151
|
Lories B, Roberfroid S, Dieltjens L, De Coster D, Foster KR, Steenackers HP. Biofilm Bacteria Use Stress Responses to Detect and Respond to Competitors. Curr Biol 2020; 30:1231-1244.e4. [PMID: 32084407 PMCID: PMC7322538 DOI: 10.1016/j.cub.2020.01.065] [Citation(s) in RCA: 72] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2019] [Revised: 10/30/2019] [Accepted: 01/17/2020] [Indexed: 12/28/2022]
Abstract
Bacteria use complex regulatory networks to cope with stress, but the function of these networks in natural habitats is poorly understood. The competition sensing hypothesis states that bacterial stress response systems can serve to detect ecological competition, but studying regulatory responses in diverse communities is challenging. Here, we solve this problem by using differential fluorescence induction to screen the Salmonella Typhimurium genome for loci that respond, at the single-cell level, to life in biofilms with competing strains of S. Typhimurium and Escherichia coli. This screening reveals the presence of competing strains drives up the expression of genes associated with biofilm matrix production (CsgD pathway), epithelial invasion (SPI1 invasion system), and, finally, chemical efflux and antibiotic tolerance (TolC efflux pump and AadA aminoglycoside 3-adenyltransferase). We validate that these regulatory changes result in the predicted phenotypic changes in biofilm, mammalian cell invasion, and antibiotic tolerance. We further show that these responses arise via activation of major stress responses, providing direct support for the competition sensing hypothesis. Moreover, inactivation of the type VI secretion system (T6SS) of a competitor annuls the responses to competition, indicating that T6SS-derived cell damage activates these stress response systems. Our work shows that bacteria use stress responses to detect and respond to competition in a manner important for major phenotypes, including biofilm formation, virulence, and antibiotic tolerance.
Collapse
Affiliation(s)
- Bram Lories
- Centre of Microbial and Plant Genetics (CMPG), Department of Microbial and Molecular Systems, KU Leuven, Kasteelpark Arenberg 20, 3001 Leuven, Belgium
| | - Stefanie Roberfroid
- Centre of Microbial and Plant Genetics (CMPG), Department of Microbial and Molecular Systems, KU Leuven, Kasteelpark Arenberg 20, 3001 Leuven, Belgium
| | - Lise Dieltjens
- Centre of Microbial and Plant Genetics (CMPG), Department of Microbial and Molecular Systems, KU Leuven, Kasteelpark Arenberg 20, 3001 Leuven, Belgium
| | - David De Coster
- Centre of Microbial and Plant Genetics (CMPG), Department of Microbial and Molecular Systems, KU Leuven, Kasteelpark Arenberg 20, 3001 Leuven, Belgium
| | - Kevin R Foster
- Department of Zoology, University of Oxford, Oxford OX1 3PS, UK.
| | - Hans P Steenackers
- Centre of Microbial and Plant Genetics (CMPG), Department of Microbial and Molecular Systems, KU Leuven, Kasteelpark Arenberg 20, 3001 Leuven, Belgium; Department of Zoology, University of Oxford, Oxford OX1 3PS, UK.
| |
Collapse
|
152
|
Khan F, Lee JW, Pham DTN, Kim YM. Chitooligosaccharides as Antibacterial, Antibiofilm, Antihemolytic and Anti-Virulence Agent against Staphylococcus aureus. Curr Pharm Biotechnol 2020; 20:1223-1233. [PMID: 31475895 DOI: 10.2174/1389201020666190902130722] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2019] [Revised: 05/16/2019] [Accepted: 08/20/2019] [Indexed: 12/11/2022]
Abstract
BACKGROUND Staphylococcus aureus nosocomial infections with a high mortality rate in human and animals have been reported to associate with bacterial biofilm formation, along with the secretion of numerous virulence factors. Therefore, the inhibition of biofilm formation and attenuation of virulence determinants are considered as a promising solution to combat the spread of S. aureus infections. Modern trends in antibiofilm therapies have opted for the active agents that are biocompatible, biodegradable, non-toxic and cost-effective. Owning the aforementioned properties, chitosan, a natural N-acetylated carbohydrate biopolymer derived from chitin, has been favorably employed. Recently, the chitosan structure has been chemically modified into Chitooligosaccharides (COS) to overcome its limited solubility in water, thus widening chitosan applications in modern antibiofilm research. In the present study, we have investigated the antibacterial, antibiofilm and anti-virulence activities against S. aureus of COS of different molecular weights dissolved in neutral water. METHODS The study of bactericidal activity was performed using the micro-dilution method while the biofilm inhibition assay was performed using crystal-violet staining method and confirmed by scanning electron microscopic analysis. The inhibition of amyloid protein production was confirmed by Congo Red staining. RESULTS Results showed that low molecular weight COS exhibited bactericidal activity and reduced the bacterial amylogenesis, hemolytic activity as well as H2O2 resistance properties, while slightly inhibiting biofilm formation. The present study provides a new insight for further applications of the water-soluble COS as a safe and cost-effective drug for the treatment of S. aureus biofilm-associated infections. CONCLUSION Reducing the molecular weight of chitosan in the form of COS has become an effective strategy to maintain chitosan biological activity while improving its water solubility. The low molecular weight COS investigated in this study have effectively performed antibacterial, antibiofilm and antivirulence properties against S. aureus.
Collapse
Affiliation(s)
- Fazlurrahman Khan
- Marine-Integrated Bionics Research Center, Pukyong National University, Busan 48513, Korea
| | - Jang-Won Lee
- Department of Food Science and Technology, Pukyong National University, Busan 48513, Korea
| | - Dung T N Pham
- Department of Food Science and Technology, Pukyong National University, Busan 48513, Korea
| | - Young-Mog Kim
- Marine-Integrated Bionics Research Center, Pukyong National University, Busan 48513, Korea.,Department of Food Science and Technology, Pukyong National University, Busan 48513, Korea
| |
Collapse
|
153
|
Rezzoagli C, Granato ET, Kümmerli R. Harnessing bacterial interactions to manage infections: a review on the opportunistic pathogen Pseudomonas aeruginosa as a case example. J Med Microbiol 2020; 69:147-161. [PMID: 31961787 PMCID: PMC7116537 DOI: 10.1099/jmm.0.001134] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
During infections, bacterial pathogens can engage in a variety of interactions with each other, ranging from the cooperative sharing of resources to deadly warfare. This is especially relevant in opportunistic infections, where different strains and species often co-infect the same patient and interact in the host. Here, we review the relevance of these social interactions during opportunistic infections using the human pathogen Pseudomonas aeruginosa as a case example. In particular, we discuss different types of pathogen-pathogen interactions, involving both cooperation and competition, and elaborate on how they impact virulence in multi-strain and multi-species infections. We then review evolutionary dynamics within pathogen populations during chronic infections. We particuarly discuss how local adaptation through niche separation, evolutionary successions and antagonistic co-evolution between pathogens can alter virulence and the damage inflicted on the host. Finally, we outline how studying bacterial social dynamics could be used to manage infections. We show that a deeper appreciation of bacterial evolution and ecology in the clinical context is important for understanding microbial infections and can inspire novel treatment strategies.
Collapse
Affiliation(s)
- Chiara Rezzoagli
- Department of Quantitative Biomedicine, University of Zurich, Zurich, Switzerland
| | - Elisa T. Granato
- Department of Zoology, University of Oxford, Oxford, United Kingdom
- Department of Biochemistry, University of Oxford, Oxford, United Kingdom
| | - Rolf Kümmerli
- Department of Quantitative Biomedicine, University of Zurich, Zurich, Switzerland
| |
Collapse
|
154
|
Oliveira JH, Bahia AC, Vale PF. How are arbovirus vectors able to tolerate infection? DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2020; 103:103514. [PMID: 31585195 DOI: 10.1016/j.dci.2019.103514] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/16/2019] [Revised: 09/20/2019] [Accepted: 10/01/2019] [Indexed: 06/10/2023]
Abstract
One of the defining features of mosquito vectors of arboviruses such as Dengue and Zika is their ability to tolerate high levels of virus proliferation without suffering significant pathology. This adaptation is central to vector competence and disease spread. The molecular mechanisms, pathways, cellular and metabolic adaptations responsible for mosquito disease tolerance are still largely unknown and may represent effective ways to control mosquito populations and prevent arboviral diseases. In this review article, we describe the key link between disease tolerance and pathogen transmission, and how vector control methods may benefit by focusing efforts on dissecting the mechanisms underlying mosquito tolerance of arboviral infections. We briefly review recent work investigating tolerance mechanisms in other insects, describe the state of the art regarding the mechanisms of disease tolerance in mosquitos, and highlight the emerging role of gut microbiota in mosquito immunity and disease tolerance.
Collapse
Affiliation(s)
- José Henrique Oliveira
- Departamento de Microbiologia, Imunologia e Parasitologia, Universidade Federal de Santa Catarina, Florianopolis, SC, Brazil.
| | - Ana Cristina Bahia
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil.
| | - Pedro F Vale
- Institute of Evolutionary Biology, School of Biological Sciences, University of Edinburgh, Edinburgh, United Kingdom.
| |
Collapse
|
155
|
Dieltjens L, Appermans K, Lissens M, Lories B, Kim W, Van der Eycken EV, Foster KR, Steenackers HP. Inhibiting bacterial cooperation is an evolutionarily robust anti-biofilm strategy. Nat Commun 2020; 11:107. [PMID: 31919364 PMCID: PMC6952394 DOI: 10.1038/s41467-019-13660-x] [Citation(s) in RCA: 110] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2019] [Accepted: 11/14/2019] [Indexed: 01/06/2023] Open
Abstract
Bacteria commonly form dense biofilms encased in extracellular polymeric substances (EPS). Biofilms are often extremely tolerant to antimicrobials but their reliance on shared EPS may also be a weakness as social evolution theory predicts that inhibiting shared traits can select against resistance. Here we show that EPS of Salmonella biofilms is a cooperative trait whose benefit is shared among cells, and that EPS inhibition reduces both cell attachment and antimicrobial tolerance. We then compare an EPS inhibitor to conventional antimicrobials in an evolutionary experiment. While resistance against conventional antimicrobials rapidly evolves, we see no evolution of resistance to EPS inhibition. We further show that a resistant strain is outcompeted by a susceptible strain under EPS inhibitor treatment, explaining why resistance does not evolve. Our work suggests that targeting cooperative traits is a viable solution to the problem of antimicrobial resistance. Bacterial biofilms rely on shared extracellular polymeric substances (EPS) and are often highly tolerant to antibiotics. Here, the authors show in in vitro experiments that Salmonella does not evolve resistance to EPS inhibition because such strains are outcompeted by a susceptible strain under inhibitor treatment.
Collapse
Affiliation(s)
- Lise Dieltjens
- Department of Microbial and Molecular Systems, Centre of Microbial and Plant Genetics (CMPG), KU Leuven, Leuven, Belgium
| | - Kenny Appermans
- Department of Microbial and Molecular Systems, Centre of Microbial and Plant Genetics (CMPG), KU Leuven, Leuven, Belgium
| | - Maries Lissens
- Department of Microbial and Molecular Systems, Centre of Microbial and Plant Genetics (CMPG), KU Leuven, Leuven, Belgium
| | - Bram Lories
- Department of Microbial and Molecular Systems, Centre of Microbial and Plant Genetics (CMPG), KU Leuven, Leuven, Belgium
| | - Wook Kim
- Department of Zoology and Department of Biochemistry, University of Oxford, Oxford, UK.,Department of Biological Sciences, Duquesne University, Pittsburgh, USA
| | - Erik V Van der Eycken
- Department of Chemistry, Laboratory for Organic & Microwave-Assisted Chemistry (LOMAC), KU Leuven, Leuven, Belgium.,Peoples' Friendship University of Russia (RUDN University), 6 Miklukho-Maklaya street, Moscow, Russia
| | - Kevin R Foster
- Department of Zoology and Department of Biochemistry, University of Oxford, Oxford, UK.
| | - Hans P Steenackers
- Department of Microbial and Molecular Systems, Centre of Microbial and Plant Genetics (CMPG), KU Leuven, Leuven, Belgium. .,Department of Zoology and Department of Biochemistry, University of Oxford, Oxford, UK.
| |
Collapse
|
156
|
Abstract
The inexorable emergence of mosquito-borne arboviruses and the failure of traditional vector control methods to prevent their transmission have triggered the development of alternative entomological interventions to render mosquito populations incapable of carrying arboviruses. Here, we use a theoretical framework to argue that decreasing mosquito tolerance to arbovirus infection could be a more evolutionarily sustainable disease control strategy than increasing mosquito resistance. Increasing resistance is predicted to select for mutant arboviruses escaping resistance, whereas reducing tolerance should lead to the death of infected vectors and thus select for mosquito-attenuated arbovirus variants that are less transmissible.
Collapse
Affiliation(s)
- Louis Lambrechts
- Institut Pasteur, Insect-Virus Interactions Unit, Department of Virology, UMR2000, CNRS, 75015 Paris, France.
| | - Maria-Carla Saleh
- Institut Pasteur, Viruses and RNA Interference Unit, Department of Virology, UMR3569, CNRS, 75015 Paris, France.
| |
Collapse
|
157
|
Virulence factors of Paracoccidioides brasiliensis as therapeutic targets: a review. Antonie van Leeuwenhoek 2020; 113:593-604. [PMID: 31902009 DOI: 10.1007/s10482-019-01382-5] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/17/2019] [Accepted: 12/26/2019] [Indexed: 12/17/2022]
Abstract
Paracoccidiodomycosis (PCM) is a systemic mycosis caused by the fungus Paracoccidioides brasiliensis and Paracoccidioides lutzii. The disease requires long and complicated treatment. The aim of this review is to address the fungal virulence factors that could be the target of the development of new drugs for PCM treatment. Virulence factors favoring the process of fungal infection and pathogenicity are considered as a microbial attribute associated with host susceptibility. P. brasiliensis has some known virulence factors which are 43 kDa glycoprotein (gp 43) which is an important fungal antigen, 70 kDa glycoprotein (gp 70), the carbohydrates constituting the fungal cell wall α-1,3, glucan and β-1,3-glucan, cell adhesion molecules and the presence of melanin pigments. The discovery and development of drugs that interact with these factors, such as inhibitors of β-1,3-glucan, reduced synthesis of gp 43, inhibitors of melanin production, is of great importance for the treatment of PCM. The study of virulence factors favors the understanding of pathogen-host relationships, aiming to evaluate the possibility of developing new therapeutic targets and mechanisms that these molecules play in the infectious process, favoring the design of a more specific treatment for this disease.
Collapse
|
158
|
Magalhães AP, Jorge P, Pereira MO. Pseudomonas aeruginosa and Staphylococcus aureus communication in biofilm infections: insights through network and database construction. Crit Rev Microbiol 2019; 45:712-728. [DOI: 10.1080/1040841x.2019.1700209] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Affiliation(s)
- Andreia Patrícia Magalhães
- CEB - Centre of Biological Engineering, LIBRO - Laboratory of Research in Biofilms Rosário Oliveira, University of Minho, Braga, Portugal
| | - Paula Jorge
- CEB - Centre of Biological Engineering, LIBRO - Laboratory of Research in Biofilms Rosário Oliveira, University of Minho, Braga, Portugal
| | - Maria Olívia Pereira
- CEB - Centre of Biological Engineering, LIBRO - Laboratory of Research in Biofilms Rosário Oliveira, University of Minho, Braga, Portugal
| |
Collapse
|
159
|
Streptomycin mediated biofilm inhibition and suppression of virulence properties in Pseudomonas aeruginosa PAO1. Appl Microbiol Biotechnol 2019; 104:799-816. [PMID: 31820066 DOI: 10.1007/s00253-019-10190-w] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2019] [Revised: 09/09/2019] [Accepted: 10/07/2019] [Indexed: 12/15/2022]
Abstract
Pseudomonas aeruginosa is known as an opportunistic pathogen whose one of the antibiotic resistance mechanisms includes biofilm formation and virulence factor production. The present study showed that the sub-minimum inhibitory concentration (sub-MIC) of streptomycin inhibited the formation of biofilm and eradicated the established mature biofilm. Streptomycin at sub-MIC was also capable of inhibiting biofilm formation on the urinary catheters. In addition, the sub-MIC of streptomycin attenuated the bacterial virulence properties as confirmed by both phenotypic and gene expression studies. The optimal conditions for streptomycin to perform anti-biofilm and anti-virulence activities were proposed as alkaline TSB media (pH 7.9) at 35 °C. However, sub-MIC of streptomycin also exhibited a comparative anti-biofilm efficacy in LB media at similar pH level and temperature. Furthermore, this condition also improved the biofilm inhibition and eradication properties of streptomycin, tobramycin and tetracycline towards the biofilm formed by a clinical isolate of P. aeruginosa. Findings from the present study provide an important insight for further studies on the mechanisms of biofilm inhibition and dispersion of pre-existing biofilm by streptomycin as well as tobramycin and tetracycline under a specific culture environment.
Collapse
|
160
|
Zender M, Witzgall F, Kiefer A, Kirsch B, Maurer CK, Kany AM, Xu N, Schmelz S, Börger C, Blankenfeldt W, Empting M. Flexible Fragment Growing Boosts Potency of Quorum-Sensing Inhibitors against Pseudomonas aeruginosa Virulence. ChemMedChem 2019; 15:188-194. [PMID: 31709767 PMCID: PMC7004148 DOI: 10.1002/cmdc.201900621] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2019] [Indexed: 12/24/2022]
Abstract
Hit-to-lead optimization is a critical phase in drug discovery. Herein, we report on the fragment-based discovery and optimization of 2-aminopyridine derivatives as a novel lead-like structure for the treatment of the dangerous opportunistic pathogen Pseudomonas aeruginosa. We pursue an innovative treatment strategy by interfering with the Pseudomonas quinolone signal (PQS) quorum sensing (QS) system leading to an abolishment of bacterial pathogenicity. Our compounds act on the PQS receptor (PqsR), a key transcription factor controlling the expression of various pathogenicity determinants. In this target-driven approach, we made use of biophysical screening via surface plasmon resonance (SPR) followed by isothermal titration calorimetry (ITC)-enabled enthalpic efficiency (EE) evaluation. Hit optimization then involved growth vector identification and exploitation. Astonishingly, the latter was successfully achieved by introducing flexible linkers rather than rigid motifs leading to a boost in activity on the target receptor and anti-virulence potency.
Collapse
Affiliation(s)
- Michael Zender
- Drug Design and Optimization (DDOP), Helmholtz-Institute for Pharmaceutical Research Saarland (HIPS)-Helmholtz Centre for Infection Research (HZI), Campus E8.1, 66123, Saarbrücken, Germany
| | - Florian Witzgall
- Structure and Function of Proteins (SFPR), Helmholtz Centre for Infection Research (HZI), Inhoffenstr. 7, 38124, Braunschweig, Germany
| | - Alexander Kiefer
- Drug Design and Optimization (DDOP), Helmholtz-Institute for Pharmaceutical Research Saarland (HIPS)-Helmholtz Centre for Infection Research (HZI), Campus E8.1, 66123, Saarbrücken, Germany
| | - Benjamin Kirsch
- Drug Design and Optimization (DDOP), Helmholtz-Institute for Pharmaceutical Research Saarland (HIPS)-Helmholtz Centre for Infection Research (HZI), Campus E8.1, 66123, Saarbrücken, Germany
| | - Christine K Maurer
- Drug Design and Optimization (DDOP), Helmholtz-Institute for Pharmaceutical Research Saarland (HIPS)-Helmholtz Centre for Infection Research (HZI), Campus E8.1, 66123, Saarbrücken, Germany
| | - Andreas M Kany
- Drug Design and Optimization (DDOP), Helmholtz-Institute for Pharmaceutical Research Saarland (HIPS)-Helmholtz Centre for Infection Research (HZI), Campus E8.1, 66123, Saarbrücken, Germany
| | - Ningna Xu
- Lehrstuhl für Biochemie, Universität Bayreuth, Universitätsstr. 30, 95447, Bayreuth, Germany
| | - Stefan Schmelz
- Structure and Function of Proteins (SFPR), Helmholtz Centre for Infection Research (HZI), Inhoffenstr. 7, 38124, Braunschweig, Germany
| | - Carsten Börger
- PharmBioTec GmbH, Science Park 1, 66123, Saarbrücken, Germany
| | - Wulf Blankenfeldt
- Structure and Function of Proteins (SFPR), Helmholtz Centre for Infection Research (HZI), Inhoffenstr. 7, 38124, Braunschweig, Germany.,Biotechnology and Bioinformatics, Institute for Biochemistry, Technische Universität Braunschweig, Spielmannstr. 7, 38106, Braunschweig, Germany
| | - Martin Empting
- Drug Design and Optimization (DDOP), Helmholtz-Institute for Pharmaceutical Research Saarland (HIPS)-Helmholtz Centre for Infection Research (HZI), Campus E8.1, 66123, Saarbrücken, Germany.,Department of Pharmacy, Saarland University, Campus E8.1, 66123, Saarbrücken, Germany
| |
Collapse
|
161
|
Annunziato G. Strategies to Overcome Antimicrobial Resistance (AMR) Making Use of Non-Essential Target Inhibitors: A Review. Int J Mol Sci 2019; 20:E5844. [PMID: 31766441 PMCID: PMC6928725 DOI: 10.3390/ijms20235844] [Citation(s) in RCA: 122] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2019] [Revised: 11/19/2019] [Accepted: 11/20/2019] [Indexed: 12/23/2022] Open
Abstract
Antibiotics have always been considered as one of the most relevant discoveries of the twentieth century. Unfortunately, the dawn of the antibiotic era has sadly corresponded to the rise of the phenomenon of antimicrobial resistance (AMR), which is a natural process whereby microbes evolve in such a way to withstand the action of drugs. In this context, the identification of new potential antimicrobial targets and/or the identification of new chemical entities as antimicrobial drugs are in great demand. To date, among the many possible approaches used to deal with antibiotic resistance is the use of antibiotic adjuvants that hit bacterial non-essential targets. In this review, the author focuses on the discovery of antibiotic adjuvants and on new tools to study and reduce the prevalence of resistant bacterial infections.
Collapse
Affiliation(s)
- Giannamaria Annunziato
- Probes for Targets Group (P4T group), Department of food and Drug, University of Parma, 43124 Parma, Italy
| |
Collapse
|
162
|
Steering Phages to Combat Bacterial Pathogens. Trends Microbiol 2019; 28:85-94. [PMID: 31744662 DOI: 10.1016/j.tim.2019.10.007] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2019] [Revised: 10/10/2019] [Accepted: 10/17/2019] [Indexed: 12/21/2022]
|
163
|
Mellini M, Di Muzio E, D’Angelo F, Baldelli V, Ferrillo S, Visca P, Leoni L, Polticelli F, Rampioni G. In silico Selection and Experimental Validation of FDA-Approved Drugs as Anti-quorum Sensing Agents. Front Microbiol 2019; 10:2355. [PMID: 31649658 PMCID: PMC6796623 DOI: 10.3389/fmicb.2019.02355] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2019] [Accepted: 09/27/2019] [Indexed: 12/13/2022] Open
Abstract
The emergence of antibiotic resistant bacterial pathogens is increasing at an unprecedented pace, calling for the development of new therapeutic options. Small molecules interfering with virulence processes rather than growth hold promise as an alternative to conventional antibiotics. Anti-virulence agents are expected to decrease bacterial virulence and to pose reduced selective pressure for the emergence of resistance. In the opportunistic pathogen Pseudomonas aeruginosa the expression of key virulence traits is controlled by quorum sensing (QS), an intercellular communication process that coordinates gene expression at the population level. Hence, QS inhibitors represent promising anti-virulence agents against P. aeruginosa. Virtual screenings allow fast and cost-effective selection of target ligands among vast libraries of molecules, thus accelerating the time and limiting the cost of conventional drug-discovery processes, while the drug-repurposing approach is based on the identification of off-target activity of FDA-approved drugs, likely endowed with low cytotoxicity and favorable pharmacological properties. This study aims at combining the advantages of virtual screening and drug-repurposing approaches to identify new QS inhibitors targeting the pqs QS system of P. aeruginosa. An in silico library of 1,467 FDA-approved drugs has been screened by molecular docking, and 5 hits showing the highest predicted binding affinity for the pqs QS receptor PqsR (also known as MvfR) have been selected. In vitro experiments have been performed by engineering ad hoc biosensor strains, which were used to verify the ability of hit compounds to decrease PqsR activity in P. aeruginosa. Phenotypic analyses confirmed the impact of the most promising hit, the antipsychotic drug pimozide, on the expression of P. aeruginosa PqsR-controlled virulence traits. Overall, this study highlights the potential of virtual screening campaigns of FDA-approved drugs to rapidly select new inhibitors of important bacterial functions.
Collapse
Affiliation(s)
- Marta Mellini
- Department of Science, University Roma Tre, Rome, Italy
| | | | | | | | | | - Paolo Visca
- Department of Science, University Roma Tre, Rome, Italy
| | - Livia Leoni
- Department of Science, University Roma Tre, Rome, Italy
| | - Fabio Polticelli
- Department of Science, University Roma Tre, Rome, Italy
- National Institute of Nuclear Physics, Roma Tre Section, Rome, Italy
| | | |
Collapse
|
164
|
The race between drug introduction and appearance of microbial resistance. Current balance and alternative approaches. Curr Opin Pharmacol 2019; 48:48-56. [DOI: 10.1016/j.coph.2019.04.016] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2019] [Revised: 04/23/2019] [Accepted: 04/23/2019] [Indexed: 11/13/2022]
|
165
|
Smith P, Cozart J, Lynn BK, Alberts E, Frangipani E, Schuster M. Bacterial Cheaters Evade Punishment by Cyanide. iScience 2019; 19:101-109. [PMID: 31357168 PMCID: PMC6664145 DOI: 10.1016/j.isci.2019.07.015] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2019] [Revised: 06/24/2019] [Accepted: 07/11/2019] [Indexed: 12/20/2022] Open
Abstract
In all domains of life, mechanisms exist that protect cooperating groups from exploitation by cheaters. Recent observations with the bacterium Pseudomonas aeruginosa have suggested a paradigmatic cheater control mechanism in which cooperator cells punish or "police" cheater cells by cyanide poisoning. These cheater cells are deficient in a pleiotropic quorum-sensing regulator that controls the production of cooperative secretions including cyanide, and presumably also cyanide resistance. In this study, we directly tested and refuted the cyanide policing model. Contrary to the hypothesis, cheater fitness was unaffected by the presence of cyanide. Cheater mutants grew equally well in co-cultures with either cyanide-proficient or cyanide-deficient cooperators, and they were as resistant to exogenous cyanide as wild-type cells. We show that these behaviors are the result of quorum-sensing-independent and cyanide-responsive resistance gene regulation. Our results highlight the role of genetic architecture in the evolution of cooperative behavior.
Collapse
Affiliation(s)
- Parker Smith
- Department of Microbiology, Oregon State University, Corvallis, OR 97331, USA
| | - Jamison Cozart
- Department of Microbiology, Oregon State University, Corvallis, OR 97331, USA
| | - Bryan K Lynn
- Department of Integrative Biology, Oregon State University, Corvallis, OR 97331, USA
| | - Erin Alberts
- Department of Microbiology, Oregon State University, Corvallis, OR 97331, USA
| | - Emanuela Frangipani
- Department of Biomolecular Sciences, University of Urbino "Carlo Bo", 61029 Urbino (PU), Italy
| | - Martin Schuster
- Department of Microbiology, Oregon State University, Corvallis, OR 97331, USA.
| |
Collapse
|
166
|
Psonis JJ, Chahales P, Henderson NS, Rigel NW, Hoffman PS, Thanassi DG. The small molecule nitazoxanide selectively disrupts BAM-mediated folding of the outer membrane usher protein. J Biol Chem 2019; 294:14357-14369. [PMID: 31391254 PMCID: PMC6768635 DOI: 10.1074/jbc.ra119.009616] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Revised: 07/30/2019] [Indexed: 12/11/2022] Open
Abstract
Bacterial pathogens assemble adhesive surface structures termed pili or fimbriae to initiate and sustain infection of host tissues. Uropathogenic Escherichia coli, the primary causative agent of urinary tract infections, expresses type 1 and P pili required for colonization of the bladder and kidney, respectively. These pili are assembled by the conserved chaperone-usher (CU) pathway, in which a periplasmic chaperone works together with an outer membrane (OM) usher protein to build and secrete the pilus fiber. Previously, we found that the small molecule and antiparasitic drug nitazoxanide (NTZ) inhibits CU pathway-mediated pilus biogenesis in E. coli by specifically interfering with proper maturation of the usher protein in the OM. The usher is folded and inserted into the OM by the β-barrel assembly machine (BAM) complex, which in E. coli comprises five proteins, BamA-E. Here, we show that sensitivity of the usher to NTZ is modulated by BAM expression levels and requires the BamB and BamE lipoproteins. Furthermore, a genetic screen for NTZ-resistant bacterial mutants isolated a mutation in the essential BamD lipoprotein. These findings suggest that NTZ selectively interferes with an usher-specific arm of the BAM complex, revealing new details of the usher folding pathway and BAM complex function. Evaluation of a set of NTZ derivatives identified compounds with increased potency and disclosed that NTZ's nitrothiazole ring is critical for usher inhibition. In summary, our findings indicate highly specific effects of NTZ on the usher folding pathway and have uncovered NTZ analogs that specifically decrease usher levels in the OM.
Collapse
Affiliation(s)
- John J Psonis
- Department of Molecular Genetics and Microbiology, Stony Brook University, Stony Brook, New York 11794
- Center for Infectious Diseases, Stony Brook University, Stony Brook, New York 11794
| | - Peter Chahales
- Department of Molecular Genetics and Microbiology, Stony Brook University, Stony Brook, New York 11794
- Center for Infectious Diseases, Stony Brook University, Stony Brook, New York 11794
| | - Nadine S Henderson
- Department of Molecular Genetics and Microbiology, Stony Brook University, Stony Brook, New York 11794
- Center for Infectious Diseases, Stony Brook University, Stony Brook, New York 11794
| | - Nathan W Rigel
- Department of Biology, Hofstra University, Hempstead, New York 11549
| | - Paul S Hoffman
- Department of Medicine, Division of Infectious Diseases and International Health, University of Virginia, Charlottesville, Virginia 22908
| | - David G Thanassi
- Department of Molecular Genetics and Microbiology, Stony Brook University, Stony Brook, New York 11794
- Center for Infectious Diseases, Stony Brook University, Stony Brook, New York 11794
| |
Collapse
|
167
|
Yan J, Monaco H, Xavier JB. The Ultimate Guide to Bacterial Swarming: An Experimental Model to Study the Evolution of Cooperative Behavior. Annu Rev Microbiol 2019; 73:293-312. [PMID: 31180806 PMCID: PMC7428860 DOI: 10.1146/annurev-micro-020518-120033] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Cooperation has fascinated biologists since Darwin. How did cooperative behaviors evolve despite the fitness cost to the cooperator? Bacteria have cooperative behaviors that make excellent models to take on this age-old problem from both proximate (molecular) and ultimate (evolutionary) angles. We delve into Pseudomonas aeruginosa swarming, a phenomenon where billions of bacteria move cooperatively across distances of centimeters in a matter of a few hours. Experiments with swarming have unveiled a strategy called metabolic prudence that stabilizes cooperation, have showed the importance of spatial structure, and have revealed a regulatory network that integrates environmental stimuli and direct cooperative behavior, similar to a machine learning algorithm. The study of swarming elucidates more than proximate mechanisms: It exposes ultimate mechanisms valid to all scales, from cells in cancerous tumors to animals in large communities.
Collapse
Affiliation(s)
- Jinyuan Yan
- Program for Computational and Systems Biology, Memorial Sloan-Kettering Cancer Center, New York, NY 10065, USA;
| | - Hilary Monaco
- Program for Computational and Systems Biology, Memorial Sloan-Kettering Cancer Center, New York, NY 10065, USA;
| | - Joao B Xavier
- Program for Computational and Systems Biology, Memorial Sloan-Kettering Cancer Center, New York, NY 10065, USA;
| |
Collapse
|
168
|
Bhattacharya A, Toro Díaz VC, Morran LT, Bashey F. Evolution of increased virulence is associated with decreased spite in the insect-pathogenic bacterium Xenorhabdus nematophila. Biol Lett 2019; 15:20190432. [PMID: 31455168 DOI: 10.1098/rsbl.2019.0432] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Disease virulence may be strongly influenced by social interactions among pathogens, both during the time course of an infection and evolutionarily. Here, we examine how spiteful bacteriocin production in the insect-pathogenic bacterium Xenorhabdus nematophila is evolutionarily linked to its virulence. We expected a negative correlation between virulence and spite owing to their inverse correlations with growth. We examined bacteriocin production and growth across 14 experimentally evolved lineages that show faster host-killing relative to their ancestral population. Consistent with expectations, these more virulent lineages showed reduced bacteriocin production and faster growth relative to the ancestor. Further, bacteriocin production was negatively correlated with growth across the examined lineages. These results strongly support an evolutionary trade-off between virulence and bacteriocin production and lend credence to the view that disease management can be improved by exploiting pathogen social interactions.
Collapse
Affiliation(s)
- Amrita Bhattacharya
- Department of Biology, Indiana University Bloomington, 1001 E 3rd Street, Bloomington, IN, USA
| | - Valeria C Toro Díaz
- Department of Biology, Indiana University Bloomington, 1001 E 3rd Street, Bloomington, IN, USA
| | - Levi T Morran
- Department of Biology, Emory University, 1510 Clifton Road NE, Atlanta, GA, USA
| | - Farrah Bashey
- Department of Biology, Indiana University Bloomington, 1001 E 3rd Street, Bloomington, IN, USA
| |
Collapse
|
169
|
Rangel-Castañeda IA, Carranza-Rosales P, Guzmán-Delgado NE, Hernández-Hernández JM, González-Pozos S, Pérez-Rangel A, Castillo-Romero A. Curcumin Attenuates the Pathogenicity of Entamoeba histolytica by Regulating the Expression of Virulence Factors in an Ex-Vivo Model Infection. Pathogens 2019; 8:pathogens8030127. [PMID: 31443160 PMCID: PMC6789811 DOI: 10.3390/pathogens8030127] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2019] [Revised: 08/08/2019] [Accepted: 08/10/2019] [Indexed: 12/12/2022] Open
Abstract
Infection with the enteric protozoan Entamoeba histolytica is still a serious public health problem, especially in developing countries. Amoebic liver abscess (ALA) is the most common extraintestinal manifestation of the amoebiasis, and it can lead to serious and potentially life-threatening complications in some people. ALA can be cured by metronidazole (MTZ); however, because it has poor activity against luminal trophozoites, 40–60% of treated patients get repeated episodes of invasive disease and require repeated treatments that can induce resistance to MTZ, this may emerge as an important public health problem. Anti-virulence strategies that impair the virulence of pathogens are one of the novel approaches to solving the problem. In this study, we found that low doses of curcumin (10 and 50 μM) attenuate the virulence of E. histolytica without affecting trophozoites growth or triggering liver injury. Curcumin (CUR) decreases the expression of genes associated with E. histolytica virulence (gal/galnac lectin, ehcp1, ehcp5, and amoebapore), and is correlated with significantly lower amoebic invasion. In addition, oxidative stress is critically involved in the etiopathology of amoebic liver abscess; our results show no changes in mRNA expression levels of superoxide dismutase (SOD) and catalase (CAT) after E. histolytica infection, with or without CUR. This study provides clear evidence that curcumin could be an anti-virulence agent against E. histolytica, and makes it an attractive potential starting point for effective treatments that reduce downstream amoebic liver abscess.
Collapse
Affiliation(s)
- Itzia Azucena Rangel-Castañeda
- Departamento de Fisiología, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara 44340, Mexico
| | - Pilar Carranza-Rosales
- Centro de Investigación Biomédica del Noreste, Instituto Mexicano del Seguro Social, Monterrey 64720, Mexico
| | | | - José Manuel Hernández-Hernández
- Departamento de Biología Celular, Centro de Investigación y Estudios Avanzados del Instituto Politécnico Nacional, Ciudad de México 07360, Mexico
| | - Sirenia González-Pozos
- Unidad de Microscopía Electrónica LaNSE, Centro de Investigación y Estudios Avanzados del Instituto Politécnico Nacional, Ciudad de México 07360, Mexico
| | - Armando Pérez-Rangel
- Departamento de Biología Celular, Centro de Investigación y Estudios Avanzados del Instituto Politécnico Nacional, Ciudad de México 07360, Mexico
| | - Araceli Castillo-Romero
- Departamento de Microbiología y Patología, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara 44340, Mexico.
| |
Collapse
|
170
|
Galdino ACM, Viganor L, de Castro AA, da Cunha EFF, Mello TP, Mattos LM, Pereira MD, Hunt MC, O'Shaughnessy M, Howe O, Devereux M, McCann M, Ramalho TC, Branquinha MH, Santos ALS. Disarming Pseudomonas aeruginosa Virulence by the Inhibitory Action of 1,10-Phenanthroline-5,6-Dione-Based Compounds: Elastase B (LasB) as a Chemotherapeutic Target. Front Microbiol 2019; 10:1701. [PMID: 31428062 PMCID: PMC6688126 DOI: 10.3389/fmicb.2019.01701] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2019] [Accepted: 07/10/2019] [Indexed: 11/13/2022] Open
Abstract
Elastase B (lasB) is a multifunctional metalloenzyme secreted by the gram-negative pathogen Pseudomonas aeruginosa, and this enzyme orchestrates several physiopathological events during bacteria-host interplays. LasB is considered to be a potential target for the development of an innovative chemotherapeutic approach, especially against multidrug-resistant strains. Recently, our group showed that 1,10-phenanthroline-5,6-dione (phendione), [Ag(phendione)2]ClO4 (Ag-phendione) and [Cu(phendione)3](ClO4)2.4H2O (Cu-phendione) had anti-P. aeruginosa action against both planktonic- and biofilm-growing cells. In the present work, we have evaluated the effects of these compounds on the (i) interaction with the lasB active site using in silico approaches, (ii) lasB proteolytic activity by using a specific fluorogenic peptide substrate, (iii) lasB gene expression by real time-polymerase chain reaction, (iv) lasB protein secretion by immunoblotting, (v) ability to block the damages induced by lasB on a monolayer of lung epithelial cells, and (vi) survivability of Galleria mellonella larvae after being challenged with purified lasB and lasB-rich bacterial secretions. Molecular docking analyses revealed that phendione and its Ag+ and Cu2+ complexes were able to interact with the amino acids forming the active site of lasB, particularly Cu-phendione which exhibited the most favorable interaction energy parameters. Additionally, the test compounds were effective inhibitors of lasB activity, blocking the in vitro cleavage of the peptide substrate, aminobenzyl-Ala-Gly-Leu-Ala-p-nitrobenzylamide, with Cu-phendione having the best inhibitory action (K i = 90 nM). Treating living bacteria with a sub-inhibitory concentration (½ × MIC value) of the test compounds caused a significant reduction in the expression of the lasB gene as well as its mature protein production/secretion. Further, Ag-phendione and Cu-phendione offered protective action for lung epithelial cells, reducing the A549 monolayer damage by approximately 32 and 42%, respectively. Interestingly, Cu-phendione mitigated the toxic effect of both purified lasB molecules and lasB-containing bacterial secretions in the in vivo model, increasing the survival time of G. mellonella larvae. Collectively, these data reinforce the concept of lasB being a veritable therapeutic target and phendione-based compounds (mainly Cu-phendione) being prospective anti-virulence drugs against P. aeruginosa.
Collapse
Affiliation(s)
- Anna Clara M Galdino
- Department of General Microbiology, Institute of Microbiology Paulo de Góes, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil.,Postgraduate Program in Biochemistry, Institute of Chemistry, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Lívia Viganor
- Department of General Microbiology, Institute of Microbiology Paulo de Góes, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil.,The Centre for Biomimetic and Therapeutic Research, Focas Research Institute, Technological University Dublin, Dublin, Ireland
| | | | | | - Thaís P Mello
- Department of General Microbiology, Institute of Microbiology Paulo de Góes, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Larissa M Mattos
- Postgraduate Program in Biochemistry, Institute of Chemistry, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Marcos D Pereira
- Postgraduate Program in Biochemistry, Institute of Chemistry, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Mary C Hunt
- The Centre for Biomimetic and Therapeutic Research, Focas Research Institute, Technological University Dublin, Dublin, Ireland
| | - Megan O'Shaughnessy
- The Centre for Biomimetic and Therapeutic Research, Focas Research Institute, Technological University Dublin, Dublin, Ireland
| | - Orla Howe
- The Centre for Biomimetic and Therapeutic Research, Focas Research Institute, Technological University Dublin, Dublin, Ireland
| | - Michael Devereux
- The Centre for Biomimetic and Therapeutic Research, Focas Research Institute, Technological University Dublin, Dublin, Ireland
| | - Malachy McCann
- Department of Chemistry, Maynooth University, Maynooth, Ireland
| | | | - Marta H Branquinha
- Department of General Microbiology, Institute of Microbiology Paulo de Góes, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - André L S Santos
- Department of General Microbiology, Institute of Microbiology Paulo de Góes, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil.,Postgraduate Program in Biochemistry, Institute of Chemistry, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| |
Collapse
|
171
|
Feng C, Huang Y, He W, Cheng X, Liu H, Huang Y, Ma B, Zhang W, Liao C, Wu W, Shao Y, Xu D, Su Z, Lu W. Tanshinones: First-in-Class Inhibitors of the Biogenesis of the Type 3 Secretion System Needle of Pseudomonas aeruginosa for Antibiotic Therapy. ACS CENTRAL SCIENCE 2019; 5:1278-1288. [PMID: 31403076 PMCID: PMC6662154 DOI: 10.1021/acscentsci.9b00452] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/07/2019] [Indexed: 05/17/2023]
Abstract
The type 3 secretion system (T3SS) found as cell-surface appendages of many pathogenic Gram-negative bacteria, although nonessential for bacterial survival, is an important therapeutic target for drug discovery and development aimed at inhibiting bacterial virulence without inducing antibiotic resistance. We designed a fluorescence-polarization-based assay for high-throughput screening as a mechanistically well-defined general strategy for antibiotic discovery targeting the T3SS and made a serendipitous discovery of a subset of tanshinones-natural herbal compounds in traditional Chinese medicine widely used for the treatment of cardiovascular and cerebrovascular diseases-as effective inhibitors of the biogenesis of the T3SS needle of multi-drug-resistant Pseudomonas aeruginosa. By inhibiting the T3SS needle assembly and, thus, cytotoxicity and pathogenicity, selected tanshinones reduced the secretion of bacterial virulence factors toxic to macrophages in vitro, and rescued experimental animals challenged with lethal doses of Pseudomonas aeruginosa in a murine model of acute pneumonia. As first-in-class inhibitors with a demonstrable safety profile in humans, tanshinones may be used directly to alleviate Pseudomonas-aeruginosa-associated pulmonary infections without inducing antibiotic resistance. Since the T3SS is highly conserved among Gram-negative bacteria, this antivirulence strategy may be applicable to the discovery and development of novel classes of antibiotics refractory to existing resistance mechanisms for the treatment of many bacterial infections.
Collapse
Affiliation(s)
- Chao Feng
- Center
for Translational Medicine, Frontier Institute of Science
and Technology, Shaanxi Institute of Pediatric Diseases, Affiliated Children’s
Hospital, and Key Laboratory of Biomedical Information Engineering of the Ministry
of Education, School of Life Science and Technology, Xi’an Jiaotong University, Xi’an 710054, China
| | - Yinong Huang
- Center
for Translational Medicine, Frontier Institute of Science
and Technology, Shaanxi Institute of Pediatric Diseases, Affiliated Children’s
Hospital, and Key Laboratory of Biomedical Information Engineering of the Ministry
of Education, School of Life Science and Technology, Xi’an Jiaotong University, Xi’an 710054, China
| | - Wangxiao He
- Center
for Translational Medicine, Frontier Institute of Science
and Technology, Shaanxi Institute of Pediatric Diseases, Affiliated Children’s
Hospital, and Key Laboratory of Biomedical Information Engineering of the Ministry
of Education, School of Life Science and Technology, Xi’an Jiaotong University, Xi’an 710054, China
| | - Xiyao Cheng
- Department
of Biological and Food Engineering, Hubei
University of Technology, Wuhan 430068, China
| | - Huili Liu
- State
Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, Wuhan Institute of Physics and Mathematics, Chinese
Academy of Sciences, Wuhan 430071, China
| | - Yongqi Huang
- Department
of Biological and Food Engineering, Hubei
University of Technology, Wuhan 430068, China
| | - Bohan Ma
- Center
for Translational Medicine, Frontier Institute of Science
and Technology, Shaanxi Institute of Pediatric Diseases, Affiliated Children’s
Hospital, and Key Laboratory of Biomedical Information Engineering of the Ministry
of Education, School of Life Science and Technology, Xi’an Jiaotong University, Xi’an 710054, China
| | - Wei Zhang
- Center
for Translational Medicine, Frontier Institute of Science
and Technology, Shaanxi Institute of Pediatric Diseases, Affiliated Children’s
Hospital, and Key Laboratory of Biomedical Information Engineering of the Ministry
of Education, School of Life Science and Technology, Xi’an Jiaotong University, Xi’an 710054, China
| | - Chongbing Liao
- Center
for Translational Medicine, Frontier Institute of Science
and Technology, Shaanxi Institute of Pediatric Diseases, Affiliated Children’s
Hospital, and Key Laboratory of Biomedical Information Engineering of the Ministry
of Education, School of Life Science and Technology, Xi’an Jiaotong University, Xi’an 710054, China
| | - Weihui Wu
- State Key
Laboratory of Medicinal Chemical Biology, Key Laboratory of Molecular
Microbiology and Technology of the Ministry of Education, Department
of Microbiology, College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Yongping Shao
- Center
for Translational Medicine, Frontier Institute of Science
and Technology, Shaanxi Institute of Pediatric Diseases, Affiliated Children’s
Hospital, and Key Laboratory of Biomedical Information Engineering of the Ministry
of Education, School of Life Science and Technology, Xi’an Jiaotong University, Xi’an 710054, China
| | - Dan Xu
- Center
for Translational Medicine, Frontier Institute of Science
and Technology, Shaanxi Institute of Pediatric Diseases, Affiliated Children’s
Hospital, and Key Laboratory of Biomedical Information Engineering of the Ministry
of Education, School of Life Science and Technology, Xi’an Jiaotong University, Xi’an 710054, China
| | - Zhengding Su
- Department
of Biological and Food Engineering, Hubei
University of Technology, Wuhan 430068, China
| | - Wuyuan Lu
- Institute
of Human Virology and Department of Biochemistry and Molecular Biology, University of Maryland School of Medicine, Baltimore, Maryland 21201, United States
| |
Collapse
|
172
|
Xue L, Chen YY, Yan Z, Lu W, Wan D, Zhu H. Staphyloxanthin: a potential target for antivirulence therapy. Infect Drug Resist 2019; 12:2151-2160. [PMID: 31410034 PMCID: PMC6647007 DOI: 10.2147/idr.s193649] [Citation(s) in RCA: 61] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2018] [Accepted: 02/13/2019] [Indexed: 12/24/2022] Open
Abstract
Staphylococcus aureus is an important and common Gram-positive bacteria which causes clinical infections and food-poisoning cases. Therapeutic schedules for treatment of S. aureus infections are facing a challenge because of the emergence of multidrug resistance strains. It is urgent to find new antiinfective drugs to control S. aureus infection. S. aureus strains are capable of producing the golden carotenoid pigment: staphyloxanthin, which acts as an important virulence factor and a potential target for antivirulence drug design. This review is aimed at presenting an updated overview of this golden carotenoid pigment of S. aureus from the biosynthesis of staphyloxanthin, its function, and the genes involved in pigment production to staphyloxanthin: a novel target for antivirulence therapy.
Collapse
Affiliation(s)
- Lijun Xue
- College of Pharmaceutical Sciences and Traditional Chinese Medicine, Southwest University, Chongqing 400715, People's Republic of China.,College of Pharmaceutical Engineering, Chongqing Chemical Industry Vocational College, Chongqing, 400020, People's Republic of China
| | - Yang Yizhi Chen
- College of Pharmaceutical Sciences and Traditional Chinese Medicine, Southwest University, Chongqing 400715, People's Republic of China
| | - Zhiyun Yan
- Wuhan Wusteel Good Life Service Co. LTD, Wuhan, 430000, People's Republic of China
| | - Wei Lu
- College of Pharmaceutical Sciences and Traditional Chinese Medicine, Southwest University, Chongqing 400715, People's Republic of China
| | - Dong Wan
- Department of Emergency and Critical Care Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, People's Republic of China
| | - Huifeng Zhu
- College of Pharmaceutical Sciences and Traditional Chinese Medicine, Southwest University, Chongqing 400715, People's Republic of China
| |
Collapse
|
173
|
Abstract
The chaperone-usher (CU) pathway is a conserved secretion system dedicated to the assembly of a superfamily of virulence-associated surface structures by a wide range of Gram-negative bacteria. Pilus biogenesis by the CU pathway requires two specialized assembly components: a dedicated periplasmic chaperone and an integral outer membrane assembly and secretion platform termed the usher. The CU pathway assembles a variety of surface fibers, ranging from thin, flexible filaments to rigid, rod-like organelles. Pili typically act as adhesins and function as virulence factors that mediate contact with host cells and colonization of host tissues. Pilus-mediated adhesion is critical for early stages of infection, allowing bacteria to establish a foothold within the host. Pili are also involved in modulation of host cell signaling pathways, bacterial invasion into host cells, and biofilm formation. Pili are critical for initiating and sustaining infection and thus represent attractive targets for the development of antivirulence therapeutics. Such therapeutics offer a promising alternative to broad-spectrum antibiotics and provide a means to combat antibiotic resistance and treat infection while preserving the beneficial microbiota. A number of strategies have been taken to develop antipilus therapeutics, including vaccines against pilus proteins, competitive inhibitors of pilus-mediated adhesion, and small molecules that disrupt pilus biogenesis. Here we provide an overview of the function and assembly of CU pili and describe current efforts aimed at interfering with these critical virulence structures.
Collapse
|
174
|
Abstract
Antibiotic resistance is a major public health threat that has stimulated the scientific community to search for nontraditional therapeutic targets. Because virulence, but not the growth, of many Gram-negative bacterial pathogens depends on the multicomponent type three secretion system injectisome (T3SSi), the T3SSi has been an attractive target for identifying small molecules, peptides, and monoclonal antibodies that inhibit its function to render the pathogen avirulent. While many small-molecule lead compounds have been identified in whole-cell-based high-throughput screens (HTSs), only a few protein targets of these compounds are known; such knowledge is an important step to developing more potent and specific inhibitors. Evaluation of the efficacy of compounds in animal studies is ongoing. Some efforts involving the development of antibodies and vaccines that target the T3SSi are further along and include an antibody that is currently in phase II clinical trials. Continued research into these antivirulence therapies, used alone or in combination with traditional antibiotics, requires combined efforts from both pharmaceutical companies and academic labs.
Collapse
|
175
|
Shehaj L, Choudary SK, Makwana KM, Gallo MC, Murphy TF, Kritzer JA. Small-Molecule Inhibitors of Haemophilus influenzae IgA1 Protease. ACS Infect Dis 2019; 5:1129-1138. [PMID: 31016966 PMCID: PMC6625846 DOI: 10.1021/acsinfecdis.9b00004] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Newly identified, nontypable Haemophilus influenzae (H. influenza) strains represent a serious threat to global health. Due to the increasing prevalence of antibiotic resistance, virulence factors have emerged as potential therapeutic targets that would be less likely to promote resistance. IgA1 proteases are secreted virulence factors of many Gram-negative human pathogens. These enzymes play important roles in tissue invasion as well as evasion of the immune response, yet there has been limited work on pharmacological inhibitors. Here, we report the discovery of the first small molecule, nonpeptidic inhibitors of H. influenzae IgA1 proteases. We screened over 47 000 compounds in a biochemical assay using recombinant protease and identified a hit compound with micromolar potency. Preliminary structure-activity relationships produced additional inhibitors, two of which showed improved inhibition and selectivity for IgA protease over other serine proteases. We further showed dose-dependent inhibition against four different IgA1 protease variants collected from clinical isolates. These data support further development of IgA protease inhibitors as potential therapeutics for antibiotic-resistant H. influenza strains. The newly discovered inhibitors also represent valuable probes for exploring the roles of these proteases in bacterial colonization, invasion, and infection of mucosal tissues.
Collapse
Affiliation(s)
- Livia Shehaj
- Department of Chemistry, Tufts University, 62 Talbot Ave, Medford, Massachusetts 02155, United States
| | - Santosh K. Choudary
- Department of Chemistry, Tufts University, 62 Talbot Ave, Medford, Massachusetts 02155, United States
| | - Kamlesh M. Makwana
- Department of Chemistry, Tufts University, 62 Talbot Ave, Medford, Massachusetts 02155, United States
| | - Mary C. Gallo
- Department of Microbiology and Immunology, University at Buffalo, Jacobs School of Medicine and Biomedical Sciences, 3435 Main St., Buffalo, NY 14203, United States
- Clinical and Translational Research Center, 875 Ellicott St., University at Buffalo, Jacobs School of Medicine and Biomedical Sciences, Buffalo, NY 14203, United States
| | - Timothy F. Murphy
- Department of Microbiology and Immunology, University at Buffalo, Jacobs School of Medicine and Biomedical Sciences, 3435 Main St., Buffalo, NY 14203, United States
- Clinical and Translational Research Center, 875 Ellicott St., University at Buffalo, Jacobs School of Medicine and Biomedical Sciences, Buffalo, NY 14203, United States
- Division of Infectious Disease, Department of Medicine, 875 Ellicott St., University at Buffalo, Jacobs School of Medicine and Biomedical Sciences, Buffalo, NY 14203, United States
| | - Joshua A. Kritzer
- Department of Chemistry, Tufts University, 62 Talbot Ave, Medford, Massachusetts 02155, United States
| |
Collapse
|
176
|
Santos FF, Yamamoto D, Abe CM, Bryant JA, Hernandes RT, Kitamura FC, Castro FS, Valiatti TB, Piazza RMF, Elias WP, Henderson IR, Gomes TAT. The Type III Secretion System (T3SS)-Translocon of Atypical Enteropathogenic Escherichia coli (aEPEC) Can Mediate Adherence. Front Microbiol 2019; 10:1527. [PMID: 31338081 PMCID: PMC6629874 DOI: 10.3389/fmicb.2019.01527] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2018] [Accepted: 06/18/2019] [Indexed: 12/18/2022] Open
Abstract
The intimin protein is the major adhesin involved in the intimate adherence of atypical enteropathogenic Escherichia coli (aEPEC) strains to epithelial cells, but little is known about the structures involved in their early colonization process. A previous study demonstrated that the type III secretion system (T3SS) plays an additional role in the adherence of an Escherichia albertii strain. Therefore, we assumed that the T3SS could be related to the adherence efficiency of aEPEC during the first stages of contact with epithelial cells. To test this hypothesis, we examined the adherence of seven aEPEC strains and their eae (intimin) isogenic mutants in the standard HeLa adherence assay and observed that all wild-type strains were adherent while five isogenic eae mutants were not. The two eae mutant strains that remained adherent were then used to generate the eae/escN double mutants (encoding intimin and the T3SS ATPase, respectively) and after the adherence assay, we observed that one strain lost its adherence capacity. This suggested a role for the T3SS in the initial adherence steps of this strain. In addition, we demonstrated that this strain expressed the T3SS at significantly higher levels when compared to the other wild-type strains and that it produced longer translocon-filaments. Our findings reveal that the T3SS-translocon can play an additional role as an adhesin at the beginning of the colonization process of aEPEC.
Collapse
Affiliation(s)
- Fernanda F Santos
- Departamento de Microbiologia, Imunologia e Parasitologia, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Denise Yamamoto
- Departamento de Microbiologia, Imunologia e Parasitologia, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Cecilia M Abe
- Laboratório de Bacteriologia, Instituto Butantan, São Paulo, Brazil
| | - Jack A Bryant
- Institute of Microbiology and Infection, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Rodrigo T Hernandes
- Departamento de Microbiologia e Imunologia, Instituto de Biociências, Universidade Estadual Paulista, Botucatu, Brazil
| | - Felipe C Kitamura
- Departamento de Diagnóstico por Imagem, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Felipe S Castro
- Departamento de Microbiologia, Imunologia e Parasitologia, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Tiago B Valiatti
- Departamento de Microbiologia, Imunologia e Parasitologia, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, Brazil
| | | | - Waldir P Elias
- Laboratório de Bacteriologia, Instituto Butantan, São Paulo, Brazil
| | - Ian R Henderson
- Institute of Microbiology and Infection, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Tânia A T Gomes
- Departamento de Microbiologia, Imunologia e Parasitologia, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, Brazil
| |
Collapse
|
177
|
Ni S, Li B, Xu Y, Mao F, Li X, Lan L, Zhu J, Li J. Targeting virulence factors as an antimicrobial approach: Pigment inhibitors. Med Res Rev 2019; 40:293-338. [PMID: 31267561 DOI: 10.1002/med.21621] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2019] [Revised: 05/30/2019] [Accepted: 06/13/2019] [Indexed: 12/19/2022]
Abstract
The fascinating and dangerous colored pathogens contain unique chemically pigmented molecules, which give varied and efficient assistance as virulence factors to the crucial reproduction and growth of microbes. Therefore, multiple novel strategies and inhibitors have been developed in recent years that target virulence factor pigments. However, despite the importance and significance of this topic, it has not yet been comprehensively reviewed. Moreover, research groups around the world have made successful progress against antibacterial infections by targeting pigment production, including our serial works on the discovery of CrtN inhibitors against staphyloxanthin production in Staphylococcus aureus. On the basis of the previous achievements and recent progress of our group in this field, this article will be the first comprehensive review of pigment inhibitors against colored pathogens, especially S. aureus infections, and this article includes design strategies, representative case studies, advantages, limitations, and perspectives to guide future research.
Collapse
Affiliation(s)
- Shuaishuai Ni
- Cancer Institute, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Baoli Li
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai, China
| | - Yixiang Xu
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai, China
| | - Fei Mao
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai, China
| | - Xiaokang Li
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai, China
| | - Lefu Lan
- State Key Laboratory of Drug Research, Shanghai Institute of Material Medical, Chinese Academy of Sciences, Shanghai, China
| | - Jin Zhu
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai, China
| | - Jian Li
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai, China.,Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai, China
| |
Collapse
|
178
|
Roberts PA, Huebinger RM, Keen E, Krachler AM, Jabbari S. Mathematical model predicts anti-adhesion-antibiotic-debridement combination therapies can clear an antibiotic resistant infection. PLoS Comput Biol 2019; 15:e1007211. [PMID: 31335907 PMCID: PMC6677339 DOI: 10.1371/journal.pcbi.1007211] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2019] [Revised: 08/02/2019] [Accepted: 06/25/2019] [Indexed: 12/26/2022] Open
Abstract
As antimicrobial resistance increases, it is crucial to develop new treatment strategies to counter the emerging threat. In this paper, we consider combination therapies involving conventional antibiotics and debridement, coupled with a novel anti-adhesion therapy, and their use in the treatment of antimicrobial resistant burn wound infections. Our models predict that anti-adhesion-antibiotic-debridement combination therapies can eliminate a bacterial infection in cases where each treatment in isolation would fail. Antibiotics are assumed to have a bactericidal mode of action, killing bacteria, while debridement involves physically cleaning a wound (e.g. with a cloth); removing free bacteria. Anti-adhesion therapy can take a number of forms. Here we consider adhesion inhibitors consisting of polystyrene microbeads chemically coupled to a protein known as multivalent adhesion molecule 7, an adhesin which mediates the initial stages of attachment of many bacterial species to host cells. Adhesion inhibitors competitively inhibit bacteria from binding to host cells, thus rendering them susceptible to removal through debridement. An ordinary differential equation model is developed and the antibiotic-related parameters are fitted against new in vitro data gathered for the present study. The model is used to predict treatment outcomes and to suggest optimal treatment strategies. Our model predicts that anti-adhesion and antibiotic therapies will combine synergistically, producing a combined effect which is often greater than the sum of their individual effects, and that anti-adhesion-antibiotic-debridement combination therapy will be more effective than any of the treatment strategies used in isolation. Further, the use of inhibitors significantly reduces the minimum dose of antibiotics required to eliminate an infection, reducing the chances that bacteria will develop increased resistance. Lastly, we use our model to suggest treatment regimens capable of eliminating bacterial infections within clinically relevant timescales.
Collapse
Affiliation(s)
- Paul A. Roberts
- School of Mathematics, University of Birmingham, Edgbaston, Birmingham, United Kingdom
- Institute of Microbiology and Infection, School of Biosciences, University of Birmingham, Edgbaston, Birmingham, United Kingdom
| | - Ryan M. Huebinger
- Department of Surgery, University of Texas Southwestern Medical Center, Dallas, Texas, United States of America
| | - Emma Keen
- Institute of Microbiology and Infection, School of Biosciences, University of Birmingham, Edgbaston, Birmingham, United Kingdom
| | - Anne-Marie Krachler
- Department of Microbiology and Molecular Genetics, University of Texas McGovern Medical School at Houston, Houston, Texas, United States of America
| | - Sara Jabbari
- School of Mathematics, University of Birmingham, Edgbaston, Birmingham, United Kingdom
- Institute of Microbiology and Infection, School of Biosciences, University of Birmingham, Edgbaston, Birmingham, United Kingdom
| |
Collapse
|
179
|
Wilson JS, Churchill-Angus AM, Davies SP, Sedelnikova SE, Tzokov SB, Rafferty JB, Bullough PA, Bisson C, Baker PJ. Identification and structural analysis of the tripartite α-pore forming toxin of Aeromonas hydrophila. Nat Commun 2019; 10:2900. [PMID: 31263098 PMCID: PMC6602965 DOI: 10.1038/s41467-019-10777-x] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2018] [Accepted: 05/29/2019] [Indexed: 02/07/2023] Open
Abstract
The alpha helical CytolysinA family of pore forming toxins (α-PFT) contains single, two, and three component members. Structures of the single component Eschericia coli ClyA and the two component Yersinia enterolytica YaxAB show both undergo conformational changes from soluble to pore forms, and oligomerization to produce the active pore. Here we identify tripartite α-PFTs in pathogenic Gram negative bacteria, including Aeromonas hydrophila (AhlABC). We show that the AhlABC toxin requires all three components for maximal cell lysis. We present structures of pore components which describe a bi-fold hinge mechanism for soluble to pore transition in AhlB and a contrasting tetrameric assembly employed by soluble AhlC to hide their hydrophobic membrane associated residues. We propose a model of pore assembly where the AhlC tetramer dissociates, binds a single membrane leaflet, recruits AhlB promoting soluble to pore transition, prior to AhlA binding to form the active hydrophilic lined pore. Pore forming toxins (PFTs) form the major group of virulence factors in many pathogenic bacteria. Here the authors identify tripartite α-helical PFTs in pathogenic Gram negative bacteria and structurally characterize AhlABC from Aeromonas hydrophila and propose a model for its pore assembly.
Collapse
Affiliation(s)
- Jason S Wilson
- Department of Molecular Biology and Biotechnology, University of Sheffield, Firth Court, Western Bank, Sheffield, South Yorkshire, S10 2TN, UK
| | - Alicia M Churchill-Angus
- Department of Molecular Biology and Biotechnology, University of Sheffield, Firth Court, Western Bank, Sheffield, South Yorkshire, S10 2TN, UK
| | - Simon P Davies
- Department of Molecular Biology and Biotechnology, University of Sheffield, Firth Court, Western Bank, Sheffield, South Yorkshire, S10 2TN, UK.,School of Biomedical Sciences, Faculty of Biological Sciences and Astbury Centre for Structural and Molecular Biology, University of Leeds, Leeds, LS2 9JT, UK
| | - Svetlana E Sedelnikova
- Department of Molecular Biology and Biotechnology, University of Sheffield, Firth Court, Western Bank, Sheffield, South Yorkshire, S10 2TN, UK
| | - Svetomir B Tzokov
- Department of Molecular Biology and Biotechnology, University of Sheffield, Firth Court, Western Bank, Sheffield, South Yorkshire, S10 2TN, UK
| | - John B Rafferty
- Department of Molecular Biology and Biotechnology, University of Sheffield, Firth Court, Western Bank, Sheffield, South Yorkshire, S10 2TN, UK
| | - Per A Bullough
- Department of Molecular Biology and Biotechnology, University of Sheffield, Firth Court, Western Bank, Sheffield, South Yorkshire, S10 2TN, UK
| | - Claudine Bisson
- Department of Molecular Biology and Biotechnology, University of Sheffield, Firth Court, Western Bank, Sheffield, South Yorkshire, S10 2TN, UK.,ISMB, Department of Biological Sciences, Birkbeck, University of London, Malet Street, London, WC1E 7HX, UK
| | - Patrick J Baker
- Department of Molecular Biology and Biotechnology, University of Sheffield, Firth Court, Western Bank, Sheffield, South Yorkshire, S10 2TN, UK.
| |
Collapse
|
180
|
|
181
|
Steenhuis M, Abdallah AM, de Munnik SM, Kuhne S, Sterk G, van den Berg van Saparoea B, Westerhausen S, Wagner S, van der Wel NN, Wijtmans M, van Ulsen P, Jong WSP, Luirink J. Inhibition of autotransporter biogenesis by small molecules. Mol Microbiol 2019; 112:81-98. [PMID: 30983025 PMCID: PMC6850105 DOI: 10.1111/mmi.14255] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/28/2019] [Indexed: 12/16/2022]
Abstract
Disarming pathogens by targeting virulence factors is a promising alternative to classic antibiotics. Many virulence factors in Gram-negative bacteria are secreted via the autotransporter (AT) pathway, also known as Type 5 secretion. These factors are secreted with the assistance of two membrane-based protein complexes: Sec and Bam. To identify inhibitors of the AT pathway, we used transcriptomics analysis to develop a fluorescence-based high-throughput assay that reports on the stress induced by the model AT hemoglobin protease (Hbp) when its secretion across the outer membrane is inhibited. Screening a library of 1600 fragments yielded the compound VUF15259 that provokes cell envelope stress and secretion inhibition of the ATs Hbp and Antigen-43. VUF15259 also impairs β-barrel folding activity of various outer membrane proteins. Furthermore, we found that mutants that are compromised in outer membrane protein biogenesis are more susceptible to VUF15259. Finally, VUF15259 induces the release of vesicles that appear to assemble in short chains. Taken together, VUF15259 is the first reported compound that inhibits AT secretion and our data are mostly consistent with VUF15259 interfering with the Bam-complex as potential mode of action. The validation of the presented assay incites its use to screen larger compound libraries with drug-like compounds.
Collapse
Affiliation(s)
- Maurice Steenhuis
- Department of Molecular Microbiology, Amsterdam Institute for Molecules Medicines and Systems (AIMMS)Vrije UniversiteitAmsterdamthe Netherlands
| | - Abdallah M. Abdallah
- Bioscience Core LaboratoryKing Abdullah University of Science and TechnologyThuwalJeddahKingdom of Saudi Arabia
| | - Sabrina M. de Munnik
- Department of Chemistry and Pharmaceutical SciencesAmsterdam Institute for Molecules Medicines and Systems (AIMMS), Vrije UniversiteitAmsterdamthe Netherlands
| | - Sebastiaan Kuhne
- Department of Chemistry and Pharmaceutical SciencesAmsterdam Institute for Molecules Medicines and Systems (AIMMS), Vrije UniversiteitAmsterdamthe Netherlands
| | - Geert‐Jan Sterk
- Department of Chemistry and Pharmaceutical SciencesAmsterdam Institute for Molecules Medicines and Systems (AIMMS), Vrije UniversiteitAmsterdamthe Netherlands
| | - Bart van den Berg van Saparoea
- Department of Molecular Microbiology, Amsterdam Institute for Molecules Medicines and Systems (AIMMS)Vrije UniversiteitAmsterdamthe Netherlands
| | - Sibel Westerhausen
- Interfaculty Institute of Microbiology and Infection Medicine (IMIT)University of TübingenTübingenGermany
| | - Samuel Wagner
- Interfaculty Institute of Microbiology and Infection Medicine (IMIT)University of TübingenTübingenGermany
- German Center for Infection Research (DZIF)TübingenGermany
| | - Nicole N. van der Wel
- Department of Medical Biology, Electron Microscopy Center Amsterdam, Academic Medical CenterUniversity of AmsterdamAmsterdamthe Netherlands
| | - Maikel Wijtmans
- Department of Chemistry and Pharmaceutical SciencesAmsterdam Institute for Molecules Medicines and Systems (AIMMS), Vrije UniversiteitAmsterdamthe Netherlands
| | - Peter van Ulsen
- Department of Molecular Microbiology, Amsterdam Institute for Molecules Medicines and Systems (AIMMS)Vrije UniversiteitAmsterdamthe Netherlands
| | - Wouter S. P. Jong
- Department of Molecular Microbiology, Amsterdam Institute for Molecules Medicines and Systems (AIMMS)Vrije UniversiteitAmsterdamthe Netherlands
| | - Joen Luirink
- Department of Molecular Microbiology, Amsterdam Institute for Molecules Medicines and Systems (AIMMS)Vrije UniversiteitAmsterdamthe Netherlands
| |
Collapse
|
182
|
Murugayah SA, Gerth ML. Engineering quorum quenching enzymes: progress and perspectives. Biochem Soc Trans 2019; 47:793-800. [PMID: 31064863 PMCID: PMC6599154 DOI: 10.1042/bst20180165] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2019] [Revised: 04/10/2019] [Accepted: 04/15/2019] [Indexed: 12/12/2022]
Abstract
Quorum sensing is a key contributor to the virulence of many important plant, animal and human pathogens. The disruption of this signalling-a process referred to as 'quorum quenching'-is a promising new approach for controlling microbial pathogens. In this mini-review, we have focused on efforts to engineer enzymes that disrupt quorum sensing by inactivating acyl-homoserine lactone signalling molecules. We review different approaches for protein engineering and provide examples of how these engineering approaches have been used to tailor the stability, specificity and activities of quorum quenching enzymes. Finally, we grapple with some of the issues around these approaches-including the disconnect between in vitro biochemistry and potential in vivo applications.
Collapse
Affiliation(s)
- Shereen A Murugayah
- Department of Biochemistry, University of Otago, Dunedin, New Zealand
- Maurice Wilkins Centre for Molecular Biodiscovery, Auckland, New Zealand
| | - Monica L Gerth
- Maurice Wilkins Centre for Molecular Biodiscovery, Auckland, New Zealand
- School of Biological Sciences, Victoria University of Wellington, Wellington, New Zealand
| |
Collapse
|
183
|
Targeting Quorum Sensing: High-Throughput Screening to Identify Novel LsrK Inhibitors. Int J Mol Sci 2019; 20:ijms20123112. [PMID: 31242708 PMCID: PMC6627609 DOI: 10.3390/ijms20123112] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2019] [Revised: 06/11/2019] [Accepted: 06/19/2019] [Indexed: 12/22/2022] Open
Abstract
Since quorum sensing (QS) is linked to the establishment of bacterial infection, its inactivation represents one of the newest strategies to fight bacterial pathogens. LsrK is a kinase playing a key role in the processing of autoinducer-2 (AI-2), a quorum-sensing mediator in gut enteric bacteria. Inhibition of LsrK might thus impair the quorum-sensing cascade and consequently reduce bacterial pathogenicity. Aiming for the development of a target-based assay for the discovery of LsrK inhibitors, we evaluated different assay set-ups based on ATP detection and optimized an automation-compatible method for the high-throughput screening of chemical libraries. The assay was then used to perform the screening of a 2000-compound library, which provided 12 active compounds with an IC50 ≤ 10 µM confirming the effectiveness and sensitivity of our assay. Follow-up studies on the positive hits led to the identification of two compounds, harpagoside and rosolic acid, active in a cell-based AI-2 QS interference assay, which are at the moment the most promising candidates for the development of a new class of antivirulence agents based on LsrK inhibition.
Collapse
|
184
|
Yang T, Liu T, Gan J, Yu K, Chen K, Xue W, Lan L, Yang S, Yang CG. Structural Insight into the Mechanism of Staphylococcus aureus Stp1 Phosphatase. ACS Infect Dis 2019; 5:841-850. [PMID: 30868877 DOI: 10.1021/acsinfecdis.8b00316] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Staphylococcus aureus Stp1, which belongs to the bacterial metal-dependent protein phosphatase (PPM) family, is a promising candidate for antivirulence targeting. How Stp1 recognizes the phosphorylated peptide remains unclear, however. In order to investigate the recognition mechanism of Stp1 in depth, we have determined a series of crystal structures of S. aureus Stp1 in different states and the structural complex of Stp1 bound with a phosphorylated peptide His12. Different phosphorylated peptides, including MgrA- and GraR-derived phosphopeptides, are substrates of Stp1, which supports the function of Stp1 as a selective Ser/Thr phosphatase. In addition, interestingly, the crystal structures of R161-Stp1 variants combined with the biochemical activity validations have uncovered that R161 residue plays a key role to control the conformation switches of the flap domain in order to facilitate substrate binding and the dephosphorylation process. Our findings provide crucial structural insight into the molecular mechanism of S. aureus Stp1 phosphatase and reveal the phosphorylated peptides for biochemistry study and inhibitor screening of Stp1.
Collapse
Affiliation(s)
- Teng Yang
- State Key Laboratory Breeding Base of Green Pesticide and Agricultural Bioengineering, Key Laboratory of Green Pesticide and Agricultural Bioengineering, Ministry of Education, Center for R&D of Fine Chemicals, Guizhou University, 2708 South Huaxi Road, Guiyang, Guizhou 550025, P. R. China
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zuchongzhi Road, Shanghai 201203, P. R. China
| | - Tingting Liu
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zuchongzhi Road, Shanghai 201203, P. R. China
- University of the Chinese Academy of Sciences, 19A Yuquan Road, Beijing 100049, P. R. China
| | - Jianhua Gan
- School of Life Sciences, Fudan University, 2005 Songhu Road, Shanghai 200433, P. R. China
| | - Kunqian Yu
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zuchongzhi Road, Shanghai 201203, P. R. China
- University of the Chinese Academy of Sciences, 19A Yuquan Road, Beijing 100049, P. R. China
| | - Kaixian Chen
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zuchongzhi Road, Shanghai 201203, P. R. China
- University of the Chinese Academy of Sciences, 19A Yuquan Road, Beijing 100049, P. R. China
| | - Wei Xue
- State Key Laboratory Breeding Base of Green Pesticide and Agricultural Bioengineering, Key Laboratory of Green Pesticide and Agricultural Bioengineering, Ministry of Education, Center for R&D of Fine Chemicals, Guizhou University, 2708 South Huaxi Road, Guiyang, Guizhou 550025, P. R. China
| | - Lefu Lan
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zuchongzhi Road, Shanghai 201203, P. R. China
- University of the Chinese Academy of Sciences, 19A Yuquan Road, Beijing 100049, P. R. China
| | - Song Yang
- State Key Laboratory Breeding Base of Green Pesticide and Agricultural Bioengineering, Key Laboratory of Green Pesticide and Agricultural Bioengineering, Ministry of Education, Center for R&D of Fine Chemicals, Guizhou University, 2708 South Huaxi Road, Guiyang, Guizhou 550025, P. R. China
| | - Cai-Guang Yang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zuchongzhi Road, Shanghai 201203, P. R. China
- University of the Chinese Academy of Sciences, 19A Yuquan Road, Beijing 100049, P. R. China
| |
Collapse
|
185
|
Raafat MM, Ali-Tammam M, Ali AE. Quorum quenching activity of Bacillus cereus isolate 30b confers antipathogenic effects in Pseudomonas aeruginosa. Infect Drug Resist 2019; 12:1583-1596. [PMID: 31239733 PMCID: PMC6559722 DOI: 10.2147/idr.s182889] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2018] [Accepted: 04/11/2019] [Indexed: 01/21/2023] Open
Abstract
Background: Quorum quenching, the interference of a Quorum sensing (QS) system that contributes to the pathogenesis through triggering the production of various virulence determinants, is among the newly suggested antivirulence strategies. Purpose: This study aimed at screening of N-Acyl homoserine lactonase activity from local bacterial isolate, and investigating its effect on Pseudomonas aeruginosa (P. aeruginosa) virulence and biofilm formation. Materials and methods: Soil bacteria were screened for aiiA gene coding for lactonase enzyme by Polymerase Chain reaction and sequencing of aiiA gene homologs. Lactonase activity and spectrum were assessed in the cell-free lysate by well diffusion assay using Agrobacterium tumafaciens KYC55. A bacterial isolate showing the highest N-acyl-homoserine lactones degradation percentage was identified by gene amplification and sequencing of the 16S rRNA gene and its aiiA gene homolog. High performance liquid chromatography was used to confirm N-acyl-homoserine lactone degradation. The effect of cell-free lysate on the biofilm formation ability and cytotoxicity of P. aeruginosa PAO1 and P. aeruginosa clinical isolates from different clinical sources were assessed by static microtiter plate and viability assay, respectively Results: Lactonase gene and activity were identified in three Bacillus spp. isolates. They showed broad catalytic activities against tested N-acyl-homoserine lactones. However, The lactonase activity in the cell- free lysate of isolate 30b showed the highest significant degradation percentage on all tested signals; N-butanoyl-L-homoserine lactone (71%), N-hexanoyl-l-homoserine lactone (100%), N-decanoyl-homoserine lactone (100%), N-(3-oxohexanoyl)-L-homoserine lactone (37.5%), N-(oxodecanoyl)-L-homoserine lactone (100%), and N-(3-oxododecanoyl)-L-homoserine lactone (100%). Alignment of the amino acid sequences of AiiA protein of isolate 30b showed 96% identity with Bacillus cereus (B. cereus) homologous lactonases in the GenBank database, and the isolate was designated as B. cereus isolate 30b. Cell-free lysate of B. cereus isolate 30b reduced biofilm formation significantly in 93% of P. aeruginosa isolates. The highest mean percentage of reduction in the biofilm was 86%. Moreover, the viability percentage of human lung carcinoma A549 cells infected by P. aeruginosa and treated with cell-free lysate of B. cereus isolate 30b increased up to 15%. Conclusion: The results of this study highlight the potential of lactonases as a promising strategy to combat Pseudomonas aeruginosa virulence.
Collapse
Affiliation(s)
- Marwa M Raafat
- Department of Microbiology & Immunology, Faculty of Pharmaceutical Sciences & Pharmaceutical Industries, Future University in Egypt (FUE), New Cairo, Egypt
| | - Marwa Ali-Tammam
- Department of Microbiology & Immunology, Faculty of Pharmaceutical Sciences & Pharmaceutical Industries, Future University in Egypt (FUE), New Cairo, Egypt
| | - Amal E Ali
- Department of Microbiology & Immunology, Faculty of Pharmaceutical Sciences & Pharmaceutical Industries, Future University in Egypt (FUE), New Cairo, Egypt
| |
Collapse
|
186
|
|
187
|
Rezzoagli C, Granato ET, Kümmerli R. In-vivo microscopy reveals the impact of Pseudomonas aeruginosa social interactions on host colonization. ISME JOURNAL 2019; 13:2403-2414. [PMID: 31123320 DOI: 10.1038/s41396-019-0442-8] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/04/2019] [Revised: 03/20/2019] [Accepted: 04/24/2019] [Indexed: 12/21/2022]
Abstract
Pathogenic bacteria engage in social interactions to colonize hosts, which include quorum-sensing-mediated communication and the secretion of virulence factors that can be shared as "public goods" between individuals. While in-vitro studies demonstrated that cooperative individuals can be displaced by "cheating" mutants freeriding on social acts, we know less about social interactions in infections. Here, we developed a live imaging system to track virulence factor expression and social strain interactions in the human pathogen Pseudomonas aeruginosa colonizing the gut of Caenorhabditis elegans. We found that shareable siderophores and quorum-sensing systems are expressed during infections, affect host gut colonization, and benefit non-producers. However, non-producers were unable to successfully cheat and outcompete producers. Our results indicate that the limited success of cheats is due to a combination of the down-regulation of virulence factors over the course of the infection, the fact that each virulence factor examined contributed to but was not essential for host colonization, and the potential for negative frequency-dependent selection. Our findings shed new light on bacterial social interactions in infections and reveal potential limits of therapeutic approaches that aim to capitalize on social dynamics between strains for infection control.
Collapse
Affiliation(s)
- Chiara Rezzoagli
- Department of Plant and Microbial Biology, University of Zurich, Zurich, Switzerland. .,Department of Quantitative Biomedicine, University of Zurich, Zurich, Switzerland.
| | | | - Rolf Kümmerli
- Department of Plant and Microbial Biology, University of Zurich, Zurich, Switzerland. .,Department of Quantitative Biomedicine, University of Zurich, Zurich, Switzerland.
| |
Collapse
|
188
|
Wang J, Lin J, Zhang Y, Zhang J, Feng T, Li H, Wang X, Sun Q, Zhang X, Wang Y. Activity Improvement and Vital Amino Acid Identification on the Marine-Derived Quorum Quenching Enzyme MomL by Protein Engineering. Mar Drugs 2019; 17:md17050300. [PMID: 31117226 PMCID: PMC6562636 DOI: 10.3390/md17050300] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2019] [Revised: 05/17/2019] [Accepted: 05/17/2019] [Indexed: 12/25/2022] Open
Abstract
MomL is a marine-derived quorum-quenching (QQ) lactonase which can degrade various N-acyl homoserine lactones (AHLs). Intentional modification of MomL may lead to a highly efficient QQ enzyme with broad application potential. In this study, we used a rapid and efficient method combining error-prone polymerase chain reaction (epPCR), high-throughput screening and site-directed mutagenesis to identify highly active MomL mutants. In this way, we obtained two candidate mutants, MomLI144V and MomLV149A. These two mutants exhibited enhanced activities and blocked the production of pathogenic factors of Pectobacterium carotovorum subsp. carotovorum (Pcc). Besides, seven amino acids which are vital for MomL enzyme activity were identified. Substitutions of these amino acids (E238G/K205E/L254R) in MomL led to almost complete loss of its QQ activity. We then tested the effect of MomL and its mutants on Pcc-infected Chinese cabbage. The results indicated that MomL and its mutants (MomLL254R, MomLI144V, MomLV149A) significantly decreased the pathogenicity of Pcc. This study provides an efficient method for QQ enzyme modification and gives us new clues for further investigation on the catalytic mechanism of QQ lactonase.
Collapse
Affiliation(s)
- Jiayi Wang
- College of Marine Life Sciences, Ocean University of China, Qingdao 266003, China.
| | - Jing Lin
- College of Marine Life Sciences, Ocean University of China, Qingdao 266003, China.
| | - Yunhui Zhang
- College of Marine Life Sciences, Ocean University of China, Qingdao 266003, China.
| | - Jingjing Zhang
- College of Marine Life Sciences, Ocean University of China, Qingdao 266003, China.
| | - Tao Feng
- College of Marine Life Sciences, Ocean University of China, Qingdao 266003, China.
| | - Hui Li
- College of Marine Life Sciences, Ocean University of China, Qingdao 266003, China.
| | - Xianghong Wang
- College of Marine Life Sciences, Ocean University of China, Qingdao 266003, China.
| | - Qingyang Sun
- College of Marine Life Sciences, Ocean University of China, Qingdao 266003, China.
| | - Xiaohua Zhang
- College of Marine Life Sciences, Ocean University of China, Qingdao 266003, China.
- Laboratory for Marine Ecology and Environmental Science, Qingdao National Laboratory for Marine Science and Technology, Qingdao 266071, China.
- Institute of Evolution & Marine Biodiversity, Ocean University of China, Qingdao 266003, China.
| | - Yan Wang
- College of Marine Life Sciences, Ocean University of China, Qingdao 266003, China.
- Laboratory for Marine Ecology and Environmental Science, Qingdao National Laboratory for Marine Science and Technology, Qingdao 266071, China.
- Institute of Evolution & Marine Biodiversity, Ocean University of China, Qingdao 266003, China.
| |
Collapse
|
189
|
Resistance diagnostics as a public health tool to combat antibiotic resistance: A model-based evaluation. PLoS Biol 2019; 17:e3000250. [PMID: 31095567 PMCID: PMC6522007 DOI: 10.1371/journal.pbio.3000250] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2018] [Accepted: 04/12/2019] [Indexed: 01/12/2023] Open
Abstract
Rapid point-of-care resistance diagnostics (POC-RD) are a key tool in the fight against antibiotic resistance. By tailoring drug choice to infection genotype, doctors can improve treatment efficacy while limiting costs of inappropriate antibiotic prescription. Here, we combine epidemiological theory and data to assess the potential of resistance diagnostics (RD) innovations in a public health context, as a means to limit or even reverse selection for antibiotic resistance. POC-RD can be used to impose a nonbiological fitness cost on resistant strains by enabling diagnostic-informed treatment and targeted interventions that reduce resistant strains' opportunities for transmission. We assess this diagnostic-imposed fitness cost in the context of a spectrum of bacterial population biologies and find that POC-RD have a greater potential against obligate pathogens than opportunistic pathogens already subject to selection under "bystander" antibiotic exposure during asymptomatic carriage (e.g., the pneumococcus). We close by generalizing the notion of RD-informed strategies to incorporate carriage surveillance information and illustrate that coupling transmission-control interventions to the discovery of resistant strains in carriage can potentially select against resistance in a broad range of opportunistic pathogens.
Collapse
|
190
|
Doyle L, Ovchinnikova OG, Myler K, Mallette E, Huang BS, Lowary TL, Kimber MS, Whitfield C. Biosynthesis of a conserved glycolipid anchor for Gram-negative bacterial capsules. Nat Chem Biol 2019; 15:632-640. [DOI: 10.1038/s41589-019-0276-8] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2018] [Accepted: 03/25/2019] [Indexed: 11/09/2022]
|
191
|
He Y, Li R, Li H, Zhang S, Dai W, Wu Q, Jiang L, Zheng Z, Shen S, Chen X, Zhu Y, Wang J, Pang Z. Erythroliposomes: Integrated Hybrid Nanovesicles Composed of Erythrocyte Membranes and Artificial Lipid Membranes for Pore-Forming Toxin Clearance. ACS NANO 2019; 13:4148-4159. [PMID: 30855941 DOI: 10.1021/acsnano.8b08964] [Citation(s) in RCA: 65] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/20/2023]
Abstract
Pore-forming toxins (PFTs) are the most common bacterial virulence proteins and play a significant role in the pathogenesis of bacterial infections; thus, PFTs are an attractive therapeutic target in bacterial infections. Inspired by the pore-forming process and mechanism of PFTs, we designed an integrated hybrid nanovesicle-the erythroliposome (called the RM-PL)-for PFT detoxification by fusing natural red blood cell (RBC) membranes with artificial lipid membranes. The lipid and RBC membranes were mutually beneficial when integrated into a hybrid nanovesicle structure. The RBC membrane endowed RM-PLs with the capacity for detoxification, while the PEGylated lipid membrane stabilized the RM-PLs and greatly improved the detoxification capacity of the RBC membrane. With α-hemolysin (Hlα) as a model PFT, we demonstrated that RM-PLs could not only significantly reduce the toxicity of Hlα to erythrocytes in vitro but also effectively sponge Hlα in vivo and rescue mice from Hlα-induced damage. Moreover, the high detoxification capacity of RM-PLs was shown to be partly related to the expression of the Hlα receptor protein, a disintegrin and metalloproteinase domain-containing protein 10 on the RBC membrane. Consequently, as a component integrating natural and artificial materials, the erythroliposome nanoplatform inspires potential strategies for antivirulence therapy.
Collapse
Affiliation(s)
- Yuwei He
- Department of Pharmaceutics, School of Pharmacy , Fudan University and Key Laboratory of Smart Drug Delivery, Ministry of Education , Shanghai 201203 , China
| | - Ruixiang Li
- Innovation Research Institute of Traditional Chinese Medicine , Shanghai University of Traditional Chinese Medicine , Shanghai 201203 , China
| | - Haichun Li
- Department of Pharmaceutics, School of Pharmacy , Fudan University and Key Laboratory of Smart Drug Delivery, Ministry of Education , Shanghai 201203 , China
| | - Shuya Zhang
- Department of Pharmaceutics, School of Pharmacy , Fudan University and Key Laboratory of Smart Drug Delivery, Ministry of Education , Shanghai 201203 , China
| | - Wentao Dai
- Shanghai Center for Bioinformation Technology , Shanghai Industrial Technology Institute , Shanghai 201203 , China
| | - Qian Wu
- Shanghai Center for Bioinformation Technology , Shanghai Industrial Technology Institute , Shanghai 201203 , China
| | - Lixian Jiang
- Innovation Research Institute of Traditional Chinese Medicine , Shanghai University of Traditional Chinese Medicine , Shanghai 201203 , China
| | - Zicong Zheng
- Department of Pharmaceutics, School of Pharmacy , Fudan University and Key Laboratory of Smart Drug Delivery, Ministry of Education , Shanghai 201203 , China
| | - Shun Shen
- Department of Pharmaceutics, School of Pharmacy , Fudan University and Key Laboratory of Smart Drug Delivery, Ministry of Education , Shanghai 201203 , China
| | - Xing Chen
- Department of Pharmaceutics, School of Pharmacy , Fudan University and Key Laboratory of Smart Drug Delivery, Ministry of Education , Shanghai 201203 , China
| | - Yuefei Zhu
- Department of Pharmaceutics, School of Pharmacy , Fudan University and Key Laboratory of Smart Drug Delivery, Ministry of Education , Shanghai 201203 , China
| | - Jianxin Wang
- Department of Pharmaceutics, School of Pharmacy , Fudan University and Key Laboratory of Smart Drug Delivery, Ministry of Education , Shanghai 201203 , China
- Institute of Integrated Chinese and Western Medicine , Fudan University , Shanghai 200040 , China
| | - Zhiqing Pang
- Department of Pharmaceutics, School of Pharmacy , Fudan University and Key Laboratory of Smart Drug Delivery, Ministry of Education , Shanghai 201203 , China
| |
Collapse
|
192
|
Martinez MN, Watts JL, Gilbert JM. Questions associated with the development of novel drugs intended for the treatment of bacterial infections in veterinary species. Vet J 2019; 248:79-85. [PMID: 31113568 DOI: 10.1016/j.tvjl.2019.04.009] [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: 06/18/2018] [Revised: 04/17/2019] [Accepted: 04/18/2019] [Indexed: 11/17/2022]
Abstract
The emergence of multi-drug resistant bacteria has limited therapeutic options for the treatment of bacterial diseases in both human and veterinary medicine. This has resulted in an urgent need for novel agents to treat infectious diseases. Veterinary medicine is further constrained by the need to ensure that our emerging therapeutics have minimal or no impact on resistance in human pathogens. Thus, there has recently been increased attention given to the development of alternative treatments for infectious disease in animals. The domain of alternative therapies, which includes antimicrobial peptides, bacteriophages, probiotics, and immunomodulators, provides a means to directly inhibit the ability of a pathogen to damage the host while optimally, not imposing a selective pressure favouring antibiotic resistance. However, it is recognized that bacterial pathogens have the capability of expressing a variety of virulence factors, necessitating a clear understanding of the specific target for that therapeutic intervention. This manuscript explores the various virulence mechanisms, the potential utility of developing novel anti-virulence agents for counteracting the expression of diseases associated with veterinary species, and some of the unique regulatory hurdles to be addressed within the framework of a new animal drug application. We conclude with the public health concerns to be considered as these agents are integrated into the veterinary therapeutic arsenal. Our hope is that this manuscript will provide a platform to stimulate discussions on the critical questions that need to be addressed.
Collapse
Affiliation(s)
- Marilyn N Martinez
- US FDA Center for Veterinary Medicine, Rockville, MD 20855, United States.
| | - Jeffrey L Watts
- Zoetis, Inc., 333 Portage Street, Kalamazoo, MI 49007, United States
| | - Jeffrey M Gilbert
- US FDA Center for Veterinary Medicine, Rockville, MD 20855, United States
| |
Collapse
|
193
|
Zheng Y, Shang W, Peng H, Rao Y, Zhao X, Hu Z, Yang Y, Hu Q, Tan L, Xiong K, Li S, Zhu J, Hu X, Zhou R, Li M, Rao X. Virulence Determinants Are Required for Brain Abscess Formation Through Staphylococcus aureus Infection and Are Potential Targets of Antivirulence Factor Therapy. Front Microbiol 2019; 10:682. [PMID: 31024479 PMCID: PMC6460967 DOI: 10.3389/fmicb.2019.00682] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2019] [Accepted: 03/19/2019] [Indexed: 11/24/2022] Open
Abstract
Bacterial brain abscesses (BAs) are difficult to treat with conventional antibiotics. Thus, the development of alternative therapeutic strategies for BAs is of high priority. Identifying the virulence determinants that contribute to BA formation induced by Staphylococcus aureus would improve the effectiveness of interventions for this disease. In this study, RT-qPCR was performed to compare the expression levels of 42 putative virulence determinants of S. aureus strains Newman and XQ during murine BA formation, ear colonization, and bacteremia. The alterations in the expression levels of 23 genes were further confirmed through specific TaqMan RT-qPCR. Eleven S. aureus genes that persistently upregulated expression levels during BA infection were identified, and their functions in BA formation were confirmed through isogenic mutant experiments. Bacterial loads and BA volumes in mice infected with isdA, isdC, lgt, hla, or spa deletion mutants and the hla/spa double mutant strain were lower than those in mice infected with the wild-type Newman strain. The therapeutic application of monoclonal antibodies against Hla and SpA decreased bacterial loads and BA volume in mice infected with Newman. This study provides insights into the virulence determinants that contribute to staphylococcal BA formation and a paradigm for antivirulence factor therapy against S. aureus infections.
Collapse
Affiliation(s)
- Ying Zheng
- Department of Microbiology, College of Basic Medical Sciences, Army Medical University, Key Laboratory of Microbial Engineering under the Educational Committee in Chongqing, Chongqing, China
| | - Weilong Shang
- Department of Microbiology, College of Basic Medical Sciences, Army Medical University, Key Laboratory of Microbial Engineering under the Educational Committee in Chongqing, Chongqing, China
| | - Huagang Peng
- Department of Microbiology, College of Basic Medical Sciences, Army Medical University, Key Laboratory of Microbial Engineering under the Educational Committee in Chongqing, Chongqing, China
| | - Yifan Rao
- Institute of Modern Biopharmaceuticals, School of Life Sciences, Southwest University, Chongqing, China
| | - Xia Zhao
- Department of Microbiology, College of Basic Medical Sciences, Army Medical University, Key Laboratory of Microbial Engineering under the Educational Committee in Chongqing, Chongqing, China
| | - Zhen Hu
- Department of Microbiology, College of Basic Medical Sciences, Army Medical University, Key Laboratory of Microbial Engineering under the Educational Committee in Chongqing, Chongqing, China
| | - Yi Yang
- Department of Microbiology, College of Basic Medical Sciences, Army Medical University, Key Laboratory of Microbial Engineering under the Educational Committee in Chongqing, Chongqing, China
| | - Qiwen Hu
- Department of Microbiology, College of Basic Medical Sciences, Army Medical University, Key Laboratory of Microbial Engineering under the Educational Committee in Chongqing, Chongqing, China
| | - Li Tan
- Department of Microbiology, College of Basic Medical Sciences, Army Medical University, Key Laboratory of Microbial Engineering under the Educational Committee in Chongqing, Chongqing, China
| | - Kun Xiong
- Department of Microbiology, College of Basic Medical Sciences, Army Medical University, Key Laboratory of Microbial Engineering under the Educational Committee in Chongqing, Chongqing, China
| | - Shu Li
- Department of Microbiology, College of Basic Medical Sciences, Army Medical University, Key Laboratory of Microbial Engineering under the Educational Committee in Chongqing, Chongqing, China
| | - Junmin Zhu
- Department of Microbiology, College of Basic Medical Sciences, Army Medical University, Key Laboratory of Microbial Engineering under the Educational Committee in Chongqing, Chongqing, China
| | - Xiaomei Hu
- Department of Microbiology, College of Basic Medical Sciences, Army Medical University, Key Laboratory of Microbial Engineering under the Educational Committee in Chongqing, Chongqing, China
| | - Renjie Zhou
- Department of Emergency, Xinqiao Hospital, Army Medical University, Chongqing, China
| | - Ming Li
- Department of Microbiology, College of Basic Medical Sciences, Army Medical University, Key Laboratory of Microbial Engineering under the Educational Committee in Chongqing, Chongqing, China
| | - Xiancai Rao
- Department of Microbiology, College of Basic Medical Sciences, Army Medical University, Key Laboratory of Microbial Engineering under the Educational Committee in Chongqing, Chongqing, China
| |
Collapse
|
194
|
Huang Y, Xue C, He W, Zhao X. Inhibition effect of Zedoary turmeric oil on Listeria monocytogenes and Staphylococcus aureus growth and exotoxin proteins production. J Med Microbiol 2019; 68:657-666. [DOI: 10.1099/jmm.0.000949] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Affiliation(s)
- Yixuan Huang
- Department of Food Quality and Safety, College of Food Science and Engineering, Tonghua Normal University, 134000 Tonghua, PR China
- Department of Food Quality and Safety, College of Food Science and Engineering, Jilin University, 130062 Changchun, PR China
| | - Changsong Xue
- Department of Chinese Medicine, College of Medicine and Pharmacy, Tonghua Normal University, 134000 Tonghua, PR China
| | - Wenbing He
- Changbai Mountain Edible Plant Resources Research and Development Engineering Center, Tonghua Normal University, 134002 Tonghua, PR China
- Department of Food Quality and Safety, College of Food Science and Engineering, Tonghua Normal University, 134000 Tonghua, PR China
| | - Xingchen Zhao
- Department of Food Quality and Safety, College of Food Science and Engineering, Tonghua Normal University, 134000 Tonghua, PR China
- Changbai Mountain Edible Plant Resources Research and Development Engineering Center, Tonghua Normal University, 134002 Tonghua, PR China
| |
Collapse
|
195
|
Aygül A, Öztürk İ, Çilli FF, Ermertcan Ş. Quercetin inhibits swarming motility and activates biofilm production of Proteus mirabilis possibly by interacting with central regulators, metabolic status or active pump proteins. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2019; 57:65-71. [PMID: 30668324 DOI: 10.1016/j.phymed.2018.12.014] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/10/2018] [Revised: 12/04/2018] [Accepted: 12/10/2018] [Indexed: 06/09/2023]
Abstract
BACKGROUND Via its virulence factors such as swarm differentiation, biofilm and hemolysin production, urease enzyme, Proteus mirabilis causes urinary tract infections (UTIs), especially in complicated cases. Anti-pathogenic compounds attenuate the virulence of bacteria without showing 'cidal' activity and carry the potential to be used in the prevention and treatment of infectious diseases. PURPOSE Search for anti-pathogenic effects of quercetin, which is a widely known and biologically active phytochemical, on Proteus mirabilis was the purpose of this study. In this context, the potential inhibitory activity of quercetin on swarming motility and biofilm production of a wild-type strain, P. mirabilis HI4320, was investigated in both phenotypically and genotypically. METHODS Quercetin's effect on swarming motility was examined on LB agar plates, containing quercetin at various concentrations, by measuring the swarming diameter. The effect on biofilm formation, on the other hand, was analyzed by staining the formed biofilm of the bacterium, exposed to quercetin at various concentrations, with crystal violet and reading spectrophotometrically. Differences in expression levels of selected genes involved in swarming regulation were determined by real-time reverse transcriptase quantitative polymerase chain reaction (RT-qPCR) to evaluate the mechanism of inhibitory action on swarming. Further investigations were carried out repeating swarming assays with the clones that derived from the wild-type strain by a TA system kit for direct one-step cloning and overexpressing the relevant genes. RESULTS Our study revealed that quercetin inhibited swarming motility while activating biofilm production of P. mirabilis in direct proportion to the dose. Although all selected genes are inhibited in the same manner in liquid medium, and no significant differences could be detected in solid medium as demonstrated by RT-qPCR, experiments repeated with the clones overexpressing flhC (a component of flagellar transcriptional activator), speB (an agmatinase enzyme) and ompF (an outer membrane porin) genes showed that the respective clones could restore swarming, compensating for the inhibitory effect of quercetin. CONCLUSION Quercetin's inhibitory effect on P. mirabilis swarming was possibly due to interactions with components of swarming regulators, the genes expressing polyamine coding enzymes that trigger swarm differentiation, or active pump proteins.
Collapse
Affiliation(s)
- Abdurrahman Aygül
- Çukurova University, Faculty of Pharmacy, Department of Pharmaceutical Microbiology, Adana 01380, Turkey.
| | - İsmail Öztürk
- İzmir Katip Çelebi University, Faculty of Pharmacy, Department of Pharmaceutical Microbiology, İzmir 35040, Turkey
| | - Fatma Feriha Çilli
- Ege University, Faculty of Medicine, Department of Medical Microbiology, İzmir 35040, Turkey
| | - Şafak Ermertcan
- Ege University, Faculty of Pharmacy, Department of Pharmaceutical Microbiology, İzmir 35040, Turkey
| |
Collapse
|
196
|
Prospective Influence of Phytotherapy on Resistant Bacterial Tonsillitis. JOURNAL OF PURE AND APPLIED MICROBIOLOGY 2019. [DOI: 10.22207/jpam.13.1.17] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
|
197
|
Small-molecule inhibitor of HlyU attenuates virulence of Vibrio species. Sci Rep 2019; 9:4346. [PMID: 30867441 PMCID: PMC6416295 DOI: 10.1038/s41598-019-39554-y] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2018] [Accepted: 01/10/2019] [Indexed: 02/08/2023] Open
Abstract
Increasing antibiotic resistance has led to the development of new strategies to combat bacterial infection. Anti-virulence strategies that impair virulence of bacterial pathogens are one of the novel approaches with less selective pressure for developing resistance than traditional strategies that impede viability. In this study, a small molecule CM14 [N-(4-oxo-4H-thieno[3,4-c]chromen-3-yl)-3-phenylprop-2-ynamide] that inhibits the activity of HlyU, a transcriptional regulator essential for the virulence of the fulminating human pathogen Vibrio vulnificus, has been identified. Without affecting bacterial growth or triggering the host cell death, CM14 reduces HlyU-dependent expression of virulence genes in V. vulnificus. In addition to the decreased hemolysis of human erythrocytes, CM14 impedes host cell rounding and lysis caused by V. vulnificus. Notably, CM14 significantly enhances survival of mice infected with V. vulnificus by alleviating hepatic and renal dysfunction and systemic inflammation. Biochemical, mass spectrometric, and mutational analyses revealed that CM14 inhibits HlyU from binding to target DNA by covalently modifying Cys30. Remarkably, CM14 decreases the expression of various virulence genes of other Vibrio species and thus attenuates their virulence phenotypes. Together, this molecule could be an anti-virulence agent against HlyU-harboring Vibrio species with a low selective pressure for the emergence of resistance.
Collapse
|
198
|
Imperi F, Fiscarelli EV, Visaggio D, Leoni L, Visca P. Activity and Impact on Resistance Development of Two Antivirulence Fluoropyrimidine Drugs in Pseudomonas aeruginosa. Front Cell Infect Microbiol 2019; 9:49. [PMID: 30915278 PMCID: PMC6422106 DOI: 10.3389/fcimb.2019.00049] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2018] [Accepted: 02/15/2019] [Indexed: 12/21/2022] Open
Abstract
The rise in antibiotic resistance among bacterial pathogens has prompted the exploitation of alternative antibacterial strategies, such as antivirulence therapy. By inhibiting virulence traits, antivirulence drugs are expected to lessen pathogenicity without affecting bacterial growth, therefore avoiding the spread of resistance. However, some studies argued against this assumption, and the lack of antivirulence drugs in clinical use hampers the empirical assessment of this concept. Here we compared the mode of action and range of activity of two drugs which have been proposed for repurposing as quorum sensing and pyoverdine inhibitors in the human pathogen Pseudomonas aeruginosa: the anticancer drug 5-fluorouracil (5-FU) and the antimycotic drug 5-fluorocytosine (5-FC), respectively. The effect on bacterial growth, emergence and spread of resistance, and activity against clinical isolates were assessed. Our results confirm that 5-FU has growth inhibitory activity on reference strains and can rapidly select for spontaneous resistant mutants with loss-of-function mutations in the upp gene, responsible for uracil conversion into UMP. These mutants were also insensitive to the anti-pyoverdine effect of 5-FC. Conversely, 5-FC did not cause relevant growth inhibition, likely because of poor enzymatic conversion into 5-FU by cytosine deaminase. However, coculturing experiments showed that 5-FU resistant mutants can outcompete sensitive cells in mixed populations, in the presence of not only 5-FU but also 5-FC. Moreover, we observed that serial passages of wild-type cells in 5-FC-containing medium leads to the appearance and spread of 5-FC insensitive sub-populations of 5-FU resistant cells. The different effect on growth of 5-FU and 5-FC was overall conserved in a large collection of cystic fibrosis (CF) isolates, corresponding to different infection stages and antibiotic resistance profiles, although high variability was observed among strains. Notably, this analysis also revealed a significant number of pyoverdine-deficient isolates, whose proportion apparently increases over the course of the CF infection. This study demonstrates that the efficacy of an antivirulence drug with no apparent effect on growth can be significantly influenced by the emergence of insensitive mutants, and highlights the importance of the assessment of resistance-associated fitness cost and activity on clinical isolates for the development of "resistance-proof" antivirulence drugs.
Collapse
Affiliation(s)
- Francesco Imperi
- Department of Science, Roma Tre University, Rome, Italy
- Laboratory affiliated to Istituto Pasteur Italia–Fondazione Cenci Bolognetti, Department of Biology and Biotechnology Charles Darwin, Sapienza University of Rome, Rome, Italy
| | | | | | - Livia Leoni
- Department of Science, Roma Tre University, Rome, Italy
| | - Paolo Visca
- Department of Science, Roma Tre University, Rome, Italy
| |
Collapse
|
199
|
Tang F, Li L, Meng XM, Li B, Wang CQ, Wang SQ, Wang TL, Tian YM. Inhibition of alpha-hemolysin expression by resveratrol attenuates Staphylococcus aureus virulence. Microb Pathog 2019; 127:85-90. [DOI: 10.1016/j.micpath.2018.11.027] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2018] [Revised: 11/19/2018] [Accepted: 11/19/2018] [Indexed: 12/24/2022]
|
200
|
Inhibition of Protein Secretion in Escherichia coli and Sub-MIC Effects of Arylomycin Antibiotics. Antimicrob Agents Chemother 2019; 63:AAC.01253-18. [PMID: 30420476 DOI: 10.1128/aac.01253-18] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2018] [Accepted: 11/04/2018] [Indexed: 12/31/2022] Open
Abstract
At sufficient concentrations, antibiotics effectively eradicate many bacterial infections. However, during therapy, bacteria are unavoidably exposed to lower antibiotic concentrations, and sub-MIC exposure can result in a wide variety of other effects, including the induction of virulence, which can complicate therapy, or horizontal gene transfer (HGT), which can accelerate the spread of resistance genes. Bacterial type I signal peptidase (SPase) is an essential protein that acts at the final step of the general secretory pathway. This pathway is required for the secretion of many proteins, including many required for virulence, and the arylomycins are a class of natural product antibiotics that target SPase. Here, we investigated the consequences of exposing Escherichia coli cultures to sub-MIC levels of an arylomycin. Using multidimensional protein identification technology mass spectrometry, we found that arylomycin treatment inhibits the proper extracytoplasmic localization of many proteins, both those that appear to be SPase substrates and several that do not. The identified proteins are involved in a broad range of extracytoplasmic processes and include a number of virulence factors. The effects of arylomycin on several processes required for virulence were then individually examined, and we found that, at even sub-MIC levels, the arylomycins potently inhibit flagellation, motility, biofilm formation, and the dissemination of antibiotic resistance via HGT. Thus, we conclude that the arylomycins represent promising novel therapeutics with the potential to eradicate infections while simultaneously reducing virulence and the dissemination of resistance.
Collapse
|