 23:5694-8. [PMID: 24011645 DOI: 10.1016/j.bmcl.2013.08.016] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2013] [Revised: 07/24/2013] [Accepted: 08/05/2013] [Indexed: 10/26/2022]
Abstract
AAC(6')-Ib is an important aminoglycoside resistance enzyme to target with enzymatic inhibitors. An in silico screening approach was used to identify potential inhibitors from the ChemBridge library. Several compounds were identified, of which two of them, 4-[(2-{[1-(3-methylphenyl)-4,6-dioxo-2-thioxotetrahydro-5(2H)-pyrimidinylidene]methyl}phenoxy)methyl]benzoic acid and 2-{5-[(4,6-dioxo-1,3-diphenyl-2-thioxotetrahydro-5(2H)-pyrimidinylidene)methyl]-2-furyl}benzoic acid, showed micromolar activity in inhibiting acetylation of kanamycin A. These compounds are predicted to bind the aminoglycoside binding site of AAC(6')-Ib and exhibited competitive inhibition against kanamycin A.
Collapse
Affiliation(s)
- David L Lin
- Center for Applied Biotechnology Studies, Department of Biological Science, College of Natural Sciences and Mathematics, California State University Fullerton, 800 N State College Boulevard, Fullerton, CA 92834-6850, United States.
| | | | | | | | | | | |
Collapse
|
35
|
Shi K, Caldwell SJ, Fong DH, Berghuis AM. Prospects for circumventing aminoglycoside kinase mediated antibiotic resistance. Front Cell Infect Microbiol 2013; 3:22. [PMID: 23805415 PMCID: PMC3691515 DOI: 10.3389/fcimb.2013.00022] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2013] [Accepted: 06/04/2013] [Indexed: 01/10/2023] Open
Abstract
Aminoglycosides are a class of antibiotics with a broad spectrum of antimicrobial activity. Unfortunately, resistance in clinical isolates is pervasive, rendering many aminoglycosides ineffective. The most widely disseminated means of resistance to this class of antibiotics is inactivation of the drug by aminoglycoside-modifying enzymes (AMEs). There are two principal strategies to overcoming the effects of AMEs. The first approach involves the design of novel aminoglycosides that can evade modification. Although this strategy has yielded a number of superior aminoglycoside variants, their efficacy cannot be sustained in the long term. The second approach entails the development of molecules that interfere with the mechanism of AMEs such that the activity of aminoglycosides is preserved. Although such a molecule has yet to enter clinical development, the search for AME inhibitors has been greatly facilitated by the wealth of structural information amassed in recent years. In particular, aminoglycoside phosphotransferases or kinases (APHs) have been studied extensively and crystal structures of a number of APHs with diverse regiospecificity and substrate specificity have been elucidated. In this review, we present a comprehensive overview of the available APH structures and recent progress in APH inhibitor development, with a focus on the structure-guided strategies.
Collapse
Affiliation(s)
- Kun Shi
- Groupe de Recherche Axé sur la Structure des Protéines, Department of Biochemistry, McGill UniversityMontreal, QC, Canada
| | - Shane J. Caldwell
- Groupe de Recherche Axé sur la Structure des Protéines, Department of Biochemistry, McGill UniversityMontreal, QC, Canada
| | - Desiree H. Fong
- Groupe de Recherche Axé sur la Structure des Protéines, Department of Biochemistry, McGill UniversityMontreal, QC, Canada
| | - Albert M. Berghuis
- Groupe de Recherche Axé sur la Structure des Protéines, Department of Biochemistry, McGill UniversityMontreal, QC, Canada
- Department of Microbiology and Immunology, McGill UniversityMontreal, QC, Canada
| |
Collapse
|
36
|
Ramirez MS, Nikolaidis N, Tolmasky ME. Rise and dissemination of aminoglycoside resistance: the aac(6')-Ib paradigm. Front Microbiol 2013; 4:121. [PMID: 23730301 PMCID: PMC3656343 DOI: 10.3389/fmicb.2013.00121] [Citation(s) in RCA: 115] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2013] [Accepted: 04/29/2013] [Indexed: 11/21/2022] Open
Abstract
Enzymatic modification is a prevalent mechanism by which bacteria defeat the action of antibiotics. Aminoglycosides are often inactivated by aminoglycoside modifying enzymes encoded by genes present in the chromosome, plasmids, and other genetic elements. The AAC(6′)-Ib (aminoglycoside 6′-N-acetyltransferase type Ib) is an enzyme of clinical importance found in a wide variety of gram-negative pathogens. The AAC(6′)-Ib enzyme is of interest not only because of his ubiquity but also because of other characteristics, it presents significant microheterogeneity at the N-termini and the aac(6′)-Ib gene is often present in integrons, transposons, plasmids, genomic islands, and other genetic structures. Excluding the highly heterogeneous N-termini, there are 45 non-identical AAC(6′)-Ib related entries in the NCBI database, 32 of which have identical name in spite of not having identical amino acid sequence. While some variants conserved similar properties, others show dramatic differences in specificity, including the case of AAC(6′)-Ib-cr that mediates acetylation of ciprofloxacin representing a rare case where a resistance enzyme acquires the ability to utilize an antibiotic of a different class as substrate. Efforts to utilize antisense technologies to turn off expression of the gene or to identify enzymatic inhibitors to induce phenotypic conversion to susceptibility are under way.
Collapse
Affiliation(s)
- María S Ramirez
- Department of Biological Science, Center for Applied Biotechnology Studies, College of Natural Sciences and Mathematics, California State University Fullerton Fullerton, CA, USA
| | | | | |
Collapse
|
37
|
Romanowska J, Reuter N, Trylska J. Comparing aminoglycoside binding sites in bacterial ribosomal RNA and aminoglycoside modifying enzymes. Proteins 2012; 81:63-80. [PMID: 22907688 DOI: 10.1002/prot.24163] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2012] [Revised: 08/02/2012] [Accepted: 08/09/2012] [Indexed: 11/10/2022]
Abstract
Aminoglycoside antibiotics are used against severe bacterial infections. They bind to the bacterial ribosomal RNA and interfere with the translation process. However, bacteria produce aminoglycoside modifying enzymes (AME) to resist aminoglycoside actions. AMEs form a variable group and yet they specifically recognize and efficiently bind aminoglycosides, which are also diverse in terms of total net charge and the number of pseudo-sugar rings. Here, we present the results of 25 molecular dynamics simulations of three AME representatives and aminoglycoside ribosomal RNA binding site, unliganded and complexed with an aminoglycoside, kanamycin A. A comparison of the aminoglycoside binding sites in these different receptors revealed that the enzymes efficiently mimic the nucleic acid environment of the ribosomal RNA binding cleft. Although internal dynamics of AMEs and their interaction patterns with aminoglycosides differ, the energetical analysis showed that the most favorable sites are virtually the same in the enzymes and RNA. The most copied interactions were of electrostatic nature, but stacking was also replicated in one AME:kanamycin complex. In addition, we found that some water-mediated interactions were very stable in the simulations of the complexes. We show that our simulations reproduce well findings from NMR or X-ray structural studies, as well as results from directed mutagenesis. The outcomes of our analyses provide new insight into aminoglycoside resistance mechanism that is related to the enzymatic modification of these drugs.
Collapse
Affiliation(s)
- Julia Romanowska
- Department of Biophysics, Faculty of Physics, University of Warsaw, Hoża 69, 00-681 Warsaw, Poland.
| | | | | |
Collapse
|
38
|
Vong K, Auclair K. Understanding and overcoming aminoglycoside resistance caused by N-6'-acetyltransferase. MEDCHEMCOMM 2012; 3:397-407. [PMID: 28018574 PMCID: PMC5179255 DOI: 10.1039/c2md00253a] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Aminoglycosides occupy a special niche amongst antibiotics in part because of their broad spectrum of action. Bacterial resistance is however menacing to render these drugs obsolete. A significant amount of work has been devoted to understand and overcome aminoglycoside resistance. This mini-review will discuss aminoglycoside-modifying enzymes (AMEs), with a special emphasis on the efforts to comprehend and block resistance caused by aminoglycoside 6'-N-acetyltransferase (AAC(6')).
Collapse
Affiliation(s)
- Kenward Vong
- Department of Chemistry, McGill University, 801 Sherbrooke Street West, Montréal, Québec, Canada H3A 2K6
| | - Karine Auclair
- Department of Chemistry, McGill University, 801 Sherbrooke Street West, Montréal, Québec, Canada H3A 2K6
| |
Collapse
|
39
|
Matesanz R, Diaz JF, Corzana F, Santana AG, Bastida A, Asensio JL. Multiple keys for a single lock: the unusual structural plasticity of the nucleotidyltransferase (4')/kanamycin complex. Chemistry 2012; 18:2875-89. [PMID: 22298309 DOI: 10.1002/chem.201101888] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2011] [Revised: 12/05/2011] [Indexed: 11/09/2022]
Abstract
The most common mode of bacterial resistance to aminoglycoside antibiotics is the enzyme-catalysed chemical modification of the drug. Over the last two decades, significant efforts in medicinal chemistry have been focused on the design of non- inactivable antibiotics. Unfortunately, this strategy has met with limited success on account of the remarkably wide substrate specificity of aminoglycoside-modifying enzymes. To understand the mechanisms behind substrate promiscuity, we have performed a comprehensive experimental and theoretical analysis of the molecular-recognition processes that lead to antibiotic inactivation by Staphylococcus aureus nucleotidyltransferase 4'(ANT(4')), a clinically relevant protein. According to our results, the ability of this enzyme to inactivate structurally diverse polycationic molecules relies on three specific features of the catalytic region. First, the dominant role of electrostatics in aminoglycoside recognition, in combination with the significant extension of the enzyme anionic regions, confers to the protein/antibiotic complex a highly dynamic character. The motion deduced for the bound antibiotic seem to be essential for the enzyme action and probably provide a mechanism to explore alternative drug inactivation modes. Second, the nucleotide recognition is exclusively mediated by the inorganic fragment. In fact, even inorganic triphosphate can be employed as a substrate. Third, ANT(4') seems to be equipped with a duplicated basic catalyst that is able to promote drug inactivation through different reactive geometries. This particular combination of features explains the enzyme versatility and renders the design of non-inactivable derivatives a challenging task.
Collapse
Affiliation(s)
- Ruth Matesanz
- Centro de Investigaciones Biológicas (CSIC), Ramiro de Maeztu 9, 28040 Madrid, Spain
| | | | | | | | | | | |
Collapse
|
40
|
The potential of antimicrobial peptides as biocides. Int J Mol Sci 2011; 12:6566-96. [PMID: 22072905 PMCID: PMC3210996 DOI: 10.3390/ijms12106566] [Citation(s) in RCA: 118] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2011] [Revised: 09/22/2011] [Accepted: 09/26/2011] [Indexed: 12/12/2022] Open
Abstract
Antimicrobial peptides constitute a diverse class of naturally occurring antimicrobial molecules which have activity against a wide range of pathogenic microorganisms. Antimicrobial peptides are exciting leads in the development of novel biocidal agents at a time when classical antibiotics are under intense pressure from emerging resistance, and the global industry in antibiotic research and development stagnates. This review will examine the potential of antimicrobial peptides, both natural and synthetic, as novel biocidal agents in the battle against multi-drug resistant pathogen infections.
Collapse
|
41
|
Abstract
The unremitting emergence of multidrug-resistant bacterial pathogens highlights a matching need for new therapeutic options. For example, new carbapenemases such as KPC (class A Klebsiella pneumoniae) and NDM-1 (New Delhi metallo-β-lactamase 1) are surfacing, resulting in almost total resistance to β-lactam antibiotics. Furthermore, resistance is quickly disseminated, not only in the healthcare sector, but also within the community at large, because many resistance determinants are carried on mobile genetic elements readily shared among pathogens. The absence of new antibiotics has led to a growing reliance on older, more toxic drugs such as colistin, but resistance to these is already arising. One approach to combat this growing problem is the use of combination drug antibiotic adjuvant therapy, which potentiates the activity of antibiotics. Here, we review the current situation and discuss potential drug combinations that may increase the potency of antibiotics in the future. Adjuvant therapies include antibiotic combinations, synergy between antibiotics and nonantibiotics, inhibition of resistance and molecules that alter the physiology of antibiotic-insensitive cells, such as those in biofilms. We provide a rationale for these multicomponent strategies, highlighting current research and important considerations for their clinical use and pharmacological properties.
Collapse
|
42
|
Affiliation(s)
- Mariya Morar
- M.G. DeGroote Institute for Infectious Disease Research and the Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario, L8N 3Z5, Canada;
| | - Gerard D. Wright
- M.G. DeGroote Institute for Infectious Disease Research and the Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario, L8N 3Z5, Canada;
| |
Collapse
|
43
|
Abstract
Aminoglycosides have been an essential component of the armamentarium in the treatment of life-threatening infections. Unfortunately, their efficacy has been reduced by the surge and dissemination of resistance. In some cases the levels of resistance reached the point that rendered them virtually useless. Among many known mechanisms of resistance to aminoglycosides, enzymatic modification is the most prevalent in the clinical setting. Aminoglycoside modifying enzymes catalyze the modification at different -OH or -NH₂ groups of the 2-deoxystreptamine nucleus or the sugar moieties and can be nucleotidyltransferases, phosphotransferases, or acetyltransferases. The number of aminoglycoside modifying enzymes identified to date as well as the genetic environments where the coding genes are located is impressive and there is virtually no bacteria that is unable to support enzymatic resistance to aminoglycosides. Aside from the development of new aminoglycosides refractory to as many as possible modifying enzymes there are currently two main strategies being pursued to overcome the action of aminoglycoside modifying enzymes. Their successful development would extend the useful life of existing antibiotics that have proven effective in the treatment of infections. These strategies consist of the development of inhibitors of the enzymatic action or of the expression of the modifying enzymes.
Collapse
Affiliation(s)
- Maria S. Ramirez
- Center for Applied Biotechnology Studies, Department of Biological Science, College of Natural Sciences and Mathematics, California State University Fullerton, Fullerton, California 92834-6850
| | - Marcelo E. Tolmasky
- Center for Applied Biotechnology Studies, Department of Biological Science, College of Natural Sciences and Mathematics, California State University Fullerton, Fullerton, California 92834-6850
| |
Collapse
|
44
|
De Pascale G, Wright GD. Antibiotic resistance by enzyme inactivation: from mechanisms to solutions. Chembiochem 2010; 11:1325-34. [PMID: 20564281 DOI: 10.1002/cbic.201000067] [Citation(s) in RCA: 56] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Affiliation(s)
- Gianfranco De Pascale
- DeGroote Institute for Infectious Disease Research, Department of Biochemistry and Biomedical Sciences, McMaster University, 1200 Main Street W, Hamilton, ON L8N 3Z5 Canada
| | | |
Collapse
|
45
|
Vacas T, Corzana F, Jiménez-Osés G, González C, Gómez AM, Bastida A, Revuelta J, Asensio JL. Role of Aromatic Rings in the Molecular Recognition of Aminoglycoside Antibiotics: Implications for Drug Design. J Am Chem Soc 2010; 132:12074-90. [DOI: 10.1021/ja1046439] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Affiliation(s)
- Tatiana Vacas
- Instituto de Química Orgánica General (CSIC), Juan de la Cierva 3, 28006 Madrid, Spain, Departamento de Química, Universidad de La Rioja, UA-CSIC, Logroño, Spain, Departamento de Química Orgánica y Química Física. Universidad de Zaragoza-CSIC, Zaragoza, Spain, and Instituto de Química Física Rocasolano (CSIC), Madrid, Spain
| | - Francisco Corzana
- Instituto de Química Orgánica General (CSIC), Juan de la Cierva 3, 28006 Madrid, Spain, Departamento de Química, Universidad de La Rioja, UA-CSIC, Logroño, Spain, Departamento de Química Orgánica y Química Física. Universidad de Zaragoza-CSIC, Zaragoza, Spain, and Instituto de Química Física Rocasolano (CSIC), Madrid, Spain
| | - Gonzalo Jiménez-Osés
- Instituto de Química Orgánica General (CSIC), Juan de la Cierva 3, 28006 Madrid, Spain, Departamento de Química, Universidad de La Rioja, UA-CSIC, Logroño, Spain, Departamento de Química Orgánica y Química Física. Universidad de Zaragoza-CSIC, Zaragoza, Spain, and Instituto de Química Física Rocasolano (CSIC), Madrid, Spain
| | - Carlos González
- Instituto de Química Orgánica General (CSIC), Juan de la Cierva 3, 28006 Madrid, Spain, Departamento de Química, Universidad de La Rioja, UA-CSIC, Logroño, Spain, Departamento de Química Orgánica y Química Física. Universidad de Zaragoza-CSIC, Zaragoza, Spain, and Instituto de Química Física Rocasolano (CSIC), Madrid, Spain
| | - Ana M. Gómez
- Instituto de Química Orgánica General (CSIC), Juan de la Cierva 3, 28006 Madrid, Spain, Departamento de Química, Universidad de La Rioja, UA-CSIC, Logroño, Spain, Departamento de Química Orgánica y Química Física. Universidad de Zaragoza-CSIC, Zaragoza, Spain, and Instituto de Química Física Rocasolano (CSIC), Madrid, Spain
| | - Agatha Bastida
- Instituto de Química Orgánica General (CSIC), Juan de la Cierva 3, 28006 Madrid, Spain, Departamento de Química, Universidad de La Rioja, UA-CSIC, Logroño, Spain, Departamento de Química Orgánica y Química Física. Universidad de Zaragoza-CSIC, Zaragoza, Spain, and Instituto de Química Física Rocasolano (CSIC), Madrid, Spain
| | - Julia Revuelta
- Instituto de Química Orgánica General (CSIC), Juan de la Cierva 3, 28006 Madrid, Spain, Departamento de Química, Universidad de La Rioja, UA-CSIC, Logroño, Spain, Departamento de Química Orgánica y Química Física. Universidad de Zaragoza-CSIC, Zaragoza, Spain, and Instituto de Química Física Rocasolano (CSIC), Madrid, Spain
| | - Juan Luis Asensio
- Instituto de Química Orgánica General (CSIC), Juan de la Cierva 3, 28006 Madrid, Spain, Departamento de Química, Universidad de La Rioja, UA-CSIC, Logroño, Spain, Departamento de Química Orgánica y Química Física. Universidad de Zaragoza-CSIC, Zaragoza, Spain, and Instituto de Química Física Rocasolano (CSIC), Madrid, Spain
| |
Collapse
|
46
|
Revuelta J, Corzana F, Bastida A, Asensio J. The Unusual Nucleotide Recognition Properties of the Resistance Enzyme ANT(4′): Inorganic Tri/Polyphosphate as a Substrate for Aminoglycoside Inactivation. Chemistry 2010; 16:8635-40. [DOI: 10.1002/chem.201000641] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
|
47
|
Akers KS, Chaney C, Barsoumian A, Beckius M, Zera W, Yu X, Guymon C, Keen EF, Robinson BJ, Mende K, Murray CK. Aminoglycoside resistance and susceptibility testing errors in Acinetobacter baumannii-calcoaceticus complex. J Clin Microbiol 2010; 48:1132-8. [PMID: 20107089 PMCID: PMC2849581 DOI: 10.1128/jcm.02006-09] [Citation(s) in RCA: 64] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2009] [Revised: 12/01/2009] [Accepted: 01/15/2010] [Indexed: 11/20/2022] Open
Abstract
Antimicrobial resistance is depleting the pharmacopeia of agents clinically useful against Gram-negative bacilli. As the number of active agents diminishes, accurate susceptibility testing becomes critical. We studied the susceptibilities of 107 isolates of the Acinetobacter baumannii-calcoaceticus complex to amikacin, gentamicin, and tobramycin using disk diffusion, Etest, as well as the Phoenix, Vitek 2, and MicroScan automated systems, and compared the results to those obtained by broth microdilution. Genes encoding aminoglycoside-modifying enzymes (AMEs) were detected by multiplex PCR, and clonal relationships were determined by pulsed-field gel electrophoresis. Tobramycin was the most active aminoglycoside (27.1% of isolates were susceptible). Disk diffusion and Etest tended to be more accurate than the Vitek 2, Phoenix, and MicroScan automated systems; but errors were noted with all methods. The Vitek 2 instrument incorrectly reported that more than one-third of the isolates were susceptible to amikacin (a very major error). Isolates were polyclonal, with 26 distinct strains, and carried multiple AME genes unrelated to the strain type. The presence of the ant(2")-Ia gene was statistically associated with resistance to each aminoglycoside. The AME genotype accounted for the resistance profile observed in a minority of isolates, suggesting the involvement of multiple resistance mechanisms. Hospital pharmacy records indicated the preferential use of amikacin over other aminoglycosides in the burn intensive care unit, where aminoglycoside resistance is prevalent. The resistance in that unit did not correlate with a predominant strain, AME genotype, or total annual aminoglycoside consumption. Susceptibility to tobramycin increased, even though susceptible isolates carried AME genotypes predicting the inactivation of tobramycin. Determination of the relative contribution of multiple concurrent resistance mechanisms may improve our understanding of aminoglycoside resistance in the Acinetobacter baumannii-calcoaceticus complex.
Collapse
Affiliation(s)
- Kevin S. Akers
- Infectious Disease Service, San Antonio Military Medical Center, 3851 Roger Brooke Drive, Fort Sam Houston, Texas 78234-6200, Department of Medicine, San Antonio Military Medical Center, 3851 Roger Brooke Drive, Fort Sam Houston, Texas 78234-6200, Department of Clinical Investigation, San Antonio Military Medical Center, 3400 Rawley E. Chambers Ave., Suite A, Fort Sam Houston, Texas 78234, U.S. Army Institute of Surgical Research, 3400 Rawley E. Chambers Ave., Bldg. 3611, Fort Sam Houston, Texas 78234, Department of Pathology and Area Laboratory Services, San Antonio Military Medical Center, 3851 Roger Brooke Drive, Fort Sam Houston, Texas 78234-6200, Infectious Disease Clinical Research Program, Uniformed Services University of the Health Sciences, 4301 Jones Bridge Drive, Bethesda, Maryland 20814
| | - Chris Chaney
- Infectious Disease Service, San Antonio Military Medical Center, 3851 Roger Brooke Drive, Fort Sam Houston, Texas 78234-6200, Department of Medicine, San Antonio Military Medical Center, 3851 Roger Brooke Drive, Fort Sam Houston, Texas 78234-6200, Department of Clinical Investigation, San Antonio Military Medical Center, 3400 Rawley E. Chambers Ave., Suite A, Fort Sam Houston, Texas 78234, U.S. Army Institute of Surgical Research, 3400 Rawley E. Chambers Ave., Bldg. 3611, Fort Sam Houston, Texas 78234, Department of Pathology and Area Laboratory Services, San Antonio Military Medical Center, 3851 Roger Brooke Drive, Fort Sam Houston, Texas 78234-6200, Infectious Disease Clinical Research Program, Uniformed Services University of the Health Sciences, 4301 Jones Bridge Drive, Bethesda, Maryland 20814
| | - Alice Barsoumian
- Infectious Disease Service, San Antonio Military Medical Center, 3851 Roger Brooke Drive, Fort Sam Houston, Texas 78234-6200, Department of Medicine, San Antonio Military Medical Center, 3851 Roger Brooke Drive, Fort Sam Houston, Texas 78234-6200, Department of Clinical Investigation, San Antonio Military Medical Center, 3400 Rawley E. Chambers Ave., Suite A, Fort Sam Houston, Texas 78234, U.S. Army Institute of Surgical Research, 3400 Rawley E. Chambers Ave., Bldg. 3611, Fort Sam Houston, Texas 78234, Department of Pathology and Area Laboratory Services, San Antonio Military Medical Center, 3851 Roger Brooke Drive, Fort Sam Houston, Texas 78234-6200, Infectious Disease Clinical Research Program, Uniformed Services University of the Health Sciences, 4301 Jones Bridge Drive, Bethesda, Maryland 20814
| | - Miriam Beckius
- Infectious Disease Service, San Antonio Military Medical Center, 3851 Roger Brooke Drive, Fort Sam Houston, Texas 78234-6200, Department of Medicine, San Antonio Military Medical Center, 3851 Roger Brooke Drive, Fort Sam Houston, Texas 78234-6200, Department of Clinical Investigation, San Antonio Military Medical Center, 3400 Rawley E. Chambers Ave., Suite A, Fort Sam Houston, Texas 78234, U.S. Army Institute of Surgical Research, 3400 Rawley E. Chambers Ave., Bldg. 3611, Fort Sam Houston, Texas 78234, Department of Pathology and Area Laboratory Services, San Antonio Military Medical Center, 3851 Roger Brooke Drive, Fort Sam Houston, Texas 78234-6200, Infectious Disease Clinical Research Program, Uniformed Services University of the Health Sciences, 4301 Jones Bridge Drive, Bethesda, Maryland 20814
| | - Wendy Zera
- Infectious Disease Service, San Antonio Military Medical Center, 3851 Roger Brooke Drive, Fort Sam Houston, Texas 78234-6200, Department of Medicine, San Antonio Military Medical Center, 3851 Roger Brooke Drive, Fort Sam Houston, Texas 78234-6200, Department of Clinical Investigation, San Antonio Military Medical Center, 3400 Rawley E. Chambers Ave., Suite A, Fort Sam Houston, Texas 78234, U.S. Army Institute of Surgical Research, 3400 Rawley E. Chambers Ave., Bldg. 3611, Fort Sam Houston, Texas 78234, Department of Pathology and Area Laboratory Services, San Antonio Military Medical Center, 3851 Roger Brooke Drive, Fort Sam Houston, Texas 78234-6200, Infectious Disease Clinical Research Program, Uniformed Services University of the Health Sciences, 4301 Jones Bridge Drive, Bethesda, Maryland 20814
| | - Xin Yu
- Infectious Disease Service, San Antonio Military Medical Center, 3851 Roger Brooke Drive, Fort Sam Houston, Texas 78234-6200, Department of Medicine, San Antonio Military Medical Center, 3851 Roger Brooke Drive, Fort Sam Houston, Texas 78234-6200, Department of Clinical Investigation, San Antonio Military Medical Center, 3400 Rawley E. Chambers Ave., Suite A, Fort Sam Houston, Texas 78234, U.S. Army Institute of Surgical Research, 3400 Rawley E. Chambers Ave., Bldg. 3611, Fort Sam Houston, Texas 78234, Department of Pathology and Area Laboratory Services, San Antonio Military Medical Center, 3851 Roger Brooke Drive, Fort Sam Houston, Texas 78234-6200, Infectious Disease Clinical Research Program, Uniformed Services University of the Health Sciences, 4301 Jones Bridge Drive, Bethesda, Maryland 20814
| | - Charles Guymon
- Infectious Disease Service, San Antonio Military Medical Center, 3851 Roger Brooke Drive, Fort Sam Houston, Texas 78234-6200, Department of Medicine, San Antonio Military Medical Center, 3851 Roger Brooke Drive, Fort Sam Houston, Texas 78234-6200, Department of Clinical Investigation, San Antonio Military Medical Center, 3400 Rawley E. Chambers Ave., Suite A, Fort Sam Houston, Texas 78234, U.S. Army Institute of Surgical Research, 3400 Rawley E. Chambers Ave., Bldg. 3611, Fort Sam Houston, Texas 78234, Department of Pathology and Area Laboratory Services, San Antonio Military Medical Center, 3851 Roger Brooke Drive, Fort Sam Houston, Texas 78234-6200, Infectious Disease Clinical Research Program, Uniformed Services University of the Health Sciences, 4301 Jones Bridge Drive, Bethesda, Maryland 20814
| | - Edward F. Keen
- Infectious Disease Service, San Antonio Military Medical Center, 3851 Roger Brooke Drive, Fort Sam Houston, Texas 78234-6200, Department of Medicine, San Antonio Military Medical Center, 3851 Roger Brooke Drive, Fort Sam Houston, Texas 78234-6200, Department of Clinical Investigation, San Antonio Military Medical Center, 3400 Rawley E. Chambers Ave., Suite A, Fort Sam Houston, Texas 78234, U.S. Army Institute of Surgical Research, 3400 Rawley E. Chambers Ave., Bldg. 3611, Fort Sam Houston, Texas 78234, Department of Pathology and Area Laboratory Services, San Antonio Military Medical Center, 3851 Roger Brooke Drive, Fort Sam Houston, Texas 78234-6200, Infectious Disease Clinical Research Program, Uniformed Services University of the Health Sciences, 4301 Jones Bridge Drive, Bethesda, Maryland 20814
| | - Brian J. Robinson
- Infectious Disease Service, San Antonio Military Medical Center, 3851 Roger Brooke Drive, Fort Sam Houston, Texas 78234-6200, Department of Medicine, San Antonio Military Medical Center, 3851 Roger Brooke Drive, Fort Sam Houston, Texas 78234-6200, Department of Clinical Investigation, San Antonio Military Medical Center, 3400 Rawley E. Chambers Ave., Suite A, Fort Sam Houston, Texas 78234, U.S. Army Institute of Surgical Research, 3400 Rawley E. Chambers Ave., Bldg. 3611, Fort Sam Houston, Texas 78234, Department of Pathology and Area Laboratory Services, San Antonio Military Medical Center, 3851 Roger Brooke Drive, Fort Sam Houston, Texas 78234-6200, Infectious Disease Clinical Research Program, Uniformed Services University of the Health Sciences, 4301 Jones Bridge Drive, Bethesda, Maryland 20814
| | - Katrin Mende
- Infectious Disease Service, San Antonio Military Medical Center, 3851 Roger Brooke Drive, Fort Sam Houston, Texas 78234-6200, Department of Medicine, San Antonio Military Medical Center, 3851 Roger Brooke Drive, Fort Sam Houston, Texas 78234-6200, Department of Clinical Investigation, San Antonio Military Medical Center, 3400 Rawley E. Chambers Ave., Suite A, Fort Sam Houston, Texas 78234, U.S. Army Institute of Surgical Research, 3400 Rawley E. Chambers Ave., Bldg. 3611, Fort Sam Houston, Texas 78234, Department of Pathology and Area Laboratory Services, San Antonio Military Medical Center, 3851 Roger Brooke Drive, Fort Sam Houston, Texas 78234-6200, Infectious Disease Clinical Research Program, Uniformed Services University of the Health Sciences, 4301 Jones Bridge Drive, Bethesda, Maryland 20814
| | - Clinton K. Murray
- Infectious Disease Service, San Antonio Military Medical Center, 3851 Roger Brooke Drive, Fort Sam Houston, Texas 78234-6200, Department of Medicine, San Antonio Military Medical Center, 3851 Roger Brooke Drive, Fort Sam Houston, Texas 78234-6200, Department of Clinical Investigation, San Antonio Military Medical Center, 3400 Rawley E. Chambers Ave., Suite A, Fort Sam Houston, Texas 78234, U.S. Army Institute of Surgical Research, 3400 Rawley E. Chambers Ave., Bldg. 3611, Fort Sam Houston, Texas 78234, Department of Pathology and Area Laboratory Services, San Antonio Military Medical Center, 3851 Roger Brooke Drive, Fort Sam Houston, Texas 78234-6200, Infectious Disease Clinical Research Program, Uniformed Services University of the Health Sciences, 4301 Jones Bridge Drive, Bethesda, Maryland 20814
| |
Collapse
|
48
|
Abstract
Acinetobacter baumannii has emerged as a highly troublesome pathogen for many institutions globally. As a consequence of its immense ability to acquire or upregulate antibiotic drug resistance determinants, it has justifiably been propelled to the forefront of scientific attention. Apart from its predilection for the seriously ill within intensive care units, A. baumannii has more recently caused a range of infectious syndromes in military personnel injured in the Iraq and Afghanistan conflicts. This review details the significant advances that have been made in our understanding of this remarkable organism over the last 10 years, including current taxonomy and species identification, issues with susceptibility testing, mechanisms of antibiotic resistance, global epidemiology, clinical impact of infection, host-pathogen interactions, and infection control and therapeutic considerations.
Collapse
|
49
|
Studies of Enzymes That Cause Resistance to Aminoglycosides Antibiotics. METHODS IN MOLECULAR MEDICINE™ 2008; 142:261-71. [DOI: 10.1007/978-1-59745-246-5_20] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/05/2022]
|
50
|
Kohl A, Amstutz P, Parizek P, Binz HK, Briand C, Capitani G, Forrer P, Plückthun A, Grütter MG. Allosteric inhibition of aminoglycoside phosphotransferase by a designed ankyrin repeat protein. Structure 2007; 13:1131-41. [PMID: 16084385 DOI: 10.1016/j.str.2005.04.020] [Citation(s) in RCA: 64] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2005] [Revised: 04/25/2005] [Accepted: 04/26/2005] [Indexed: 10/25/2022]
Abstract
Aminoglycoside phosphotransferase (3')-IIIa (APH) is a bacterial kinase that confers antibiotic resistance to many pathogenic bacteria and shares structural homology with eukaryotic protein kinases. We report here the crystal structure of APH, trapped in an inactive conformation by a tailor-made inhibitory ankyrin repeat (AR) protein, at 2.15 A resolution. The inhibitor was selected from a combinatorial library of designed AR proteins. The AR protein binds the C-terminal lobe of APH and thereby stabilizes three alpha helices, which are necessary for substrate binding, in a significantly displaced conformation. BIAcore analysis and kinetic enzyme inhibition experiments are consistent with the proposed allosteric inhibition mechanism. In contrast to most small-molecule kinase inhibitors, the AR proteins are not restricted to active site binding, allowing for higher specificity. Inactive conformations of pharmaceutically relevant enzymes, as can be elucidated with the approach presented here, represent powerful starting points for rational drug design.
Collapse
Affiliation(s)
- Andreas Kohl
- Department of Biochemistry, University of Zürich, Winterthurerstrasse 190, CH-8057 Zürich, Switzerland
| | | | | | | | | | | | | | | | | |
Collapse
|