1
|
Yaeger LN, French S, Brown ED, Côté JP, Burrows LL. Central metabolism is a key player in E. coli biofilm stimulation by sub-MIC antibiotics. PLoS Genet 2023; 19:e1011013. [PMID: 37917668 PMCID: PMC10645362 DOI: 10.1371/journal.pgen.1011013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2023] [Revised: 11/14/2023] [Accepted: 10/10/2023] [Indexed: 11/04/2023] Open
Abstract
Exposure of Escherichia coli to sub-inhibitory antibiotics stimulates biofilm formation through poorly characterized mechanisms. Using a high-throughput Congo Red binding assay to report on biofilm matrix production, we screened ~4000 E. coli K12 deletion mutants for deficiencies in this biofilm stimulation response. We screened using three different antibiotics to identify core components of the biofilm stimulation response. Mutants lacking acnA, nuoE, or lpdA failed to respond to sub-MIC cefixime and novobiocin, implicating central metabolism and aerobic respiration in biofilm stimulation. These genes are members of the ArcA/B regulon-controlled by a respiration-sensitive two-component system. Mutants of arcA and arcB had a 'pre-activated' phenotype, where biofilm formation was already high relative to wild type in vehicle control conditions, and failed to increase further with the addition of sub-MIC cefixime. Using a tetrazolium dye and an in vivo NADH sensor, we showed spatial co-localization of increased metabolic activity with sub-lethal concentrations of the bactericidal antibiotics cefixime and novobiocin. Supporting a role for respiratory stress, the biofilm stimulation response to cefixime and novobiocin was inhibited when nitrate was provided as an alternative electron acceptor. Deletion of a gene encoding part of the machinery for respiring nitrate abolished its ameliorating effects, and nitrate respiration increased during growth with sub-MIC cefixime. Finally, in probing the generalizability of biofilm stimulation, we found that the stimulation response to translation inhibitors, unlike other antibiotic classes, was minimally affected by nitrate supplementation, suggesting that targeting the ribosome stimulates biofilm formation in distinct ways. By characterizing the biofilm stimulation response to sub-MIC antibiotics at a systems level, we identified multiple avenues for design of therapeutics that impair bacterial stress management.
Collapse
Affiliation(s)
- Luke N. Yaeger
- Department of Biochemistry and Biomedical Sciences, and the Michael G. DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, Ontario, Canada
| | - Shawn French
- Department of Biochemistry and Biomedical Sciences, and the Michael G. DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, Ontario, Canada
| | - Eric D. Brown
- Department of Biochemistry and Biomedical Sciences, and the Michael G. DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, Ontario, Canada
| | - Jean Philippe Côté
- Department of Biochemistry and Biomedical Sciences, and the Michael G. DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, Ontario, Canada
- Département de Biologie, Université de Sherbrooke, Sherbrooke, Quebec, Canada
| | - Lori L. Burrows
- Department of Biochemistry and Biomedical Sciences, and the Michael G. DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, Ontario, Canada
| |
Collapse
|
2
|
Pino-Otín MR, Gan C, Terrado E, Sanz MA, Ballestero D, Langa E. Antibiotic properties of Satureja montana L. hydrolate in bacteria and fungus of clinical interest and its impact in non-target environmental microorganisms. Sci Rep 2022; 12:18460. [PMID: 36323748 PMCID: PMC9630514 DOI: 10.1038/s41598-022-22419-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Accepted: 10/14/2022] [Indexed: 11/20/2022] Open
Abstract
The aim of this study was to analyse the microbicidal and microbiostatic activity of S. montana hydrolate L., the water-soluble fraction of the hydro-distillation process used to obtain the essential oil, on 14 Gram-positive and Gram-negative bacteria and a fungus of clinical interest. To consider whether this hydrolate is a more environmentally friendly alternative to traditional antibiotics, its effect on non-target microorganisms in the aquatic and terrestrial environment was analysed using natural soil and river microorganism communities, characterized through 16S rRNA gene sequencing. Results showed that S. montana hydrolate was especially effective (25% v/v concentration) against Pasteurella aerogenes, Streptococcus agalactiae and Acinetobacter baumannii (priority 1, WHO). It was also a microbicide for a further 7 bacterial strains and the fungus Candida albicans (50% v/v concentration). The river and soil communities exposed to the hydrolate showed a decrease in their growth, as well as a decrease in their ability to metabolize polymers and carbohydrates (soil microorganisms) and polymers, carboxylic and ketone acids (river microorganisms). Hydrolates could be an alternative to conventional antibiotics, but their impact on the environment must be taken into account.
Collapse
Affiliation(s)
- María Rosa Pino-Otín
- Universidad San Jorge, Campus Universitario Villanueva de Gállego Autovía A-23 Zaragoza-Huesca, Km. 510, Villanueva de Gállego, 50830, Zaragoza, Spain.
| | - Cristina Gan
- Universidad San Jorge, Campus Universitario Villanueva de Gállego Autovía A-23 Zaragoza-Huesca, Km. 510, Villanueva de Gállego, 50830, Zaragoza, Spain
| | - Eva Terrado
- Universidad de Zaragoza, C. de Pedro Cerbuna, 12, 50009, Zaragoza, Spain
| | - María Angeles Sanz
- CITA, Área de Laboratorios de Análisis y Asistencia Tecnológica, Avda. Montañana 930, 50059, Zaragoza, Spain
| | - Diego Ballestero
- Universidad San Jorge, Campus Universitario Villanueva de Gállego Autovía A-23 Zaragoza-Huesca, Km. 510, Villanueva de Gállego, 50830, Zaragoza, Spain
| | - Elisa Langa
- Universidad San Jorge, Campus Universitario Villanueva de Gállego Autovía A-23 Zaragoza-Huesca, Km. 510, Villanueva de Gállego, 50830, Zaragoza, Spain
| |
Collapse
|
3
|
Coles VE, Darveau P, Zhang X, Harvey H, Henriksbo BD, Yang A, Schertzer JD, Magolan J, Burrows LL. Exploration of BAY 11-7082 as a Potential Antibiotic. ACS Infect Dis 2022; 8:170-182. [PMID: 34860493 DOI: 10.1021/acsinfecdis.1c00522] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Exposure of the Gram-negative pathogen Pseudomonas aeruginosa to subinhibitory concentrations of antibiotics increases the formation of biofilms. We exploited this phenotype to identify molecules with potential antimicrobial activity in a biofilm-based high-throughput screen. The anti-inflammatory compound BAY 11-7082 induced dose-dependent biofilm stimulation, indicative of antibacterial activity. We confirmed that BAY 11-7082 inhibits the growth of P. aeruginosa and other priority pathogens, including methicillin-resistant Staphylococcus aureus (MRSA). We synthesized 27 structural analogues, including a series based on the related scaffold 3-(phenylsulfonyl)-2-pyrazinecarbonitrile (PSPC), 10 of which displayed increased anti-Staphylococcal activity. Because the parent molecule inhibits the NLR Family Pyrin Domain Containing 3 (NLRP3) inflammasome, we measured the ability of select analogues to reduce interleukin-1β (IL-1β) production in mammalian macrophages, identifying minor differences in the structure-activity relationship for the anti-inflammatory and antibacterial properties of this scaffold. Although we could evolve stably resistant MRSA mutants with cross-resistance to BAY 11-7082 and PSPC, their lack of shared mutations suggested that the two molecules could have multiple targets. Finally, we showed that BAY 11-7082 and its analogues synergize with penicillin G against MRSA, suggesting that this scaffold may serve as an interesting starting point for the development of antibiotic adjuvants.
Collapse
Affiliation(s)
- Victoria E. Coles
- Department of Biochemistry and Biomedical Sciences, McMaster University, 1280 Main Street West Hamilton, Ontario L8S 4K1, Canada
- Michael G. DeGroote Institute for Infectious Disease Research, McMaster University, 1280 Main Street West Hamilton, Ontario L8S 4K1, Canada
| | - Patrick Darveau
- Department of Chemistry and Chemical Biology, McMaster University, 1280 Main Street West Hamilton, Ontario L8S 4L8, Canada
| | - Xiong Zhang
- Department of Biochemistry and Biomedical Sciences, McMaster University, 1280 Main Street West Hamilton, Ontario L8S 4K1, Canada
| | - Hanjeong Harvey
- Department of Biochemistry and Biomedical Sciences, McMaster University, 1280 Main Street West Hamilton, Ontario L8S 4K1, Canada
- Michael G. DeGroote Institute for Infectious Disease Research, McMaster University, 1280 Main Street West Hamilton, Ontario L8S 4K1, Canada
| | - Brandyn D. Henriksbo
- Department of Biochemistry and Biomedical Sciences, McMaster University, 1280 Main Street West Hamilton, Ontario L8S 4K1, Canada
| | - Angela Yang
- Department of Biochemistry and Biomedical Sciences, McMaster University, 1280 Main Street West Hamilton, Ontario L8S 4K1, Canada
- Michael G. DeGroote Institute for Infectious Disease Research, McMaster University, 1280 Main Street West Hamilton, Ontario L8S 4K1, Canada
| | - Jonathan D. Schertzer
- Department of Biochemistry and Biomedical Sciences, McMaster University, 1280 Main Street West Hamilton, Ontario L8S 4K1, Canada
| | - Jakob Magolan
- Department of Biochemistry and Biomedical Sciences, McMaster University, 1280 Main Street West Hamilton, Ontario L8S 4K1, Canada
- Michael G. DeGroote Institute for Infectious Disease Research, McMaster University, 1280 Main Street West Hamilton, Ontario L8S 4K1, Canada
- Department of Chemistry and Chemical Biology, McMaster University, 1280 Main Street West Hamilton, Ontario L8S 4L8, Canada
| | - Lori L. Burrows
- Department of Biochemistry and Biomedical Sciences, McMaster University, 1280 Main Street West Hamilton, Ontario L8S 4K1, Canada
- Michael G. DeGroote Institute for Infectious Disease Research, McMaster University, 1280 Main Street West Hamilton, Ontario L8S 4K1, Canada
| |
Collapse
|
4
|
Janež N, Škrlj B, Sterniša M, Klančnik A, Sabotič J. The role of the Listeria monocytogenes surfactome in biofilm formation. Microb Biotechnol 2021; 14:1269-1281. [PMID: 34106516 PMCID: PMC8313260 DOI: 10.1111/1751-7915.13847] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2021] [Revised: 05/12/2021] [Accepted: 05/17/2021] [Indexed: 12/11/2022] Open
Abstract
Listeria monocytogenes is a highly pathogenic foodborne bacterium that is ubiquitous in the natural environment and capable of forming persistent biofilms in food processing environments. This species has a rich repertoire of surface structures that enable it to survive, adapt and persist in various environments and promote biofilm formation. We review current understanding and advances on how L. monocytogenes organizes its surface for biofilm formation on surfaces associated with food processing settings, because they may be an important target for development of novel antibiofilm compounds. A synthesis of the current knowledge on the role of Listeria surfactome, comprising peptidoglycan, teichoic acids and cell wall proteins, during biofilm formation on abiotic surfaces is provided. We consider indications gained from genome-wide studies and discuss surfactome structures with established mechanistic aspects in biofilm formation. Additionally, we look at the analogies to the species L. innocua, which is closely related to L. monocytogenes and often used as its model (surrogate) organism.
Collapse
Affiliation(s)
- Nika Janež
- Department of BiotechnologyJožef Stefan InstituteLjubljanaSlovenia
| | - Blaž Škrlj
- Department of Knowledge TechnologiesJožef Stefan InstituteLjubljanaSlovenia
- Jožef Stefan International Postgraduate SchoolLjubljanaSlovenia
| | - Meta Sterniša
- Department of Food Science and TechnologyBiotechnical FacultyUniversity of LjubljanaLjubljanaSlovenia
| | - Anja Klančnik
- Department of Food Science and TechnologyBiotechnical FacultyUniversity of LjubljanaLjubljanaSlovenia
| | - Jerica Sabotič
- Department of BiotechnologyJožef Stefan InstituteLjubljanaSlovenia
| |
Collapse
|
5
|
Sandala JL, Eichar BW, Kuo LG, Hahn MM, Basak AK, Huggins WM, Woolard K, Melander C, Gunn JS. A dual-therapy approach for the treatment of biofilm-mediated Salmonella gallbladder carriage. PLoS Pathog 2020; 16:e1009192. [PMID: 33370414 PMCID: PMC7793255 DOI: 10.1371/journal.ppat.1009192] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Revised: 01/08/2021] [Accepted: 11/29/2020] [Indexed: 01/12/2023] Open
Abstract
Asymptomatic carriage of Salmonella Typhi continues to facilitate the transmission of typhoid fever, resulting in 14 million new infections and 136,000 fatalities each year. Asymptomatic chronic carriage of S. Typhi is facilitated by the formation of biofilms on gallstones that protect the bacteria from environmental insults and immune system clearance. Here, we identified two unique small molecules capable of both inhibiting Salmonella biofilm growth and disrupting pre-formed biofilm structures without affecting bacterial viability. In a mouse model of chronic gallbladder Salmonella carriage, treatment with either compound reduced bacterial burden in the gallbladder by 1–2 logs resulting in bacterial dissemination to peripheral organs that was associated with increased mortality. Co-administration of either compound with ciprofloxacin not only enhanced compound efficacy in the gallbladder by a further 1–1.5 logs for a total of 3–4.5 log reduction, but also prevented bacterial dissemination to peripheral organs. These data suggest a dual-therapy approach targeting both biofilm and planktonic populations can be further developed as a safe and efficient treatment of biofilm-mediated chronic S. Typhi infections. Typhoid fever is an infectious disease caused by Salmonella Typhi (S. Typhi), a bacterium that causes as many as 14 million new infections and 136,000 deaths annually. Asymptomatic chronic carriers of S. Typhi play a major role in the transmission of typhoid fever, as they intermittently shed the bacteria and can unknowingly infect surrounding individuals. Here, we characterized novel compounds that target biofilm formation, a process utilized by S. Typhi to establish and maintain chronic carriage in the gallbladder, in hopes that they may be eventually used in conjunction with traditional antibiotics to prevent and/or cure chronic infections more efficiently than antibiotics alone.
Collapse
Affiliation(s)
- Jenna L. Sandala
- Center for Microbial Pathogenesis, Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus, Ohio, United States of America
- Department of Microbial Infection and Immunity, The Ohio State University College of Medicine, Columbus, Ohio, United States of America
- Infectious Diseases Institute, The Ohio State University, Columbus, Ohio, United States of America
| | - Bradley W. Eichar
- Department of Microbial Infection and Immunity, The Ohio State University College of Medicine, Columbus, Ohio, United States of America
| | - Laura G. Kuo
- Department of Microbial Infection and Immunity, The Ohio State University College of Medicine, Columbus, Ohio, United States of America
| | - Mark M. Hahn
- Center for Microbial Pathogenesis, Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus, Ohio, United States of America
- Department of Microbial Infection and Immunity, The Ohio State University College of Medicine, Columbus, Ohio, United States of America
- Infectious Diseases Institute, The Ohio State University, Columbus, Ohio, United States of America
| | - Akash K. Basak
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, Indiana, United States of America
| | - William M. Huggins
- Department of Chemistry, North Carolina State University, Raleigh, North Carolina, United States of America
| | - Katherine Woolard
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, Indiana, United States of America
| | - Christian Melander
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, Indiana, United States of America
| | - John S. Gunn
- Center for Microbial Pathogenesis, Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus, Ohio, United States of America
- Department of Microbial Infection and Immunity, The Ohio State University College of Medicine, Columbus, Ohio, United States of America
- Infectious Diseases Institute, The Ohio State University, Columbus, Ohio, United States of America
- Department of Pediatrics, The Ohio State University College of Medicine, Columbus, Ohio, United States of America
- * E-mail:
| |
Collapse
|
6
|
Kumar P, Lee JH, Beyenal H, Lee J. Fatty Acids as Antibiofilm and Antivirulence Agents. Trends Microbiol 2020; 28:753-768. [DOI: 10.1016/j.tim.2020.03.014] [Citation(s) in RCA: 77] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2019] [Revised: 03/09/2020] [Accepted: 03/25/2020] [Indexed: 12/21/2022]
|
7
|
Shao X, Fang K, Medina D, Wan J, Lee J, Hong SH. The probiotic,
Leuconostoc mesenteroides
, inhibits
Listeria monocytogenes
biofilm formation. J Food Saf 2020. [DOI: 10.1111/jfs.12750] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Affiliation(s)
- Xinhao Shao
- Department of Chemical and Biological EngineeringIllinois Institute of Technology Chicago Illinois
| | - Kuili Fang
- Department of Chemical and Biological EngineeringIllinois Institute of Technology Chicago Illinois
| | - Daniel Medina
- Department of Biomedical EngineeringIllinois Institute of Technology Chicago Illinois
| | - Jason Wan
- Institute for Food Safety and HealthIllinois Institute of Technology Bedford Park Illinois
| | - Jung‐Lim Lee
- Department of Human Ecology, Food Science and Biotechnology Program, Food Microbiology LaboratoryDelaware State University Dover Delaware
| | - Seok Hoon Hong
- Department of Chemical and Biological EngineeringIllinois Institute of Technology Chicago Illinois
| |
Collapse
|
8
|
Lee YJ, Wang C. Links between S-adenosylmethionine and Agr-based quorum sensing for biofilm development in Listeria monocytogenes EGD-e. Microbiologyopen 2020; 9:e1015. [PMID: 32134563 PMCID: PMC7221448 DOI: 10.1002/mbo3.1015] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Revised: 02/05/2020] [Accepted: 02/06/2020] [Indexed: 12/20/2022] Open
Abstract
Listeria monocytogenes is the causative agent of human listeriosis which has high hospitalization and mortality rates for individuals with weakened immune systems. The survival and dissemination of L. monocytogenes in adverse environments can be reinforced by the formation of biofilms. Therefore, this study aimed to understand the mechanisms underlying listerial biofilm development. Given that both nutrient availability and quorum sensing (QS) have been known as the factors influencing biofilm development, we hypothesized that the signal from a sentinel metabolite S‐adenosylmethionine (SAM) and Agr‐based QS could be synchronous in L. monocytogenes to modulate nutrient availability, the synthesis of extracellular polymeric substances (EPSs), and biofilm formation. We performed biofilm assays and quantitative real‐time PCR to investigate how biofilm volumes and the expression of genes for the synthesis of EPS were affected by SAM supplementation, agr deletion, or both. We found that exogenously applied SAM induced biofilm formation and that the expression of genes encoding the EPS synthesis machineries was regulated by SAM and/or Agr QS. Moreover, the gene transcription of components acting in the methyl cycle for SAM synthesis and Agr QS was affected by the signals from the other system. In summary, we reveal an interconnection at the transcriptional level between metabolism and QS in L. monocytogenes and highlight the critical role of metabolite‐oriented QS in biofilm development.
Collapse
Affiliation(s)
- Yue-Jia Lee
- Department of Basic Sciences, College of Veterinary Medicine, Mississippi State University, Mississippi State, MS, USA
| | - Chinling Wang
- Department of Basic Sciences, College of Veterinary Medicine, Mississippi State University, Mississippi State, MS, USA
| |
Collapse
|
9
|
Barker WT, Nemeth AM, Brackett SM, Basak AK, Chandler CE, Jania LA, Zuercher WJ, Melander RJ, Koller BH, Ernst RK, Melander C. Repurposing Eukaryotic Kinase Inhibitors as Colistin Adjuvants in Gram-Negative Bacteria. ACS Infect Dis 2019; 5:1764-1771. [PMID: 31434474 DOI: 10.1021/acsinfecdis.9b00212] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Kinase inhibitors comprise a diverse cohort of chemical scaffolds that are active in multiple biological systems. Currently, thousands of eukaryotic kinase inhibitors are commercially available, have well-characterized targets, and often carry pharmaceutically favorable toxicity profiles. Recently, our group disclosed that derivatives of the natural product meridianin D, a known inhibitor of eukaryotic kinases, modulated behaviors of both Gram-positive and Gram-negative bacteria. Herein, we expand our exploration of kinase inhibitors in Gram-negative bacilli utilizing three commercially available kinase inhibitor libraries and, ultimately, identify two chemical structures that potentiate colistin (polymyxin E) in multiple strains. We report IMD-0354, an inhibitor of IKK-β, as a markedly effective adjuvant in colistin-resistant bacteria and also describe AR-12 (OSU-03012), an inhibitor of pyruvate dehydrogenase kinase-1 (PDK-1), as a potentiator in colistin-sensitive strains. This report comprises the first description of the novel cross-reactivity of these molecules.
Collapse
Affiliation(s)
- William T. Barker
- Department of Chemistry and Biochemistry, University of Notre Dame, 240 McCourtney Hall, Notre Dame, Indiana 46556, United States
| | - Ansley M. Nemeth
- Department of Chemistry and Biochemistry, University of Notre Dame, 240 McCourtney Hall, Notre Dame, Indiana 46556, United States
| | - Sara M. Brackett
- Department of Chemistry and Biochemistry, University of Notre Dame, 240 McCourtney Hall, Notre Dame, Indiana 46556, United States
| | - Akash K. Basak
- Department of Chemistry and Biochemistry, University of Notre Dame, 240 McCourtney Hall, Notre Dame, Indiana 46556, United States
| | - Courtney E. Chandler
- Department of Microbial Pathogenesis, University of Maryland-Baltimore, 650 W. Baltimore Street, Baltimore, Maryland 21201, United States
| | - Leigh A. Jania
- Department of Genetics, University of North Carolina at Chapel Hill, 120 Mason Farm Road, Chapel Hill, North Carolina 27599, United States
| | - William J. Zuercher
- Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, 120 Mason Farm Road, Chapel Hill, North Carolina 27599, United States
| | - Roberta J. Melander
- Department of Chemistry and Biochemistry, University of Notre Dame, 240 McCourtney Hall, Notre Dame, Indiana 46556, United States
| | - Beverly H. Koller
- Department of Genetics, University of North Carolina at Chapel Hill, 120 Mason Farm Road, Chapel Hill, North Carolina 27599, United States
| | - Robert K. Ernst
- Department of Microbial Pathogenesis, University of Maryland-Baltimore, 650 W. Baltimore Street, Baltimore, Maryland 21201, United States
| | - Christian Melander
- Department of Chemistry and Biochemistry, University of Notre Dame, 240 McCourtney Hall, Notre Dame, Indiana 46556, United States
| |
Collapse
|
10
|
Young M, Ozcan A, Lee B, Maxwell T, Andl T, Rajasekaran P, Beazley MJ, Tetard L, Santra S. N-acetyl Cysteine Coated Gallium Particles Demonstrate High Potency against Pseudomonas aeruginosa PAO1. Pathogens 2019; 8:pathogens8030120. [PMID: 31374947 PMCID: PMC6789799 DOI: 10.3390/pathogens8030120] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2019] [Revised: 07/28/2019] [Accepted: 07/30/2019] [Indexed: 12/19/2022] Open
Abstract
Nosocomial infections pose serious health concerns with over 2 million reported annually in the United States. Many of these infections are associated with bacterial resistance to antibiotics and hence, alternative treatments are critically needed. The objective of this study was to assess the antimicrobial efficacy of a gallium (Ga)-based particle coated with N-Acetyl Cysteine (Ga-NAC) against Pseudomonas aeruginosa PAO1. Our studies showed the Minimum Inhibitory Concentration (MIC) of PAO1 treated with Ga-NAC was 1 µg/mL. Cytotoxicity of Ga-NAC against multiple cell lines was determined with no cytotoxicity observed up to concentrations of 2000 µg/mL (metal concentration), indicating a high therapeutic window. To elucidate potential antibacterial modes of action, Inductively Coupled Plasma—Mass Spectrometry (ICP-MS), infrared spectroscopy, and atomic force microscopy (AFM) were used. The results suggest improved Ga3+ interaction with PAO1 through Ga-NAC particles. No significant change in cell membrane chemistry or roughening was detected. As cell membrane integrity remained intact, the antimicrobial mode of action was linked to cellular internalization of Ga and subsequent iron metabolic disruption. Furthermore, Ga-NAC inhibited and disrupted biofilms seen with crystal violet assay and microscopy. Our findings suggest the Ga-NAC particle can potentially be used as an alternative to antibiotics for treatment of Pseudomonas aeruginosa infections.
Collapse
Affiliation(s)
- Mikaeel Young
- Burnett School of Biomedical Sciences, University of Central Florida, Orlando, FL 32816, USA
- NanoScience Technology Center, University of Central Florida, Orlando, FL 32816, USA
| | - Ali Ozcan
- NanoScience Technology Center, University of Central Florida, Orlando, FL 32816, USA
- Department of Chemistry, University of Central Florida, Orlando, FL 32816, USA
| | - Briana Lee
- NanoScience Technology Center, University of Central Florida, Orlando, FL 32816, USA
| | - Tyler Maxwell
- NanoScience Technology Center, University of Central Florida, Orlando, FL 32816, USA
- Department of Chemistry, University of Central Florida, Orlando, FL 32816, USA
| | - Thomas Andl
- Burnett School of Biomedical Sciences, University of Central Florida, Orlando, FL 32816, USA
| | - Parthiban Rajasekaran
- NanoScience Technology Center, University of Central Florida, Orlando, FL 32816, USA
| | - Melanie J Beazley
- Department of Chemistry, University of Central Florida, Orlando, FL 32816, USA
| | - Laurene Tetard
- NanoScience Technology Center, University of Central Florida, Orlando, FL 32816, USA.
- Department of Physics, University of Central Florida, Orlando, FL 32816, USA.
| | - Swadeshmukul Santra
- Burnett School of Biomedical Sciences, University of Central Florida, Orlando, FL 32816, USA.
- NanoScience Technology Center, University of Central Florida, Orlando, FL 32816, USA.
- Department of Chemistry, University of Central Florida, Orlando, FL 32816, USA.
- Department of Materials Science and Engineering, University of Central Florida, Orlando, FL 32816, USA.
| |
Collapse
|
11
|
Haque S, Yadav DK, Bisht SC, Yadav N, Singh V, Dubey KK, Jawed A, Wahid M, Dar SA. Quorum sensing pathways in Gram-positive and -negative bacteria: potential of their interruption in abating drug resistance. J Chemother 2019; 31:161-187. [DOI: 10.1080/1120009x.2019.1599175] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Affiliation(s)
- Shafiul Haque
- Research and Scientific Studies Unit, College of Nursing and Allied Health Sciences, Jazan University, Jazan, Saudi Arabia
- Gene Expression Laboratory, Department of Biosciences, Faculty of Natural Sciences, Jamia Millia Islamia, New Delhi, India
| | - Dinesh K. Yadav
- Department of Botany, University of Allahabad, Allahabad, Uttar Pradesh, India
| | - Shekhar C. Bisht
- Department of Biotechnology, H.N.B Garhwal University, Srinagar, Uttarakhand, India
| | - Neelam Yadav
- Department of Botany, University of Allahabad, Allahabad, Uttar Pradesh, India
| | - Vineeta Singh
- Microbiology Division, CSIR-Central Drug Research Institute, Lucknow, Uttar Pradesh, India
| | - Kashyap Kumar Dubey
- Industrial Biotechnology Laboratory, University Institute of Engineering and Technology, M.D. University, Rohtak, Haryana, India
| | - Arshad Jawed
- Research and Scientific Studies Unit, College of Nursing and Allied Health Sciences, Jazan University, Jazan, Saudi Arabia
| | - Mohd Wahid
- Research and Scientific Studies Unit, College of Nursing and Allied Health Sciences, Jazan University, Jazan, Saudi Arabia
| | - Sajad Ahmad Dar
- Research and Scientific Studies Unit, College of Nursing and Allied Health Sciences, Jazan University, Jazan, Saudi Arabia
- Departments of Microbiology, University College of Medical Sciences (University of Delhi), Delhi, India
| |
Collapse
|
12
|
Young M, Ozcan A, Rajasekaran P, Kumrah P, Myers ME, Johnson E, Graham JH, Santra S. Fixed-Quat: An Attractive Nonmetal Alternative to Copper Biocides against Plant Pathogens. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2018; 66:13056-13064. [PMID: 30511854 DOI: 10.1021/acs.jafc.8b04189] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/17/2023]
Abstract
In this paper, we report a nonphytotoxic bactericide and fungicide formulation containing a composite of silica and quaternary ammonium compound (quat). The composite material was prepared using an acid-catalyzed sol-gel method. Positively charged quat was associated with a negatively charged silica-gel matrix, producing a stable suspension of fixed-quat gel (FQ-G). The morphology of FQ-G and the interaction of quat with silica were characterized using SEM and FTIR, respectively. Silica gel significantly reduced quat phytotoxicity when tested at 500 and 1000 μg/mL foliar-application rates. The in vitro antimicrobial efficacy of FQ-G was evaluated against Xanthomonas alfalfae, Pseudomonas syringae, and Clavibacter michiganensis, showing comparable efficacies to that of quat itself. In field conditions, its efficacy in controlling the bacterial and fungal diseases citrus canker, scab, and melanose on 'Ray Ruby' red grapefruit was evaluated. Foliar application rates at 100 and 200 μg/mL provided comparable disease control to those of several copper standards, demonstrating the potential for use as an alternative agricultural biocide.
Collapse
Affiliation(s)
| | | | | | | | - Monty E Myers
- Citrus Research and Education Center , University of Florida , 700 Experiment Station Road , Lake Alfred , Florida 33850 , United States
| | - Evan Johnson
- Citrus Research and Education Center , University of Florida , 700 Experiment Station Road , Lake Alfred , Florida 33850 , United States
| | - James H Graham
- Citrus Research and Education Center , University of Florida , 700 Experiment Station Road , Lake Alfred , Florida 33850 , United States
| | | |
Collapse
|
13
|
Palmieri G, Tatè R, Gogliettino M, Balestrieri M, Rea I, Terracciano M, Proroga YT, Capuano F, Anastasio A, De Stefano L. Small Synthetic Peptides Bioconjugated to Hybrid Gold Nanoparticles Destroy Potentially Deadly Bacteria at Submicromolar Concentrations. Bioconjug Chem 2018; 29:3877-3885. [PMID: 30352512 DOI: 10.1021/acs.bioconjchem.8b00706] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Synthetic antibacterial peptides are advanced weapons that scientists design and produce to confront current threats of harmful and mortal pathogens, which could affect humans in everyday life. Recently, many small amino acid sequences, greatly efficient in their antibacterial action, have been reported in the literature. To date, only a few synthetic peptides, acting at micromolar or even tenths of micromolar concentrations, are on the market as commercial products, mainly because of their high cost of production. In this context, materials science can provide fundamental help by engineering small synthetic peptides, powered by hybrid gold nanoparticles, which have been found to strongly enhance antimicrobial activity against bacterial infections. Submicromolar concentrations of the 1018K6 peptide, bioconjugated to hybrid polymer-gold nanoparticles, kill almost 100% of pathogen bacteria, such as Listeria and Salmonella genera, paving the way for economically sustainable commercial products based on this synthetic nanocomplex.
Collapse
Affiliation(s)
- Gianna Palmieri
- Institute of Biosciences and BioResources, UOS Na , National Research Council , Via Pietro Castellino 111 , 80131 Naples , Italy.,Materias S.r.l. , Corso N. Protopisani n. 50 , 80146 Naples , Italy
| | - Rosarita Tatè
- Institute of Genetics and Biophysics , National Research Council , Via Pietro Castellino 111 , 80131 Naples , Italy
| | - Marta Gogliettino
- Institute of Biosciences and BioResources, UOS Na , National Research Council , Via Pietro Castellino 111 , 80131 Naples , Italy
| | - Marco Balestrieri
- Institute of Biosciences and BioResources, UOS Na , National Research Council , Via Pietro Castellino 111 , 80131 Naples , Italy
| | - Ilaria Rea
- Institute for Microelectronics and Microsystems , National Research Council , Via Pietro Castellino 111 , 80131 Naples , Italy
| | - Monica Terracciano
- Institute for Microelectronics and Microsystems , National Research Council , Via Pietro Castellino 111 , 80131 Naples , Italy.,Materias S.r.l. , Corso N. Protopisani n. 50 , 80146 Naples , Italy
| | - Yolande Therese Proroga
- Department of Food Microbiology , Istituto Zooprofilattico Sperimentale del Mezzogiorno , via della Salute, 2 , 80055 Portici, Naples , Italy
| | - Federico Capuano
- Department of Food Microbiology , Istituto Zooprofilattico Sperimentale del Mezzogiorno , via della Salute, 2 , 80055 Portici, Naples , Italy
| | - Aniello Anastasio
- Department of Veterinary Medicine and Animal Production , University of Naples Federico II , Via Federico Delpino 1 , 80137 Naples , Italy
| | - Luca De Stefano
- Institute for Microelectronics and Microsystems , National Research Council , Via Pietro Castellino 111 , 80131 Naples , Italy.,Materias S.r.l. , Corso N. Protopisani n. 50 , 80146 Naples , Italy
| |
Collapse
|
14
|
The Pseudomonas aeruginosa PilSR Two-Component System Regulates Both Twitching and Swimming Motilities. mBio 2018; 9:mBio.01310-18. [PMID: 30042200 PMCID: PMC6058289 DOI: 10.1128/mbio.01310-18] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
Abstract
Motility is an important virulence trait for many bacterial pathogens, allowing them to position themselves in appropriate locations at appropriate times. The motility structures type IV pili and flagella are also involved in sensing surface contact, which modulates pathogenicity. In Pseudomonas aeruginosa, the PilS-PilR two-component system (TCS) regulates expression of the type IV pilus (T4P) major subunit PilA, while biosynthesis of the single polar flagellum is regulated by a hierarchical system that includes the FleSR TCS. Previous studies of Geobacter sulfurreducens and Dichelobacter nodosus implicated PilR in regulation of non-T4P-related genes, including some involved in flagellar biosynthesis. Here we used transcriptome sequencing (RNA-seq) analysis to identify genes in addition to pilA with changes in expression in the absence of pilR. Among the genes identified were 10 genes whose transcription increased in the pilA mutant but decreased in the pilR mutant, despite both mutants lacking T4P and pilus-related phenotypes. The products of these inversely dysregulated genes, many of which were hypothetical, may be important for virulence and surface-associated behaviors, as mutants had altered swarming motility, biofilm formation, type VI secretion system expression, and pathogenicity in a nematode model. Further, the PilSR TCS positively regulated transcription of fleSR, and thus many genes in the FleSR regulon. As a result, pilSR deletion mutants had defects in swimming motility that were independent of the loss of PilA. Together, these data suggest that in addition to controlling T4P expression, PilSR could have a broader role in the regulation of P. aeruginosa motility and surface sensing behaviors. Surface appendages such as type IV pili and flagella are important for establishing surface attachment and infection in a host in response to appropriate cues. The PilSR regulatory system that controls type IV pilus expression in Pseudomonas aeruginosa has an established role in expression of the major pilin PilA. Here we provide evidence supporting a new role for PilSR in regulating flagellum-dependent swimming motility in addition to pilus-dependent twitching motility. Further, even though both pilA and pilR mutants lack PilA and pili, we identified sets of genes downregulated in the pilR mutant and upregulated in a pilA mutant as well as genes downregulated only in a pilR mutant, independent of pilus expression. This finding suggests that change in the inner membrane levels of PilA is only one of the cues to which PilR responds to modulate gene expression. Identification of PilR as a regulator of multiple motility pathways may make it an interesting therapeutic target for antivirulence compounds.
Collapse
|
15
|
Haque S, Ahmad F, Dar SA, Jawed A, Mandal RK, Wahid M, Lohani M, Khan S, Singh V, Akhter N. Developments in strategies for Quorum Sensing virulence factor inhibition to combat bacterial drug resistance. Microb Pathog 2018; 121:293-302. [PMID: 29857121 DOI: 10.1016/j.micpath.2018.05.046] [Citation(s) in RCA: 61] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2018] [Revised: 05/27/2018] [Accepted: 05/28/2018] [Indexed: 12/22/2022]
Abstract
Quorum sensing (QS) is a complex bacterial intercellular communication system. It is mediated by molecules called auto-inducers (AIs) and allows coordinated responses to a variety of environmental signals by inducing alterations in gene expression. Communication through QS can tremendously stimulate the pathogenicity and virulence via multiple mechanisms in pathogenic bacteria. The present review explores the major types of multitudinous QS systems known in Gram-positive and Gram-negative bacteria and their roles in bacterial pathogenesis and drug resistance. Because bacterial resistance to antibiotics is increasingly becoming a significant clinical challenge to human health; alternate strategies to combat drug resistance are warranted. Targeting bacterial pathogenicity by interruptions in QS using natural QS inhibitors and synthetic quorum-quenching analogs are being increasingly considered for development of next generation antimicrobials. The review highlights the recent advancements in discovery of promising new QS modulators and their efficiency in controlling infections caused by multidrug-resistant bacterial pathogens.
Collapse
Affiliation(s)
- Shafiul Haque
- Research and Scientific Studies Unit, College of Nursing & Allied Health Sciences, Jazan University, Jazan, 45142, Saudi Arabia.
| | - Faraz Ahmad
- Department of Public Health, College of Public Health, Imam Abdulrahman Bin Faisal University, Dammam, 31441, Saudi Arabia
| | - Sajad A Dar
- Research and Scientific Studies Unit, College of Nursing & Allied Health Sciences, Jazan University, Jazan, 45142, Saudi Arabia
| | - Arshad Jawed
- Research and Scientific Studies Unit, College of Nursing & Allied Health Sciences, Jazan University, Jazan, 45142, Saudi Arabia
| | - Raju K Mandal
- Research and Scientific Studies Unit, College of Nursing & Allied Health Sciences, Jazan University, Jazan, 45142, Saudi Arabia
| | - Mohd Wahid
- Research and Scientific Studies Unit, College of Nursing & Allied Health Sciences, Jazan University, Jazan, 45142, Saudi Arabia
| | - Mohtashim Lohani
- Department of Emergency Medical Services, College of Applied Medical Sciences, Jazan University, Jazan, 45142, Saudi Arabia
| | - Saif Khan
- Department of Clinical Laboratory Science, College of Applied Medical Sciences, University of Ha'il, Ha'il, 2440, Saudi Arabia
| | - Vineeta Singh
- Department of Biotechnology, Institute of Engineering & Technology, Lucknow, 226021, Uttar Pradesh, India
| | - Naseem Akhter
- Department of Laboratory Medicine, Faculty of Applied Medical Sciences, Albaha University, Albaha, 65431, Saudi Arabia
| |
Collapse
|
16
|
Abstract
Salmonella typhimurium is an important biofilm-forming bacteria. It is known to be resistant to a wide range of antimicrobials. The present study was carried out to evaluate the effects of dimethyl sulfoxide (DMSO) against S. typhimurium biofilm and investigate whole-cell protein expression by biofilm cells following treatment with DMSO. Antibiofilm activities were assessed using pellicle assay, crystal violet assay, colony-forming unit counting and extracellular polymeric substance (EPS) matrix assay whilst differential protein expression was investigated using a combination of one dimensional sodium dodecyl sulfate polyacrylamide gel electrophoresis, tandem mass spectrometry and bioinformatics. Treatment with 32% DMSO inhibited pellicle formation, biofilm viability, biofilm biomass and several important components of EPS matrix. Subtractive protein profiling identified two unique protein bands (25.4 and 51.2 kDa) which were present only in control biofilm and not in 32% DMSO-treated biofilm. In turn, 29 and 46 proteins were successfully identified from the protein bands of 25.4 and 51.2 kDa respectively. Protein interaction network analysis identified several biological pathways to be affected, including glycolysis, PhoP-PhoQ phosphorelay signalling and flagellar biosynthesis. The present study suggests that DMSO may inhibit multiple biological pathways to control biofilm formation.
Collapse
|
17
|
Oloketuyi SF, Khan F. Inhibition strategies of Listeria monocytogenes biofilms-current knowledge and future outlooks. J Basic Microbiol 2017; 57:728-743. [PMID: 28594071 DOI: 10.1002/jobm.201700071] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2017] [Revised: 05/12/2017] [Accepted: 05/12/2017] [Indexed: 12/30/2022]
Abstract
There is an increasing trend in the food industry on the Listeria monocytogenes biofilm formation and inhibition. This is attributed to its easy survival on contact surfaces, resistance to disinfectants or antibiotics and growth under the stringent condition used for food processing and preservation thereby leading to food contamination products by direct or indirect exposure. Though, there is a lack of conclusive evidences about the mechanism of biofilm formation, in this review, the concept of biofilm formation and various chemical, physical, and green technology approaches to prevent or control the biofilm formed is discussed. State-of-the-art approaches ranging from the application of natural to synthetic molecules with high effectiveness and non-toxicity targeted at the different steps of biofilm formation could positively influence the biofilm inhibition in the future.
Collapse
Affiliation(s)
- Sandra F Oloketuyi
- Department of Biotechnology, School of Engineering and Technology, Sharda University, Greater Noida, U.P., India
| | - Fazlurrahman Khan
- Department of Biotechnology, School of Engineering and Technology, Sharda University, Greater Noida, U.P., India
| |
Collapse
|
18
|
Mass spectrometry imaging identifies palmitoylcarnitine as an immunological mediator during Salmonella Typhimurium infection. Sci Rep 2017; 7:2786. [PMID: 28584281 PMCID: PMC5459799 DOI: 10.1038/s41598-017-03100-5] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2016] [Accepted: 04/25/2017] [Indexed: 12/19/2022] Open
Abstract
Salmonella Typhimurium causes a self-limiting gastroenteritis that may lead to systemic disease. Bacteria invade the small intestine, crossing the intestinal epithelium from where they are transported to the mesenteric lymph nodes (MLNs) within migrating immune cells. MLNs are an important site at which the innate and adaptive immune responses converge but their architecture and function is severely disrupted during S. Typhimurium infection. To further understand host-pathogen interactions at this site, we used mass spectrometry imaging (MSI) to analyse MLN tissue from a murine model of S. Typhimurium infection. A molecule, identified as palmitoylcarnitine (PalC), was of particular interest due to its high abundance at loci of S. Typhimurium infection and MLN disruption. High levels of PalC localised to sites within the MLNs where B and T cells were absent and where the perimeter of CD169+ sub capsular sinus macrophages was disrupted. MLN cells cultured ex vivo and treated with PalC had reduced CD4+CD25+ T cells and an increased number of B220+CD19+ B cells. The reduction in CD4+CD25+ T cells was likely due to apoptosis driven by increased caspase-3/7 activity. These data indicate that PalC significantly alters the host response in the MLNs, acting as a decisive factor in infection outcome.
Collapse
|
19
|
Yahya MFZR, Alias Z, Karsani SA. Antibiofilm activity and mode of action of DMSO alone and its combination with afatinib against Gram-negative pathogens. Folia Microbiol (Praha) 2017; 63:23-30. [PMID: 28540585 DOI: 10.1007/s12223-017-0532-9] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2016] [Accepted: 05/10/2017] [Indexed: 12/20/2022]
Abstract
Biofilms are complex microbial communities that tend to attach to either biotic or abiotic surface. Enclosed in a self-produced extracellular polymeric substance (EPS) matrix, the biofilms often cause persistent infections. The objective of this study was to investigate the antibiofilm activity of dimethyl sulfoxide (DMSO) and afatinib against Gram-negative pathogens. Test microorganisms used in this study were Escherichia coli ATCC 1299, Pseudomonas aeruginosa ATCC 10145, and Salmonella typhimurium ATCC 14028. Biofilms were developed in 96-well microplate at 37°C for 24 h. Following removal of non-adherent cells, analysis of biofilm viability, biofilm biomass, and extracellular polymeric substances (EPS) matrix were performed using resazurin assay, crystal violet assay, and attenuated total reflectance fourier transform infrared (ATR-FTIR) spectroscopy, respectively. Bradford protein assay was conducted to determine the total amount of EPS proteins. The results demonstrated that both 32% DMSO alone and its combination with 3.2 μg/mL afatinib were effective in killing biofilm cells and reducing biofilm biomass. IR spectral variations of EPS matrix of biofilms in the range between 1700 and 900 cm-1 were also observed. Reduction in EPS proteins verified the chemical modifications of EPS matrix. In conclusion, 32% DMSO alone and its combination with 3.2 μg/mL afatinib showed remarkable antibiofilm activities against Gram-negative pathogens. It was suggested that the biofilm inhibition was mediated by the chemical modification of EPS matrix.
Collapse
Affiliation(s)
- Mohd Fakharul Zaman Raja Yahya
- Institute of Biological Sciences, Faculty of Science, University of Malaya, 50603, Kuala Lumpur, Malaysia. .,Faculty of Applied Sciences, Universiti Teknologi MARA Shah Alam, 40450, Shah Alam, Malaysia.
| | - Zazali Alias
- Institute of Biological Sciences, Faculty of Science, University of Malaya, 50603, Kuala Lumpur, Malaysia
| | - Saiful Anuar Karsani
- Institute of Biological Sciences, Faculty of Science, University of Malaya, 50603, Kuala Lumpur, Malaysia.
| |
Collapse
|
20
|
Palmieri G, Balestrieri M, Proroga YT, Falcigno L, Facchiano A, Riccio A, Capuano F, Marrone R, Neglia G, Anastasio A. New antimicrobial peptides against foodborne pathogens: From in silico design to experimental evidence. Food Chem 2016; 211:546-54. [DOI: 10.1016/j.foodchem.2016.05.100] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2016] [Revised: 04/19/2016] [Accepted: 05/16/2016] [Indexed: 10/21/2022]
|
21
|
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: 248] [Impact Index Per Article: 31.0] [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]
|
22
|
Silva LN, Zimmer KR, Macedo AJ, Trentin DS. Plant Natural Products Targeting Bacterial Virulence Factors. Chem Rev 2016; 116:9162-236. [PMID: 27437994 DOI: 10.1021/acs.chemrev.6b00184] [Citation(s) in RCA: 263] [Impact Index Per Article: 32.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Decreased antimicrobial efficiency has become a global public health issue. The paucity of new antibacterial drugs is evident, and the arsenal against infectious diseases needs to be improved urgently. The selection of plants as a source of prototype compounds is appropriate, since plant species naturally produce a wide range of secondary metabolites that act as a chemical line of defense against microorganisms in the environment. Although traditional approaches to combat microbial infections remain effective, targeting microbial virulence rather than survival seems to be an exciting strategy, since the modulation of virulence factors might lead to a milder evolutionary pressure for the development of resistance. Additionally, anti-infective chemotherapies may be successfully achieved by combining antivirulence and conventional antimicrobials, extending the lifespan of these drugs. This review presents an updated discussion of natural compounds isolated from plants with chemically characterized structures and activity against the major bacterial virulence factors: quorum sensing, bacterial biofilms, bacterial motility, bacterial toxins, bacterial pigments, bacterial enzymes, and bacterial surfactants. Moreover, a critical analysis of the most promising virulence factors is presented, highlighting their potential as targets to attenuate bacterial virulence. The ongoing progress in the field of antivirulence therapy may therefore help to translate this promising concept into real intervention strategies in clinical areas.
Collapse
Affiliation(s)
- Laura Nunes Silva
- Faculdade de Farmácia, Universidade Federal do Rio Grande do Sul , Porto Alegre, Rio Grande do Sul 90610-000, Brazil.,Centro de Biotecnologia, Universidade Federal do Rio Grande do Sul , Porto Alegre, Rio Grande do Sul 91501-970, Brazil
| | - Karine Rigon Zimmer
- Departamento de Ciências Básicas da Saúde, Universidade Federal de Ciências da Saúde de Porto Alegre , Porto Alegre, Rio Grande do Sul 90050-170, Brazil
| | - Alexandre José Macedo
- Faculdade de Farmácia, Universidade Federal do Rio Grande do Sul , Porto Alegre, Rio Grande do Sul 90610-000, Brazil.,Centro de Biotecnologia, Universidade Federal do Rio Grande do Sul , Porto Alegre, Rio Grande do Sul 91501-970, Brazil.,Instituto Nacional do Semiárido , Campina Grande, Paraı́ba 58429-970, Brazil
| | - Danielle Silva Trentin
- Faculdade de Farmácia, Universidade Federal do Rio Grande do Sul , Porto Alegre, Rio Grande do Sul 90610-000, Brazil.,Centro de Biotecnologia, Universidade Federal do Rio Grande do Sul , Porto Alegre, Rio Grande do Sul 91501-970, Brazil
| |
Collapse
|
23
|
Gowrishankar S, Sivaranjani M, Kamaladevi A, Ravi AV, Balamurugan K, Karutha Pandian S. Cyclic dipeptide cyclo(l-leucyl-l-prolyl) from marineBacillus amyloliquefaciensmitigates biofilm formation and virulence inListeria monocytogenes. Pathog Dis 2016; 74:ftw017. [DOI: 10.1093/femspd/ftw017] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/29/2016] [Indexed: 01/18/2023] Open
|
24
|
Meadows JA, Wargo MJ. Carnitine in bacterial physiology and metabolism. MICROBIOLOGY (READING, ENGLAND) 2015; 161:1161-74. [PMID: 25787873 PMCID: PMC4635513 DOI: 10.1099/mic.0.000080] [Citation(s) in RCA: 103] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/05/2015] [Accepted: 03/17/2015] [Indexed: 12/23/2022]
Abstract
Carnitine is a quaternary amine compound found at high concentration in animal tissues, particularly muscle, and is most well studied for its contribution to fatty acid transport into mitochondria. In bacteria, carnitine is an important osmoprotectant, and can also enhance thermotolerance, cryotolerance and barotolerance. Carnitine can be transported into the cell or acquired from metabolic precursors, where it can serve directly as a compatible solute for stress protection or be metabolized through one of a few distinct pathways as a nutrient source. In this review, we summarize what is known about carnitine physiology and metabolism in bacteria. In particular, recent advances in the aerobic and anaerobic metabolic pathways as well as the use of carnitine as an electron acceptor have addressed some long-standing questions in the field.
Collapse
Affiliation(s)
- Jamie A. Meadows
- Department of Microbiology and Molecular Genetics, University of Vermont College of Medicine, 95 Carrigan Drive, Burlington, VT, 05405, USA
| | - Matthew J. Wargo
- Department of Microbiology and Molecular Genetics, University of Vermont College of Medicine, 95 Carrigan Drive, Burlington, VT, 05405, USA
| |
Collapse
|
25
|
Gorityala BK, Guchhait G, Schweizer F. Amphiphilic Aminoglycoside Antimicrobials in Antibacterial Discovery. CARBOHYDRATES IN DRUG DESIGN AND DISCOVERY 2015. [DOI: 10.1039/9781849739993-00255] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
Amphiphilic aminoglycoside antimicrobials (AAAs) are an emerging class of polycationic antibacterial agents with broad-spectrum antibacterial activity. In contrast to aminoglycosides, which interfere with protein synthesis by interacting with the 30S ribosomal subunit, AAAs appear to target the bacterial cell wall by interactions with extracellular lipids or proteins or by enhancing the permeability of the bacterial cell wall. The physicochemical similarities between amphiphilic aminoglycosides and antimicrobial peptides, another class of polycationic amphiphiles with broad-spectrum antibacterial activity, suggest similar mode(s) of action. However, in contrast to antimicrobial peptides, AAAs are not composed of peptide bonds and as such promise to display superior metabolic stability. As a result, AAAs may be considered to be a novel class of antimicrobial peptidomimetics. Many AAAs possess impressive potent antibacterial activity against Gram-positive and Gram-negative bacteria, especially against bacterial strains that are resistant to clinically used antibiotics. In summary, AAAs promise to provide a new and rich source of antibacterial lead structures to combat antibiotic-resistant and multidrug-resistant pathogens.
Collapse
Affiliation(s)
| | - Goutam Guchhait
- Department of Chemistry, University of Manitoba Winnipeg, MB R3T 2N2 Canada
| | - Frank Schweizer
- Department of Chemistry, University of Manitoba Winnipeg, MB R3T 2N2 Canada
| |
Collapse
|
26
|
Melander RJ, Melander C. Innovative strategies for combating biofilm-based infections. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2015; 831:69-91. [PMID: 25384664 DOI: 10.1007/978-3-319-09782-4_6] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Affiliation(s)
- Roberta J Melander
- Department of Chemistry, North Carolina State University, Raleigh, NC, USA
| | | |
Collapse
|
27
|
Nguyen UT, Harvey H, Hogan AJ, Afonso ACF, Wright GD, Burrows LL. Role of PBPD1 in stimulation of Listeria monocytogenes biofilm formation by subminimal inhibitory β-lactam concentrations. Antimicrob Agents Chemother 2014; 58:6508-17. [PMID: 25136010 PMCID: PMC4249420 DOI: 10.1128/aac.03671-14] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2014] [Accepted: 08/10/2014] [Indexed: 12/19/2022] Open
Abstract
Disinfectant-tolerant Listeria monocytogenes biofilms can colonize surfaces that come into contact with food, leading to contamination and, potentially, food-borne illnesses. To better understand the process of L. monocytogenes biofilm formation and dispersal, we screened 1,120 off-patent FDA-approved drugs and identified several that modulate Listeria biofilm development. Among the hits were more than 30 β-lactam antibiotics, with effects ranging from inhibiting (≤50%) to stimulating (≥200%) biofilm formation compared to control. Most β-lactams also dispersed a substantial proportion of established biofilms. This phenotype did not necessarily involve killing, as >50% dispersal could be achieved with concentrations as low as 1/20 of the MIC of some cephalosporins. Penicillin-binding protein (PBP) profiling using a fluorescent penicillin analogue showed similar inhibition patterns for most β-lactams, except that biofilm-stimulatory drugs did not bind PBPD1, a low-molecular-weight d,d-carboxypeptidase. Compared to the wild type, a pbpD1 mutant had an attenuated biofilm response to stimulatory β-lactams. The cephalosporin-responsive CesRK two-component regulatory system, whose regulon includes PBPs, was not required for the response. The requirement for PBPD1 activity for β-lactam stimulation of L. monocytogenes biofilms shows that the specific set of PBPs that are inactivated by a particular drug dictates whether a protective biofilm response is provoked.
Collapse
Affiliation(s)
- Uyen T Nguyen
- Department of Biochemistry and Biomedical Sciences and the Michael G. DeGroote Institute for Infectious Diseases Research, McMaster University, Hamilton, Ontario, Canada
| | - Hanjeong Harvey
- Department of Biochemistry and Biomedical Sciences and the Michael G. DeGroote Institute for Infectious Diseases Research, McMaster University, Hamilton, Ontario, Canada
| | - Andrew J Hogan
- Department of Biochemistry and Biomedical Sciences and the Michael G. DeGroote Institute for Infectious Diseases Research, McMaster University, Hamilton, Ontario, Canada
| | - Alexandria C F Afonso
- Department of Biochemistry and Biomedical Sciences and the Michael G. DeGroote Institute for Infectious Diseases Research, McMaster University, Hamilton, Ontario, Canada
| | - Gerard D Wright
- Department of Biochemistry and Biomedical Sciences and the Michael G. DeGroote Institute for Infectious Diseases Research, McMaster University, Hamilton, Ontario, Canada
| | - Lori L Burrows
- Department of Biochemistry and Biomedical Sciences and the Michael G. DeGroote Institute for Infectious Diseases Research, McMaster University, Hamilton, Ontario, Canada
| |
Collapse
|
28
|
Nguyen UT, Burrows LL. DNase I and proteinase K impair Listeria monocytogenes biofilm formation and induce dispersal of pre-existing biofilms. Int J Food Microbiol 2014; 187:26-32. [PMID: 25043896 DOI: 10.1016/j.ijfoodmicro.2014.06.025] [Citation(s) in RCA: 100] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2014] [Revised: 05/19/2014] [Accepted: 06/28/2014] [Indexed: 12/15/2022]
Abstract
Current sanitation methods in the food industry are not always sufficient for prevention or dispersal of Listeria monocytogenes biofilms. Here, we determined if prevention of adherence or dispersal of existing biofilms could occur if biofilm matrix components were disrupted enzymatically. Addition of DNase during biofilm formation reduced attachment (<50% of control) to polystyrene. Treatment of established 72h biofilms with 100μg/ml of DNase for 24h induced incomplete biofilm dispersal, with <25% biofilm remaining compared to control. In contrast, addition of proteinase K completely inhibited biofilm formation, and 72h biofilms-including those grown under stimulatory conditions-were completely dispersed with 100μg/ml proteinase K. Generally-regarded-as-safe proteases bromelain and papain were less effective dispersants than proteinase K. In a time course assay, complete dispersal of L. monocytogenes biofilms from both polystyrene and type 304H food-grade stainless steel occurred within 5min at proteinase K concentrations above 25μg/ml. These data confirm that both DNA and proteins are required for L. monocytogenes biofilm development and maintenance, and that these components of the biofilm matrix can be targeted for effective prevention and removal of biofilms.
Collapse
Affiliation(s)
- Uyen T Nguyen
- Department of Biochemistry and Biomedical Sciences and the Michael G. DeGroote Institute for Infectious Diseases Research, McMaster University, Hamilton, ON, Canada
| | - Lori L Burrows
- Department of Biochemistry and Biomedical Sciences and the Michael G. DeGroote Institute for Infectious Diseases Research, McMaster University, Hamilton, ON, Canada.
| |
Collapse
|
29
|
Wright GD. Something old, something new: revisiting natural products in antibiotic drug discovery. Can J Microbiol 2014; 60:147-54. [DOI: 10.1139/cjm-2014-0063] [Citation(s) in RCA: 160] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Antibiotic discovery is in crisis. Despite a growing need for new drugs resulting from the increasing number of multi-antibiotic-resistant pathogens, there have been only a handful of new antibiotics approved for clinical use in the past 2 decades. Faced with scientific, economic, and regulatory challenges, the pharmaceutical sector seems unable to respond to what has been called an “apocalyptic” threat. Natural products produced by bacteria and fungi are genetically encoded products of natural selection that have been the mainstay sources of the antibiotics in current clinical use. The pharmaceutical industry has largely abandoned these compounds in favor of large libraries of synthetic molecules because of difficulties in identifying new natural product antibiotics scaffolds. Advances in next-generation genome sequencing, bioinformatics, and analytical chemistry are combining to overcome barriers to natural products. Coupled with new strategies in antibiotic discovery, including inhibition of resistance, novel drug combinations, and new targets, natural products are poised for a renaissance to address what is a pressing health care crisis.
Collapse
Affiliation(s)
- Gerard D. Wright
- Michael G. DeGroote Institute for Infectious Disease Research, Department of Biochemistry and Biomedical Sciences, McMaster University, 1280 Main Street West, Hamilton, ON L8S 4K1, Canada
| |
Collapse
|
30
|
Szczepanski S, Lipski A. Essential oils show specific inhibiting effects on bacterial biofilm formation. Food Control 2014. [DOI: 10.1016/j.foodcont.2013.08.023] [Citation(s) in RCA: 54] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
31
|
The dynamics of peptidoglycan structure and function: conference report on the 3rd Great Wall Symposium. Res Microbiol 2013; 165:60-7. [PMID: 24239960 DOI: 10.1016/j.resmic.2013.11.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
|
32
|
Herzog IM, Feldman M, Eldar-Boock A, Satchi-Fainaro R, Fridman M. Design of membrane targeting tobramycin-based cationic amphiphiles with reduced hemolytic activity. MEDCHEMCOMM 2013. [DOI: 10.1039/c2md20162c] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
Hemolysis caused by amphiphilic tobramycin antimicrobial agents is significantly reduced by altering the bond linking their hydrophobic and hydrophilic parts.
Collapse
Affiliation(s)
- Ido M. Herzog
- School of Chemistry
- Tel Aviv University
- Tel Aviv 69978
- Israel
| | - Mark Feldman
- School of Chemistry
- Tel Aviv University
- Tel Aviv 69978
- Israel
| | - Anat Eldar-Boock
- Department of Physiology and Pharmacology
- Sackler School of Medicine
- Tel Aviv University
- Tel Aviv 69978
- Israel
| | - Ronit Satchi-Fainaro
- Department of Physiology and Pharmacology
- Sackler School of Medicine
- Tel Aviv University
- Tel Aviv 69978
- Israel
| | - Micha Fridman
- School of Chemistry
- Tel Aviv University
- Tel Aviv 69978
- Israel
| |
Collapse
|
33
|
Current knowledge and perspectives on biofilm formation: the case of Listeria monocytogenes. Appl Microbiol Biotechnol 2012; 97:957-68. [DOI: 10.1007/s00253-012-4611-1] [Citation(s) in RCA: 82] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2012] [Revised: 11/22/2012] [Accepted: 11/22/2012] [Indexed: 01/10/2023]
|
34
|
Abstract
Bacterial biofilms are defined as a surface attached community of bacteria embedded in a matrix of extracellular polymeric substances that they have produced. When in the biofilm state, bacteria are more resistant to antibiotics and the host immune response than are their planktonic counterparts. Biofilms are increasingly recognized as being significant in human disease, accounting for 80% of bacterial infections in the body and diseases associated with bacterial biofilms include: lung infections of cystic fibrosis patients, colitis, urethritis, conjunctivitis, otitis, endocarditis and periodontitis. Additionally, biofilm infections of indwelling medical devices are of particular concern, as once the device is colonized infection is virtually impossible to eradicate. Given the prominence of biofilms in infectious diseases, there has been an increased effort toward the development of small molecules that will modulate bacterial biofilm development and maintenance. In this review, we highlight the development of small molecules that inhibit and/or disperse bacterial biofilms through non-microbicidal mechanisms. The review discuses the numerous approaches that have been applied to the discovery of lead small molecules that mediate biofilm development. These approaches are grouped into: (1) the identification and development of small molecules that target one of the bacterial signaling pathways involved in biofilm regulation, (2) chemical library screening for compounds with anti-biofilm activity, and (3) the identification of natural products that possess anti-biofilm activity, and the chemical manipulation of these natural products to obtain analogues with increased activity.
Collapse
|