1
|
Babar J, Ahmad S, Parveen B, Ali KG, Mushtaq A, Zahoor AF, Ahmad R, Mansha A, Irfan A. Exploring the Synthetic Potential of Horner-Wadsworth-Emmons Reaction Toward the Synthesis of Polyketide Based Natural Products: A Review. Top Curr Chem (Cham) 2025; 383:20. [PMID: 40286003 DOI: 10.1007/s41061-025-00504-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Accepted: 04/06/2025] [Indexed: 04/29/2025]
Abstract
The Horner-Wadsworth-Emmons (HWE) reaction is a commonly used and reliable phenomenon for carbon-carbon olefination in organic chemistry, carried out by treating aldehyde or ketones with phosphonate esters to afford the substituted alkenes. HWE reaction has also been observed to be involved in the stereo-controlled syntheses of naturally occurring compounds that acquire pharmaceutical profiles against various diseases. In this article, recent implementations of Horner-Wadsworth-Emmons reaction towards the notable total syntheses of naturally occurring compounds such as polyketides have been summarized.
Collapse
Affiliation(s)
- Javeria Babar
- Department of Chemistry, Government College University Faisalabad, Faisalabad, 38000, Pakistan
| | - Sajjad Ahmad
- Department of Chemistry, University of Engineering and Technology Lahore, Faisalabad Campus, Faisalabad, 38000, Pakistan
| | - Bushra Parveen
- Department of Chemistry, Government College University Faisalabad, Faisalabad, 38000, Pakistan
| | - Kulsoom Ghulam Ali
- Department of Chemistry, Government College University Faisalabad, Faisalabad, 38000, Pakistan
| | - Aqsa Mushtaq
- Department of Chemistry, Government College University Faisalabad, Faisalabad, 38000, Pakistan
| | - Ameer Fawad Zahoor
- Department of Chemistry, Government College University Faisalabad, Faisalabad, 38000, Pakistan.
| | - Raheel Ahmad
- Department of Chemistry, Government College University Faisalabad, Faisalabad, 38000, Pakistan
| | - Asim Mansha
- Department of Chemistry, Government College University Faisalabad, Faisalabad, 38000, Pakistan
| | - Ahmad Irfan
- Department of Chemistry, College of Science, King Khalid University, P.O. Box 9004, 61413, Abha, Saudi Arabia
| |
Collapse
|
2
|
McHugh RE, Rooney LM, Mark DR, Wale KR, Clapperton M, McConnell G, Hoskisson PA, Douce GR, Roe AJ. Evaluating the protective effects of Aurodox in a murine model of Shiga toxin-producing Escherichia coli. NPJ ANTIMICROBIALS AND RESISTANCE 2025; 3:23. [PMID: 40169888 PMCID: PMC11962119 DOI: 10.1038/s44259-025-00094-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/01/2024] [Accepted: 03/14/2025] [Indexed: 04/03/2025]
Abstract
Shiga Toxin-Producing E. coli (STEC) are a group of acute small intestine pathogens responsible for foodborne outbreaks of bloody diarrhoea. The expression of Shiga toxins (Stx) carried by STEC can initiate Haemolytic Uremic Syndrome (HUS), a major cause of acute renal failure in children. Here, we investigate the anti-virulence potential of Aurodox - a natural product of Streptomyces goldiniensis. Previously, we have shown that Aurodox downregulates the expression of the T3SS, inhibiting epithelial cell colonisation in vitro. Here, we use the Citrobacter rodentium DBS770 (Cr Stx2dact) model of STEC infection to demonstrate that Aurodox protects mice against Citrobacter rodentium-associated colonic hyperplasia and Stx-mediated renal injury. Given antibiotic-associated dysbiosis of the gut is associated with inflammation and the emergence of opportunistic pathogens, we examined the effect of Aurodox on the faecal bacteriome. We show that although the microbial community is altered following Aurodox treatment, changes are distinct from those associated with traditional antibiotic therapies.
Collapse
Affiliation(s)
- Rebecca E McHugh
- School of Infection and Immunity, University of Glasgow, Glasgow, UK
| | - Liam M Rooney
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, 161 Cathedral Street, Glasgow, UK
- School of Infection and Immunity, University of Glasgow, Glasgow, UK
| | - David R Mark
- School of Infection and Immunity, University of Glasgow, Glasgow, UK
| | - Kabo R Wale
- School of Infection and Immunity, University of Glasgow, Glasgow, UK
- Biological Sciences, University of Botswana, Gaborone, Botswana
| | - Megan Clapperton
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, 161 Cathedral Street, Glasgow, UK
| | - Gail McConnell
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, 161 Cathedral Street, Glasgow, UK
| | - Paul A Hoskisson
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, 161 Cathedral Street, Glasgow, UK
| | - Gillian R Douce
- School of Infection and Immunity, University of Glasgow, Glasgow, UK
| | - Andrew J Roe
- School of Infection and Immunity, University of Glasgow, Glasgow, UK.
| |
Collapse
|
3
|
Zhou L, Zhang Y, Wu S, Kuang Y, Jiang P, Zhu X, Yin K. Type III Secretion System in Intestinal Pathogens and Metabolic Diseases. J Diabetes Res 2024; 2024:4864639. [PMID: 39544522 PMCID: PMC11561183 DOI: 10.1155/2024/4864639] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Revised: 10/08/2024] [Accepted: 10/21/2024] [Indexed: 11/17/2024] Open
Abstract
Modern lifestyle changes, especially the consumption of a diet high in salt, sugar, and fat, have contributed to the increasing incidence and prevalence of chronic metabolic diseases such as diabetes, obesity, and gout. Changing lifestyles continuously shape the gut microbiota which is closely related to the occurrence and development of metabolic diseases due to its specificity of composition and structural diversity. A large number of pathogenic bacteria such as Yersinia, Salmonella, Shigella, and pathogenic E. coli in the gut utilize the type III secretion system (T3SS) to help them resist host defenses and cause disease. Although the T3SS is critical for the virulence of many important human pathogens, its relationship with metabolic diseases remains unknown. This article reviews the structure and function of the T3SS, the disruption of intestinal barrier integrity by the T3SS, the changes in intestinal flora containing the T3SS in metabolic diseases, the possible mechanisms of the T3SS affecting metabolic diseases, and the application of the T3SS in the treatment of metabolic diseases. The aim is to provide insights into metabolic diseases targeting the T3SS, thereby serving as a valuable reference for future research on disease diagnosis, prevention, and treatment.
Collapse
Affiliation(s)
- Le Zhou
- Guangxi Key Laboratory of Diabetic Systems Medicine, Guilin Medical University, Guilin 541100, China
| | - Yaoyuan Zhang
- Department of General Practice, The Fifth Affiliated Hospital of Southern Medical University, Guangzhou 510900, China
| | - Shiqi Wu
- Guangxi Key Laboratory of Diabetic Systems Medicine, Guilin Medical University, Guilin 541100, China
| | - Yiyu Kuang
- Guangxi Key Laboratory of Diabetic Systems Medicine, Guilin Medical University, Guilin 541100, China
| | - Pengfei Jiang
- Guangxi Key Laboratory of Diabetic Systems Medicine, Guilin Medical University, Guilin 541100, China
| | - Xiao Zhu
- Guangxi Key Laboratory of Diabetic Systems Medicine, Guilin Medical University, Guilin 541100, China
| | - Kai Yin
- Department of General Practice, The Fifth Affiliated Hospital of Southern Medical University, Guangzhou 510900, China
| |
Collapse
|
4
|
Mark DR, O’Boyle N, Wale KR, Tucker SK, McHugh RE, Roe AJ. Aurodox inhibits type III secretion in multiple Gram-negative pathogens. Open Biol 2024; 14:240240. [PMID: 39591989 PMCID: PMC11597412 DOI: 10.1098/rsob.240240] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2024] [Revised: 10/21/2024] [Accepted: 10/24/2024] [Indexed: 11/28/2024] Open
Abstract
Gram-negative pathogens pose a significant threat due to their propensity for causing various infections, often coupled with formidable resistance to conventional antibiotic treatments. The development of antivirulence (AV) compounds emerges as a promising alternative strategy by disrupting virulence mechanisms rather than targeting bacterial viability. Aurodox has exhibited promising AV properties in previous studies by blocking the expression and function of the LEE-encoded type 3 secretion system (T3SS) in enterohaemorrhagic Escherichia coli, an injectosome that translocates effector proteins directly into host target cells. However, aurodox's efficacy against the T3SS of other pathogens remained unanswered. Using quantitative real-time polymerase chain reaction, we show that aurodox exerts inhibitory effects on selected T3SS including those of Salmonella Typhimurium, Yersinia pseudotuberculosis and Vibrio parahaemolyticus. Imaging of RAW 264.7 cells infected with S. Typhimurium showed that aurodox protects against late stages of infection by blocking the expression of the SPI-2 T3SS. To elucidate a conserved mechanism of action, we compared transcriptomic datasets from both E. coli and S. Typhimurium treated with aurodox to identify orthologous genes differentially expressed in response to aurodox treatment across both pathogens. This study sheds light on potential mechanisms driving the action of this promising AV compound.
Collapse
Affiliation(s)
- David R. Mark
- School of Infection and Immunity, University of Glasgow, GlasgowG12 8TA, UK
| | - Nicky O’Boyle
- Department of Pathology, School of Microbiology & School of Medicine, University College Cork, National University of Ireland, CorkT12 K8AF, Ireland
- Department of Microbiology, School of Genetics & Microbiology, Moyne Institute of Preventive Medicine, Trinity College Dublin, DublinD02 A2H0, Ireland
| | - Kabo R. Wale
- School of Infection and Immunity, University of Glasgow, GlasgowG12 8TA, UK
- Biological Sciences, University of Botswana, Gaborone, Botswana
| | - Samantha K. Tucker
- School of Infection and Immunity, University of Glasgow, GlasgowG12 8TA, UK
| | - Rebecca E. McHugh
- School of Infection and Immunity, University of Glasgow, GlasgowG12 8TA, UK
| | - Andrew J. Roe
- School of Infection and Immunity, University of Glasgow, GlasgowG12 8TA, UK
| |
Collapse
|
5
|
Benyamini P. Beyond Antibiotics: What the Future Holds. Antibiotics (Basel) 2024; 13:919. [PMID: 39452186 PMCID: PMC11504868 DOI: 10.3390/antibiotics13100919] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Revised: 09/23/2024] [Accepted: 09/24/2024] [Indexed: 10/26/2024] Open
Abstract
The prevalence of multidrug resistance (MDR) and stagnant drug-development pipelines have led to the rapid rise of hard-to-treat antibiotic-resistant bacterial infections. These infectious diseases are no longer just nosocomial but are also becoming community-acquired. The spread of MDR has reached a crisis level that needs immediate attention. The landmark O'Neill report projects that by 2050, mortality rates associated with MDR bacterial infections will surpass mortality rates associated with individuals afflicted with cancer. Since conventional antimicrobials are no longer very reliable, it is of great importance to investigate different strategies to combat these life-threatening infectious diseases. Here, we provide an overview of recent advances in viable alternative treatment strategies mainly targeting a pathogen's virulence capability rather than viability. Topics include small molecule and immune inhibition of virulence factors, quorum sensing (QS) quenching, inhibition of biofilm development, bacteriophage-mediated therapy, and manipulation of an individual's macroflora to combat MDR bacterial infections.
Collapse
Affiliation(s)
- Payam Benyamini
- Department of Health Sciences at Extension, University of California Los Angeles, 1145 Gayley Ave., Los Angeles, CA 90024, USA
| |
Collapse
|
6
|
Pylkkö T, Schneider YKH, Rämä T, Andersen JH, Tammela P. Bioprospecting of inhibitors of EPEC virulence from metabolites of marine actinobacteria from the Arctic Sea. Front Microbiol 2024; 15:1432475. [PMID: 39282555 PMCID: PMC11392781 DOI: 10.3389/fmicb.2024.1432475] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Accepted: 06/28/2024] [Indexed: 09/19/2024] Open
Abstract
A considerable number of antibacterial agents are derived from bacterial metabolites. Similarly, numerous known compounds that impede bacterial virulence stem from bacterial metabolites. Enteropathogenic Escherichia coli (EPEC) is a notable human pathogen causing intestinal infections, particularly affecting infant mortality in developing regions. These infections are characterized by microvilli effacement and intestinal epithelial lesions linked with aberrant actin polymerization. This study aimed to identify potential antivirulence compounds for EPEC infections among bacterial metabolites harvested from marine actinobacteria (Kocuria sp. and Rhodococcus spp.) from the Arctic Sea by the application of virulence-based screening assays. Moreover, we demonstrate the suitability of these antivirulence assays to screen actinobacteria extract fractions for the bioassay-guided identification of metabolites. We discovered a compound in the fifth fraction of a Kocuria strain that interferes with EPEC-induced actin polymerization without affecting growth. Furthermore, a growth-inhibiting compound was identified in the fifth fraction of a Rhodococcus strain. Our findings include the bioassay-guided identification, HPLC-MS-based dereplication, and isolation of a large phospholipid and a likely antimicrobial peptide, demonstrating the usefulness of this approach in screening for compounds capable of inhibiting EPEC virulence.
Collapse
Affiliation(s)
- Tuomas Pylkkö
- Drug Research Program, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland
| | | | - Teppo Rämä
- Marbio, Faculty for Fisheries, Biosciences and Economy, UiT-The Arctic University of Norway, Tromsø, Norway
| | - Jeanette Hammer Andersen
- Marbio, Faculty for Fisheries, Biosciences and Economy, UiT-The Arctic University of Norway, Tromsø, Norway
| | - Päivi Tammela
- Drug Research Program, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland
| |
Collapse
|
7
|
Watanabe Y, Haneda T, Kimishima A, Kuwae A, Suga T, Suzuki T, Iwabuchi Y, Honsho M, Honma S, Iwatsuki M, Matsui H, Hanaki H, Kanoh N, Abe A, Asami Y, Ōmura S. PurA is the main target of aurodox, a type III secretion system inhibitor. Proc Natl Acad Sci U S A 2024; 121:e2322363121. [PMID: 38640341 PMCID: PMC11046696 DOI: 10.1073/pnas.2322363121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2023] [Accepted: 03/18/2024] [Indexed: 04/21/2024] Open
Abstract
Anti-microbial resistance (AMR) is one of the greatest threats to global health. The continual battle between the emergence of AMR and the development of drugs will be extremely difficult to stop as long as traditional anti-biotic approaches are taken. In order to overcome this impasse, we here focused on the type III secretion system (T3SS), which is highly conserved in many Gram-negative pathogenic bacteria. The T3SS is known to be indispensable in establishing disease processes but not essential for pathogen survival. Therefore, T3SS inhibitors may be innovative anti-infective agents that could dramatically reduce the evolutionary selective pressure on strains resistant to treatment. Based on this concept, we previously identified a polyketide natural product, aurodox (AD), as a specific T3SS inhibitor using our original screening system. However, despite its promise as a unique anti-infective drug of AD, the molecular target of AD has remained unclear. In this paper, using an innovative chemistry and genetic biology-based approach, we show that AD binds to adenylosuccinate synthase (PurA), which suppresses the production of the secreted proteins from T3SS, resulting in the expression of bacterial virulence both in vitro and in vivo experiments. Our findings illuminate the potential of PurA as a target of anti-infective drugs and vaccination and could open a avenue for application of PurA in the regulation of T3SS.
Collapse
Affiliation(s)
- Yoshihiro Watanabe
- Ōmura Satoshi Memorial Institute, Kitasato University, Minato-ku, Tokyo108-8641, Japan
- Graduate School of Infection Control Sciences, Kitasato University, Minato-ku, Tokyo108-8641, Japan
| | - Takeshi Haneda
- Laboratory of Microbiology, School of Pharmacy, Kitasato University, Minato-ku, Tokyo108-8641, Japan
| | - Aoi Kimishima
- Ōmura Satoshi Memorial Institute, Kitasato University, Minato-ku, Tokyo108-8641, Japan
- Graduate School of Infection Control Sciences, Kitasato University, Minato-ku, Tokyo108-8641, Japan
| | - Asaomi Kuwae
- Ōmura Satoshi Memorial Institute, Kitasato University, Minato-ku, Tokyo108-8641, Japan
- Graduate School of Infection Control Sciences, Kitasato University, Minato-ku, Tokyo108-8641, Japan
| | - Takuya Suga
- Ōmura Satoshi Memorial Institute, Kitasato University, Minato-ku, Tokyo108-8641, Japan
- Graduate School of Infection Control Sciences, Kitasato University, Minato-ku, Tokyo108-8641, Japan
| | - Takahiro Suzuki
- Department of Organic Chemistry, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai980-8578, Japan
| | - Yoshiharu Iwabuchi
- Department of Organic Chemistry, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai980-8578, Japan
| | - Masako Honsho
- Ōmura Satoshi Memorial Institute, Kitasato University, Minato-ku, Tokyo108-8641, Japan
- Graduate School of Infection Control Sciences, Kitasato University, Minato-ku, Tokyo108-8641, Japan
| | - Sota Honma
- Ōmura Satoshi Memorial Institute, Kitasato University, Minato-ku, Tokyo108-8641, Japan
- Graduate School of Infection Control Sciences, Kitasato University, Minato-ku, Tokyo108-8641, Japan
| | - Masato Iwatsuki
- Ōmura Satoshi Memorial Institute, Kitasato University, Minato-ku, Tokyo108-8641, Japan
- Graduate School of Infection Control Sciences, Kitasato University, Minato-ku, Tokyo108-8641, Japan
| | - Hidehito Matsui
- Ōmura Satoshi Memorial Institute, Kitasato University, Minato-ku, Tokyo108-8641, Japan
- Graduate School of Infection Control Sciences, Kitasato University, Minato-ku, Tokyo108-8641, Japan
| | - Hideaki Hanaki
- Ōmura Satoshi Memorial Institute, Kitasato University, Minato-ku, Tokyo108-8641, Japan
- Graduate School of Infection Control Sciences, Kitasato University, Minato-ku, Tokyo108-8641, Japan
| | - Naoki Kanoh
- Department of Organic Chemistry, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai980-8578, Japan
- School of Pharmacy and Pharmaceutical Sciences, and Institute of Medicinal Chemistry, Hoshi University, Shinagawa-ku, Tokyo142-8501, Japan
| | - Akio Abe
- Ōmura Satoshi Memorial Institute, Kitasato University, Minato-ku, Tokyo108-8641, Japan
- Graduate School of Infection Control Sciences, Kitasato University, Minato-ku, Tokyo108-8641, Japan
| | - Yukihiro Asami
- Ōmura Satoshi Memorial Institute, Kitasato University, Minato-ku, Tokyo108-8641, Japan
- Graduate School of Infection Control Sciences, Kitasato University, Minato-ku, Tokyo108-8641, Japan
| | - Satoshi Ōmura
- Ōmura Satoshi Memorial Institute, Kitasato University, Minato-ku, Tokyo108-8641, Japan
| |
Collapse
|
8
|
Hibbert T, Krpetic Z, Latimer J, Leighton H, McHugh R, Pottenger S, Wragg C, James CE. Antimicrobials: An update on new strategies to diversify treatment for bacterial infections. Adv Microb Physiol 2024; 84:135-241. [PMID: 38821632 DOI: 10.1016/bs.ampbs.2023.12.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/02/2024]
Abstract
Ninety-five years after Fleming's discovery of penicillin, a bounty of antibiotic compounds have been discovered, modified, or synthesised. Diversification of target sites, improved stability and altered activity spectra have enabled continued antibiotic efficacy, but overwhelming reliance and misuse has fuelled the global spread of antimicrobial resistance (AMR). An estimated 1.27 million deaths were attributable to antibiotic resistant bacteria in 2019, representing a major threat to modern medicine. Although antibiotics remain at the heart of strategies for treatment and control of bacterial diseases, the threat of AMR has reached catastrophic proportions urgently calling for fresh innovation. The last decade has been peppered with ground-breaking developments in genome sequencing, high throughput screening technologies and machine learning. These advances have opened new doors for bioprospecting for novel antimicrobials. They have also enabled more thorough exploration of complex and polymicrobial infections and interactions with the healthy microbiome. Using models of infection that more closely resemble the infection state in vivo, we are now beginning to measure the impacts of antimicrobial therapy on host/microbiota/pathogen interactions. However new approaches are needed for developing and standardising appropriate methods to measure efficacy of novel antimicrobial combinations in these contexts. A battery of promising new antimicrobials is now in various stages of development including co-administered inhibitors, phages, nanoparticles, immunotherapy, anti-biofilm and anti-virulence agents. These novel therapeutics need multidisciplinary collaboration and new ways of thinking to bring them into large scale clinical use.
Collapse
Affiliation(s)
- Tegan Hibbert
- Department of Clinical Infection, Microbiology and Immunology, Institute of Infection, Veterinary and Ecological Sciences (IVES), University of Liverpool, Liverpool, UK
| | - Zeljka Krpetic
- School of Science, Engineering, and Environment, University of Salford, Salford, UK
| | - Joe Latimer
- School of Science, Engineering, and Environment, University of Salford, Salford, UK
| | - Hollie Leighton
- Department of Clinical Infection, Microbiology and Immunology, Institute of Infection, Veterinary and Ecological Sciences (IVES), University of Liverpool, Liverpool, UK
| | - Rebecca McHugh
- School of Infection and Immunity, University of Glasgow, Glasgow, UK
| | - Sian Pottenger
- Department of Clinical Infection, Microbiology and Immunology, Institute of Infection, Veterinary and Ecological Sciences (IVES), University of Liverpool, Liverpool, UK
| | - Charlotte Wragg
- Department of Clinical Infection, Microbiology and Immunology, Institute of Infection, Veterinary and Ecological Sciences (IVES), University of Liverpool, Liverpool, UK
| | - Chloë E James
- School of Science, Engineering, and Environment, University of Salford, Salford, UK.
| |
Collapse
|
9
|
Wang Y, Jia X, Li Y, Ma S, Ma C, Xin D, Wang J, Chen Q, Liu C. NopAA and NopD Signaling Association-Related Gene GmNAC27 Promotes Nodulation in Soybean ( Glycine max). Int J Mol Sci 2023; 24:17498. [PMID: 38139327 PMCID: PMC10744329 DOI: 10.3390/ijms242417498] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2023] [Revised: 12/02/2023] [Accepted: 12/12/2023] [Indexed: 12/24/2023] Open
Abstract
Rhizobia secrete effectors that are essential for the effective establishment of their symbiotic interactions with leguminous host plants. However, the signaling pathways governing rhizobial type III effectors have yet to be sufficiently characterized. In the present study, the type III effectors, NopAA and NopD, which perhaps have signaling pathway crosstalk in the regulation of plant defense responses, have been studied together for the first time during nodulation. Initial qRT-PCR experiments were used to explore the impact of NopAA and NopD on marker genes associated with symbiosis and defense responses. The effects of these effectors on nodulation were then assessed by generating bacteria in which both NopAA and NopD were mutated. RNA-sequencing analyses of soybean roots were further utilized to assess signaling crosstalk between NopAA and NopD. NopAA mutant and NopD mutant were both found to repress GmPR1, GmPR2, and GmPR5 expression in these roots. The two mutants also significantly reduced nodules dry weight and the number of nodules and infection threads, although these changes were not significantly different from those observed following inoculation with double-mutant (HH103ΩNopAA&NopD). NopAA and NopD co-mutant inoculation was primarily found to impact the plant-pathogen interaction pathway. Common differentially expressed genes (DEGs) associated with both NopAA and NopD were enriched in the plant-pathogen interaction, plant hormone signal transduction, and MAPK signaling pathways, and no further changes in these common DEGs were noted in response to inoculation with HH103ΩNopAA&NopD. Glyma.13G279900 (GmNAC27) was ultimately identified as being significantly upregulated in the context of HH103ΩNopAA&NopD inoculation, serving as a positive regulator of nodulation. These results provide new insight into the synergistic impact that specific effectors can have on the establishment of symbiosis and the responses of host plant proteins.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Qingshan Chen
- Key Laboratory of Soybean Biology in Chinese Ministry of Education, National Key Laboratory of Smart Farm Technology and System, College of Agriculture, Northeast Agricultural University, Harbin 150030, China; (Y.W.); (X.J.); (Y.L.); (S.M.); (C.M.); (D.X.); (J.W.)
| | - Chunyan Liu
- Key Laboratory of Soybean Biology in Chinese Ministry of Education, National Key Laboratory of Smart Farm Technology and System, College of Agriculture, Northeast Agricultural University, Harbin 150030, China; (Y.W.); (X.J.); (Y.L.); (S.M.); (C.M.); (D.X.); (J.W.)
| |
Collapse
|
10
|
Tahara A, Tani K, Wakatsuki M, Tokiwa T, Higo M, Nonaka K, Hirose T, Hokari R, Ishiyama A, Iwatsuki M, Watanabe Y, Honsho M, Asami Y, Matsui H, Sunazuka T, Hanaki H, Teruya T, Ishii T. A novel aromatic compound from the fungus Synnemellisia sp. FKR-0921. J Antibiot (Tokyo) 2023; 76:706-710. [PMID: 37758818 DOI: 10.1038/s41429-023-00657-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2023] [Revised: 08/15/2023] [Accepted: 09/11/2023] [Indexed: 09/29/2023]
Abstract
The filamentous fungus Synnemellisia sp. strain FKR-0921 was obtained from soil collected on Kume Island, Okinawa. The MeOH extract of FKR-0921 cultured on a solid rice medium yielded a new aromatic compound, synnemellisitriol A (1). The structure, including the absolute configuration, was elucidated by spectroscopic analysis (FT-IR, NMR, and HR-ESI-MS), and the absolute configuration at C-9 of 1 was determined using the modified Mosher's method. Additionally, 1 was evaluated for its biological activities, including metallo-β-lactamase inhibitory activity, type III secretion system inhibitory activity, antimicrobial activity, antimalarial activity, and cytotoxicity.
Collapse
Affiliation(s)
- Arisu Tahara
- Department of Biosciences and Biotechnology, Faculty of Agriculture, University of the Ryukyus, 1 Senbaru, Nishihara, Okinawa, 903-0213, Japan
| | - Kazuki Tani
- Department of Biosciences and Biotechnology, Faculty of Agriculture, University of the Ryukyus, 1 Senbaru, Nishihara, Okinawa, 903-0213, Japan
| | - Miyu Wakatsuki
- Department of Biosciences and Biotechnology, Faculty of Agriculture, University of the Ryukyus, 1 Senbaru, Nishihara, Okinawa, 903-0213, Japan
| | - Toshiyuki Tokiwa
- Ōmura Satoshi Memorial Institute, Kitasato University, 5-9-1 Shirokane, Minato-ku, Tokyo, 108-8641, Japan
| | - Mayuka Higo
- Ōmura Satoshi Memorial Institute, Kitasato University, 5-9-1 Shirokane, Minato-ku, Tokyo, 108-8641, Japan
| | - Kenichi Nonaka
- Ōmura Satoshi Memorial Institute, Kitasato University, 5-9-1 Shirokane, Minato-ku, Tokyo, 108-8641, Japan
| | - Tomoyasu Hirose
- Ōmura Satoshi Memorial Institute, Kitasato University, 5-9-1 Shirokane, Minato-ku, Tokyo, 108-8641, Japan
| | - Rei Hokari
- Ōmura Satoshi Memorial Institute, Kitasato University, 5-9-1 Shirokane, Minato-ku, Tokyo, 108-8641, Japan
| | - Aki Ishiyama
- Ōmura Satoshi Memorial Institute, Kitasato University, 5-9-1 Shirokane, Minato-ku, Tokyo, 108-8641, Japan
| | - Masato Iwatsuki
- Ōmura Satoshi Memorial Institute, Kitasato University, 5-9-1 Shirokane, Minato-ku, Tokyo, 108-8641, Japan
| | - Yoshihiro Watanabe
- Ōmura Satoshi Memorial Institute, Kitasato University, 5-9-1 Shirokane, Minato-ku, Tokyo, 108-8641, Japan
| | - Masako Honsho
- Ōmura Satoshi Memorial Institute, Kitasato University, 5-9-1 Shirokane, Minato-ku, Tokyo, 108-8641, Japan
| | - Yukihiro Asami
- Ōmura Satoshi Memorial Institute, Kitasato University, 5-9-1 Shirokane, Minato-ku, Tokyo, 108-8641, Japan
| | - Hidehito Matsui
- Ōmura Satoshi Memorial Institute, Kitasato University, 5-9-1 Shirokane, Minato-ku, Tokyo, 108-8641, Japan
| | - Toshiaki Sunazuka
- Ōmura Satoshi Memorial Institute, Kitasato University, 5-9-1 Shirokane, Minato-ku, Tokyo, 108-8641, Japan
| | - Hideaki Hanaki
- Ōmura Satoshi Memorial Institute, Kitasato University, 5-9-1 Shirokane, Minato-ku, Tokyo, 108-8641, Japan
| | - Toshiaki Teruya
- Faculty of Education, University of the Ryukyus, 1 Senbaru, Nishihara, Okinawa, 903-0213, Japan
| | - Takahiro Ishii
- Department of Biosciences and Biotechnology, Faculty of Agriculture, University of the Ryukyus, 1 Senbaru, Nishihara, Okinawa, 903-0213, Japan.
| |
Collapse
|
11
|
Blasey N, Rehrmann D, Riebisch AK, Mühlen S. Targeting bacterial pathogenesis by inhibiting virulence-associated Type III and Type IV secretion systems. Front Cell Infect Microbiol 2023; 12:1065561. [PMID: 36704108 PMCID: PMC9872159 DOI: 10.3389/fcimb.2022.1065561] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2022] [Accepted: 12/19/2022] [Indexed: 01/12/2023] Open
Abstract
Infections caused by Gram-negative pathogens pose a major health burden. Both respiratory and gastrointestinal infections are commonly associated with these pathogens. With the increase in antimicrobial resistance (AMR) over the last decades, bacterial infections may soon become the threat they have been before the discovery of antibiotics. Many Gram-negative pathogens encode virulence-associated Type III and Type IV secretion systems, which they use to inject bacterial effector proteins across bacterial and host cell membranes into the host cell cytosol, where they subvert host cell functions in favor of bacterial replication and survival. These secretion systems are essential for the pathogens to cause disease, and secretion system mutants are commonly avirulent in infection models. Hence, these structures present attractive targets for anti-virulence therapies. Here, we review previously and recently identified inhibitors of virulence-associated bacterial secretions systems and discuss their potential as therapeutics.
Collapse
|
12
|
Synthesis of resveratrol derivatives and their anti-virulence activity as T3SS inhibitors of Salmonella. J Mol Struct 2022. [DOI: 10.1016/j.molstruc.2022.134826] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
|
13
|
Research Progress on Small Molecular Inhibitors of the Type 3 Secretion System. Molecules 2022; 27:molecules27238348. [PMID: 36500441 PMCID: PMC9740592 DOI: 10.3390/molecules27238348] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Revised: 11/22/2022] [Accepted: 11/23/2022] [Indexed: 12/05/2022] Open
Abstract
The overuse of antibiotics has led to severe bacterial drug resistance. Blocking pathogen virulence devices is a highly effective approach to combating bacterial resistance worldwide. Type three secretion systems (T3SSs) are significant virulence factors in Gram-negative pathogens. Inhibition of these systems can effectively weaken infection whilst having no significant effect on bacterial growth. Therefore, T3SS inhibitors may be a powerful weapon against resistance in Gram-negative bacteria, and there has been increasing interest in the research and development of T3SS inhibitors. This review outlines several reported small-molecule inhibitors of the T3SS, covering those of synthetic and natural origin, including their sources, structures, and mechanisms of action.
Collapse
|
14
|
McHugh RE, Munnoch JT, Braes RE, McKean IJW, Giard J, Taladriz-Sender A, Peschke F, Burley GA, Roe AJ, Hoskisson PA. Biosynthesis of Aurodox, a Type III Secretion System Inhibitor from Streptomyces goldiniensis. Appl Environ Microbiol 2022; 88:e0069222. [PMID: 35867559 PMCID: PMC9361827 DOI: 10.1128/aem.00692-22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Accepted: 06/26/2022] [Indexed: 11/20/2022] Open
Abstract
The global increase in antimicrobial-resistant infections means that there is a need to develop new antimicrobial molecules and strategies to combat the issue. Aurodox is a linear polyketide natural product that is produced by Streptomyces goldiniensis, yet little is known about aurodox biosynthesis or the nature of the biosynthetic gene cluster (BGC) that encodes its production. To gain a deeper understanding of aurodox biosynthesis by S. goldiniensis, the whole genome of the organism was sequenced, revealing the presence of an 87 kb hybrid polyketide synthase/non-ribosomal peptide synthetase (PKS/NRPS) BGC. The aurodox BGC shares significant homology with the kirromycin BGC from S. collinus Tϋ 365. However, the genetic organization of the BGC differs significantly. The candidate aurodox gene cluster was cloned and expressed in a heterologous host to demonstrate that it was responsible for aurodox biosynthesis and disruption of the primary PKS gene (aurAI) abolished aurodox production. These data supported a model whereby the initial core biosynthetic reactions involved in aurodox biosynthesis followed that of kirromycin. Cloning aurM* from S. goldiniensis and expressing this in the kirromycin producer S. collinus Tϋ 365 enabled methylation of the pyridone group, suggesting this is the last step in biosynthesis. This methylation step is also sufficient to confer the unique type III secretion system inhibitory properties to aurodox. IMPORTANCE Enterohemorrhagic Escherichia coli (EHEC) is a significant global pathogen for which traditional antibiotic treatment is not recommended. Aurodox inhibits the ability of EHEC to establish infection in the host gut through the specific targeting of the type III secretion system while circumventing the induction of toxin production associated with traditional antibiotics. These properties suggest aurodox could be a promising anti-virulence compound for EHEC, which merits further investigation. Here, we characterized the aurodox biosynthetic gene cluster from Streptomyces goldiniensis and established the key enzymatic steps of aurodox biosynthesis that give rise to the unique anti-virulence activity. These data provide the basis for future chemical and genetic approaches to produce aurodox derivatives with increased efficacy and the potential to engineer novel elfamycins.
Collapse
Affiliation(s)
- Rebecca E. McHugh
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, UK
- Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, UK
| | - John T. Munnoch
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, UK
| | - Robyn E. Braes
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, UK
| | - Iain J. W. McKean
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, UK
- Department of Pure and Applied Chemistry, University of Strathclyde, Glasgow, UK
| | - Josephine Giard
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, UK
| | | | - Frederik Peschke
- Department of Pure and Applied Chemistry, University of Strathclyde, Glasgow, UK
| | - Glenn A. Burley
- Department of Pure and Applied Chemistry, University of Strathclyde, Glasgow, UK
| | - Andrew J. Roe
- Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, UK
| | - Paul A. Hoskisson
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, UK
| |
Collapse
|
15
|
Kimishima A, Hagimoto D, Honsho M, Watanabe Y, Iwatsuki M, Tsutsumi H, Inahashi Y, Naher K, Sakai K, Kuwae A, Abe A, Asami Y. Insights into the structure-activity relationship of a type III secretion system inhibitor, aurodox. Bioorg Med Chem Lett 2022; 69:128779. [PMID: 35545199 DOI: 10.1016/j.bmcl.2022.128779] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Revised: 04/28/2022] [Accepted: 05/01/2022] [Indexed: 11/02/2022]
Abstract
Aurodox was originally isolated in 1972 as a linear polyketide compound exhibiting antibacterial activity against Gram-positive bacteria. We have since identified aurodox as a specific inhibitor of the bacterial type III secretion system (T3SS) using our original screening system for inhibition of T3SS-mediated hemolysis in enteropathogenic Escherichia coli (EPEC). In this research, we synthesized 15 derivatives of aurodox and evaluated EPEC T3SS inhibitory activity as well as antibacterial activity against EPEC. One of the derivatives was highly selective for T3SS inhibition, equivalent to that of aurodox, but without exhibiting antibacterial activity (69-fold selectivity). This work revealed the structure-activity relationship for the inhibition of T3SS by aurodox and suggests that the target of T3SS is distinct from the target for antibacterial activity.
Collapse
Affiliation(s)
- Aoi Kimishima
- Graduate School of Infection Control Sciences, Kitasato University, Tokyo, Japan; Ōmura Satoshi Memorial Institute, Kitasato University, Tokyo, Japan
| | - Daichi Hagimoto
- Graduate School of Infection Control Sciences, Kitasato University, Tokyo, Japan
| | - Masako Honsho
- Ōmura Satoshi Memorial Institute, Kitasato University, Tokyo, Japan
| | - Yoshihiro Watanabe
- Graduate School of Infection Control Sciences, Kitasato University, Tokyo, Japan; Ōmura Satoshi Memorial Institute, Kitasato University, Tokyo, Japan
| | - Masato Iwatsuki
- Graduate School of Infection Control Sciences, Kitasato University, Tokyo, Japan; Ōmura Satoshi Memorial Institute, Kitasato University, Tokyo, Japan
| | - Hayama Tsutsumi
- Graduate School of Infection Control Sciences, Kitasato University, Tokyo, Japan; Ōmura Satoshi Memorial Institute, Kitasato University, Tokyo, Japan
| | - Yuki Inahashi
- Graduate School of Infection Control Sciences, Kitasato University, Tokyo, Japan; Ōmura Satoshi Memorial Institute, Kitasato University, Tokyo, Japan
| | - Kamrun Naher
- Ōmura Satoshi Memorial Institute, Kitasato University, Tokyo, Japan
| | - Kazunari Sakai
- Graduate School of Infection Control Sciences, Kitasato University, Tokyo, Japan; Ōmura Satoshi Memorial Institute, Kitasato University, Tokyo, Japan
| | - Asaomi Kuwae
- Graduate School of Infection Control Sciences, Kitasato University, Tokyo, Japan; Ōmura Satoshi Memorial Institute, Kitasato University, Tokyo, Japan
| | - Akio Abe
- Graduate School of Infection Control Sciences, Kitasato University, Tokyo, Japan; Ōmura Satoshi Memorial Institute, Kitasato University, Tokyo, Japan
| | - Yukihiro Asami
- Graduate School of Infection Control Sciences, Kitasato University, Tokyo, Japan; Ōmura Satoshi Memorial Institute, Kitasato University, Tokyo, Japan.
| |
Collapse
|
16
|
McHugh RE, Munnoch JT, Roe AJ, Hoskisson PA. Genome sequence of the aurodox-producing bacterium Streptomyces goldiniensis ATCC 21386. Access Microbiol 2022; 4:acmi000358. [PMID: 36003359 PMCID: PMC9394534 DOI: 10.1099/acmi.0.000358] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Revised: 04/01/2022] [Accepted: 04/08/2022] [Indexed: 11/18/2022] Open
Abstract
We report the genome sequence of Streptomyces goldiniensis ATCC 21386, a strain which produces the anti-bacterial and anti-virulence polyketide, aurodox. The genome of S. goldiniensis ATCC 21386 was sequenced using a multiplatform hybrid approach, revealing a linear genome of ~10 Mbp with a G+C content of 71%. The genome sequence revealed 36 putative biosynthetic gene clusters (BGCs), including a large region of 271 Kbp that was rich in biosynthetic capability. The genome sequence is deposited in DDBJ/EMBL/GenBank with the accession number PRJNA602141.
Collapse
Affiliation(s)
- Rebecca E. McHugh
- *Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, G12 8TA, UK
- Strathclyde Institute of Pharmacy and Biomedical Science, University of Strathclyde, 161 Cathedral Street, Glasgow, G4 0RE, UK
| | - John T. Munnoch
- Strathclyde Institute of Pharmacy and Biomedical Science, University of Strathclyde, 161 Cathedral Street, Glasgow, G4 0RE, UK
| | - Andrew J. Roe
- *Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, G12 8TA, UK
| | - Paul A. Hoskisson
- Strathclyde Institute of Pharmacy and Biomedical Science, University of Strathclyde, 161 Cathedral Street, Glasgow, G4 0RE, UK
| |
Collapse
|
17
|
Martínez OF, Duque HM, Franco OL. Peptidomimetics as Potential Anti-Virulence Drugs Against Resistant Bacterial Pathogens. Front Microbiol 2022; 13:831037. [PMID: 35516442 PMCID: PMC9062693 DOI: 10.3389/fmicb.2022.831037] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Accepted: 02/28/2022] [Indexed: 12/11/2022] Open
Abstract
The uncontrollable spread of superbugs calls for new approaches in dealing with microbial-antibiotic resistance. Accordingly, the anti-virulence approach has arisen as an attractive unconventional strategy to face multidrug-resistant pathogens. As an emergent strategy, there is an imperative demand for discovery, design, and development of anti-virulence drugs. In this regard, peptidomimetic compounds could be a valuable source of anti-virulence drugs, since these molecules circumvent several shortcomings of natural peptide-based drugs like proteolytic instability, immunogenicity, toxicity, and low bioavailability. Some emerging evidence points to the feasibility of peptidomimetics to impair pathogen virulence. Consequently, in this review, we shed some light on the potential of peptidomimetics as anti-virulence drugs to overcome antibiotic resistance. Specifically, we address the anti-virulence activity of peptidomimetics against pathogens' secretion systems, biofilms, and quorum-sensing systems.
Collapse
Affiliation(s)
- Osmel Fleitas Martínez
- Programa de Pós-Graduação em Ciências Genômicas e Biotecnologia, Centro de Análises Proteômicas e Bioquímicas, Universidade Católica de Brasília, Brasília, Brazil.,Programa de Pós-Graduação em Biotecnologia, S-Inova Biotech, Universidade Católica Dom Bosco, Campo Grande, Brazil
| | - Harry Morales Duque
- Programa de Pós-Graduação em Ciências Genômicas e Biotecnologia, Centro de Análises Proteômicas e Bioquímicas, Universidade Católica de Brasília, Brasília, Brazil
| | - Octávio Luiz Franco
- Programa de Pós-Graduação em Ciências Genômicas e Biotecnologia, Centro de Análises Proteômicas e Bioquímicas, Universidade Católica de Brasília, Brasília, Brazil.,Programa de Pós-Graduação em Biotecnologia, S-Inova Biotech, Universidade Católica Dom Bosco, Campo Grande, Brazil
| |
Collapse
|
18
|
A small molecule, C24H17ClN4O2S, inhibits the function of the type III secretion system in Salmonella Typhimurium. J Genet Eng Biotechnol 2022; 20:54. [PMID: 35380331 PMCID: PMC8982747 DOI: 10.1186/s43141-022-00336-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Accepted: 03/22/2022] [Indexed: 11/17/2022]
Abstract
Background Salmonella enterica serovar Typhimurium (S. Typhimurium) causes gastroenteritis and diarrhea in humans and food-producing animals. The type III secretion system (T3SS) has been known to be a potent virulence mechanism by injecting effector proteins into the cytosol of host cells. S. Typhimurium encodes two T3SSs by Salmonella pathogenicity islands 1 and 2. Previous studies showed that T3SS shared a potent virulence mechanism and molecular structure among several gram-negative bacteria. Therefore, T3SS has been identified as an attractive target in the development of novel therapeutics for the treatment of bacterial infections. Several studies reported that small-molecule compounds are able to inhibit functions of bacterial T3SSs. A small molecule, C24H17ClN4O2S, has been shown the ability to inhibit the activity of Yersinia pestis T3SS ATPase, YscN, resulting to block the secretion of effector proteins. In this study, we studied the effects and mechanism for SPI-1 T3SS inhibition of this compound in S. Typhimurium. Results We demonstrated that this compound prohibited the secretion of effector proteins from Salmonella via SPI-1 T3SS at 100 μM. As the result, bacterial invasion ability into epithelial cell cultures was reduced. In contrast with previous study, the C24H17ClN4O2S molecule did not inactivate the activity of SPI-1 T3SS ATPase, InvC, in Salmonella. However, we studied the global cellular effects of S. Typhimurium after being treated with this compound using a quantitative proteomic technique. These proteomic results showed that the main SPI-1 transcription regulator, InvF, and two effector proteins, SipA and SipC, were reduced in bacterial cells treated with the compound. Conclusions It may explain that action of the small-molecule compound, C24H17ClN4O2S, for blocking the secretion of SPI-1 T3SS in Salmonella is through inhibition of SPI-1 regulator, InvF, expression. Further studies are necessary to identify specific mechanisms for inhibition between this small-compound and InvF SPI-1 regulator protein.
Collapse
|
19
|
Song Y, Xu G, Li C, Li Z, Lu C, Shen Y. Structural optimization of natural product fusaric acid to discover novel T3SS inhibitors of Salmonella. Biochem Biophys Res Commun 2021; 582:72-76. [PMID: 34695753 DOI: 10.1016/j.bbrc.2021.10.035] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2021] [Accepted: 10/14/2021] [Indexed: 11/17/2022]
Abstract
Type III secretion system (T3SS) plays a critical role in host cell invasion and pathogenesis of Salmonella. We recently identified the mycotoxin fusaric acid (FA) as a T3SS inhibitor of Salmonella. Herein, twenty-two diphenylsulfane derivatives were designed and synthesized using FA as a lead compound through scaffold hopping. Among them, SL-8 and SL-19 possessing strong anti-T3SS and anti-invasion activity were identified as T3SS inhibitors with improvement in potency as compared to FA. The inhibitory mechanisms on SPI-1 did not depend on the HilD-HilC-RtsA-HilA or PhoP-PhoQ pathway or the assembly of T3SS needle complex. Accordingly, we proposed that the inhibitory effects of SL-8 and SL-19 on SPI-1 probably influence the formation of SicA/InvF-effector complex or other related proteins.
Collapse
Affiliation(s)
- Yuliang Song
- Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, China
| | - Guangsen Xu
- Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, China
| | - Chaoqun Li
- Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, China
| | - Zhiying Li
- Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, China
| | - Chunhua Lu
- Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, China.
| | - Yuemao Shen
- Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, China
| |
Collapse
|
20
|
Identification of Translocation Inhibitors Targeting the Type III Secretion System of Enteropathogenic Escherichia coli. Antimicrob Agents Chemother 2021; 65:e0095821. [PMID: 34543097 DOI: 10.1128/aac.00958-21] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Infections with enteropathogenic Escherichia coli (EPEC) cause severe diarrhea in children. The noninvasive bacteria adhere to enterocytes of the small intestine and use a type III secretion system (T3SS) to inject effector proteins into host cells to modify and exploit cellular processes in favor of bacterial survival and replication. Several studies have shown that the T3SSs of bacterial pathogens are essential for virulence. Furthermore, the loss of T3SS-mediated effector translocation results in increased immune recognition and clearance of the bacteria. The T3SS is, therefore, considered a promising target for antivirulence strategies and novel therapeutics development. Here, we report the results of a high-throughput screening assay based on the translocation of the EPEC effector protein Tir (translocated intimin receptor). Using this assay, we screened more than 13,000 small molecular compounds of six different compound libraries and identified three substances which showed a significant dose-dependent effect on translocation without adverse effects on bacterial or eukaryotic cell viability. In addition, these substances reduced bacterial binding to host cells, effector-dependent cell detachment, and abolished attaching and effacing lesion formation without affecting the expression of components of the T3SS or associated effector proteins. Moreover, no effects of the inhibitors on bacterial motility or Shiga-toxin expression were observed. In summary, we have identified three new compounds that strongly inhibit T3SS-mediated translocation of effectors into mammalian cells, which could be valuable as lead substances for treating EPEC and enterohemorrhagic E. coli infections.
Collapse
|
21
|
Hotinger JA, Morris ST, May AE. The Case against Antibiotics and for Anti-Virulence Therapeutics. Microorganisms 2021; 9:2049. [PMID: 34683370 PMCID: PMC8537500 DOI: 10.3390/microorganisms9102049] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2021] [Revised: 09/22/2021] [Accepted: 09/24/2021] [Indexed: 12/14/2022] Open
Abstract
Although antibiotics have been indispensable in the advancement of modern medicine, there are downsides to their use. Growing resistance to broad-spectrum antibiotics is leading to an epidemic of infections untreatable by first-line therapies. Resistance is exacerbated by antibiotics used as growth factors in livestock, over-prescribing by doctors, and poor treatment adherence by patients. This generates populations of resistant bacteria that can then spread resistance genes horizontally to other bacterial species, including commensals. Furthermore, even when antibiotics are used appropriately, they harm commensal bacteria leading to increased secondary infection risk. Effective antibiotic treatment can induce bacterial survival tactics, such as toxin release and increasing resistance gene transfer. These problems highlight the need for new approaches to treating bacterial infection. Current solutions include combination therapies, narrow-spectrum therapeutics, and antibiotic stewardship programs. These mediate the issues but do not address their root cause. One emerging solution to these problems is anti-virulence treatment: preventing bacterial pathogenesis instead of using bactericidal agents. In this review, we discuss select examples of potential anti-virulence targets and strategies that could be developed into bacterial infection treatments: the bacterial type III secretion system, quorum sensing, and liposomes.
Collapse
Affiliation(s)
| | | | - Aaron E. May
- Department of Medicinal Chemistry, School of Pharmacy, Virginia Commonwealth University, Richmond, VA 23219, USA; (J.A.H.); (S.T.M.)
| |
Collapse
|
22
|
Developing Cyclic Peptomers as Broad-Spectrum Type III Secretion System Inhibitors in Gram-Negative Bacteria. Antimicrob Agents Chemother 2021; 65:e0169020. [PMID: 33875435 PMCID: PMC8373237 DOI: 10.1128/aac.01690-20] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Antibiotic-resistant bacteria are an emerging global health threat. New antimicrobials are urgently needed. The injectisome type III secretion system (T3SS), required by dozens of Gram-negative bacteria for virulence but largely absent from nonpathogenic bacteria, is an attractive antimicrobial target. We previously identified synthetic cyclic peptomers, inspired by the natural product phepropeptin D, that inhibit protein secretion through the Yersinia Ysc and Pseudomonas aeruginosa Psc T3SSs but do not inhibit bacterial growth. Here, we describe the identification of an isomer, 4EpDN, that is 2-fold more potent (50% inhibitory concentration [IC50] of 4 μM) than its parental compound. Furthermore, 4EpDN inhibited the Yersinia Ysa and the Salmonella SPI-1 T3SSs, suggesting that this cyclic peptomer has broad efficacy against evolutionarily distant injectisome T3SSs. Indeed, 4EpDN strongly inhibited intracellular growth of Chlamydia trachomatis in HeLa cells, which requires the T3SS. 4EpDN did not inhibit the unrelated twin arginine translocation (Tat) system, nor did it impact T3SS gene transcription. Moreover, although the injectisome and flagellar T3SSs are evolutionarily and structurally related, the 4EpDN cyclic peptomer did not inhibit secretion of substrates through the Salmonella flagellar T3SS, indicating that cyclic peptomers broadly but specifically target the injectisome T3SS. 4EpDN reduced the number of T3SS needles detected on the surface of Yersinia pseudotuberculosis as detected by microscopy. Collectively, these data suggest that cyclic peptomers specifically inhibit the injectisome T3SS from a variety of Gram-negative bacteria, possibly by preventing complete T3SS assembly.
Collapse
|
23
|
Sharma P, Elofsson M, Roy S. Attenuation of Pseudomonas aeruginosa infection by INP0341, a salicylidene acylhydrazide, in a murine model of keratitis. Virulence 2021; 11:795-804. [PMID: 32507000 PMCID: PMC7567437 DOI: 10.1080/21505594.2020.1776979] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
PSEUDOMONAS AERUGINOSA is an opportunistic pathogen and a major cause of corneal infections worldwide. The bacterium secretes several toxins through its type III secretion system (T3SS) to subvert host immune responses. In addition, it is armed with intrinsic as well as acquired antibiotic resistance mechanisms that make treatment a significant challenge and new therapeutic interventions are needed. Type III secretion inhibitors have been studied as an alternative or in accompaniment to traditional antibiotics to inhibit virulence of bacteria. In this study, INP0341, a T3SS inhibitor, inhibited cytotoxicity by P. aeruginosa toward human corneal epithelial cells (HCEC) at 100 μM without affecting bacterial growth in the liquid media. An increased expression of antimicrobial peptides and reactive oxygen species generation was also observed in cells exposed to P. aeruginosa in the presence of INP0341. Furthermore, INP0341 efficiently attenuated corneal infection by P. aeruginosa in an experimental model of murine keratitis as evident from corneal opacity, clinical score and bacterial load. Thus, INP0341 appears to be a promising candidate to treat corneal infection caused by P. aeruginosa and can be further considered as an alternative therapeutic intervention.
Collapse
Affiliation(s)
- Prerana Sharma
- Prof. Brien Holden Eye Research Centre, LV Prasad Eye Institute , Hyderabad, India.,Department of Animal Biology, University of Hyderabad , Hyderabad, India
| | | | - Sanhita Roy
- Prof. Brien Holden Eye Research Centre, LV Prasad Eye Institute , Hyderabad, India
| |
Collapse
|
24
|
Zigangirova NA, Nesterenko LN, Sheremet AB, Soloveva AV, Luyksaar SI, Zayakin ES, Balunets DV, Gintsburg AL. Fluorothiazinon, a small-molecular inhibitor of T3SS, suppresses salmonella oral infection in mice. J Antibiot (Tokyo) 2021; 74:244-254. [PMID: 33479520 DOI: 10.1038/s41429-020-00396-w] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2020] [Revised: 11/05/2020] [Accepted: 11/18/2020] [Indexed: 01/29/2023]
Abstract
Therapeutic strategies that target bacterial virulence have received considerable attention. The type III secretion system (T3SS) is important for bacterial virulence and represents an attractive therapeutic target. Recently, we developed a new small-molecule inhibitor belonging to a class 2,4-disubstituted-4H-[1,3,4]-thiadiazine-5-ones, Fluorothiazinon (FT-previously called CL-55). FT effectively suppressed T3SS of Chlamydia spp., Pseudomonas aeruginosa, and Salmonella without affecting bacterial growth in vitro. FT was previously characterized by low toxicity, stability, and therapeutic efficacy in animal models. Salmonella T3SS inhibition by FT was studied using in vitro assays for effector proteins detection and estimation of salmonella replication in peritoneal macrophages. The antibacterial effect of FT in vivo was investigated in murine models of salmonella chronic systemic and acute infection. Oral administration of the virulent strain of Salmonella enterica serovar Typhimurium to mice-induced chronic systemic infection with the pathogen persistence in different lymphoid organs such as spleens, Peyer's plaques, and mesenteric lymph nodes. We found that FT suppressed orally induced salmonella infection both with therapeutic and prophylactic administration. Treatment by FT at a dose of 50 mg/kg for 4 days starting from day 7 post-infection (therapy) as well as for 4 days before infection (prevention) led to practically complete eradication of salmonella in mice. FT shows a strong potential for antibacterial therapy and could be used as a substance in the design of antibacterial drugs for pharmaceutical intervention including therapy of antibiotic-resistant infections.
Collapse
Affiliation(s)
- Nailya A Zigangirova
- Gamaleya National Research Center of Epidemiology and Microbiology, Ministry of Health Russian Federation, Gamaleya str.18, Moscow, 123098, Russia.
| | - Ludmila N Nesterenko
- Gamaleya National Research Center of Epidemiology and Microbiology, Ministry of Health Russian Federation, Gamaleya str.18, Moscow, 123098, Russia
| | - Anna B Sheremet
- Gamaleya National Research Center of Epidemiology and Microbiology, Ministry of Health Russian Federation, Gamaleya str.18, Moscow, 123098, Russia
| | - Anna V Soloveva
- Gamaleya National Research Center of Epidemiology and Microbiology, Ministry of Health Russian Federation, Gamaleya str.18, Moscow, 123098, Russia
| | - Sergey I Luyksaar
- Gamaleya National Research Center of Epidemiology and Microbiology, Ministry of Health Russian Federation, Gamaleya str.18, Moscow, 123098, Russia
| | - Egor S Zayakin
- Gamaleya National Research Center of Epidemiology and Microbiology, Ministry of Health Russian Federation, Gamaleya str.18, Moscow, 123098, Russia
| | - Denis V Balunets
- Gamaleya National Research Center of Epidemiology and Microbiology, Ministry of Health Russian Federation, Gamaleya str.18, Moscow, 123098, Russia
| | - Alexandr L Gintsburg
- Gamaleya National Research Center of Epidemiology and Microbiology, Ministry of Health Russian Federation, Gamaleya str.18, Moscow, 123098, Russia
| |
Collapse
|
25
|
Pylkkö T, Ilina P, Tammela P. Development and validation of a high-content screening assay for inhibitors of enteropathogenic E. coli adhesion. J Microbiol Methods 2021; 184:106201. [PMID: 33713725 DOI: 10.1016/j.mimet.2021.106201] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2021] [Revised: 03/05/2021] [Accepted: 03/05/2021] [Indexed: 11/25/2022]
Abstract
Enteropathogenic E. coli (EPEC) causes intestinal infections leading to severe diarrhea. EPEC attaches to the host cell causing lesions to the intestinal epithelium coupled with the effacement of microvilli. In the process, actin accumulates into a pedestal-like structure under bacterial microcolonies. We designed an automated fluorescence microscopy-based screening method for discovering compounds capable of inhibiting EPEC adhesion and virulence using aurodox, a type three secretion system (T3SS) inhibitor, as a positive control. The screening assay employs an EPEC strain (2348/69) expressing a fluorescent protein and actin staining for monitoring the bacteria and their pedestals respectively, analyzing these with a custom image analysis pipeline. The assay allows for the discovery of compounds capable of preventing the formation of pathogenic actin rearrangements. These compounds may be interfering with virulence-related molecular pathways relevant for developing antivirulence leads.
Collapse
Affiliation(s)
- Tuomas Pylkkö
- Drug Research Program, Division of Pharmaceutical Biosciences, Faculty of Pharmacy, P.O. Box 56, FI-00014, University of Helsinki, Finland
| | - Polina Ilina
- Drug Research Program, Division of Pharmaceutical Biosciences, Faculty of Pharmacy, P.O. Box 56, FI-00014, University of Helsinki, Finland
| | - Päivi Tammela
- Drug Research Program, Division of Pharmaceutical Biosciences, Faculty of Pharmacy, P.O. Box 56, FI-00014, University of Helsinki, Finland.
| |
Collapse
|
26
|
Ohara M, Ikeda A, Nakajima A, Ōno T, Noguchi Y, Watanabe A, Hirose T, Sunazuka T. Towards the total synthesis of aurodox: Preparation of the key hemiacetal-lactone. Tetrahedron Lett 2021. [DOI: 10.1016/j.tetlet.2020.152799] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
27
|
Pendergrass HA, Johnson AL, Hotinger JA, May AE. Fluorescence Detection of Type III Secretion Using a Glu-CyFur Reporter System in Citrobacter rodentium. Microorganisms 2020; 8:microorganisms8121953. [PMID: 33316970 PMCID: PMC7764322 DOI: 10.3390/microorganisms8121953] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2020] [Revised: 12/02/2020] [Accepted: 12/06/2020] [Indexed: 12/21/2022] Open
Abstract
Enteropathogenic Escherichia coli (EPEC) is a major cause of infantile diarrhea worldwide. EPEC and the closely related murine model of EPEC infection, Citrobacter rodentium, utilize a type III secretion system (T3SS) to propagate the infection. Since the T3SS is not essential for the bacteria to survive or propagate, inhibiting the virulence factor with a therapeutic would treat the infection without causing harm to commensal bacteria. Studying inhibitors of the T3SS usually requires a BSL-2 laboratory designation and eukaryotic host cells while not indicating the mechanism of inhibition. We have designed a BSL-1 assay using the murine model C. rodentium that does not require mammalian cell culture. This CPG2-reporter assay allows for more rapid analysis of secretion efficiency than Western blotting and is sensitive enough to differentiate between partial and total inhibition of the T3SS. Here we present our method and the results of a small collection of compounds we have screened, including known T3SS inhibitors EGCG, regacin, and aurodox and related quorum sensing inhibitors tannic acid and ellagic acid. We have further characterized EGCG as a T3SS inhibitor and established its IC50 of 1.8 ± 0.4 μM. We also establish tannic acid as a potent inhibitor of the T3SS with an IC50 of 0.65 ± 0.09 μM.
Collapse
|
28
|
Forrestall KL, Burley DE, Cash MK, Pottie IR, Darvesh S. 2-Pyridone natural products as inhibitors of SARS-CoV-2 main protease. Chem Biol Interact 2020; 335:109348. [PMID: 33278462 PMCID: PMC7710351 DOI: 10.1016/j.cbi.2020.109348] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2020] [Revised: 10/05/2020] [Accepted: 11/26/2020] [Indexed: 12/16/2022]
Abstract
The disease, COVID-19, is caused by the severe acute respiratory coronavirus 2 (SARS-CoV-2) for which there is currently no treatment. The SARS-CoV-2 main protease (Mpro) is an important enzyme for viral replication. Small molecules that inhibit this protease could lead to an effective COVID-19 treatment. The 2-pyridone scaffold was previously identified as a possible key pharmacophore to inhibit SARS-CoV-2 Mpro. A search for natural, antimicrobial products with the 2-pyridone moiety was undertaken herein, and their calculated potency as inhibitors of SARS-CoV-2 Mpro was investigated. Thirty-three natural products containing the 2-pyridone scaffold were identified from the literature. An in silico methodology using AutoDock was employed to predict the binding energies and inhibition constants (Ki values) for each 2-pyridone-containing compound with SARS-CoV-2 Mpro. This consisted of molecular optimization of the 2-pyridone compound, docking of the compound with a crystal structure of SARS-CoV-2 Mpro, and evaluation of the predicted interactions and ligand-enzyme conformations. All compounds investigated bound to the active site of SARS-CoV-2 Mpro, close to the catalytic dyad (His-41 and Cys-145). Thirteen molecules had predicted Ki values <1 μM. Glu-166 formed a key hydrogen bond in the majority of the predicted complexes, while Met-165 had some involvement in the complex binding as a close contact to the ligand. Prominent 2-pyridone compounds were further evaluated for their ADMET properties. This work has identified 2-pyridone natural products with calculated potent inhibitory activity against SARS-CoV-2 Mpro and with desirable drug-like properties, which may lead to the rapid discovery of a treatment for COVID-19. 2-pyridone-scaffold is an inhibitory pharmacophore for SARS-CoV-2 Mpro. Thirty-three natural, antimicrobial products identified with 2-pyridone moiety. All 2-pyridone natural products bind to active site of SARS-CoV-2 Mproin silico. Thirteen molecules found to have potent inhibitory activity against SARS-CoV-2 Mpro. Inhibition of SARS-CoV-2 by natural 2-pyridones may lead to treatment of COVID-19.
Collapse
Affiliation(s)
- Katrina L Forrestall
- Department of Medical Neuroscience, Faculty of Medicine, Dalhousie University, Halifax, Nova Scotia, B3H 4R2, Canada
| | - Darcy E Burley
- Department of Medical Neuroscience, Faculty of Medicine, Dalhousie University, Halifax, Nova Scotia, B3H 4R2, Canada
| | - Meghan K Cash
- Department of Medical Neuroscience, Faculty of Medicine, Dalhousie University, Halifax, Nova Scotia, B3H 4R2, Canada
| | - Ian R Pottie
- Department of Chemistry and Physics, Faculty of Arts and Science, Mount Saint Vincent University, Halifax, Nova Scotia, B3M 2J6, Canada; Department of Chemistry, Faculty of Science, Saint Mary's University, Halifax, Nova Scotia, B3H 3C3, Canada
| | - Sultan Darvesh
- Department of Medical Neuroscience, Faculty of Medicine, Dalhousie University, Halifax, Nova Scotia, B3H 4R2, Canada; Department of Chemistry and Physics, Faculty of Arts and Science, Mount Saint Vincent University, Halifax, Nova Scotia, B3M 2J6, Canada; Department of Medicine (Neurology), Faculty of Medicine, Dalhousie University, Halifax, Nova Scotia, B3H 4R2, Canada.
| |
Collapse
|
29
|
Pollock J, Low AS, McHugh RE, Muwonge A, Stevens MP, Corbishley A, Gally DL. Alternatives to antibiotics in a One Health context and the role genomics can play in reducing antimicrobial use. Clin Microbiol Infect 2020; 26:1617-1621. [PMID: 32220638 DOI: 10.1016/j.cmi.2020.02.028] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2019] [Revised: 02/19/2020] [Accepted: 02/22/2020] [Indexed: 01/16/2023]
Abstract
BACKGROUND This review follows on from the International Conference on One Health Antimicrobial Resistance (ICOHAR 2019), where strategies to improve the fundamental understanding and management of antimicrobial resistance at the interface between humans, animals and the environment were discussed. OBJECTIVE This review identifies alternatives to antimicrobials in a One Health context, noting how advances in genomic technologies are assisting their development and enabling more targeted use of antimicrobials. SOURCES Key articles on the use of microbiota modulation, livestock breeding and gene editing, vaccination, antivirulence strategies and bacteriophage therapy are discussed. CONTENT Antimicrobials are central for disease control, but reducing their use is paramount as a result of the rise of transmissible antimicrobial resistance. This review discusses antimicrobial alternatives in the context of improved understanding of fundamental host-pathogen and microbiota interactions using genomic tools. IMPLICATIONS Host and microbial genomics and other novel technologies play an important role in devising disease control strategies for healthier animals and humans that in turn reduce our reliance on antimicrobials.
Collapse
Affiliation(s)
- J Pollock
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, Edinburgh, UK
| | - A S Low
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, Edinburgh, UK
| | - R E McHugh
- Institute of Infection, Immunity & Inflammation, University of Glasgow, Glasgow, Scotland, UK; Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, Scotland, UK
| | - A Muwonge
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, Edinburgh, UK
| | - M P Stevens
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, Edinburgh, UK
| | - A Corbishley
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, Edinburgh, UK
| | - D L Gally
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, Edinburgh, UK.
| |
Collapse
|
30
|
Yoshihara A, Shimatani M, Sakata M, Takemura C, Senuma W, Hikichi Y, Kai K. Quorum Sensing Inhibition Attenuates the Virulence of the Plant Pathogen Ralstonia solanacearum Species Complex. ACS Chem Biol 2020; 15:3050-3059. [PMID: 33172253 DOI: 10.1021/acschembio.0c00752] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Strains of Ralstonia solanacearum species complex (RSSC) cause "bacterial wilt" on a wide range of plant species and thus lead to marked economic losses in agriculture. Quorum sensing (QS), a bacterial cell-cell communication mechanism, controls the virulence of RSSC strains by regulating the production of extracellular polysaccharide (EPS) and secondary metabolites, biofilm formation, and cellular motility. R. solanacearum strain OE1-1 employs (R)-methyl 3-hydroxymyristate (3-OH MAME) as a QS signal, which is synthesized by the PhcB methyltransferase and sensed by the PhcS/PhcRQ two-component system. We describe the design, synthesis, and biological evaluation of inhibitors of the phc QS system. Initial screening of a small set of QS signal analogues revealed that methyl 3-hydroxy-8-phenyloctanoate, named, PQI-1 (phc quorum sensing inhibitor-1), inhibited biofilm formation by strain OE1-1. To improve its inhibitory activity, the derivatives of PQI-1 were synthesized, and their QS inhibition activities were evaluated. PQIs-2-5 evolved from PQI-1 more strongly inhibited not only biofilm formation but also the production of ralfuranone and EPS. Furthermore, RNA-Seq analysis revealed that the PQIs effectively inhibited QS-dependent gene expression and repression in strain OE1-1. On the other hand, the PQIs did not affect the canonical QS systems of the representative reporter bacteria. These antagonists, especially PQI-5, reduced wilting symptoms of the tomato plants infected with strain OE1-1. Taken together, we suggest that targeting the phc QS system has potential for the development of chemicals that protect agricultural crops from bacterial wilt disease.
Collapse
Affiliation(s)
- Ayaka Yoshihara
- Graduate School of Life and Environmental Sciences, Osaka Prefecture University, 1-1 Gakuen-cho, Naka-ku, Sakai, Osaka 599-8531, Japan
| | - Mika Shimatani
- Graduate School of Life and Environmental Sciences, Osaka Prefecture University, 1-1 Gakuen-cho, Naka-ku, Sakai, Osaka 599-8531, Japan
| | - Megumi Sakata
- Graduate School of Life and Environmental Sciences, Osaka Prefecture University, 1-1 Gakuen-cho, Naka-ku, Sakai, Osaka 599-8531, Japan
| | - Chika Takemura
- Laboratory of Plant Pathology and Biotechnology, Kochi University, 200 Otsu, Monobe, Nanko-ku, Kochi 783-8502, Japan
| | - Wakana Senuma
- Laboratory of Plant Pathology and Biotechnology, Kochi University, 200 Otsu, Monobe, Nanko-ku, Kochi 783-8502, Japan
| | - Yasufumi Hikichi
- Laboratory of Plant Pathology and Biotechnology, Kochi University, 200 Otsu, Monobe, Nanko-ku, Kochi 783-8502, Japan
| | - Kenji Kai
- Graduate School of Life and Environmental Sciences, Osaka Prefecture University, 1-1 Gakuen-cho, Naka-ku, Sakai, Osaka 599-8531, Japan
| |
Collapse
|
31
|
Kongkham B, Prabakaran D, Puttaswamy H. Opportunities and challenges in managing antibiotic resistance in bacteria using plant secondary metabolites. Fitoterapia 2020; 147:104762. [PMID: 33069839 DOI: 10.1016/j.fitote.2020.104762] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2020] [Revised: 10/08/2020] [Accepted: 10/12/2020] [Indexed: 12/11/2022]
Abstract
Development of antibiotic resistance (ABR) in bacteria and its multidimensional spread is an emerging global threat that needs immediate attention. Extensive antibiotics (AB) usage results in development of ABR in bacteria by target modification, production of AB degrading enzymes, porin modifications, efflux pumps overexpression, etc. To counter this, apart from strict regulation of AB use and behavioural changes, research and development (R&D) of newer antimicrobials are in place. One such emerging approach to combat ABR is the use of structurally and functionally diverse plant secondary metabolites (PSMs) in combination with the conventional AB. Either the PSMs are themselves antimicrobial or they potentiate the activity of the AB through a range of mechanisms. However, their use is lagging due to poor knowledge of mode of action, structure-activity relationships, pharmacokinetics, etc. This review paper discussed the opportunities and challenges in managing ABR using PSMs. Mechanisms of ABR development in bacteria and current strategies to counter them were studied and the areas where PSMs can play an important role were highlighted. The use of PSMs, both as an anti-resistance and anti-virulence agent in combination therapy to counter multi-drug resistance along with their mechanisms of action, has been discussed in detail. The difficulties in the commercialisation of PSMs and strategies to overcome them along with future priority areas of research have also been given. Following the given R&D path will definitely help in better understanding and utilising the full potential of PSMs in solving the problem of antimicrobial resistance (AMR).
Collapse
Affiliation(s)
- Bhani Kongkham
- Environmental Biotechnology Laboratory, Centre for Rural Development and Technology, Indian Institute of Technology Delhi, Delhi 110016, India
| | - Duraivadivel Prabakaran
- Environmental Biotechnology Laboratory, Centre for Rural Development and Technology, Indian Institute of Technology Delhi, Delhi 110016, India
| | - Hariprasad Puttaswamy
- Environmental Biotechnology Laboratory, Centre for Rural Development and Technology, Indian Institute of Technology Delhi, Delhi 110016, India.
| |
Collapse
|
32
|
Hotinger JA, May AE. Antibodies Inhibiting the Type III Secretion System of Gram-Negative Pathogenic Bacteria. Antibodies (Basel) 2020; 9:antib9030035. [PMID: 32726928 PMCID: PMC7551047 DOI: 10.3390/antib9030035] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Revised: 07/16/2020] [Accepted: 07/22/2020] [Indexed: 12/11/2022] Open
Abstract
Pathogenic bacteria are a global health threat, with over 2 million infections caused by Gram-negative bacteria every year in the United States. This problem is exacerbated by the increase in resistance to common antibiotics that are routinely used to treat these infections, creating an urgent need for innovative ways to treat and prevent virulence caused by these pathogens. Many Gram-negative pathogenic bacteria use a type III secretion system (T3SS) to inject toxins and other effector proteins directly into host cells. The T3SS has become a popular anti-virulence target because it is required for pathogenesis and knockouts have attenuated virulence. It is also not required for survival, which should result in less selective pressure for resistance formation against T3SS inhibitors. In this review, we will highlight selected examples of direct antibody immunizations and the use of antibodies in immunotherapy treatments that target the bacterial T3SS. These examples include antibodies targeting the T3SS of Pseudomonas aeruginosa, Yersinia pestis, Escherichia coli, Salmonella enterica, Shigella spp., and Chlamydia trachomatis.
Collapse
|
33
|
Possible drugs for the treatment of bacterial infections in the future: anti-virulence drugs. J Antibiot (Tokyo) 2020; 74:24-41. [PMID: 32647212 DOI: 10.1038/s41429-020-0344-z] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Revised: 05/26/2020] [Accepted: 05/29/2020] [Indexed: 12/19/2022]
Abstract
Antibiotic resistance is a global threat that should be urgently resolved. Finding a new antibiotic is one way, whereas the repression of the dissemination of virulent pathogenic bacteria is another. From this point of view, this paper summarizes first the mechanisms of conjugation and transformation, two important processes of horizontal gene transfer, and then discusses the approaches for disarming virulent pathogenic bacteria, that is, virulence factor inhibitors. In contrast to antibiotics, anti-virulence drugs do not impose a high selective pressure on a bacterial population, and repress the dissemination of antibiotic resistance and virulence genes. Disarmed virulence factors make virulent pathogens avirulent bacteria or pathobionts, so that we human will be able to coexist with these disarmed bacteria peacefully.
Collapse
|
34
|
Prudence SMM, Addington E, Castaño-Espriu L, Mark DR, Pintor-Escobar L, Russell AH, McLean TC. Advances in actinomycete research: an ActinoBase review of 2019. MICROBIOLOGY-SGM 2020; 166:683-694. [PMID: 32558638 PMCID: PMC7641383 DOI: 10.1099/mic.0.000944] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The actinomycetes are Gram-positive bacteria belonging to the order Actinomycetales within the phylum Actinobacteria. They include members with significant economic and medical importance, for example filamentous actinomycetes such as Streptomyces species, which have a propensity to produce a plethora of bioactive secondary metabolites and form symbioses with higher organisms, such as plants and insects. Studying these bacteria is challenging, but also fascinating and very rewarding. As a Microbiology Society initiative, members of the actinomycete research community have been developing a Wikipedia-style resource, called ActinoBase, the purpose of which is to aid in the study of these filamentous bacteria. This review will highlight 10 publications from 2019 that have been of special interest to the ActinoBase community, covering 4 major components of actinomycete research: (i) development and regulation; (ii) specialized metabolites; (iii) ecology and host interactions; and (iv) technology and methodology.
Collapse
Affiliation(s)
- Samuel M M Prudence
- School of Biological Sciences, University of East Anglia, Norwich Research Park, Norwich, Norfolk NR4 7TJ, UK
| | - Emily Addington
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, 161 Cathedral Street, Glasgow, G4 0RE, UK
| | - Laia Castaño-Espriu
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, 161 Cathedral Street, Glasgow, G4 0RE, UK
| | - David R Mark
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, 161 Cathedral Street, Glasgow, G4 0RE, UK
| | | | - Alicia H Russell
- Department of Molecular Microbiology, John Innes Centre, Norwich, NR4 7UH, UK
| | - Thomas C McLean
- School of Biological Sciences, University of East Anglia, Norwich Research Park, Norwich, Norfolk NR4 7TJ, UK
| |
Collapse
|
35
|
Sheremet AB, Nesterenko LN, Zigangirova NA. The Type Three Secretion System of Pseudomonas aeruginosa as a Target for Development of Antivirulence Drugs. MOLECULAR GENETICS, MICROBIOLOGY AND VIROLOGY 2020. [DOI: 10.3103/s0891416820010073] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
|
36
|
Delivery of Heterologous Proteins, Enzymes, and Antigens via the Bacterial Type III Secretion System. Microorganisms 2020; 8:microorganisms8050777. [PMID: 32455678 PMCID: PMC7285344 DOI: 10.3390/microorganisms8050777] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Revised: 05/14/2020] [Accepted: 05/19/2020] [Indexed: 12/27/2022] Open
Abstract
The Type III Secretion System (T3SS) is a multimeric protein complex composed of over 20 different proteins, utilized by Gram-negative bacteria to infect eukaryotic host cells. The T3SS has been implicated as a virulence factor by which pathogens cause infection and has recently been characterized as a communication tool between bacteria and plant cells in the rhizosphere. The T3SS has been repurposed to be used as a tool for the delivery of non-native or heterologous proteins to eukaryotic cells or the extracellular space for a variety of purposes, including drug discovery and drug delivery. This review covers the methodology of heterologous protein secretion as well as multiple cases of utilizing the T3SS to deliver heterologous proteins or artificial materials. The research covered in this review will serve to outline the scope and limitations of utilizing the T3SS as a tool for protein delivery.
Collapse
|
37
|
Exploring resveratrol dimers as virulence blocking agents - Attenuation of type III secretion in Yersinia pseudotuberculosis and Pseudomonas aeruginosa. Sci Rep 2020; 10:2103. [PMID: 32034212 PMCID: PMC7005745 DOI: 10.1038/s41598-020-58872-0] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2019] [Accepted: 01/20/2020] [Indexed: 12/25/2022] Open
Abstract
Bacterial infections continue to threaten humankind and the rapid spread of antibiotic resistant bacteria is alarming. Current antibiotics target essential bacterial processes and thereby apply a strong selective pressure on pathogenic and non-pathogenic bacteria alike. One alternative strategy is to block bacterial virulence systems that are essential for the ability to cause disease but not for general bacterial viability. We have previously show that the plant natural product (-)-hopeaphenol blocks the type III secretion system (T3SS) in the Gram-negative pathogens Yersinia pseudotuberculosis and Pseudomonas aeruginosa. (-)-Hopeaphenol is a resveratrol tetramer and in the present study we explore various resveratrol dimers, including partial structures of (-)-hopeaphenol, as T3SS inhibitors. To allow rapid and efficient assessment of T3SS inhibition in P. aeruginosa, we developed a new screening method by using a green fluorescent protein reporter under the control of the ExoS promoter. Using a panel of assays we showed that compounds with a benzofuran core structure i.e. viniferifuran, dehydroampelopsin B, anigopreissin A, dehydro-δ-viniferin and resveratrol-piceatannol hybrid displayed significant to moderate activities towards the T3SS in Y. pseudotuberculosis and P. aeruginosa.
Collapse
|
38
|
Hotinger JA, May AE. Animal Models of Type III Secretion System-Mediated Pathogenesis. Pathogens 2019; 8:pathogens8040257. [PMID: 31766664 PMCID: PMC6963218 DOI: 10.3390/pathogens8040257] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2019] [Revised: 11/18/2019] [Accepted: 11/20/2019] [Indexed: 01/22/2023] Open
Abstract
The type III secretion system (T3SS) is a conserved virulence factor used by many Gram-negative pathogenic bacteria and has become an important target for anti-virulence drugs. Most T3SS inhibitors to date have been discovered using in vitro screening assays. Pharmacokinetics and other important characteristics of pharmaceuticals cannot be determined with in vitro assays alone. In vivo assays are required to study pathogens in their natural environment and are an important step in the development of new drugs and vaccines. Animal models are also required to understand whether T3SS inhibition will enable the host to clear the infection. This review covers selected animal models (mouse, rat, guinea pig, rabbit, cat, dog, pig, cattle, primates, chicken, zebrafish, nematode, wax moth, flea, fly, and amoeba), where T3SS activity and infectivity have been studied in relation to specific pathogens (Escherichia coli, Salmonella spp., Pseudomonas spp., Shigella spp., Bordetella spp., Vibrio spp., Chlamydia spp., and Yersinia spp.). These assays may be appropriate for those researching T3SS inhibition.
Collapse
|
39
|
Pendergrass HA, May AE. Natural Product Type III Secretion System Inhibitors. Antibiotics (Basel) 2019; 8:antibiotics8040162. [PMID: 31554164 PMCID: PMC6963908 DOI: 10.3390/antibiotics8040162] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2019] [Revised: 09/18/2019] [Accepted: 09/19/2019] [Indexed: 01/05/2023] Open
Abstract
Many known inhibitors of the bacterial type III secretion system (T3SS), a virulence factor used by pathogenic bacteria to infect host cells, are natural products. These compounds, produced by bacteria, fungi, and plants, may have developed as prophylactic treatments for potential attack by bacterial pathogens or as an attempt by symbiotic organisms to protect their hosts. Regardless, better understanding of the structures and mechanisms of action of these compounds may open opportunities for drug development against diseases caused by pathogens utilizing the T3SS. This review will cover selected known natural products of the T3SS and detail what is known of their origin and mechanism of action. These inhibitors highlight nature’s ability to modulate interactions between organisms at a cellular level.
Collapse
Affiliation(s)
- Heather A Pendergrass
- Department of Medicinal Chemistry, Virginia Commonwealth University, Richmond, VA 23284, USA.
| | - Aaron E May
- Department of Medicinal Chemistry, Virginia Commonwealth University, Richmond, VA 23284, USA.
| |
Collapse
|
40
|
Ma YN, Chen L, Si NG, Jiang WJ, Zhou ZG, Liu JL, Zhang LQ. Identification of Benzyloxy Carbonimidoyl Dicyanide Derivatives as Novel Type III Secretion System Inhibitors via High-Throughput Screening. FRONTIERS IN PLANT SCIENCE 2019; 10:1059. [PMID: 31543889 PMCID: PMC6739442 DOI: 10.3389/fpls.2019.01059] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/15/2019] [Accepted: 08/05/2019] [Indexed: 06/10/2023]
Abstract
The type III secretion system (T3SS) in many Gram-negative bacterial pathogens is regarded as the most critical virulence determinant and an attractive target for novel anti-virulence drugs. In this study, we constructed a T3SS secretion reporter containing the β-lactamase gene fused with a signal peptide sequence of the T3SS effector gene, and established a high-throughput screening system for T3SS inhibitors in the plant pathogenic bacterium Acidovorax citrulli. From a library of 12,000 chemical compounds, we identified a series of benzyloxy carbonimidoyl dicyanide (BCD) derivatives that effectively blocked T3SS-dependent β-lactamase secretion. Substitution of halogens or nitro groups at the para-position on the benzene ring contributed to an increased inhibitory activity. One representative compound, BCD03 (3,4-dichloro-benzyloxy carbonimidoyl dicyanide), dramatically reduced pathogenicity of A. citrulli on melon seedlings, and attenuated hypersensitive responses in the non-host Nicotiana tabacum caused by pathogenic bacteria A. citrulli, Xanthomonas oryzae pv. oryzae and Pseudomonas syringae pv. tomato at sub-MIC concentrations. Western blotting assay further confirmed that BCD03 inhibited effector secretion from the above bacteria via T3SS in the liquid medium. Taken together, our data suggest that BCD derivatives act as novel inhibitors of T3SS in multiple plant bacterial pathogens.
Collapse
Affiliation(s)
- Yi-Nan Ma
- Department of Plant Pathology and MOA Key Laboratory of Pest Monitoring and Green Management, China Agricultural University, Beijing, China
| | - Liang Chen
- Department of Plant Pathology and MOA Key Laboratory of Pest Monitoring and Green Management, China Agricultural University, Beijing, China
- State Key Laboratory of the Discovery and Development of Novel Pesticide, Shenyang Sinochem Agrochemicals R&D Co., Ltd, Shenyang, China
| | - Nai-Guo Si
- State Key Laboratory of the Discovery and Development of Novel Pesticide, Shenyang Sinochem Agrochemicals R&D Co., Ltd, Shenyang, China
| | - Wen-Jun Jiang
- Department of Plant Pathology and MOA Key Laboratory of Pest Monitoring and Green Management, China Agricultural University, Beijing, China
| | - Zhi-Gang Zhou
- China-Norway Joint Lab on Fish Gut Microbiota, Feed Research Institute, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Jun-Li Liu
- State Key Laboratory of the Discovery and Development of Novel Pesticide, Shenyang Sinochem Agrochemicals R&D Co., Ltd, Shenyang, China
| | - Li-Qun Zhang
- Department of Plant Pathology and MOA Key Laboratory of Pest Monitoring and Green Management, China Agricultural University, Beijing, China
| |
Collapse
|
41
|
Lyons BJE, Strynadka NCJ. On the road to structure-based development of anti-virulence therapeutics targeting the type III secretion system injectisome. MEDCHEMCOMM 2019; 10:1273-1289. [PMID: 31534650 PMCID: PMC6748289 DOI: 10.1039/c9md00146h] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/11/2019] [Accepted: 06/07/2019] [Indexed: 12/19/2022]
Abstract
The type III secretion system injectisome is a syringe-like multimembrane spanning nanomachine that is essential to the pathogenicity but not viability of many clinically relevant Gram-negative bacteria, such as enteropathogenic Escherichia coli, Salmonella enterica and Pseudomonas aeruginosa. Due to the rise in antibiotic resistance, new strategies must be developed to treat the growing spectre of drug resistant infections. Targeting the injectisome via an 'anti-virulence strategy' is a promising avenue to pursue as an alternative to the more commonly used bactericidal therapeutics, which have a high propensity for resulting resistance development and often more broad killing profile, including unwanted side effects in eliminating favourable members of the microbiome. Building on more than a decade of crystallographic work of truncated or isolated forms of the more than two dozen components of the secretion apparatus, recent advances in the field of single-particle cryo-electron microscopy have allowed for the elucidation of atomic resolution structures for many of the type III secretion system components in their assembled, oligomerized state including the needle complex, export apparatus and ATPase. Cryo-electron tomography studies have also advanced our understanding of the direct pathogen-host interaction between the type III secretion system translocon and host cell membrane. These new structural works that further our understanding of the myriad of protein-protein interactions that promote injectisome function will be highlighted in this review, with a focus on those that yield promise for future anti-virulence drug discovery and design. Recently developed inhibitors, including both synthetic, natural product and peptide inhibitors, as well as promising new developments of immunotherapeutics will be discussed. As our understanding of this intricate molecular machinery advances, the development of anti-virulence inhibitors can be enhanced through structure-guided drug design.
Collapse
Affiliation(s)
- Bronwyn J E Lyons
- Department of Biochemistry and Molecular Biology and Center for Blood Research , University of British Columbia , 2350 Health Sciences Mall , Vancouver , British Columbia V6T 1Z3 , Canada .
| | - Natalie C J Strynadka
- Department of Biochemistry and Molecular Biology and Center for Blood Research , University of British Columbia , 2350 Health Sciences Mall , Vancouver , British Columbia V6T 1Z3 , Canada .
| |
Collapse
|
42
|
Larzábal M, Baldoni HA, Suvire FD, Curto LM, Gomez GE, Da Silva WM, Giudicessi SL, Camperi SA, Delfino JM, Cataldi AA, Enriz D. An inhibitory mechanism of action of coiled‐coil peptides against type three secretion system from enteropathogenicEscherichia coli. J Pept Sci 2019; 25:e3149. [DOI: 10.1002/psc.3149] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2018] [Revised: 12/28/2018] [Accepted: 01/08/2019] [Indexed: 12/17/2022]
Affiliation(s)
| | - Hector A. Baldoni
- Facultad de Química, Bioquímica y FarmaciaUniversidad Nacional de San Luis San Luis Argentina
- IMASL‐CONICET San Luis Argentina
| | - Fernando D. Suvire
- Facultad de Química, Bioquímica y FarmaciaUniversidad Nacional de San Luis San Luis Argentina
- IMIBIO‐CONICET San Luis Argentina
| | - Lucrecia M. Curto
- Facultad de Farmacia y Bioquímica, Departamento de Química BiológicaUniversidad de Buenos Aires (UBA) Buenos Aires Argentina
- Universidad de Buenos Aires‐CONICET, Instituto de Química y Fisicoquímica Biológicas (IQUIFIB) Buenos Aires Argentina
| | - Gabriela E. Gomez
- Facultad de Farmacia y Bioquímica, Departamento de Química BiológicaUniversidad de Buenos Aires (UBA) Buenos Aires Argentina
- Universidad de Buenos Aires‐CONICET, Instituto de Química y Fisicoquímica Biológicas (IQUIFIB) Buenos Aires Argentina
| | | | - Silvana L. Giudicessi
- Facultad de Farmacia y Bioquímica, Cátedra de BiotecnologíaUniversidad de Buenos Aires (UBA) Buenos Aires Argentina
- CONICET—Universidad de Buenos Aires, Instituto de Nanobiotecnología (NANOBIOTEC) Buenos Aires Argentina
| | - Silvia A. Camperi
- Facultad de Farmacia y Bioquímica, Cátedra de BiotecnologíaUniversidad de Buenos Aires (UBA) Buenos Aires Argentina
- CONICET—Universidad de Buenos Aires, Instituto de Nanobiotecnología (NANOBIOTEC) Buenos Aires Argentina
| | - Jose M. Delfino
- Facultad de Farmacia y Bioquímica, Departamento de Química BiológicaUniversidad de Buenos Aires (UBA) Buenos Aires Argentina
- Universidad de Buenos Aires‐CONICET, Instituto de Química y Fisicoquímica Biológicas (IQUIFIB) Buenos Aires Argentina
| | | | - Daniel Enriz
- Facultad de Química, Bioquímica y FarmaciaUniversidad Nacional de San Luis San Luis Argentina
- IMIBIO‐CONICET San Luis Argentina
| |
Collapse
|
43
|
Characterization of the Mode of Action of Aurodox, a Type III Secretion System Inhibitor from Streptomyces goldiniensis. Infect Immun 2019; 87:IAI.00595-18. [PMID: 30455200 PMCID: PMC6346137 DOI: 10.1128/iai.00595-18] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2018] [Accepted: 11/12/2018] [Indexed: 11/20/2022] Open
Abstract
Recent work has demonstrated that the polyketide natural product Aurodox from Streptomyces goldiniensis is able to block the pathogenesis of the murine pathogen Citrobacter rodentium. In this work, we aimed to gain a better understanding of the mechanism of action of the compound. Recent work has demonstrated that the polyketide natural product Aurodox from Streptomyces goldiniensis is able to block the pathogenesis of the murine pathogen Citrobacter rodentium. In this work, we aimed to gain a better understanding of the mechanism of action of the compound. We show that Aurodox downregulates the expression of the type III secretion systems of enteropathogenic and enterohemorrhagic Escherichia coli. Furthermore, we have used transcriptomic analysis to show that Aurodox inhibits the expression at the transcriptional level by repressing the master regulator, ler. Our data support a model in which Aurodox acts upstream of ler and not directly on the secretion system itself. Finally, we have shown that Aurodox, unlike some traditional antibiotics, does not induce expression of RecA, which is essential for the production of Shiga toxin. We propose that these properties nominate Aurodox as a promising antivirulence therapy for the treatment of these infections.
Collapse
|
44
|
Puigvert M, Solé M, López‐Garcia B, Coll NS, Beattie KD, Davis RA, Elofsson M, Valls M. Type III secretion inhibitors for the management of bacterial plant diseases. MOLECULAR PLANT PATHOLOGY 2019; 20:20-32. [PMID: 30062690 PMCID: PMC6430469 DOI: 10.1111/mpp.12736] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/26/2023]
Abstract
The identification of chemical compounds that prevent and combat bacterial diseases is fundamental for crop production. Bacterial virulence inhibitors are a promising alternative to classical control treatments, because they have a low environmental impact and are less likely to generate bacterial resistance. The major virulence determinant of most animal and plant bacterial pathogens is the type III secretion system (T3SS). In this work, we screened nine plant extracts and 12 isolated compounds-including molecules effective against human pathogens-for their capacity to inhibit the T3SS of plant pathogens and for their applicability as virulence inhibitors for crop protection. The screen was performed using a luminescent reporter system developed in the model pathogenic bacterium Ralstonia solanacearum. Five synthetic molecules, one natural product and two plant extracts were found to down-regulate T3SS transcription, most through the inhibition of the regulator hrpB. In addition, for three of the molecules, corresponding to salicylidene acylhydrazide derivatives, the inhibitory effect caused a dramatic decrease in the secretion capacity, which was translated into impaired plant responses. These candidate virulence inhibitors were then tested for their ability to protect plants. We demonstrated that salicylidene acylhydrazides can limit R. solanacearum multiplication in planta and protect tomato plants from bacterial speck caused by Pseudomonas syringae pv. tomato. Our work validates the efficiency of transcription reporters to discover compounds or natural product extracts that can be potentially applied to prevent bacterial plant diseases.
Collapse
Affiliation(s)
- Marina Puigvert
- Department of GeneticsUniversity of BarcelonaBarcelona08028CataloniaSpain
- Centre for Research in Agricultural Genomics (CSIC‐IRTA‐UAB‐UB)Bellaterra08193CataloniaSpain
| | - Montserrat Solé
- Centre for Research in Agricultural Genomics (CSIC‐IRTA‐UAB‐UB)Bellaterra08193CataloniaSpain
| | - Belén López‐Garcia
- Centre for Research in Agricultural Genomics (CSIC‐IRTA‐UAB‐UB)Bellaterra08193CataloniaSpain
| | - Núria S. Coll
- Centre for Research in Agricultural Genomics (CSIC‐IRTA‐UAB‐UB)Bellaterra08193CataloniaSpain
| | - Karren D. Beattie
- Griffith Institute for Drug DiscoveryGriffith UniversityQld4111Australia
| | - Rohan A. Davis
- Griffith Institute for Drug DiscoveryGriffith UniversityQld4111Australia
| | | | - Marc Valls
- Department of GeneticsUniversity of BarcelonaBarcelona08028CataloniaSpain
- Centre for Research in Agricultural Genomics (CSIC‐IRTA‐UAB‐UB)Bellaterra08193CataloniaSpain
| |
Collapse
|
45
|
El Qaidi S, Zhu C, McDonald P, Roy A, Maity PK, Rane D, Perera C, Hardwidge PR. High-Throughput Screening for Bacterial Glycosyltransferase Inhibitors. Front Cell Infect Microbiol 2018; 8:435. [PMID: 30619781 PMCID: PMC6305410 DOI: 10.3389/fcimb.2018.00435] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2018] [Accepted: 12/05/2018] [Indexed: 11/25/2022] Open
Abstract
The enteropathogenic and enterohemorrhagic Escherichia coli NleB proteins as well as the Salmonella enterica SseK proteins are type III secretion system effectors that function as glycosyltransferase enzymes to post-translationally modify host substrates on arginine residues. This modification is unusual because it occurs on the guanidinium groups of arginines, which are poor nucleophiles, and is distinct from the activity of the mammalian O-linked N-acetylglucosaminyltransferase. We conducted high-throughput screening assays to identify small molecules that inhibit NleB/SseK activity. Two compounds, 100066N and 102644N, both significantly inhibited NleB1, SseK1, and SseK2 activities. Addition of these compounds to cultured mammalian cells was sufficient to inhibit NleB1 glycosylation of the tumor necrosis factor receptor type 1-associated DEATH domain protein. These compounds were also capable of inhibiting Salmonella enterica strain ATCC 14028 replication in mouse macrophage-like cells. Neither inhibitor was significantly toxic to mammalian cells, nor showed in vitro cross-reactivity with the mammalian O-linked N-acetylglucosaminyltransferase. These compounds or derivatives generated from medicinal chemistry refinements may have utility as a potential alternative therapeutic strategy to antibiotics or as reagents to further the study of bacterial glycosyltransferases.
Collapse
Affiliation(s)
- Samir El Qaidi
- College of Veterinary Medicine, Kansas State University, Manhattan, KS, United States
| | - Congrui Zhu
- College of Veterinary Medicine, Kansas State University, Manhattan, KS, United States
| | - Peter McDonald
- High Throughput Screening Laboratory, University of Kansas, Lawrence, KS, United States
| | - Anuradha Roy
- High Throughput Screening Laboratory, University of Kansas, Lawrence, KS, United States
| | - Pradip Kumar Maity
- Synthetic Chemical Biology Core Laboratory, University of Kansas, Lawrence, KS, United States
| | - Digamber Rane
- Synthetic Chemical Biology Core Laboratory, University of Kansas, Lawrence, KS, United States
| | - Chamani Perera
- Synthetic Chemical Biology Core Laboratory, University of Kansas, Lawrence, KS, United States
| | - Philip R Hardwidge
- College of Veterinary Medicine, Kansas State University, Manhattan, KS, United States
| |
Collapse
|
46
|
Li T, Zhang D, Oo TN, San MM, Mon AM, Hein PP, Wang Y, Lu C, Yang X. Investigation on the Antibacterial and Anti-T3SS Activity of Traditional Myanmar Medicinal Plants. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE : ECAM 2018; 2018:2812908. [PMID: 30402120 PMCID: PMC6198585 DOI: 10.1155/2018/2812908] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/29/2018] [Accepted: 09/27/2018] [Indexed: 01/18/2023]
Abstract
Myanmar has a rich pool of, but less known, medicinal plants with traditional knowledge. In this study, we aimed to investigate the inhibitory activity of traditional Myanmar medicinal plants against the type III secretion system (T3SS) of Salmonella enterica serovar Typhimurium UK-1 χ8956 and the intestinal disease-caused by microbes including S. enterica serovar Typhimurium UK-1 χ8956, Proteusbacillus vulgaris CPCC 160013, Escherichia coli CICC 10003, and Staphylococcus aureus ATCC 25923. The EtOH extracts of 93 samples were used to screen the inhibitory activities against the secretion of T3SS effector proteins SipA/B/C/D of S. enterica and the antibacterial activity against S. enterica, P. vulgaris, E. coli, and S. aureus. Out of 71 crude drugs traditionally used, 18 were proofed to be effective either on the growth inhibition of tested bacteria and/or as inhibitors for the T3SS. The EtOH extracts of five plants, Luvunga scandens (Roxb.) Buch.-Ham. ex Wight & Arn. (My7), Myrica nagi Thunb. (My11), Terminalia citrina Roxb. ex Fleming (My21), Thymus vulgaris L. (My49), and Cinnamomum bejolghota (Buch.-Ham.) Sweet (My104), showed potent inhibitory activities against the secretion of T3SS proteins SipA/B/C/D of S. enterica serovar Typhimurium UK-1 χ 8956. Mansonia gagei J.R.Drumm (My3) and Mesua ferrea (Roxb.) L. (My10) showed strong antibacterial activities against P. vulgaris and S. aureus. This study provided the first scientific evidence of T3SS prohibiting and antibacterial properties for the traditional knowledge in Myanmar of using plants as medicines for treating infections and gastrointestinal disease. Further researches are proposed to discover the active chemical compounds and mechanism of L. scandens (Roxb.) Buch.-Ham. ex Wight & Arn, M. nagi Thunb., T. citrina Roxb. ex Fleming, T. vulgaris L., and C. bejolghota (Buch.-Ham.) Sweet as antivirulence drugs and the potential of M. gagei J.R.Drumm and M. ferrea L. as new broad spectrum plant antibiotics.
Collapse
Affiliation(s)
- Tianhong Li
- Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Shandong University, Jinan 250012, China
| | - Dongdong Zhang
- Key Laboratory of Economic Plants and Biotechnology, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming 650201, China
- Southeast Asia Biodiversity Research Institute, Chinese Academy of Sciences, Yezin, Nay Pyi Taw 05282, Myanmar
| | - Thaung Naing Oo
- Forest Research Institute, Yezin, Nay Pyi Taw 05282, Myanmar
| | - Myint Myint San
- Forest Research Institute, Yezin, Nay Pyi Taw 05282, Myanmar
| | - Aye Mya Mon
- Key Laboratory of Economic Plants and Biotechnology, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming 650201, China
- Southeast Asia Biodiversity Research Institute, Chinese Academy of Sciences, Yezin, Nay Pyi Taw 05282, Myanmar
| | - Pyae Phyo Hein
- Key Laboratory of Economic Plants and Biotechnology, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming 650201, China
- Southeast Asia Biodiversity Research Institute, Chinese Academy of Sciences, Yezin, Nay Pyi Taw 05282, Myanmar
| | - Yuehu Wang
- Key Laboratory of Economic Plants and Biotechnology, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming 650201, China
| | - Chunhua Lu
- Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Shandong University, Jinan 250012, China
| | - Xuefei Yang
- Key Laboratory of Economic Plants and Biotechnology, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming 650201, China
- Southeast Asia Biodiversity Research Institute, Chinese Academy of Sciences, Yezin, Nay Pyi Taw 05282, Myanmar
| |
Collapse
|
47
|
Nakasone N, Ogura Y, Higa N, Toma C, Koizumi Y, Takaesu G, Suzuki T, Yamashiro T. Effects of Psidium guajava leaf extract on secretion systems of Gram-negative enteropathogenic bacteria. Microbiol Immunol 2018; 62:444-453. [PMID: 29790584 DOI: 10.1111/1348-0421.12604] [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: 06/13/2017] [Revised: 04/25/2018] [Accepted: 05/16/2018] [Indexed: 01/22/2023]
Abstract
We screened a total of 672 plant-tissue extracts to search for phytochemicals that inhibit the function of the type III secretion system (T3SS) of enteropathogenic Escherichia coli (EPEC) and enterohemorrhagic E. coli (EHEC). Among candidates examined, we found that an extract from the leaves of Psidium guajava (guava) inhibited the secretion of the EspB protein from EPEC and EHEC without affecting bacterial growth. The guava extract (GE) also inhibited EPEC and EHEC from adhering to and injecting EspB protein into HEp-2 cells. GE seemed to block the translocation of EspB from the bacterial cells to the culture medium. In addition to EPEC and EHEC, GE also inhibited the T3SS of Yersinia pseudotuberculosis and Salmonella enterica serovar Typhimurium. After exposure to GE, Y. pseudotuberculosis stopped the secretion of Yop proteins and lost its ability to induce the apoptosis of mouse bone marrow-derived macrophages. S. Typhimurium exposed to GE ceased the secretion of Sip proteins and lost its ability to invade HEp-2 cells. GE inhibited EspC secretion, the type V secretion protein of EPEC, but not Shiga toxin2 from EHEC. Thus, our results suggest that guava leaves contain a novel type of antimicrobial compound that could be used for the therapeutic treatment and prevention of gram-negative enteropathogenic bacterial infections.
Collapse
Affiliation(s)
- Noboru Nakasone
- Department of Bacteriology, Graduate School of Medicine, University of the Ryukyus, Nishihara, Okinawa 903- 0215, Japan
| | - Yasunori Ogura
- Department of Food Science and Nutrition, Nara Women's University, Nara 630-8506
| | - Naomi Higa
- Department of Bacteriology, Graduate School of Medicine, University of the Ryukyus, Nishihara, Okinawa 903- 0215, Japan
| | - Claudia Toma
- Department of Bacteriology, Graduate School of Medicine, University of the Ryukyus, Nishihara, Okinawa 903- 0215, Japan
| | - Yukiko Koizumi
- Department of Pathology and Comprehensive Cancer Center, University of Michigan Medical School, 3852 E Medical Center Dr, Ann Arbor, MI 48109, USA
| | - Giichi Takaesu
- Tropical Biosphere Research Center University of the Ryukyus, Nishihara, Okinawa 903-0213, Japan
| | - Toshihiko Suzuki
- Department of Bacterial Pathogenesis, Infection and Host Response Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Yushima, bunkyo-ku 1130034, Tokyo, Japan
| | - Tetsu Yamashiro
- Department of Bacteriology, Graduate School of Medicine, University of the Ryukyus, Nishihara, Okinawa 903- 0215, Japan
| |
Collapse
|
48
|
Zhang Y, Liu Y, Wang T, Deng X, Chu X. Natural compound sanguinarine chloride targets the type III secretion system of Salmonella enterica Serovar Typhimurium. Biochem Biophys Rep 2018; 14:149-154. [PMID: 29761161 PMCID: PMC5948472 DOI: 10.1016/j.bbrep.2018.04.011] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2018] [Revised: 04/24/2018] [Accepted: 04/26/2018] [Indexed: 11/24/2022] Open
Abstract
The type III secretion system (T3SS) is a key virulence mechanism of many Gram-negative bacterial pathogens. Upon contact between bacteria and host cells, T3SS transfers a series of effectors from the bacterial cytosol to host cells. It is widely known that a mutation in T3SS does not impair bacterial growth, thereby avoiding any subsequent development of resistance. Thus, T3SS is expected to be a candidate therapeutic target. While developing the T3SS screening method, we discovered that sanguinarine chloride, a natural compound, could decrease the production of the SPI-1 type III secretion system main virulence proteins SipA and SipB and prevent the invasion of HeLa cells by Salmonella enterica serovar Typhimurium without affecting the growth of Salmonella. Furthermore, sanguinarine chloride downregulated the transcription of HilA and consequently regulated the expression of the SPI-1 apparatus and effector genes. In summary, our study directly demonstrated that this putative SPI-1 inhibitor belongs to a novel class of anti-Salmonella compounds. Sanguinarine chloride effectively inhibits the translocation of a SipA-Lactamase fusion into mammalian cells. Sanguinarine chloride inhibits the invasion of Hela cells by Salmonella enterica serovar Typhimurium. Sanguinarine chloride inhibits the secretion of SPI-1 virulence proteins. Sanguinarine chloride inhibits SPI-1 effectors through SPI-1 transcription regulate.
Collapse
Affiliation(s)
- Yong Zhang
- Center of Infection and Immunity, First Hospital, Jilin University, Changchun 130061, China.,Key Laboratory of Zoonosis, Ministry of Education, Institute of Zoonosis, College of Veterinary Medicine, Jilin University, Changchun 130062, China
| | - Yan Liu
- Key Laboratory of Zoonosis, Ministry of Education, Institute of Zoonosis, College of Veterinary Medicine, Jilin University, Changchun 130062, China
| | - Tingting Wang
- Key Laboratory of Zoonosis, Ministry of Education, Institute of Zoonosis, College of Veterinary Medicine, Jilin University, Changchun 130062, China
| | - Xuming Deng
- Center of Infection and Immunity, First Hospital, Jilin University, Changchun 130061, China.,Key Laboratory of Zoonosis, Ministry of Education, Institute of Zoonosis, College of Veterinary Medicine, Jilin University, Changchun 130062, China
| | - Xiao Chu
- Center of Infection and Immunity, First Hospital, Jilin University, Changchun 130061, China
| |
Collapse
|
49
|
Nakasone N, Higa N, Toma C, Ogura Y, Suzuki T, Yamashiro T. Epigallocatechin gallate inhibits the type III secretion system of Gram-negative enteropathogenic bacteria under model conditions. FEMS Microbiol Lett 2018. [PMID: 28651361 DOI: 10.1093/femsle/fnx111] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Epigallocatechin gallate (EGCG), a major polyphenol in green tea, inhibits the type III secretion system (T3SS) of enteropathogenic and enterohemorrhagic Escherichia coli (EPEC and EHEC, respectively), Salmonella enterica serovar Typhimurium, and Yersinia pseudotuberculosis. The inhibitory effect causes the inhibition of hemolysis, cell invasion, cell adhesion and apoptosis, which are functions of the type III secretion device. In the case of EPEC, EspB accumulates in the cells. RT-PCR showed that the translation of EspB was not blocked. The transcription of escN, which supplies energy for the injection of the effector factor into the host cells, was also not inhibited. EGCG does not suppress the transcription and translation of T3SS constitutive protein in bacterial cells, but it seems to suppress the normal construction or secretion of T3SS. When Luria-Bertani (LB) medium was used to visualize the EGCG-induced inhibition of T3SS, the inhibitory effect disappeared. The inhibition of T3SS was partially canceled when the T3SS inhibitory potency of EGCG was examined by adding yeast extract, which is a component of LB medium, to DMEM. These results suggest that EGCG probably inhibits secretion by suppressing some metabolic mechanisms of T3SS.
Collapse
Affiliation(s)
- Noboru Nakasone
- Department of Bacteriology, Graduate School of Medicine, University of the Ryukyus, Nishihara, Okinawa 903-0215, Japan
| | - Naomi Higa
- Department of Bacteriology, Graduate School of Medicine, University of the Ryukyus, Nishihara, Okinawa 903-0215, Japan
| | - Claudia Toma
- Department of Bacteriology, Graduate School of Medicine, University of the Ryukyus, Nishihara, Okinawa 903-0215, Japan
| | - Yasunori Ogura
- Department of Food Science and Nutrition, Nara Women's University, Nara 630-8506, Japan
| | - Toshihiko Suzuki
- Department of Bacterial Pathogenesis, Infection and Host Response Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Yushima, Bunkyo-ku 1130034, Tokyo, Japan
| | - Tetsu Yamashiro
- Department of Bacteriology, Graduate School of Medicine, University of the Ryukyus, Nishihara, Okinawa 903-0215, Japan
| |
Collapse
|
50
|
Charousová I, Medo J, Hleba L, Javoreková S. Streptomyces globosus DK15 and Streptomyces ederensis ST13 as new producers of factumycin and tetrangomycin antibiotics. Braz J Microbiol 2018; 49:816-822. [PMID: 29705162 PMCID: PMC6175699 DOI: 10.1016/j.bjm.2017.12.007] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2017] [Revised: 11/17/2017] [Accepted: 12/13/2017] [Indexed: 12/08/2022] Open
Abstract
Fifty seven soil-borne actinomycete strains were assessed for the antibiotic production. Two of the most active isolates, designed as Streptomyces ST-13 and DK-15 exhibited a broad range of antimicrobial activity and therefore they were selected for HPLC fractionation against the most suppressed bacteria Staphylococcus aureus (ST-13) and Chromobacterium violaceum (DK-15). LC/MS analysis of extracts showed the presence of polyketides factumycin (DK15) and tetrangomycin (ST13). The taxonomic position of the antibiotic-producing actinomycetes was determined using a polyphasic approach. Phenotypic characterization and 16S rRNA gene sequence analysis of the isolates matched those described for members of the genus Streptomyces. DK-15 strain exhibited the highest 16S rRNA gene sequence similarity to Streptomyces globosus DSM-40815 (T) and Streptomyces toxytricini DSM-40178 (T) and ST-13 strain to Streptomyces ederensis DSM-40741 (T) and Streptomyces phaeochromogenes DSM-40073 (T). For the proper identification, MALDI-TOF/MS profile of whole-cell proteins led to the identification of S. globosus DK-15 (accession number: KX527570) and S. ederensis ST13 (accession number: KX527568). To our knowledge, there is no report about the production of these antibiotics by S.globosus and S. ederensis, thus isolates DK15 and ST13 identified as S. globosus DK-15 and S.ederensis ST-13 can be considered as new sources of these unique antibacterial metabolites.
Collapse
Affiliation(s)
- Ivana Charousová
- Slovak University of Agriculture in Nitra, Faculty of Biotechnology and Food Sciences, Department of Microbiology, Tr. A. Hlinku 2, 949 76, Nitra, Slovak Republic.
| | - Juraj Medo
- Slovak University of Agriculture in Nitra, Faculty of Biotechnology and Food Sciences, Department of Microbiology, Tr. A. Hlinku 2, 949 76, Nitra, Slovak Republic
| | - Lukáš Hleba
- Slovak University of Agriculture in Nitra, Faculty of Biotechnology and Food Sciences, Department of Microbiology, Tr. A. Hlinku 2, 949 76, Nitra, Slovak Republic
| | - Soňa Javoreková
- Slovak University of Agriculture in Nitra, Faculty of Biotechnology and Food Sciences, Department of Microbiology, Tr. A. Hlinku 2, 949 76, Nitra, Slovak Republic
| |
Collapse
|