401
|
Seydlová G, Pohl R, Zborníková E, Ehn M, Šimák O, Panova N, Kolář M, Bogdanová K, Večeřová R, Fišer R, Šanderová H, Vítovská D, Sudzinová P, Pospíšil J, Benada O, Křížek T, Sedlák D, Bartůněk P, Krásný L, Rejman D. Lipophosphonoxins II: Design, Synthesis, and Properties of Novel Broad Spectrum Antibacterial Agents. J Med Chem 2017; 60:6098-6118. [PMID: 28654257 DOI: 10.1021/acs.jmedchem.7b00355] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
The increase in the number of bacterial strains resistant to known antibiotics is alarming. In this study we report the synthesis of novel compounds termed Lipophosphonoxins II (LPPO II). We show that LPPO II display excellent activities against Gram-positive and -negative bacteria, including pathogens and multiresistant strains. We describe their mechanism of action-plasmatic membrane pore-forming activity selective for bacteria. Importantly, LPPO II neither damage nor cross the eukaryotic plasmatic membrane at their bactericidal concentrations. Further, we demonstrate LPPO II have low propensity for resistance development, likely due to their rapid membrane-targeting mode of action. Finally, we reveal that LPPO II are not toxic to either eukaryotic cells or model animals when administered orally or topically. Collectively, these results suggest that LPPO II are highly promising compounds for development into pharmaceuticals.
Collapse
Affiliation(s)
- Gabriela Seydlová
- Department of Genetics and Microbiology, Faculty of Science, Charles University , Viničná 5, 128 43 Prague 2, Czech Republic
| | - Radek Pohl
- Institute of Organic Chemistry and Biochemistry, Czech Academy of Sciences v.v.i. , Flemingovo nám. 2, 166 10 Prague 6, Czech Republic
| | - Eva Zborníková
- Institute of Organic Chemistry and Biochemistry, Czech Academy of Sciences v.v.i. , Flemingovo nám. 2, 166 10 Prague 6, Czech Republic.,Department of Analytical Chemistry, Faculty of Science, Charles University , Albertov 6, 128 43 Prague 2, Czech Republic
| | - Marcel Ehn
- Institute of Organic Chemistry and Biochemistry, Czech Academy of Sciences v.v.i. , Flemingovo nám. 2, 166 10 Prague 6, Czech Republic
| | - Ondřej Šimák
- Institute of Organic Chemistry and Biochemistry, Czech Academy of Sciences v.v.i. , Flemingovo nám. 2, 166 10 Prague 6, Czech Republic
| | - Natalya Panova
- Institute of Microbiology, Czech Academy of Sciences v.v.i. , Vídeňská 1083, 142 20 Prague 4, Czech Republic
| | - Milan Kolář
- Department of Microbiology, Faculty of Medicine and Dentistry, Palacký University Olomouc , Hněvotínská 3, 775 15 Olomouc, Czech Republic
| | - Kateřina Bogdanová
- Department of Microbiology, Faculty of Medicine and Dentistry, Palacký University Olomouc , Hněvotínská 3, 775 15 Olomouc, Czech Republic
| | - Renata Večeřová
- Department of Microbiology, Faculty of Medicine and Dentistry, Palacký University Olomouc , Hněvotínská 3, 775 15 Olomouc, Czech Republic
| | - Radovan Fišer
- Department of Genetics and Microbiology, Faculty of Science, Charles University , Viničná 5, 128 43 Prague 2, Czech Republic
| | - Hana Šanderová
- Institute of Microbiology, Czech Academy of Sciences v.v.i. , Vídeňská 1083, 142 20 Prague 4, Czech Republic
| | - Dragana Vítovská
- Institute of Microbiology, Czech Academy of Sciences v.v.i. , Vídeňská 1083, 142 20 Prague 4, Czech Republic
| | - Petra Sudzinová
- Department of Genetics and Microbiology, Faculty of Science, Charles University , Viničná 5, 128 43 Prague 2, Czech Republic.,Institute of Microbiology, Czech Academy of Sciences v.v.i. , Vídeňská 1083, 142 20 Prague 4, Czech Republic
| | - Jiří Pospíšil
- Department of Genetics and Microbiology, Faculty of Science, Charles University , Viničná 5, 128 43 Prague 2, Czech Republic.,Institute of Microbiology, Czech Academy of Sciences v.v.i. , Vídeňská 1083, 142 20 Prague 4, Czech Republic
| | - Oldřich Benada
- Institute of Microbiology, Czech Academy of Sciences v.v.i. , Vídeňská 1083, 142 20 Prague 4, Czech Republic
| | - Tomáš Křížek
- Department of Analytical Chemistry, Faculty of Science, Charles University , Albertov 6, 128 43 Prague 2, Czech Republic
| | - David Sedlák
- Institute of Molecular Genetics, Czech Academy of Sciences v.v.i. , Vídeňská 1083, 142 20 Prague 4, Czech Republic
| | - Petr Bartůněk
- Institute of Molecular Genetics, Czech Academy of Sciences v.v.i. , Vídeňská 1083, 142 20 Prague 4, Czech Republic
| | - Libor Krásný
- Institute of Microbiology, Czech Academy of Sciences v.v.i. , Vídeňská 1083, 142 20 Prague 4, Czech Republic
| | - Dominik Rejman
- Institute of Organic Chemistry and Biochemistry, Czech Academy of Sciences v.v.i. , Flemingovo nám. 2, 166 10 Prague 6, Czech Republic
| |
Collapse
|
402
|
Yavvari PS, Gupta S, Arora D, Nandicoori VK, Srivastava A, Bajaj A. Clathrin-Independent Killing of Intracellular Mycobacteria and Biofilm Disruptions Using Synthetic Antimicrobial Polymers. Biomacromolecules 2017; 18:2024-2033. [DOI: 10.1021/acs.biomac.7b00106] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Affiliation(s)
- Prabhu S. Yavvari
- Department
of Chemistry, Indian Institute of Science Education and Research, Bhopal By-pass Road, Bhopal-462066, Madhya Pradesh, India
| | - Siddhi Gupta
- Laboratory
of Nanotechnology and Chemical Biology, Regional Centre for Biotechnology, Third Milestone Faridabad-Gurgaon Expressway, NCR Biotech Cluster, Faridabad-121001, Haryana, India
| | - Divya Arora
- National Institute of Immunology, Aruna Asaf Ali Marg, New Delhi, Delhi-110067, India
| | - Vinay K. Nandicoori
- National Institute of Immunology, Aruna Asaf Ali Marg, New Delhi, Delhi-110067, India
| | - Aasheesh Srivastava
- Department
of Chemistry, Indian Institute of Science Education and Research, Bhopal By-pass Road, Bhopal-462066, Madhya Pradesh, India
| | - Avinash Bajaj
- Laboratory
of Nanotechnology and Chemical Biology, Regional Centre for Biotechnology, Third Milestone Faridabad-Gurgaon Expressway, NCR Biotech Cluster, Faridabad-121001, Haryana, India
| |
Collapse
|
403
|
Identification and synthesis of novel inhibitors of mycobacterium ATP synthase. Bioorg Med Chem Lett 2017; 27:3454-3459. [PMID: 28587823 DOI: 10.1016/j.bmcl.2017.05.081] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2017] [Revised: 04/24/2017] [Accepted: 05/26/2017] [Indexed: 12/17/2022]
Abstract
A non-diaryl quinoline scaffold 6,7-dihydropyrazolo[1,5-a]pyrazin-4-one was identified by screening of diverse set of compounds against M. smegmatis ATP synthase. Herein, we disclose our efforts to develop the structure activity relationship against Mycobacterium tuberculosis (Mtb.H37Rv strain) around the identified hit 1. A scaffold hopping approach was used to identify compounds 14a, 14b and 24a with improved activity against MTb.H37Rv.
Collapse
|
404
|
Meir O, Zaknoon F, Cogan U, Mor A. A broad-spectrum bactericidal lipopeptide with anti-biofilm properties. Sci Rep 2017; 7:2198. [PMID: 28526864 PMCID: PMC5438364 DOI: 10.1038/s41598-017-02373-0] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2016] [Accepted: 04/10/2017] [Indexed: 11/30/2022] Open
Abstract
Previous studies of the oligoacyllysyl (OAK) series acyl-lysyl-lysyl-aminoacyl-lysine-amide, suggested their utility towards generating robust linear lipopeptide-like alternatives to antibiotics, although to date, none exhibited potent broad-spectrum bactericidal activity. To follow up on this premise, we produced a new analog (C14KKc12K) and investigated its properties in various media. Mechanistic studies suggest that C14KKc12K uses a non-specific membrane-disruptive mode of action for rapidly reducing viability of Gram-negative bacteria (GNB) similarly to polymyxin B (PMB), a cyclic lipopeptide used as last resort antibiotic. Indeed, C14KKc12K displayed similar affinity for lipopolysaccharides and induced cell permeabilization associated with rapid massive membrane depolarization. Unlike PMB however, C14KKc12K was also bactericidal to Gram-positive bacteria (GPB) at or near the minimal inhibitory concentration (MIC), as assessed against a multispecies panel of >50 strains, displaying MIC50 at 3 and 6 µM, respectively for GPB and GNB. C14KKc12K retained activity in human saliva, reducing the viability of cultivable oral microflora by >99% within two minutes of exposure, albeit at higher concentrations, which, nonetheless, were similar to the commercial gold standard, chlorhexidine. This equipotent bactericidal activity was also observed in pre-formed biofilms of Streptococcus mutans, a major periodontal pathogen. Such compounds therefore, may be useful for eradication of challenging poly-microbial infections.
Collapse
Affiliation(s)
- Ohad Meir
- Department of Biotechnology & Food Engineering, Technion-Israel Institute of Technology, Haifa, 32000, Israel
| | - Fadia Zaknoon
- Department of Biotechnology & Food Engineering, Technion-Israel Institute of Technology, Haifa, 32000, Israel
| | - Uri Cogan
- Department of Biotechnology & Food Engineering, Technion-Israel Institute of Technology, Haifa, 32000, Israel
| | - Amram Mor
- Department of Biotechnology & Food Engineering, Technion-Israel Institute of Technology, Haifa, 32000, Israel.
| |
Collapse
|
405
|
Yuan Y, Zhang Y. Synthesis of Imidazolium Oligomers with Planar and Stereo Cores and Their Antimicrobial Applications. ChemMedChem 2017; 12:835-840. [DOI: 10.1002/cmdc.201700167] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2017] [Revised: 05/03/2017] [Indexed: 12/19/2022]
Affiliation(s)
- Yuan Yuan
- Institute of Bioengineering and Nanotechnology; 31 Biopolis Way, The Nanos Singapore 138669 Singapore
| | - Yugen Zhang
- Institute of Bioengineering and Nanotechnology; 31 Biopolis Way, The Nanos Singapore 138669 Singapore
| |
Collapse
|
406
|
Kalily E, Hollander A, Korin B, Cymerman I, Yaron S. Adaptation of Salmonella enterica Serovar Senftenberg to Linalool and Its Association with Antibiotic Resistance and Environmental Persistence. Appl Environ Microbiol 2017; 83:e03398-16. [PMID: 28258149 PMCID: PMC5411494 DOI: 10.1128/aem.03398-16] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2016] [Accepted: 02/22/2017] [Indexed: 11/20/2022] Open
Abstract
A clinical isolate of Salmonella enterica serovar Senftenberg, isolated from an outbreak linked to the herb Ocimum basilicum L. (basil), has been shown to be resistant to basil oil and to the terpene alcohol linalool. To better understand how human pathogens might develop resistance to linalool and to investigate the association of this resistance with resistance to different antimicrobial agents, selective pressure was applied to the wild-type strain by sequential exposure to increasing concentrations of linalool. The results demonstrated that S Senftenberg adapted to linalool with a MIC increment of at least 8-fold, which also resulted in better resistance to basil oil and better survival on harvested basil leaves. Adaptation to linalool was shown to confer cross protection against the antibiotics trimethoprim, sulfamethoxazole, piperacillin, chloramphenicol, and tetracycline, increasing their MICs by 2- to 32-fold. The improved resistance was shown to correlate with multiple phenotypes that included changes in membrane fatty acid composition, induced efflux, reduced influx, controlled motility, and the ability to form larger aggregates in the presence of linalool. The adaptation to linalool obtained in vitro did not affect survival on the basil phyllosphere in planta and even diminished survival in soil, suggesting that development of extreme resistance to linalool may be accompanied by a loss of fitness. Altogether, this report notes the concern regarding the ability of human pathogens to develop resistance to commercial essential oils, a resistance that is also associated with cross-resistance to antibiotics and may endanger public health.IMPORTANCE Greater consumer awareness and concern regarding synthetic chemical additives have led producers to control microbial spoilage and hazards by the use of natural preservatives, such as plant essential oils with antimicrobial activity. This report establishes, however, that these compounds may provoke the emergence of resistant human pathogens. Herein, we demonstrate the acquisition of resistance to basil oil by Salmonella Senftenberg. Exposure to linalool, a component of basil oil, resulted in adaptation to the basil oil mixture, as well as cross protection against several antibiotics and better survival on harvested basil leaves. Collectively, this work highlights the hazard to public health while using plant essential oils without sufficient knowledge about their influence on pathogens at subinhibitory concentrations.
Collapse
Affiliation(s)
- Emmanuel Kalily
- Faculty of Biotechnology and Food Engineering and the Russell Berrie Nanotechnology Institute, Technion-Israel Institute of Technology, Haifa, Israel
| | - Amit Hollander
- Faculty of Biotechnology and Food Engineering and the Russell Berrie Nanotechnology Institute, Technion-Israel Institute of Technology, Haifa, Israel
| | - Ben Korin
- Faculty of Biotechnology and Food Engineering and the Russell Berrie Nanotechnology Institute, Technion-Israel Institute of Technology, Haifa, Israel
| | - Itamar Cymerman
- Faculty of Biotechnology and Food Engineering and the Russell Berrie Nanotechnology Institute, Technion-Israel Institute of Technology, Haifa, Israel
| | - Sima Yaron
- Faculty of Biotechnology and Food Engineering and the Russell Berrie Nanotechnology Institute, Technion-Israel Institute of Technology, Haifa, Israel
| |
Collapse
|
407
|
Zhang S, Shi W, Feng J, Zhang W, Zhang Y. Varying effects of common tuberculosis drugs on enhancing clofazimine activity in vitro. Emerg Microbes Infect 2017; 6:e28. [PMID: 28442751 PMCID: PMC5457683 DOI: 10.1038/emi.2017.24] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2017] [Revised: 03/09/2017] [Accepted: 03/10/2017] [Indexed: 11/29/2022]
Affiliation(s)
- Shuo Zhang
- Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD 21205, USA.,Shandong Medicinal Biotechnology Centre, Shandong Academy of Medical Sciences, Jinan 250062, China
| | - Wanliang Shi
- Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Jie Feng
- Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Wenhong Zhang
- Department of Infectious Diseases, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Ying Zhang
- Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD 21205, USA.,Department of Infectious Diseases, Huashan Hospital, Fudan University, Shanghai 200040, China
| |
Collapse
|
408
|
Niu H, Ma C, Cui P, Shi W, Zhang S, Feng J, Sullivan D, Zhu B, Zhang W, Zhang Y. Identification of drug candidates that enhance pyrazinamide activity from a clinical compound library. Emerg Microbes Infect 2017; 6:e27. [PMID: 28442749 PMCID: PMC5457674 DOI: 10.1038/emi.2017.23] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2016] [Revised: 03/05/2017] [Accepted: 03/14/2017] [Indexed: 12/04/2022]
Affiliation(s)
- Hongxia Niu
- Lanzhou Center for Tuberculosis Research and Institute of Pathogenic Biology, School of Basic Medical Sciences, Lanzhou University, Lanzhou 730000, China.,Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Chao Ma
- College of Biological Sciences and Technology, Beijing Forestry University, Beijing 100083, China
| | - Peng Cui
- Key Laboratory of Medical Molecular Virology, Department of Infectious Diseases, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai 200040, China
| | - Wanliang Shi
- Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Shuo Zhang
- Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Jie Feng
- Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD 21205, USA
| | - David Sullivan
- Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Bingdong Zhu
- Lanzhou Center for Tuberculosis Research and Institute of Pathogenic Biology, School of Basic Medical Sciences, Lanzhou University, Lanzhou 730000, China
| | - Wenhong Zhang
- Key Laboratory of Medical Molecular Virology, Department of Infectious Diseases, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai 200040, China
| | - Ying Zhang
- Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD 21205, USA.,Key Laboratory of Medical Molecular Virology, Department of Infectious Diseases, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai 200040, China
| |
Collapse
|
409
|
Yang X, Goswami S, Gorityala BK, Domalaon R, Lyu Y, Kumar A, Zhanel GG, Schweizer F. A Tobramycin Vector Enhances Synergy and Efficacy of Efflux Pump Inhibitors against Multidrug-Resistant Gram-Negative Bacteria. J Med Chem 2017; 60:3913-3932. [DOI: 10.1021/acs.jmedchem.7b00156] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Affiliation(s)
- Xuan Yang
- Department
of Chemistry, University of Manitoba, Winnipeg, MB R3T 2N2, Canada
| | - Sudeep Goswami
- Department
of Chemistry, University of Manitoba, Winnipeg, MB R3T 2N2, Canada
| | | | - Ronald Domalaon
- Department
of Chemistry, University of Manitoba, Winnipeg, MB R3T 2N2, Canada
| | - Yinfeng Lyu
- Department
of Chemistry, University of Manitoba, Winnipeg, MB R3T 2N2, Canada
- Institute
of Animal Nutrition, Northeast Agricultural University, Harbin, Heilongjiang 150030, P.R. China
| | - Ayush Kumar
- Department
of Microbiology, University of Manitoba, Winnipeg, MB R3T 2N2, Canada
- Department
of Medical Microbiology, University of Manitoba, Winnipeg, MB R3T 1R9, Canada
| | - George G. Zhanel
- Department
of Medical Microbiology, University of Manitoba, Winnipeg, MB R3T 1R9, Canada
| | - Frank Schweizer
- Department
of Chemistry, University of Manitoba, Winnipeg, MB R3T 2N2, Canada
- Department
of Medical Microbiology, University of Manitoba, Winnipeg, MB R3T 1R9, Canada
| |
Collapse
|
410
|
Kim W, Hendricks GL, Lee K, Mylonakis E. An update on the use of C. elegans for preclinical drug discovery: screening and identifying anti-infective drugs. Expert Opin Drug Discov 2017; 12:625-633. [PMID: 28402221 DOI: 10.1080/17460441.2017.1319358] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
INTRODUCTION The emergence of antibiotic-resistant and -tolerant bacteria is a major threat to human health. Although efforts for drug discovery are ongoing, conventional bacteria-centered screening strategies have thus far failed to yield new classes of effective antibiotics. Therefore, new paradigms for discovering novel antibiotics are of critical importance. Caenorhabditis elegans, a model organism used for in vivo, offers a promising solution for identification of anti-infective compounds. Areas covered: This review examines the advantages of C. elegans-based high-throughput screening over conventional, bacteria-centered in vitro screens. It discusses major anti-infective compounds identified from large-scale C. elegans-based screens and presents the first clinically-approved drugs, then known bioactive compounds, and finally novel small molecules. Expert opinion: There are clear advantages of using a C. elegans-infection based screening method. A C. elegans-based screen produces an enriched pool of non-toxic, efficacious, potential anti-infectives, covering: conventional antimicrobial agents, immunomodulators, and anti-virulence agents. Although C. elegans-based screens do not denote the mode of action of hit compounds, this can be elucidated in secondary studies by comparing the results to target-based screens, or conducting subsequent target-based screens, including the genetic knock-down of host or bacterial genes.
Collapse
Affiliation(s)
- Wooseong Kim
- a Division of Infectious Diseases , Rhode Island Hospital, Alpert Medical School of Brown University , Providence , RI , USA
| | - Gabriel Lambert Hendricks
- a Division of Infectious Diseases , Rhode Island Hospital, Alpert Medical School of Brown University , Providence , RI , USA
| | - Kiho Lee
- a Division of Infectious Diseases , Rhode Island Hospital, Alpert Medical School of Brown University , Providence , RI , USA
| | - Eleftherios Mylonakis
- a Division of Infectious Diseases , Rhode Island Hospital, Alpert Medical School of Brown University , Providence , RI , USA
| |
Collapse
|
411
|
Takahashi H, Caputo GA, Vemparala S, Kuroda K. Synthetic Random Copolymers as a Molecular Platform To Mimic Host-Defense Antimicrobial Peptides. Bioconjug Chem 2017; 28:1340-1350. [PMID: 28379682 DOI: 10.1021/acs.bioconjchem.7b00114] [Citation(s) in RCA: 99] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Synthetic polymers have been used as a molecular platform to develop host-defense antimicrobial peptide (AMP) mimetics which are effective in killing drug-resistant bacteria. In this topical review, we will discuss the AMP-mimetic design and chemical optimization strategies as well as the biological and biophysical implications of AMP mimicry by synthetic polymers. Traditionally, synthetic polymers have been used as a chemical means to replicate the chemical functionalities and physicochemical properties of AMPs (e.g., cationic charge, hydrophobicity) to recapitulate their mode of action. However, we propose a new perception that AMP-mimetic polymers are an inherently bioactive platform as whole molecules, which mimic more than the side chain functionalities of AMPs. The tunable nature and chemical simplicity of synthetic random polymers facilitate the development of potent, cost-effective, broad-spectrum antimicrobials. The polymer-based approach offers the potential for many antimicrobial applications to be used directly in solution or attached to surfaces to fight against drug-resistant bacteria.
Collapse
Affiliation(s)
- Haruko Takahashi
- Center for International Research on Integrative Biomedical Systems, Institute of Industrial Science, The University of Tokyo , Tokyo, 153-8505, Japan
| | | | - Satyavani Vemparala
- The Institute of Mathematical Sciences , C.I.T. Campus, Taramani, Chennai, 600113, India
| | - Kenichi Kuroda
- Department of Biologic and Materials Sciences, School of Dentistry, University of Michigan , Ann Arbor, Michigan 48109, United States
| |
Collapse
|
412
|
Novel therapeutics for bacterial infections. Emerg Top Life Sci 2017; 1:85-92. [PMID: 33525811 DOI: 10.1042/etls20160017] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2017] [Revised: 02/22/2017] [Accepted: 02/22/2017] [Indexed: 01/28/2023]
Abstract
The relentless increase in antibiotic resistance among all major groups of bacterial pathogens shows no sign of abating. The situation is exacerbated by a marked decline in the number of new antibiotics entering the marketplace. It is essential that new ways to treat severe bacterial infections are investigated before the antibiotic well runs dry. This review covers many promising approaches, some novel and some based on old ideas that were not considered viable when clinicians were able to exploit a wide palette of cheap and effective antibacterial chemotherapeutics. These approaches include the use of photosensitive dyes, bacteriophage and phage-encoded proteins, and agents that compromise virulence and antibiotic-resistance machineries. I also make a case for continuing in some form with tried and trusted platforms for drug discovery that served society well in the past.
Collapse
|
413
|
Zeng X, Wu P, Yao C, Liang J, Zhang S, Yin H. Small Molecule and Peptide Recognition of Protein Transmembrane Domains. Biochemistry 2017; 56:2076-2085. [DOI: 10.1021/acs.biochem.6b00909] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Affiliation(s)
- Xianfeng Zeng
- Center
of Basic Molecular Science, Department of Chemistry, Tsinghua University, Beijing 100082, China
| | - Peiyao Wu
- Center
of Basic Molecular Science, Department of Chemistry, Tsinghua University, Beijing 100082, China
| | - Chengbo Yao
- Center
of Basic Molecular Science, Department of Chemistry, Tsinghua University, Beijing 100082, China
| | - Jiaqi Liang
- Center
of Basic Molecular Science, Department of Chemistry, Tsinghua University, Beijing 100082, China
| | - Shuting Zhang
- Center
of Basic Molecular Science, Department of Chemistry, Tsinghua University, Beijing 100082, China
- School
of Chemistry and Chemical Engineering, Henan Normal University, Xinxiang 453007, China
| | - Hang Yin
- Center
of Basic Molecular Science, Department of Chemistry, Tsinghua University, Beijing 100082, China
| |
Collapse
|
414
|
The antimicrobial effects of the alginate oligomer OligoG CF-5/20 are independent of direct bacterial cell membrane disruption. Sci Rep 2017; 7:44731. [PMID: 28361894 PMCID: PMC5374485 DOI: 10.1038/srep44731] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2016] [Accepted: 02/13/2017] [Indexed: 12/19/2022] Open
Abstract
Concerns about acquisition of antibiotic resistance have led to increasing demand for new antimicrobial therapies. OligoG CF-5/20 is an alginate oligosaccharide previously shown to have antimicrobial and antibiotic potentiating activity. We investigated the structural modification of the bacterial cell wall by OligoG CF-5/20 and its effect on membrane permeability. Binding of OligoG CF-5/20 to the bacterial cell surface was demonstrated in Gram-negative bacteria. Permeability assays revealed that OligoG CF-5/20 had virtually no membrane-perturbing effects. Lipopolysaccharide (LPS) surface charge and aggregation were unaltered in the presence of OligoG CF-5/20. Small angle neutron scattering and circular dichroism spectroscopy showed no substantial change to the structure of LPS in the presence of OligoG CF-5/20, however, isothermal titration calorimetry demonstrated a weak calcium-mediated interaction. Metabolomic analysis confirmed no change in cellular metabolic response to a range of osmolytes when treated with OligoG CF-5/20. This data shows that, although weak interactions occur between LPS and OligoG CF-5/20 in the presence of calcium, the antimicrobial effects of OligoG CF-5/20 are not related to the induction of structural alterations in the LPS or cell permeability. These results suggest a novel mechanism of action that may avoid the common route in acquisition of resistance via LPS structural modification.
Collapse
|
415
|
Yang T, Moreira W, Nyantakyi SA, Chen H, Aziz DB, Go ML, Dick T. Amphiphilic Indole Derivatives as Antimycobacterial Agents: Structure–Activity Relationships and Membrane Targeting Properties. J Med Chem 2017; 60:2745-2763. [DOI: 10.1021/acs.jmedchem.6b01530] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Affiliation(s)
- Tianming Yang
- Department
of Pharmacy and ‡Department of Microbiology and Immunology, National University of Singapore, Singapore 117543, Singapore
| | - Wilfried Moreira
- Department
of Pharmacy and ‡Department of Microbiology and Immunology, National University of Singapore, Singapore 117543, Singapore
| | - Samuel Agyei Nyantakyi
- Department
of Pharmacy and ‡Department of Microbiology and Immunology, National University of Singapore, Singapore 117543, Singapore
| | - Huan Chen
- Department
of Pharmacy and ‡Department of Microbiology and Immunology, National University of Singapore, Singapore 117543, Singapore
| | - Dinah binte Aziz
- Department
of Pharmacy and ‡Department of Microbiology and Immunology, National University of Singapore, Singapore 117543, Singapore
| | - Mei-Lin Go
- Department
of Pharmacy and ‡Department of Microbiology and Immunology, National University of Singapore, Singapore 117543, Singapore
| | - Thomas Dick
- Department
of Pharmacy and ‡Department of Microbiology and Immunology, National University of Singapore, Singapore 117543, Singapore
| |
Collapse
|
416
|
Ran R, Zeng H, Zhao D, Liu R, Xu X. The Novel Property of Heptapeptide of Microcin C7 in Affecting the Cell Growth of Escherichia coli. Molecules 2017; 22:E432. [PMID: 28282893 PMCID: PMC6155343 DOI: 10.3390/molecules22030432] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2016] [Accepted: 03/06/2017] [Indexed: 12/01/2022] Open
Abstract
Microcin C7 (McC), widely distributed in enterobacteria, is a promising antibiotic against antibiotic resistance [...].
Collapse
Affiliation(s)
- Rensen Ran
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing 100190, China.
- School of Chemistry and Chemical Engineering, University of Chinese Academy of Sciences, Beijing 100049, China.
| | - Huan Zeng
- College of Chemical Engineering, Beijing University of Chemical Technology, Beijing 100029, China.
| | - Dong Zhao
- College of Chemical Engineering, Beijing University of Chemical Technology, Beijing 100029, China.
| | - Ruiyuan Liu
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing 100190, China.
- School of Chemistry and Chemical Engineering, University of Chinese Academy of Sciences, Beijing 100049, China.
| | - Xia Xu
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing 100190, China.
| |
Collapse
|
417
|
Jammal J, Zaknoon F, Kaneti G, Hershkovits AS, Mor A. Sensitization of Gram-Negative Bacilli to Host Antibacterial Proteins. J Infect Dis 2017; 215:1599-1607. [DOI: 10.1093/infdis/jix119] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2016] [Accepted: 03/02/2017] [Indexed: 12/17/2022] Open
|
418
|
Repurposing Toremifene for Treatment of Oral Bacterial Infections. Antimicrob Agents Chemother 2017; 61:AAC.01846-16. [PMID: 27993858 DOI: 10.1128/aac.01846-16] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2016] [Accepted: 12/11/2016] [Indexed: 12/24/2022] Open
Abstract
The spread of antibiotic resistance and the challenges associated with antiseptics such as chlorhexidine have necessitated a search for new antibacterial agents against oral bacterial pathogens. As a result of failing traditional approaches, drug repurposing has emerged as a novel paradigm to find new antibacterial agents. In this study, we examined the effects of the FDA-approved anticancer agent toremifene against the oral bacteria Porphyromonas gingivalis and Streptococcus mutans We found that the drug was able to inhibit the growth of both pathogens, as well as prevent biofilm formation, at concentrations ranging from 12.5 to 25 μM. Moreover, toremifene was shown to eradicate preformed biofilms at concentrations ranging from 25 to 50 μM. In addition, we found that toremifene prevents P. gingivalis and S. mutans biofilm formation on titanium surfaces. A time-kill study indicated that toremifene is bactericidal against S. mutans Macromolecular synthesis assays revealed that treatment with toremifene does not cause preferential inhibition of DNA, RNA, or protein synthesis pathways, indicating membrane-damaging activity. Biophysical studies using fluorescent probes and fluorescence microscopy further confirmed the membrane-damaging mode of action. Taken together, our results suggest that the anticancer agent toremifene is a suitable candidate for further investigation for the development of new treatment strategies for oral bacterial infections.
Collapse
|
419
|
Van den Bergh B, Fauvart M, Michiels J. Formation, physiology, ecology, evolution and clinical importance of bacterial persisters. FEMS Microbiol Rev 2017; 41:219-251. [DOI: 10.1093/femsre/fux001] [Citation(s) in RCA: 217] [Impact Index Per Article: 27.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2016] [Accepted: 01/12/2017] [Indexed: 12/19/2022] Open
|
420
|
Hubbard ATM, Barker R, Rehal R, Vandera KKA, Harvey RD, Coates ARM. Mechanism of Action of a Membrane-Active Quinoline-Based Antimicrobial on Natural and Model Bacterial Membranes. Biochemistry 2017; 56:1163-1174. [PMID: 28156093 DOI: 10.1021/acs.biochem.6b01135] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
HT61 is a quinoline-derived antimicrobial, which exhibits bactericidal potency against both multiplying and quiescent methicillin resistant and sensitive Staphylococcus aureus, and has been proposed as an adjunct for other antimicrobials to extend their usefulness in the face of increasing antimicrobial resistance. In this study, we have examined HT61's effect on the permeability of S. aureus membranes and whether this putative activity can be attributed to an interaction with lipid bilayers. Using membrane potential and ATP release assays, we have shown that HT61 disrupts the membrane enough to result in depolarization of the membrane and release of intercellular constituents at concentrations above and below the minimum inhibitory concentration of the drug. Utilizing both monolayer subphase injection and neutron reflectometry, we have shown that increasing the anionic lipid content of the membrane leads to a more marked effect of the drug. In bilayers containing 25 mol % phosphatidylglycerol, neutron reflectometry data suggest that exposure to HT61 increases the level of solvent in the hydrophobic region of the membrane, which is indicative of gross structural damage. Increasing the proportion of PG elicits a concomitant level of membrane damage, resulting in almost total destruction when 75 mol % phosphatidylglycerol is present. We therefore propose that HT61's primary action is directed toward the cytoplasmic membrane of Gram-positive bacteria.
Collapse
Affiliation(s)
- Alasdair T M Hubbard
- Medical Microbiology, Institute for Infection and Immunity, St George's, University of London , Cranmer Terrace, London SW17 ORE, U.K
| | - Robert Barker
- Institut Laue Langevin , 71 avenue des Martyrs, 38042 Grenoble, France
| | - Reg Rehal
- Institute of Pharmaceutical Science, King's College London , Franklin Wilkins Building, 150 Stamford Street, London SE1 9NH, U.K
| | - Kalliopi-Kelli A Vandera
- Institute of Pharmaceutical Science, King's College London , Franklin Wilkins Building, 150 Stamford Street, London SE1 9NH, U.K
| | - Richard D Harvey
- Institute of Pharmaceutical Science, King's College London , Franklin Wilkins Building, 150 Stamford Street, London SE1 9NH, U.K
| | - Anthony R M Coates
- Medical Microbiology, Institute for Infection and Immunity, St George's, University of London , Cranmer Terrace, London SW17 ORE, U.K
| |
Collapse
|
421
|
Zhang M, Zhu PP, Xin P, Si W, Li ZT, Hou JL. Synthetic Channel Specifically Inserts into the Lipid Bilayer of Gram-Positive Bacteria but not that of Mammalian Erythrocytes. Angew Chem Int Ed Engl 2017. [DOI: 10.1002/ange.201612093] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Affiliation(s)
- Min Zhang
- Department of Chemistry; Fudan University; 220 Handan Road Shanghai 200433 China
| | - Ping-Ping Zhu
- Department of Chemistry; Fudan University; 220 Handan Road Shanghai 200433 China
| | - Pengyang Xin
- Department of Chemistry; Fudan University; 220 Handan Road Shanghai 200433 China
| | - Wen Si
- Department of Chemistry; Fudan University; 220 Handan Road Shanghai 200433 China
| | - Zhan-Ting Li
- Department of Chemistry; Fudan University; 220 Handan Road Shanghai 200433 China
| | - Jun-Li Hou
- Department of Chemistry; Fudan University; 220 Handan Road Shanghai 200433 China
| |
Collapse
|
422
|
Zhang M, Zhu PP, Xin P, Si W, Li ZT, Hou JL. Synthetic Channel Specifically Inserts into the Lipid Bilayer of Gram-Positive Bacteria but not that of Mammalian Erythrocytes. Angew Chem Int Ed Engl 2017; 56:2999-3003. [DOI: 10.1002/anie.201612093] [Citation(s) in RCA: 78] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2016] [Revised: 12/29/2016] [Indexed: 11/09/2022]
Affiliation(s)
- Min Zhang
- Department of Chemistry; Fudan University; 220 Handan Road Shanghai 200433 China
| | - Ping-Ping Zhu
- Department of Chemistry; Fudan University; 220 Handan Road Shanghai 200433 China
| | - Pengyang Xin
- Department of Chemistry; Fudan University; 220 Handan Road Shanghai 200433 China
| | - Wen Si
- Department of Chemistry; Fudan University; 220 Handan Road Shanghai 200433 China
| | - Zhan-Ting Li
- Department of Chemistry; Fudan University; 220 Handan Road Shanghai 200433 China
| | - Jun-Li Hou
- Department of Chemistry; Fudan University; 220 Handan Road Shanghai 200433 China
| |
Collapse
|
423
|
Liu Y, Ding S, Dietrich R, Märtlbauer E, Zhu K. A Biosurfactant‐Inspired Heptapeptide with Improved Specificity to Kill MRSA. Angew Chem Int Ed Engl 2017; 56:1486-1490. [PMID: 28106348 DOI: 10.1002/anie.201609277] [Citation(s) in RCA: 86] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2016] [Revised: 11/16/2016] [Indexed: 02/06/2023]
Affiliation(s)
- Yuan Liu
- Beijing Advanced Innovation Center for Food Nutrition and Human HealthNational Center for Veterinary Drug Safety EvaluationCollege of Veterinary MedicineChina Agricultural University Yuanmingyuan West Road No.2 Beijing 100193 China
| | - Shuangyang Ding
- Beijing Advanced Innovation Center for Food Nutrition and Human HealthNational Center for Veterinary Drug Safety EvaluationCollege of Veterinary MedicineChina Agricultural University Yuanmingyuan West Road No.2 Beijing 100193 China
| | - Richard Dietrich
- Institute of Food SafetyDepartment of Veterinary SciencesLudwig-Maximilians-University Munich 85764 Oberschleißheim Germany
| | - Erwin Märtlbauer
- Institute of Food SafetyDepartment of Veterinary SciencesLudwig-Maximilians-University Munich 85764 Oberschleißheim Germany
| | - Kui Zhu
- Beijing Advanced Innovation Center for Food Nutrition and Human HealthNational Center for Veterinary Drug Safety EvaluationCollege of Veterinary MedicineChina Agricultural University Yuanmingyuan West Road No.2 Beijing 100193 China
- Department of Biomedical EngineeringDuke University Durham NC 27708 USA
| |
Collapse
|
424
|
Hamoen LW, Wenzel M. Editorial: Antimicrobial Peptides - Interaction with Membrane Lipids and Proteins. Front Cell Dev Biol 2017; 5:4. [PMID: 28203562 PMCID: PMC5285327 DOI: 10.3389/fcell.2017.00004] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2016] [Accepted: 01/18/2017] [Indexed: 01/18/2023] Open
Affiliation(s)
- Leendert W Hamoen
- Bacterial Cell Biology, Swammerdam Institute for Life Sciences, University of Amsterdam Amsterdam, Netherlands
| | - Michaela Wenzel
- Bacterial Cell Biology, Swammerdam Institute for Life Sciences, University of Amsterdam Amsterdam, Netherlands
| |
Collapse
|
425
|
Targeting biofilms and persisters of ESKAPE pathogens with P14KanS, a kanamycin peptide conjugate. Biochim Biophys Acta Gen Subj 2017; 1861:848-859. [PMID: 28132897 DOI: 10.1016/j.bbagen.2017.01.029] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2016] [Revised: 01/19/2017] [Accepted: 01/26/2017] [Indexed: 12/12/2022]
Abstract
BACKGROUND The worldwide emergence of antibiotic resistance represents a serious medical threat. The ability of these resistant pathogens to form biofilms that are highly tolerant to antibiotics further aggravates the situation and leads to recurring infections. Thus, new therapeutic approaches that adopt novel mechanisms of action are urgently needed. To address this significant problem, we conjugated the antibiotic kanamycin with a novel antimicrobial peptide (P14LRR) to develop a kanamycin peptide conjugate (P14KanS). METHODS Antibacterial activities were evaluated in vitro and in vivo using a Caenorhabditis elegans model. Additionally, the mechanism of action, antibiofilm activity and anti-inflammatory effect of P14KanS were investigated. RESULTS P14KanS exhibited potent antimicrobial activity against ESKAPE pathogens. P14KanS demonstrated a ≥128-fold improvement in MIC relative to kanamycin against kanamycin-resistant strains. Mechanistic studies confirmed that P14KanS exerts its antibacterial effect by selectively disrupting the bacterial cell membrane. Unlike many antibiotics, P14KanS demonstrated rapid bactericidal activity against stationary phases of both Gram-positive and Gram-negative pathogens. Moreover, P14KanS was superior in disrupting adherent bacterial biofilms and in killing intracellular pathogens as compared to conventional antibiotics. Furthermore, P14KanS demonstrated potent anti-inflammatory activity via the suppression of LPS-induced proinflammatory cytokines. Finally, P14KanS protected C. elegans from lethal infections of both Gram-positive and Gram-negative pathogens. CONCLUSIONS The potent in vitro and in vivo activity of P14KanS warrants further investigation as a potential therapeutic agent for bacterial infections. GENERAL SIGNIFICANCE This study demonstrates that equipping kanamycin with an antimicrobial peptide is a promising method to tackle bacterial biofilms and address bacterial resistance to aminoglycosides.
Collapse
|
426
|
Hijazi S, Visca P, Frangipani E. Gallium-Protoporphyrin IX Inhibits Pseudomonas aeruginosa Growth by Targeting Cytochromes. Front Cell Infect Microbiol 2017; 7:12. [PMID: 28184354 PMCID: PMC5266731 DOI: 10.3389/fcimb.2017.00012] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2016] [Accepted: 01/10/2017] [Indexed: 01/08/2023] Open
Abstract
Pseudomonas aeruginosa is a challenging pathogen due to both innate and acquired resistance to antibiotics. It is capable of causing a variety of infections, including chronic lung infection in cystic fibrosis (CF) patients. Given the importance of iron in bacterial physiology and pathogenicity, iron-uptake and metabolism have become attractive targets for the development of new antibacterial compounds. P. aeruginosa can acquire iron from a variety of sources to fulfill its nutritional requirements both in the environment and in the infected host. The adaptation of P. aeruginosa to heme iron acquisition in the CF lung makes heme utilization pathways a promising target for the development of new anti-Pseudomonas drugs. Gallium [Ga(III)] is an iron mimetic metal which inhibits P. aeruginosa growth by interfering with iron-dependent metabolism. The Ga(III) complex of the heme precursor protoporphyrin IX (GaPPIX) showed enhanced antibacterial activity against several bacterial species, although no inhibitory effect has been reported on P. aeruginosa. Here, we demonstrate that GaPPIX is indeed capable of inhibiting the growth of clinical P. aeruginosa strains under iron-deplete conditions, as those encountered by bacteria during infection, and that GaPPIX inhibition is reversed by iron. Using P. aeruginosa PAO1 as model organism, we show that GaPPIX enters cells through both the heme-uptake systems has and phu, primarily via the PhuR receptor which plays a crucial role in P. aeruginosa adaptation to the CF lung. We also demonstrate that intracellular GaPPIX inhibits the aerobic growth of P. aeruginosa by targeting cytochromes, thus interfering with cellular respiration.
Collapse
Affiliation(s)
- Sarah Hijazi
- Department of Science, Roma Tre University Rome, Italy
| | - Paolo Visca
- Department of Science, Roma Tre University Rome, Italy
| | | |
Collapse
|
427
|
Chen Y, Moloney JG, Christensen KE, Moloney MG. Fused-Ring Oxazolopyrrolopyridopyrimidine Systems with Gram-Negative Activity. Antibiotics (Basel) 2017; 6:antibiotics6010002. [PMID: 28098784 PMCID: PMC5372982 DOI: 10.3390/antibiotics6010002] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2016] [Revised: 01/03/2017] [Accepted: 01/05/2017] [Indexed: 02/07/2023] Open
Abstract
Fused polyheterocyclic derivatives are available by annulation of a tetramate scaffold, and been shown to have antibacterial activity against a Gram-negative, but not a Gram-positive, bacterial strain. While the activity is not potent, these systems are structurally novel showing, in particular, a high level of polarity, and offer potential for the optimization of antibacterial activity.
Collapse
Affiliation(s)
- Yiyuan Chen
- Chemistry Research Laboratory, Department of Chemistry, University of Oxford, 12 Mansfield Road, Oxford OX1 3TA, UK.
| | - Jonathan G Moloney
- Chemistry Research Laboratory, Department of Chemistry, University of Oxford, 12 Mansfield Road, Oxford OX1 3TA, UK.
| | - Kirsten E Christensen
- Chemistry Research Laboratory, Department of Chemistry, University of Oxford, 12 Mansfield Road, Oxford OX1 3TA, UK.
| | - Mark G Moloney
- Chemistry Research Laboratory, Department of Chemistry, University of Oxford, 12 Mansfield Road, Oxford OX1 3TA, UK.
| |
Collapse
|
428
|
Liu Y, Ding S, Dietrich R, Märtlbauer E, Zhu K. A Biosurfactant-Inspired Heptapeptide with Improved Specificity to Kill MRSA. Angew Chem Int Ed Engl 2017. [DOI: 10.1002/ange.201609277] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Affiliation(s)
- Yuan Liu
- Beijing Advanced Innovation Center for Food Nutrition and Human Health; National Center for Veterinary Drug Safety Evaluation; College of Veterinary Medicine; China Agricultural University; Yuanmingyuan West Road No.2 Beijing 100193 China
| | - Shuangyang Ding
- Beijing Advanced Innovation Center for Food Nutrition and Human Health; National Center for Veterinary Drug Safety Evaluation; College of Veterinary Medicine; China Agricultural University; Yuanmingyuan West Road No.2 Beijing 100193 China
| | - Richard Dietrich
- Institute of Food Safety; Department of Veterinary Sciences; Ludwig-Maximilians-University Munich; 85764 Oberschleißheim Germany
| | - Erwin Märtlbauer
- Institute of Food Safety; Department of Veterinary Sciences; Ludwig-Maximilians-University Munich; 85764 Oberschleißheim Germany
| | - Kui Zhu
- Beijing Advanced Innovation Center for Food Nutrition and Human Health; National Center for Veterinary Drug Safety Evaluation; College of Veterinary Medicine; China Agricultural University; Yuanmingyuan West Road No.2 Beijing 100193 China
- Department of Biomedical Engineering; Duke University; Durham NC 27708 USA
| |
Collapse
|
429
|
Zhu R, Gao F, Piao JG, Yang L. Skin-safe photothermal therapy enabled by responsive release of acid-activated membrane-disruptive polymer from polydopamine nanoparticle upon very low laser irradiation. Biomater Sci 2017; 5:1596-1602. [DOI: 10.1039/c7bm00256d] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
How to ablate tumor without damaging skin is a challenge for photothermal therapy.
Collapse
Affiliation(s)
- Rui Zhu
- CAS Key Laboratory of Soft Matter Chemistry
- University of Science and Technology of China
- Hefei
- China
- School of Chemistry and Materials Science
| | - Feng Gao
- CAS Key Laboratory of Soft Matter Chemistry
- University of Science and Technology of China
- Hefei
- China
- School of Chemistry and Materials Science
| | - Ji-Gang Piao
- CAS Key Laboratory of Soft Matter Chemistry
- University of Science and Technology of China
- Hefei
- China
- School of Chemistry and Materials Science
| | - Lihua Yang
- CAS Key Laboratory of Soft Matter Chemistry
- University of Science and Technology of China
- Hefei
- China
- School of Chemistry and Materials Science
| |
Collapse
|
430
|
Singha Mahapatra T, Roy A, Chaudhury S, Dasgupta S, Lal Shrivastava S, Bertolasi V, Ray D. Trapping of a Methanoato Bridge in µ‐1,1,3,3 Mode for [Cu
4
] Aggregate Formation: Synthesis, Steric Control on Nuclearity, Antimicrobial Activity, and DNA‐Interaction Properties. Eur J Inorg Chem 2016. [DOI: 10.1002/ejic.201601092] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Affiliation(s)
| | - Anupam Roy
- Agricultural and Food Engineering Department Indian Institute of Technology Kharagpur West Bengal India
| | | | - Swagata Dasgupta
- Department of Chemistry Indian Institute of Technology 721302 Kharagpur India
| | - Shankar Lal Shrivastava
- Agricultural and Food Engineering Department Indian Institute of Technology Kharagpur West Bengal India
| | - Valerio Bertolasi
- Dipartimento di Scienze Chimiche e Farmaceutiche and Centro di Strutturistica Diffrattometrica Università di Ferrara Via L. Borsari 46 44121 Ferrara Italy
| | - Debashis Ray
- Department of Chemistry Indian Institute of Technology 721302 Kharagpur India
| |
Collapse
|
431
|
Nanostructured polysaccharide-based carriers for antimicrobial peptide delivery. JOURNAL OF PHARMACEUTICAL INVESTIGATION 2016. [DOI: 10.1007/s40005-016-0289-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
|
432
|
Narayanan M, Sekar P, Pasupathi M, Mukhopadhyay T. Self-preserving personal care products. Int J Cosmet Sci 2016; 39:301-309. [PMID: 27761899 DOI: 10.1111/ics.12376] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2016] [Accepted: 10/18/2016] [Indexed: 11/28/2022]
Abstract
OBJECTIVE As questions on the safety of some popular preservatives are on the rise, there is a growing interest in developing 'self-preserving' personal care products. Use of multifunctional ingredients/actives with antimicrobial properties has been explored as replacements for conventional preservatives. This study explores the use of combinations of multifunctional actives (MFA) and other cosmetic ingredients in various personal care formulations, to deliver microbiologically safe self-preserving products. Products studied in this study include face wash, gel-based leave-on skin care product and face mask. METHODS Minimum inhibitory concentration (MIC) of several cosmetic ingredients was determined to identify multifunctional actives with antimicrobial activity. Personal care formulations made with multifunctional actives and other cosmetic ingredients were studied for preservative efficacy by challenging the product with six multiple cycles of microbial challenge. RESULTS Formulations with combinations of multifunctional actives with antioxidant (AO) and chelators (CHL) were found to work synergistically and were highly efficacious in controlling multiple microbial challenges as observed in the preservative efficacy test (PET) studies. The effective combinations were able to withstand up to six multiple microbial challenges without product degradation. The preservative efficacy profile was similar to control formula containing preservatives. CONCLUSION Self-preserving personal care/cosmetic products can be developed which are as efficacious as preserved products by a prudent selection of multifunctional actives, antioxidants and chelators as a part of the formulation.
Collapse
Affiliation(s)
- M Narayanan
- Research Scholar, Bharathiar University, Coimbatore, 641046, India.,CavinKare Research Center, 12 Poonamallee Road, Chennai, 600032, India
| | - P Sekar
- Department of Zoology, Arignar Anna Government Arts College, Namakkal, 637002, India
| | - M Pasupathi
- CavinKare Research Center, 12 Poonamallee Road, Chennai, 600032, India
| | - T Mukhopadhyay
- CavinKare Research Center, 12 Poonamallee Road, Chennai, 600032, India
| |
Collapse
|
433
|
Malik E, Dennison SR, Harris F, Phoenix DA. pH Dependent Antimicrobial Peptides and Proteins, Their Mechanisms of Action and Potential as Therapeutic Agents. Pharmaceuticals (Basel) 2016; 9:ph9040067. [PMID: 27809281 PMCID: PMC5198042 DOI: 10.3390/ph9040067] [Citation(s) in RCA: 87] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2016] [Revised: 10/25/2016] [Accepted: 10/26/2016] [Indexed: 12/16/2022] Open
Abstract
Antimicrobial peptides (AMPs) are potent antibiotics of the innate immune system that have been extensively investigated as a potential solution to the global problem of infectious diseases caused by pathogenic microbes. A group of AMPs that are increasingly being reported are those that utilise pH dependent antimicrobial mechanisms, and here we review research into this area. This review shows that these antimicrobial molecules are produced by a diverse spectrum of creatures, including vertebrates and invertebrates, and are primarily cationic, although a number of anionic examples are known. Some of these molecules exhibit high pH optima for their antimicrobial activity but in most cases, these AMPs show activity against microbes that present low pH optima, which reflects the acidic pH generally found at their sites of action, particularly the skin. The modes of action used by these molecules are based on a number of major structure/function relationships, which include metal ion binding, changes to net charge and conformational plasticity, and primarily involve the protonation of histidine, aspartic acid and glutamic acid residues at low pH. The pH dependent activity of pore forming antimicrobial proteins involves mechanisms that generally differ fundamentally to those used by pH dependent AMPs, which can be described by the carpet, toroidal pore and barrel-stave pore models of membrane interaction. A number of pH dependent AMPs and antimicrobial proteins have been developed for medical purposes and have successfully completed clinical trials, including kappacins, LL-37, histatins and lactoferrin, along with a number of their derivatives. Major examples of the therapeutic application of these antimicrobial molecules include wound healing as well as the treatment of multiple cancers and infections due to viruses, bacteria and fungi. In general, these applications involve topical administration, such as the use of mouth washes, cream formulations and hydrogel delivery systems. Nonetheless, many pH dependent AMPs and antimicrobial proteins have yet to be fully characterized and these molecules, as a whole, represent an untapped source of novel biologically active agents that could aid fulfillment of the urgent need for alternatives to conventional antibiotics, helping to avert a return to the pre-antibiotic era.
Collapse
Affiliation(s)
- Erum Malik
- School of Forensic and Applied Sciences, University of Central Lancashire, Preston PR1 2HE, UK.
| | - Sarah R Dennison
- School of Pharmacy and Biological Sciences, University of Central Lancashire, Preston PR1 2HE, UK.
| | - Frederick Harris
- School of Forensic and Applied Sciences, University of Central Lancashire, Preston PR1 2HE, UK.
| | - David A Phoenix
- Office of the Vice Chancellor, London South Bank University, 103 Borough Road, London SE1 0AA, UK.
| |
Collapse
|
434
|
Daptomycin inhibits cell envelope synthesis by interfering with fluid membrane microdomains. Proc Natl Acad Sci U S A 2016; 113:E7077-E7086. [PMID: 27791134 DOI: 10.1073/pnas.1611173113] [Citation(s) in RCA: 290] [Impact Index Per Article: 32.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
Abstract
Daptomycin is a highly efficient last-resort antibiotic that targets the bacterial cell membrane. Despite its clinical importance, the exact mechanism by which daptomycin kills bacteria is not fully understood. Different experiments have led to different models, including (i) blockage of cell wall synthesis, (ii) membrane pore formation, and (iii) the generation of altered membrane curvature leading to aberrant recruitment of proteins. To determine which model is correct, we carried out a comprehensive mode-of-action study using the model organism Bacillus subtilis and different assays, including proteomics, ionomics, and fluorescence light microscopy. We found that daptomycin causes a gradual decrease in membrane potential but does not form discrete membrane pores. Although we found no evidence for altered membrane curvature, we confirmed that daptomycin inhibits cell wall synthesis. Interestingly, using different fluorescent lipid probes, we showed that binding of daptomycin led to a drastic rearrangement of fluid lipid domains, affecting overall membrane fluidity. Importantly, these changes resulted in the rapid detachment of the membrane-associated lipid II synthase MurG and the phospholipid synthase PlsX. Both proteins preferentially colocalize with fluid membrane microdomains. Delocalization of these proteins presumably is a key reason why daptomycin blocks cell wall synthesis. Finally, clustering of fluid lipids by daptomycin likely causes hydrophobic mismatches between fluid and more rigid membrane areas. This mismatch can facilitate proton leakage and may explain the gradual membrane depolarization observed with daptomycin. Targeting of fluid lipid domains has not been described before for antibiotics and adds another dimension to our understanding of membrane-active antibiotics.
Collapse
|
435
|
Zimmermann L, Das I, Désiré J, Sautrey G, Barros R. S. V, El Khoury M, Mingeot-Leclercq MP, Décout JL. New Broad-Spectrum Antibacterial Amphiphilic Aminoglycosides Active against Resistant Bacteria: From Neamine Derivatives to Smaller Neosamine Analogues. J Med Chem 2016; 59:9350-9369. [DOI: 10.1021/acs.jmedchem.6b00818] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Affiliation(s)
- Louis Zimmermann
- Département
de Pharmacochimie Moléculaire, ICMG FR 2607, University Grenoble Alpes/CNRS, UMR 5063, 470 Rue de la Chimie, BP 53, F-38041 Grenoble, France
| | - Indrajit Das
- Département
de Pharmacochimie Moléculaire, ICMG FR 2607, University Grenoble Alpes/CNRS, UMR 5063, 470 Rue de la Chimie, BP 53, F-38041 Grenoble, France
| | - Jérôme Désiré
- Département
de Pharmacochimie Moléculaire, ICMG FR 2607, University Grenoble Alpes/CNRS, UMR 5063, 470 Rue de la Chimie, BP 53, F-38041 Grenoble, France
| | - Guillaume Sautrey
- Unité
de Pharmacologie Cellulaire et Moléculaire, Louvain Drug Research
Institute, Université Catholique de Louvain, Avenue E.
Mounier 73, B1.73.05, B-1200 Brussels, Belgium
| | - Vinicius Barros R. S.
- Département
de Pharmacochimie Moléculaire, ICMG FR 2607, University Grenoble Alpes/CNRS, UMR 5063, 470 Rue de la Chimie, BP 53, F-38041 Grenoble, France
| | - Micheline El Khoury
- Unité
de Pharmacologie Cellulaire et Moléculaire, Louvain Drug Research
Institute, Université Catholique de Louvain, Avenue E.
Mounier 73, B1.73.05, B-1200 Brussels, Belgium
| | - Marie-Paule Mingeot-Leclercq
- Unité
de Pharmacologie Cellulaire et Moléculaire, Louvain Drug Research
Institute, Université Catholique de Louvain, Avenue E.
Mounier 73, B1.73.05, B-1200 Brussels, Belgium
| | - Jean-Luc Décout
- Département
de Pharmacochimie Moléculaire, ICMG FR 2607, University Grenoble Alpes/CNRS, UMR 5063, 470 Rue de la Chimie, BP 53, F-38041 Grenoble, France
| |
Collapse
|
436
|
Ghosh C, Konai MM, Sarkar P, Samaddar S, Haldar J. Designing Simple Lipidated Lysines: Bifurcation Imparts Selective Antibacterial Activity. ChemMedChem 2016; 11:2367-2371. [DOI: 10.1002/cmdc.201600400] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2016] [Indexed: 11/07/2022]
Affiliation(s)
- Chandradhish Ghosh
- Chemical Biology and Medicinal Chemistry Laboratory, New Chemistry Unit; Jawaharlal Nehru Centre for Advanced Scientific Research (JNCASR), Jakkur, Bengaluru; 560064 Karnataka India
| | - Mohini Mohan Konai
- Chemical Biology and Medicinal Chemistry Laboratory, New Chemistry Unit; Jawaharlal Nehru Centre for Advanced Scientific Research (JNCASR), Jakkur, Bengaluru; 560064 Karnataka India
| | - Paramita Sarkar
- Chemical Biology and Medicinal Chemistry Laboratory, New Chemistry Unit; Jawaharlal Nehru Centre for Advanced Scientific Research (JNCASR), Jakkur, Bengaluru; 560064 Karnataka India
| | - Sandip Samaddar
- Chemical Biology and Medicinal Chemistry Laboratory, New Chemistry Unit; Jawaharlal Nehru Centre for Advanced Scientific Research (JNCASR), Jakkur, Bengaluru; 560064 Karnataka India
| | - Jayanta Haldar
- Chemical Biology and Medicinal Chemistry Laboratory, New Chemistry Unit; Jawaharlal Nehru Centre for Advanced Scientific Research (JNCASR), Jakkur, Bengaluru; 560064 Karnataka India
| |
Collapse
|
437
|
Wang KC, Huang CH, Ding SM, Chen CK, Fang HW, Huang MT, Fang SB. Role of yqiC in the Pathogenicity of Salmonella and Innate Immune Responses of Human Intestinal Epithelium. Front Microbiol 2016; 7:1614. [PMID: 27777572 PMCID: PMC5056187 DOI: 10.3389/fmicb.2016.01614] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2016] [Accepted: 09/27/2016] [Indexed: 01/12/2023] Open
Abstract
The yqiC gene of Salmonella enterica serovar Typhimurium (S. Typhimurium) regulates bacterial growth at different temperatures and mice survival after infection. However, the role of yqiC in bacterial colonization and host immunity remains unknown. We infected human LS174T, Caco-2, HeLa, and THP-1 cells with S. Typhimurium wild-type SL1344, its yqiC mutant, and its complemented strain. Bacterial colonization and internalization in the four cell lines significantly reduced on yqiC depletion. Post-infection production of interleukin-8 and human β-defensin-3 in LS174T cells significantly reduced because of yqiC deleted in S. Typhimurium. The phenotype of yqiC mutant exhibited few and short flagella, fimbriae on the cell surface, enhanced biofilm formation, upregulated type-1 fimbriae expression, and reduced bacterial motility. Type-1 fimbriae, flagella, SPI-1, and SPI-2 gene expression was quantified using real-time PCR. The data show that deletion of yqiC upregulated fimA and fimZ expression and downregulated flhD, fliZ, invA, and sseB expression. Furthermore, thin-layer chromatography and high-performance liquid chromatography revealed the absence of menaquinone in the yqiC mutant, thus validating the importance of yqiC in the bacterial electron transport chain. Therefore, YqiC can negatively regulate FimZ for type-1 fimbriae expression and manipulate the functions of its downstream virulence factors including flagella, SPI-1, and SPI-2 effectors.
Collapse
Affiliation(s)
- Ke-Chuan Wang
- Division of Pediatric Gastroenterology and Hepatology, Department of Pediatrics, Shuang Ho Hospital, Taipei Medical UniversityTaipei, Taiwan; Department of Pediatrics, School of Medicine, College of Medicine, Taipei Medical UniversityTaipei, Taiwan
| | - Chih-Hung Huang
- Graduate Institute of Biochemical and Biomedical Engineering, National Taipei University of Technology Taipei, Taiwan
| | - Shih-Min Ding
- Division of Pediatric Gastroenterology and Hepatology, Department of Pediatrics, Shuang Ho Hospital, Taipei Medical UniversityTaipei, Taiwan; Graduate Institute of Biochemical and Biomedical Engineering, National Taipei University of TechnologyTaipei, Taiwan
| | - Ching-Kuo Chen
- Graduate Institute of Biochemical and Biomedical Engineering, National Taipei University of Technology Taipei, Taiwan
| | - Hsu-Wei Fang
- Graduate Institute of Biochemical and Biomedical Engineering, National Taipei University of TechnologyTaipei, Taiwan; Institute of Biomedical Engineering and Nanomedicine - National Health Research InstitutesZhunan, Taiwan
| | - Ming-Te Huang
- Department of Surgery, Shuang Ho Hospital, Taipei Medical UniversityTaipei, Taiwan; Department of Surgery, School of Medicine, College of Medicine, Taipei Medical UniversityTaipei, Taiwan
| | - Shiuh-Bin Fang
- Division of Pediatric Gastroenterology and Hepatology, Department of Pediatrics, Shuang Ho Hospital, Taipei Medical UniversityTaipei, Taiwan; Department of Pediatrics, School of Medicine, College of Medicine, Taipei Medical UniversityTaipei, Taiwan
| |
Collapse
|
438
|
Rathna J, Bakkiyaraj D, Pandian SK. Anti-biofilm mechanisms of 3,5-di-tert-butylphenol against clinically relevant fungal pathogens. BIOFOULING 2016; 32:979-993. [PMID: 27535698 DOI: 10.1080/08927014.2016.1216103] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/07/2016] [Accepted: 07/15/2016] [Indexed: 06/06/2023]
Abstract
The methanolic extract (PFME) of Pleurotus florida was assessed for anti-biofilm activity against Candida species. 3,5-Di-tert-butylphenol (3,5-DTB) was identified as the major antifungal constituent in PFME. In its pure form 3,5-DTB inhibits, disrupts, and reduces the viability of biofilm cells as seen from scanning electron and confocal microscopy studies. Microscopic studies and propidium iodide uptake assays confirmed that 3,5-DTB damages the cell membrane of Candida cells. In addition, 3,5-DTB induces accumulation of reactive oxygen species (ROS) which contribute to its pronounced anti-biofilm activity. The results of the present study show that 3,5-DTB exhibits combined anti-biofilm and conventional fungicidal activity against Candida species and elucidate the underlying mechanisms.
Collapse
|
439
|
Leite de Souza E. The effects of sublethal doses of essential oils and their constituents on antimicrobial susceptibility and antibiotic resistance among food-related bacteria: A review. Trends Food Sci Technol 2016. [DOI: 10.1016/j.tifs.2016.07.012] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
|
440
|
Kumar SV, Scottwell SØ, Waugh E, McAdam CJ, Hanton LR, Brooks HJL, Crowley JD. Antimicrobial Properties of Tris(homoleptic) Ruthenium(II) 2-Pyridyl-1,2,3-triazole “Click” Complexes against Pathogenic Bacteria, Including Methicillin-Resistant Staphylococcus aureus (MRSA). Inorg Chem 2016; 55:9767-9777. [DOI: 10.1021/acs.inorgchem.6b01574] [Citation(s) in RCA: 57] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Affiliation(s)
- Sreedhar V. Kumar
- Department
of Chemistry, University of Otago, PO Box 56, Dunedin, New Zealand
- Department
of Microbiology and Immunology, Otago School of Medical Sciences, University of Otago, PO Box 56, Dunedin, New Zealand
| | - Synøve Ø. Scottwell
- Department
of Chemistry, University of Otago, PO Box 56, Dunedin, New Zealand
- Department
of Microbiology and Immunology, Otago School of Medical Sciences, University of Otago, PO Box 56, Dunedin, New Zealand
| | - Emily Waugh
- Department
of Microbiology and Immunology, Otago School of Medical Sciences, University of Otago, PO Box 56, Dunedin, New Zealand
| | - C. John McAdam
- Department
of Chemistry, University of Otago, PO Box 56, Dunedin, New Zealand
| | - Lyall R. Hanton
- Department
of Chemistry, University of Otago, PO Box 56, Dunedin, New Zealand
| | - Heather J. L. Brooks
- Department
of Microbiology and Immunology, Otago School of Medical Sciences, University of Otago, PO Box 56, Dunedin, New Zealand
| | - James D. Crowley
- Department
of Chemistry, University of Otago, PO Box 56, Dunedin, New Zealand
| |
Collapse
|
441
|
Gooyit M, Janda KD. Reprofiled anthelmintics abate hypervirulent stationary-phase Clostridium difficile. Sci Rep 2016; 6:33642. [PMID: 27633064 PMCID: PMC5025651 DOI: 10.1038/srep33642] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2016] [Accepted: 08/31/2016] [Indexed: 01/28/2023] Open
Abstract
Prolonged use of broad-spectrum antibiotics disrupts the indigenous gut microbiota, which consequently enables toxigenic Clostridium difficile species to proliferate and cause infection. The burden of C. difficile infections was exacerbated with the outbreak of hypervirulent strains that produce copious amounts of enterotoxins and spores. In recent past, membrane-active agents have generated a surge of interest due to their bactericidal property with a low propensity for resistance. In this study, we capitalized on the antimicrobial property and low oral bioavailability of salicylanilide anthelmintics (closantel, rafoxanide, niclosamide, oxyclozanide) to target the gut pathogen. By broth microdilution techniques, we determined the MIC values of the anthelmintics against 16 C. difficile isolates of defined PCR-ribotype. The anthelmintics broadly inhibited C. difficile growth in vitro via a membrane depolarization mechanism. Interestingly, the salicylanilides were bactericidal against logarithmic- and stationary-phase cultures of the BI/NAP1/027 strain 4118. The salicylanilides were poorly active against select gut commensals (Bacteroides, Bifidobacterium and Lactobacillus species), and were non-hemolytic and non-toxic to mammalian cell lines HepG2 and HEK 293T/17 within the range of their in vitro MICs and MBCs. The salicylanilide anthelmintics exhibit desirable properties for repositioning as anti-C. difficile agents.
Collapse
Affiliation(s)
- Major Gooyit
- Departments of Chemistry and Immunology and Microbial Science, The Skaggs Institute for Chemical Biology, and The Worm Institute of Research and Medicine (WIRM), The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, California 92037, United States
| | - Kim D Janda
- Departments of Chemistry and Immunology and Microbial Science, The Skaggs Institute for Chemical Biology, and The Worm Institute of Research and Medicine (WIRM), The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, California 92037, United States
| |
Collapse
|
442
|
Gorityala BK, Guchhait G, Goswami S, Fernando DM, Kumar A, Zhanel GG, Schweizer F. Hybrid Antibiotic Overcomes Resistance in P. aeruginosa by Enhancing Outer Membrane Penetration and Reducing Efflux. J Med Chem 2016; 59:8441-55. [DOI: 10.1021/acs.jmedchem.6b00867] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Affiliation(s)
- Bala Kishan Gorityala
- Department
of Chemistry, University of Manitoba, 144 Dysart Road, 450 Parker Building, Winnipeg, Manitoba R3T 2N2, Canada
| | - Goutam Guchhait
- Department
of Chemistry, University of Manitoba, 144 Dysart Road, 450 Parker Building, Winnipeg, Manitoba R3T 2N2, Canada
| | - Sudeep Goswami
- Department
of Chemistry, University of Manitoba, 144 Dysart Road, 450 Parker Building, Winnipeg, Manitoba R3T 2N2, Canada
| | - Dinesh M. Fernando
- Department
of Microbiology, University of Manitoba, Winnipeg, Manitoba R3T 2N2, Canada
| | - Ayush Kumar
- Department
of Microbiology, University of Manitoba, Winnipeg, Manitoba R3T 2N2, Canada
- Department
of Medical Microbiology, University of Manitoba, Winnipeg, Manitoba R3T 1R9, Canada
| | - George G. Zhanel
- Department
of Medical Microbiology, University of Manitoba, Winnipeg, Manitoba R3T 1R9, Canada
| | - Frank Schweizer
- Department
of Chemistry, University of Manitoba, 144 Dysart Road, 450 Parker Building, Winnipeg, Manitoba R3T 2N2, Canada
- Department
of Medical Microbiology, University of Manitoba, Winnipeg, Manitoba R3T 1R9, Canada
| |
Collapse
|
443
|
Talukdar M, Bordoloi M, Dutta P, Saikia S, Kolita B, Talukdar S, Nath S, Yadav A, Saikia R, Jha D, Bora T. Structure elucidation and biological activity of antibacterial compound from Micromonospora auratinigra
, a soil Actinomycetes. J Appl Microbiol 2016; 121:973-87. [DOI: 10.1111/jam.13233] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2016] [Revised: 05/20/2016] [Accepted: 07/04/2016] [Indexed: 11/28/2022]
Affiliation(s)
- M. Talukdar
- Biotechnology Division; CSIR-North East Institute of Science and Technology; Jorhat Assam India
| | - M. Bordoloi
- Natural Product Chemistry Division; CSIR-North East Institute of Science and Technology; Jorhat Assam India
| | - P.P. Dutta
- Natural Product Chemistry Division; CSIR-North East Institute of Science and Technology; Jorhat Assam India
| | - S. Saikia
- Natural Product Chemistry Division; CSIR-North East Institute of Science and Technology; Jorhat Assam India
| | - B. Kolita
- Natural Product Chemistry Division; CSIR-North East Institute of Science and Technology; Jorhat Assam India
| | - S. Talukdar
- Biotechnology Division; CSIR-North East Institute of Science and Technology; Jorhat Assam India
| | - S. Nath
- Natural Product Chemistry Division; CSIR-North East Institute of Science and Technology; Jorhat Assam India
| | - A. Yadav
- Biotechnology Division; CSIR-North East Institute of Science and Technology; Jorhat Assam India
| | - R. Saikia
- Biotechnology Division; CSIR-North East Institute of Science and Technology; Jorhat Assam India
| | - D.K. Jha
- Microbial Ecology Laboratory; Department of Botany; Gauhati University; Guwahati Assam India
| | - T.C. Bora
- Biotechnology Division; CSIR-North East Institute of Science and Technology; Jorhat Assam India
| |
Collapse
|
444
|
Sarkar P, Acharyya S, Banerjee A, Patra A, Thankamani K, Koley H, Bag PK. Intracellular, biofilm-inhibitory and membrane-damaging activities of nimbolide isolated from Azadirachta indica A. Juss (Meliaceae) against meticillin-resistant Staphylococcus aureus. J Med Microbiol 2016; 65:1205-1214. [PMID: 27553840 DOI: 10.1099/jmm.0.000343] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
Staphylococcus aureus is a leading aetiologic agent of nosocomial- and community-acquired infectious diseases worldwide. The public health concern regarding staphylococcal infections is inflated by the increasing occurrence of multidrug-resistant strains, e.g. multidrug- and meticillin-resistant S.aureus (MDR MRSA). This study was designed to evaluate the intracellular killing, membrane-damaging and biofilm-inhibitory activities of nimbolide isolated from Azadirachta indica against MDR MRSA. In vitro antibacterial activity of nimbolide was determined by performing MIC, minimal bactericidal concentration (MBC) and time-kill kinetic studies. Bacterial membrane-damaging activity was determined by membrane perturbation and scanning electron microscopy (SEM) examination. Biofilm-inhibitory activities were determined by SEM. Cellular drug accumulation and assessments of intracellular activities were performed using Vero cell culture. SEM revealed that nimbolide caused significant membrane damage and lysis of the S. aureus cells. The biofilm structure was disrupted, and the biofilm formation was greatly reduced in the presence of nimbolide as examined by SEM. The level of accumulation of nimbolide in Vero cells incubated for 24 h is relatively higher than that of ciprofloxacin and nalidixic acid (Cc/Ce for nimbolide > ciprofloxacin and nalidixic acid). The viable number of intracellular S. aureus was decreased [reduction of ~2 log10 c.f.u. (mg Vero cell protein)-1] in a time-dependent manner in the presence of nimbolide (4× MBC) that was comparable to that of tetracycline and nalidixic acid. The significant intracellular, biofilm-inhibitory and bacterial membrane-damaging activities of nimbolide demonstrated here suggested that it has potential as an effective antibacterial agent for the treatment of severe infections caused by MDR MRSA.
Collapse
Affiliation(s)
- Prodipta Sarkar
- Department of Biochemistry, University of Calcutta, 35 Ballygunge, Circular Road, Kolkata 700019, India
| | - Saurabh Acharyya
- Department of Biochemistry, University of Calcutta, 35 Ballygunge, Circular Road, Kolkata 700019, India
| | - Anirban Banerjee
- Department of Biochemistry, University of Calcutta, 35 Ballygunge, Circular Road, Kolkata 700019, India
| | - Amarendra Patra
- Department of Chemistry, University of Calcutta, 92 Acharya Prafulla Chandra Road, Kolkata 700009, India
| | - Karthika Thankamani
- Department of Biochemistry, University of Calcutta, 35 Ballygunge, Circular Road, Kolkata 700019, India
| | - Hemanta Koley
- National Institute of Cholera and Enteric Diseases, P-33 C.I.T. Road, Scheme-XM, Beliaghata, Kolkata 700010, India
| | - Prasanta K Bag
- Department of Biochemistry, University of Calcutta, 35 Ballygunge, Circular Road, Kolkata 700019, India
| |
Collapse
|
445
|
Abstract
UNLABELLED The outer membrane (OM) of Gram-negative bacteria provides protection against toxic molecules, including reactive oxygen species (ROS). Decreased OM permeability can promote bacterial survival under harsh circumstances and protects against antibiotics. To better understand the regulation of OM permeability, we studied the real-time influx of hydrogen peroxide in Salmonella bacteria and discovered two novel mechanisms by which they rapidly control OM permeability. We found that pores in two major OM proteins, OmpA and OmpC, could be rapidly opened or closed when oxidative stress is encountered and that the underlying mechanisms rely on the formation of disulfide bonds in the periplasmic domain of OmpA and TrxA, respectively. Additionally, we found that a Salmonella mutant showing increased OM permeability was killed more effectively by treatment with antibiotics. Together, these results demonstrate that Gram-negative bacteria regulate the influx of ROS for defense against oxidative stress and reveal novel targets that can be therapeutically targeted to increase bacterial killing by conventional antibiotics. IMPORTANCE Pathogenic bacteria have evolved ways to circumvent inflammatory immune responses. A decrease in bacterial outer membrane permeability during infection helps protect bacteria from toxic molecules produced by the host immune system and allows for effective colonization of the host. In this report, we reveal molecular mechanisms that rapidly alter outer membrane pores and their permeability in response to hydrogen peroxide and oxidative stress. These mechanisms are the first examples of pores that are rapidly opened or closed in response to reactive oxygen species. Moreover, one of these mechanisms can be targeted to artificially increase membrane permeability and thereby increase bacterial killing by the antibiotic cefotaxime during in vitro experiments and in a mouse model of infection. We envision that a better understanding of the regulation of membrane permeability will lead to new targets and treatment options for multidrug-resistant infections.
Collapse
|
446
|
Moloney MG. Natural Products as a Source for Novel Antibiotics. Trends Pharmacol Sci 2016; 37:689-701. [DOI: 10.1016/j.tips.2016.05.001] [Citation(s) in RCA: 185] [Impact Index Per Article: 20.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2016] [Revised: 05/02/2016] [Accepted: 05/02/2016] [Indexed: 01/04/2023]
|
447
|
Amphiphilic xanthones as a potent chemical entity of anti-mycobacterial agents with membrane-targeting properties. Eur J Med Chem 2016; 123:684-703. [PMID: 27517813 DOI: 10.1016/j.ejmech.2016.07.068] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2016] [Revised: 07/26/2016] [Accepted: 07/27/2016] [Indexed: 12/31/2022]
Abstract
Tuberculosis (TB) remains a deadly disease and infects one-third of the world's population. Given the low success rates encountered in clinical development, there is an urgent need to identify structurally novel antimicrobials for tuberculosis. The present report details the anti-mycobacterial activities, structure-activity relationships (SARs) and mechanism of action of amphiphilic xanthone derivatives. The xanthones exhibited potent MIC, rapid time-kill and no cross-resistance with the current anti-TB drugs. Evidence is presented that these compounds disrupted the inner membrane and led to ATP depletion. Amphiphilic xanthone derivatives exhibited superior metabolic stability, low cytotoxicity and low activity against the common cytochrome P450. Compound 5 was selected for an in vivo pharmacokinetic study. Its bioavailability at an oral dose of 2 mg/kg was 15%. The xanthones thuse provide valuable insight for the development of a new class of membrane targeting antimycobacterial agents that may assist in overcoming the limitations of the current TB medications.
Collapse
|
448
|
Mohamed MF, Abdelkhalek A, Seleem MN. Evaluation of short synthetic antimicrobial peptides for treatment of drug-resistant and intracellular Staphylococcus aureus. Sci Rep 2016; 6:29707. [PMID: 27405275 PMCID: PMC4942614 DOI: 10.1038/srep29707] [Citation(s) in RCA: 202] [Impact Index Per Article: 22.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2016] [Accepted: 06/21/2016] [Indexed: 02/07/2023] Open
Abstract
Methicillin-resistant Staphylococcus aureus (MRSA) infections present a serious challenge because of the emergence of resistance to numerous conventional antibiotics. Due to their unique mode of action, antimicrobial peptides are novel alternatives to traditional antibiotics for tackling the issue of bacterial multidrug resistance. Herein, we investigated the antibacterial activity of two short novel peptides (WR12, a 12 residue peptide composed exclusively of arginine and tryptophan, and D-IK8, an eight residue β-sheet peptide) against multidrug resistant staphylococci. In vitro, both peptides exhibited good antibacterial activity against MRSA, vancomycin-resistant S. aureus, linezolid-resistant S. aureus, and methicillin-resistant S. epidermidis. WR12 and D-IK8 were able to eradicate persisters, MRSA in stationary growth phase, and showed significant clearance of intracellular MRSA in comparison to both vancomycin and linezolid. In vivo, topical WR12 and D-IK8 significantly reduced both the bacterial load and the levels of the pro-inflammatory cytokines including tumor necrosis factor-α (TNF-α) and interleukin-6 (IL-6) in MRSA-infected skin lesions. Moreover, both peptides disrupted established in vitro biofilms of S. aureus and S. epidermidis significantly more so than traditional antimicrobials tested. Taken together, these results support the potential of WR12 and D-IK8 to be used as a topical antimicrobial agent for the treatment of staphylococcal skin infections.
Collapse
Affiliation(s)
- Mohamed F. Mohamed
- Department of Comparative Pathobiology, College of Veterinary Medicine, Purdue University, West Lafayette, IN 47907, USA
| | - Ahmed Abdelkhalek
- Department of Comparative Pathobiology, College of Veterinary Medicine, Purdue University, West Lafayette, IN 47907, USA
| | - Mohamed N. Seleem
- Department of Comparative Pathobiology, College of Veterinary Medicine, Purdue University, West Lafayette, IN 47907, USA
- Purdue Institute for Inflammation, Immunology, and Infectious Disease, Purdue University, West Lafayette, IN 47907, USA
| |
Collapse
|
449
|
Wang CM, Jhan YL, Tsai SJ, Chou CH. The Pleiotropic Antibacterial Mechanisms of Ursolic Acid against Methicillin-Resistant Staphylococcus aureus (MRSA). Molecules 2016; 21:E884. [PMID: 27399657 PMCID: PMC6273082 DOI: 10.3390/molecules21070884] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2016] [Revised: 06/23/2016] [Accepted: 06/30/2016] [Indexed: 01/20/2023] Open
Abstract
(1) BACKGROUND: Several triterpenoids were found to act synergistically with classes of antibiotic, indicating that plant-derived chemicals have potential to be used as therapeutics to enhance the activity of antibiotics against multidrug-resistant pathogens. However, the mode of action of triterpenoids against bacterial pathogens remains unclear. The objective of this study is to evaluate the interaction between ursolic acid against methicillin-resistant Staphylococcus aureus (MRSA); (2) METHODS: The ability of ursolic acid to damage mammalian and bacterial membranes was examined. The proteomic response of methicillin-resistant S. aureus in ursolic acid treatment was investigated using two-dimensional (2D) proteomic analysis; (3) RESULTS: Ursolic acid caused the loss of staphylococcal membrane integrity without hemolytic activity. The comparison of the protein pattern of ursolic acid-treated and normal MRSA cells revealed that ursolic acid affected a variety of proteins involved in the translation process with translational accuracy, ribonuclease and chaperon subunits, glycolysis and oxidative responses; (4) CONCLUSION: The mode of action of ursolic acid appears to be the influence on the integrity of the bacterial membrane initially, followed by inhibition of protein synthesis and the metabolic pathway. These findings reflect that the pleiotropic effects of ursolic acid against MRSA make it a promising antibacterial agent in pharmaceutical research.
Collapse
Affiliation(s)
- Chao-Min Wang
- Research Center for Biodiversity, China Medical University, Taichung 40402, Taiwan.
| | - Yun-Lian Jhan
- Research Center for Biodiversity, China Medical University, Taichung 40402, Taiwan.
| | - Shang-Jie Tsai
- Research Center for Biodiversity, China Medical University, Taichung 40402, Taiwan.
| | - Chang-Hung Chou
- Research Center for Biodiversity, China Medical University, Taichung 40402, Taiwan.
| |
Collapse
|
450
|
Eroshenko DV, Polyudova TV, Korobov VP. Role of proton-motive force in adhesion and biofilm formation by staphylococcus epidermidis. Microbiology (Reading) 2016. [DOI: 10.1134/s0026261716040044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
|