1
|
Al-Tohamy A, Grove A. Targeting bacterial transcription factors for infection control: opportunities and challenges. Transcription 2025; 16:141-168. [PMID: 38126125 PMCID: PMC11970743 DOI: 10.1080/21541264.2023.2293523] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Revised: 11/13/2023] [Accepted: 12/07/2023] [Indexed: 12/23/2023] Open
Abstract
The rising threat of antibiotic resistance in pathogenic bacteria emphasizes the need for new therapeutic strategies. This review focuses on bacterial transcription factors (TFs), which play crucial roles in bacterial pathogenesis. We discuss the regulatory roles of these factors through examples, and we outline potential therapeutic strategies targeting bacterial TFs. Specifically, we discuss the use of small molecules to interfere with TF function and the development of transcription factor decoys, oligonucleotides that compete with promoters for TF binding. We also cover peptides that target the interaction between the bacterial TF and other factors, such as RNA polymerase, and the targeting of sigma factors. These strategies, while promising, come with challenges, from identifying targets to designing interventions, managing side effects, and accounting for changing bacterial resistance patterns. We also delve into how Artificial Intelligence contributes to these efforts and how it may be exploited in the future, and we touch on the roles of multidisciplinary collaboration and policy to advance this research domain.Abbreviations: AI, artificial intelligence; CNN, convolutional neural networks; DTI: drug-target interaction; HTH, helix-turn-helix; IHF, integration host factor; LTTRs, LysR-type transcriptional regulators; MarR, multiple antibiotic resistance regulator; MRSA, methicillin resistant Staphylococcus aureus; MSA: multiple sequence alignment; NAP, nucleoid-associated protein; PROTACs, proteolysis targeting chimeras; RNAP, RNA polymerase; TF, transcription factor; TFD, transcription factor decoying; TFTRs, TetR-family transcriptional regulators; wHTH, winged helix-turn-helix.
Collapse
Affiliation(s)
- Ahmed Al-Tohamy
- Department of Biological Sciences, Louisiana State University, Baton Rouge, LA, USA
- Department of Cell Biology, Biotechnology Research Institute, National Research Centre, Cairo, Egypt
| | - Anne Grove
- Department of Biological Sciences, Louisiana State University, Baton Rouge, LA, USA
| |
Collapse
|
2
|
Sajeevan A, Ramamurthy T, Solomon AP. Vibrio cholerae virulence and its suppression through the quorum-sensing system. Crit Rev Microbiol 2025; 51:22-43. [PMID: 38441045 DOI: 10.1080/1040841x.2024.2320823] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2023] [Revised: 12/21/2023] [Accepted: 02/10/2024] [Indexed: 03/06/2024]
Abstract
Vibrio cholerae is a cholera-causing pathogen known to instigate severe contagious diarrhea that affects millions globally. Survival of vibrios depend on a combination of multicellular responses and adapt to changes that prevail in the environment. This process is achieved through a strong communication at the cellular level, the process has been recognized as quorum sensing (QS). The severity of infection is highly dependent on the QS of vibrios in the gut milieu. The quorum may exist in a low/high cell density (LCD/HCD) state to exert a positive or negative response to control the regulatory pathogenic networks. The impact of this regulation reflects on the transition of pathogenic V. cholerae from the environment to infect humans and cause outbreaks or epidemics of cholera. In this context, the review portrays various regulatory processes and associated virulent pathways, which maneuver and control LCD and HCD states for their survival in the host. Although several treatment options are existing, promotion of therapeutics by exploiting the virulence network may potentiate ineffective antibiotics to manage cholera. In addition, this approach is also useful in resource-limited settings, where the accessibility to antibiotics or conventional therapeutic options is limited.
Collapse
Affiliation(s)
- Anusree Sajeevan
- Quorum Sensing Laboratory, Centre for Research in Infectious Diseases (CRID), School of Chemical and Biotechnology, SASTRA Deemed to be University, Thanjavur, India
| | - Thandavarayan Ramamurthy
- Division of Bacteriology, ICMR-National Institute of Cholera and Enteric Disease, Kolkata, India
| | - Adline Princy Solomon
- Quorum Sensing Laboratory, Centre for Research in Infectious Diseases (CRID), School of Chemical and Biotechnology, SASTRA Deemed to be University, Thanjavur, India
| |
Collapse
|
3
|
Bhalerao SE, Sen H, Raychaudhuri S. Administration of novobiocin and apomorphine mitigates cholera toxin mediated cellular toxicity: Lessons from cholera toxin yeast model system. PLoS One 2024; 19:e0315052. [PMID: 39637178 PMCID: PMC11620602 DOI: 10.1371/journal.pone.0315052] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Accepted: 11/20/2024] [Indexed: 12/07/2024] Open
Abstract
Cholera is a dreadful disease. The scourge of this deadly disease is still evident in the developing world. Though several therapeutic strategies are in practice to combat and contain the disease, there is still a need for new drugs to control the disease safely and effectively. Keeping in view the concern, we first successfully established an inducible yeast model to express cholera toxin subunit A, and then used this yeast model, to screen a small molecule library against cholera toxin A subunit. Our effort resulted in the discovery of a small molecule, apomorphine (a Parkinson's disease drug) effective in reducing the lethality of toxic subunit in yeast model. In addition, novobiocin, an inhibitor of ADP ribosylation process, a key biochemical event through which cholera toxin exerts its action on host, was also found to rescue yeast cells from cholera toxin A subunit mediated toxicity. Finally, the effects of both molecules were tested on the cholera toxin-treated human gut epithelial cell line HT29, and it was observed that both apomorphine and novobiocin prevented cholera toxin-mediated cellular toxicity on HT29 intestinal epithelial cells.
Collapse
Affiliation(s)
| | - Himanshu Sen
- CSIR-Institute of Microbial Technology, Chandigarh, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| | - Saumya Raychaudhuri
- CSIR-Institute of Microbial Technology, Chandigarh, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| |
Collapse
|
4
|
Su HL, Lai SJ, Tsai KC, Fung KM, Lung TL, Hsu HM, Wu YC, Liu CH, Lai HX, Lin JH, Tseng TS. Structure-guided identification and characterization of potent inhibitors targeting PhoP and MtrA to combat mycobacteria. Comput Struct Biotechnol J 2024; 23:1477-1488. [PMID: 38623562 PMCID: PMC11016868 DOI: 10.1016/j.csbj.2024.04.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2023] [Revised: 04/01/2024] [Accepted: 04/01/2024] [Indexed: 04/17/2024] Open
Abstract
Mycobacteria are causative agents of tuberculosis (TB), which is a global health concern. Drug-resistant TB strains are rapidly emerging, thereby necessitating the urgent development of new drugs. Two-component signal transduction systems (TCSs) are signaling pathways involved in the regulation of various bacterial behaviors and responses to environmental stimuli. Applying specific inhibitors of TCSs can disrupt bacterial signaling, growth, and virulence, and can help combat drug-resistant TB. We conducted a comprehensive pharmacophore-based inhibitor screening and biochemical and biophysical examinations to identify, characterize, and validate potential inhibitors targeting the response regulators PhoP and MtrA of mycobacteria. The constructed pharmacophore model Phar-PR-n4 identified effective inhibitors of formation of the PhoP-DNA complex: ST132 (IC50 = 29 ± 1.6 µM) and ST166 (IC50 = 18 ± 1.3 µM). ST166 (KD = 18.4 ± 4.3 μM) and ST132 (KD = 14.5 ± 0.1 μM) strongly targeted PhoP in a slow-on, slow-off manner. The inhibitory potency and binding affinity of ST166 and ST132 for MtrAC were comparable to those of PhoP. Structural analyses and molecular dynamics simulations revealed that ST166 and ST132 mainly interact with the α8-helix and C-terminal β-hairpin of PhoP, with functionally essential residue hotspots for structure-based inhibitor optimization. Moreover, ST166 has in vitro antibacterial activity against Macrobacterium marinum. Thus, ST166, with its characteristic 1,2,5,6-tetrathiocane and terminal sulphonic groups, has excellent potential as a candidate for the development of novel antimicrobial agents to combat pathogenic mycobacteria.
Collapse
Affiliation(s)
- Han-Li Su
- Department of Emergency Medicine, Ditmanson Medical Foundation Chia-Yi Christian Hospital, Chiayi City 600, Taiwan
| | - Shu-Jung Lai
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung, Taiwan
- Research Center for Cancer Biology, China Medical University, Taichung, Taiwan
| | - Keng-Chang Tsai
- National Research Institute of Chinese Medicine, Ministry of Health and Welfare, Taipei, Taiwan
- Ph.D. Program in Medical Biotechnology, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan
| | - Kit-Man Fung
- Biomedical Translation Research Center (BioTReC), Academia Sinica, Taipei 11529, Taiwan
| | - Tse-Lin Lung
- Institute of Molecular Biology, National Chung Hsing University, Taichung,Taiwan
| | - Hsing-Mien Hsu
- Institute of Molecular Biology, National Chung Hsing University, Taichung,Taiwan
| | - Yi-Chen Wu
- Institute of Molecular Biology, National Chung Hsing University, Taichung,Taiwan
| | - Ching-Hui Liu
- Institute of Molecular Biology, National Chung Hsing University, Taichung,Taiwan
| | - Hui-Xiang Lai
- Institute of Molecular Biology, National Chung Hsing University, Taichung,Taiwan
| | - Jiun-Han Lin
- Department of Industrial Technology, Ministry of Economic Affairs, Taipei, Taiwan
- Food Industry Research and Development Institute, Hsinchu City, Taiwan
| | - Tien-Sheng Tseng
- Institute of Molecular Biology, National Chung Hsing University, Taichung,Taiwan
| |
Collapse
|
5
|
Lee D, Joo J, Choi H, Son S, Bae J, Kim DW, Kim EJ. Variations in the Antivirulence Effects of Fatty Acids and Virstatin against Vibrio cholerae Strains. J Microbiol Biotechnol 2024; 34:1757-1768. [PMID: 39187456 PMCID: PMC11485679 DOI: 10.4014/jmb.2405.05002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Revised: 06/27/2024] [Accepted: 06/28/2024] [Indexed: 08/28/2024]
Abstract
The expression of two major virulence factors of Vibrio cholerae, cholera toxin (CT) and toxin co-regulated pilus (TCP), is induced by environmental stimuli through a cascade of interactions among regulatory proteins known as the ToxR regulon when the bacteria reach the human small intestine. ToxT is produced via the ToxR regulon and acts as the direct transcriptional activator of CT (ctxAB), TCP (tcp gene cluster), and other virulence genes. Unsaturated fatty acids (UFAs) and several small-molecule inhibitors of ToxT have been developed as antivirulence agents against V. cholerae. This study reports the inhibitory effects of fatty acids and virstatin (a small-molecule inhibitor of ToxT) on the transcriptional activation functions of ToxT in isogenic derivatives of V. cholerae strains containing various toxT alleles. The fatty acids and virstatin had discrete effects depending on the ToxT allele (different by 2 amino acids), V. cholerae strain, and culture conditions, indicating that V. cholerae strains could overcome the effects of UFAs and small-molecule inhibitors by acquiring point mutations in toxT. Our results suggest that small-molecule inhibitors should be examined thoroughly against various V. cholerae strains and toxT alleles during development.
Collapse
Affiliation(s)
- Donghyun Lee
- Department of Pharmacy, College of Pharmacy, Hanyang University, Ansan 15588, Republic of Korea
- Institute of Pharmacological Research, Hanyang University, Ansan 15588, Republic of Korea
| | - Jayun Joo
- Department of Pharmacy, College of Pharmacy, Hanyang University, Ansan 15588, Republic of Korea
- Institute of Pharmacological Research, Hanyang University, Ansan 15588, Republic of Korea
| | - Hunseok Choi
- Department of Pharmacy, College of Pharmacy, Hanyang University, Ansan 15588, Republic of Korea
- Institute of Pharmacological Research, Hanyang University, Ansan 15588, Republic of Korea
| | - Seonghyeon Son
- Department of Pharmacy, College of Pharmacy, Hanyang University, Ansan 15588, Republic of Korea
- Institute of Pharmacological Research, Hanyang University, Ansan 15588, Republic of Korea
| | - Jonghyun Bae
- Department of Pharmacy, College of Pharmacy, Hanyang University, Ansan 15588, Republic of Korea
- Institute of Pharmacological Research, Hanyang University, Ansan 15588, Republic of Korea
| | - Dong Wook Kim
- Department of Pharmacy, College of Pharmacy, Hanyang University, Ansan 15588, Republic of Korea
- Institute of Pharmacological Research, Hanyang University, Ansan 15588, Republic of Korea
| | - Eun Jin Kim
- Department of Pharmacy, College of Pharmacy, Hanyang University, Ansan 15588, Republic of Korea
- Institute of Pharmacological Research, Hanyang University, Ansan 15588, Republic of Korea
| |
Collapse
|
6
|
Zhang T, Nickerson R, Zhang W, Peng X, Shang Y, Zhou Y, Luo Q, Wen G, Cheng Z. The impacts of animal agriculture on One Health-Bacterial zoonosis, antimicrobial resistance, and beyond. One Health 2024; 18:100748. [PMID: 38774301 PMCID: PMC11107239 DOI: 10.1016/j.onehlt.2024.100748] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Accepted: 05/02/2024] [Indexed: 05/24/2024] Open
Abstract
The industrialization of animal agriculture has undoubtedly contributed to the improvement of human well-being by increasing the efficiency of food animal production. At the same time, it has also drastically impacted the natural environment and human society. The One Health initiative emphasizes the interdependency of the health of ecosystems, animals, and humans. In this paper, we discuss some of the most profound consequences of animal agriculture practices from a One Health perspective. More specifically, we focus on impacts to host-microbe interactions by elaborating on how modern animal agriculture affects zoonotic infections, specifically those of bacterial origin, and the concomitant emergence of antimicrobial resistance (AMR). A key question underlying these deeply interconnected issues is how to better prevent, monitor, and manage infections in animal agriculture. To address this, we outline approaches to mitigate the impacts of agricultural bacterial zoonoses and AMR, including the development of novel treatments as well as non-drug approaches comprising integrated surveillance programs and policy and education regarding agricultural practices and antimicrobial stewardship. Finally, we touch upon additional major environmental and health factors impacted by animal agriculture within the One Health context, including animal welfare, food security, food safety, and climate change. Charting how these issues are interwoven to comprise the complex web of animal agriculture's broad impacts on One Health will allow for the development of concerted, multidisciplinary interventions which are truly necessary to tackle these issues from a One Health perspective.
Collapse
Affiliation(s)
- Tengfei Zhang
- Key Laboratory of Prevention and Control Agents for Animal Bacteriosis (Ministry of Agriculture and Rural Affairs), Hubei Provincial Key Laboratory of Animal Pathogenic Microbiology, Institute of Animal Husbandry and Veterinary, Hubei Academy of Agricultural Sciences, Wuhan 430064, China
| | - Rhea Nickerson
- Department of Microbiology and Immunology, Dalhousie University, Halifax, NS, Canada
| | - Wenting Zhang
- Key Laboratory of Prevention and Control Agents for Animal Bacteriosis (Ministry of Agriculture and Rural Affairs), Hubei Provincial Key Laboratory of Animal Pathogenic Microbiology, Institute of Animal Husbandry and Veterinary, Hubei Academy of Agricultural Sciences, Wuhan 430064, China
| | - Xitian Peng
- Institute of Quality Standard and Testing Technology for Agro-Products, Hubei Academy of Agricultural Sciences, Wuhan 430064, Hubei, China
- Hubei Key Laboratory of Nutritional Quality and Safety of Agro-products, Wuhan 430064, Hubei, China
- Ministry of Agriculture and Rural Affairs Laboratory of Quality and Safe Risk Assessment for Agro-products (Wuhan), Wuhan 430064, Hubei, China
| | - Yu Shang
- Key Laboratory of Prevention and Control Agents for Animal Bacteriosis (Ministry of Agriculture and Rural Affairs), Hubei Provincial Key Laboratory of Animal Pathogenic Microbiology, Institute of Animal Husbandry and Veterinary, Hubei Academy of Agricultural Sciences, Wuhan 430064, China
| | - Youxiang Zhou
- Institute of Quality Standard and Testing Technology for Agro-Products, Hubei Academy of Agricultural Sciences, Wuhan 430064, Hubei, China
- Hubei Key Laboratory of Nutritional Quality and Safety of Agro-products, Wuhan 430064, Hubei, China
- Ministry of Agriculture and Rural Affairs Laboratory of Quality and Safe Risk Assessment for Agro-products (Wuhan), Wuhan 430064, Hubei, China
| | - Qingping Luo
- Key Laboratory of Prevention and Control Agents for Animal Bacteriosis (Ministry of Agriculture and Rural Affairs), Hubei Provincial Key Laboratory of Animal Pathogenic Microbiology, Institute of Animal Husbandry and Veterinary, Hubei Academy of Agricultural Sciences, Wuhan 430064, China
- Hubei Hongshan Laboratory, Wuhan 430064, China
| | - Guoyuan Wen
- Key Laboratory of Prevention and Control Agents for Animal Bacteriosis (Ministry of Agriculture and Rural Affairs), Hubei Provincial Key Laboratory of Animal Pathogenic Microbiology, Institute of Animal Husbandry and Veterinary, Hubei Academy of Agricultural Sciences, Wuhan 430064, China
| | - Zhenyu Cheng
- Department of Microbiology and Immunology, Dalhousie University, Halifax, NS, Canada
| |
Collapse
|
7
|
Joiner JD, Steinchen W, Mozer N, Kronenberger T, Bange G, Poso A, Wagner S, Hartmann MD. HilE represses the activity of the Salmonella virulence regulator HilD via a mechanism distinct from that of intestinal long-chain fatty acids. J Biol Chem 2023; 299:105387. [PMID: 37890783 PMCID: PMC10696396 DOI: 10.1016/j.jbc.2023.105387] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2023] [Revised: 10/11/2023] [Accepted: 10/16/2023] [Indexed: 10/29/2023] Open
Abstract
The expression of virulence factors essential for the invasion of host cells by Salmonella enterica is tightly controlled by a network of transcription regulators. The AraC/XylS transcription factor HilD is the main integration point of environmental signals into this regulatory network, with many factors affecting HilD activity. Long-chain fatty acids, which are highly abundant throughout the host intestine, directly bind to and repress HilD, acting as environmental cues to coordinate virulence gene expression. The regulatory protein HilE also negatively regulates HilD activity, through a protein-protein interaction. Both of these regulators inhibit HilD dimerization, preventing HilD from binding to target DNA. We investigated the structural basis of these mechanisms of HilD repression. Long-chain fatty acids bind to a conserved pocket in HilD, in a comparable manner to that reported for other AraC/XylS regulators, whereas HilE forms a stable heterodimer with HilD by binding to the HilD dimerization interface. Our results highlight two distinct, mutually exclusive mechanisms by which HilD activity is repressed, which could be exploited for the development of new antivirulence leads.
Collapse
Affiliation(s)
- Joe D Joiner
- Department of Protein Evolution, Max Planck Institute for Biology Tübingen, Tübingen, Germany
| | - Wieland Steinchen
- Center for Synthetic Microbiology, Philipps University of Marburg, Marburg, Germany; Department of Chemistry, Philipps University of Marburg, Marburg, Germany
| | - Nick Mozer
- Department of Protein Evolution, Max Planck Institute for Biology Tübingen, Tübingen, Germany
| | - Thales Kronenberger
- Department of Internal Medicine VIII, University Hospital Tübingen, Tübingen, Germany; Institute of Pharmacy, Pharmaceutical/Medicinal Chemistry and Tübingen Center for Academic Drug Discovery & Development (TüCAD2), Eberhard Karls University Tübingen, Tübingen, Germany; School of Pharmacy, Faculty of Health Sciences, University of Eastern Finland, Kuopio, Finland; Excellence Cluster "Controlling Microbes to Fight Infections" (CMFI), Tübingen, Germany
| | - Gert Bange
- Center for Synthetic Microbiology, Philipps University of Marburg, Marburg, Germany; Department of Chemistry, Philipps University of Marburg, Marburg, Germany
| | - Antti Poso
- Department of Internal Medicine VIII, University Hospital Tübingen, Tübingen, Germany; Institute of Pharmacy, Pharmaceutical/Medicinal Chemistry and Tübingen Center for Academic Drug Discovery & Development (TüCAD2), Eberhard Karls University Tübingen, Tübingen, Germany; School of Pharmacy, Faculty of Health Sciences, University of Eastern Finland, Kuopio, Finland
| | - Samuel Wagner
- Excellence Cluster "Controlling Microbes to Fight Infections" (CMFI), Tübingen, Germany; Interfaculty Institute of Microbiology and Infection Medicine (IMIT), University of Tübingen, Tübingen, Germany; Partner-site Tübingen, German Center for Infection Research (DZIF), Tübingen, Germany
| | - Marcus D Hartmann
- Department of Protein Evolution, Max Planck Institute for Biology Tübingen, Tübingen, Germany; Interfaculty Institute of Biochemistry, University of Tübingen, Tübingen, Germany.
| |
Collapse
|
8
|
Bertschi A, Wang P, Galvan S, Teixeira AP, Fussenegger M. Combinatorial protein dimerization enables precise multi-input synthetic computations. Nat Chem Biol 2023; 19:767-777. [PMID: 36894721 DOI: 10.1038/s41589-023-01281-x] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Accepted: 02/01/2023] [Indexed: 03/11/2023]
Abstract
Bacterial transcription factors (TFs) with helix-turn-helix (HTH) DNA-binding domains have been widely explored to build orthogonal transcriptional regulation systems in mammalian cells. Here we capitalize on the modular structure of these proteins to build a framework for multi-input logic gates relying on serial combinations of inducible protein-protein interactions. We found that for some TFs, their HTH domain alone is sufficient for DNA binding. By fusing the HTH domain to TFs, we established dimerization dependent rather than DNA-binding-dependent activation. This enabled us to convert gene switches from OFF-type into more widely applicable ON-type systems and to create mammalian gene switches responsive to new inducers. By combining both OFF and ON modes of action, we built a compact, high-performance bandpass filter. Furthermore, we were able to show cytosolic and extracellular dimerization. Cascading up to five pairwise fusion proteins yielded robust multi-input AND logic gates. Combinations of different pairwise fusion proteins afforded a variety of 4-input 1-output AND and OR logic gate configurations.
Collapse
Affiliation(s)
- Adrian Bertschi
- Department of Biosystems Science and Engineering, ETH Zurich, Basel, Switzerland
| | - Pengli Wang
- Department of Biosystems Science and Engineering, ETH Zurich, Basel, Switzerland
| | - Silvia Galvan
- Department of Biosystems Science and Engineering, ETH Zurich, Basel, Switzerland
| | - Ana Palma Teixeira
- Department of Biosystems Science and Engineering, ETH Zurich, Basel, Switzerland
| | - Martin Fussenegger
- Department of Biosystems Science and Engineering, ETH Zurich, Basel, Switzerland. .,University of Basel, Faculty of Science, Basel, Switzerland.
| |
Collapse
|
9
|
Midgett CR, Kull FJ. Structural Insights into Regulation of Vibrio Virulence Gene Networks. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2023; 1404:269-294. [PMID: 36792881 DOI: 10.1007/978-3-031-22997-8_14] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/17/2023]
Abstract
One of the best studied aspects of pathogenic Vibrios are the virulence cascades that lead to the production of virulence factors and, ultimately, clinical outcomes. In this chapter, we will examine the regulation of Vibrio virulence gene networks from a structural and biochemical perspective. We will discuss the recent research into the numerous proteins that contribute to regulating virulence in Vibrio spp such as quorum sensing regulator HapR, the transcription factors AphA and AphB, or the virulence regulators ToxR and ToxT. We highlight how insights gained from these studies are already illuminating the basic molecular mechanisms by which the virulence cascade of pathogenic Vibrios unfold and contend that understanding how protein interactions contribute to the host-pathogen communications will enable the development of new antivirulence compounds that can effectively target these pathogens.
Collapse
Affiliation(s)
| | - F Jon Kull
- Chemistry Department, Dartmouth College, Hanover, NH, USA.
| |
Collapse
|
10
|
Yang X, Stein KR, Hang HC. Anti-infective bile acids bind and inactivate a Salmonella virulence regulator. Nat Chem Biol 2023; 19:91-100. [PMID: 36175659 PMCID: PMC9805502 DOI: 10.1038/s41589-022-01122-3] [Citation(s) in RCA: 30] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2021] [Accepted: 07/26/2022] [Indexed: 01/03/2023]
Abstract
Bile acids are prominent host and microbiota metabolites that modulate host immunity and microbial pathogenesis. However, the mechanisms by which bile acids suppress microbial virulence are not clear. To identify the direct protein targets of bile acids in bacterial pathogens, we performed activity-guided chemical proteomic studies. In Salmonella enterica serovar Typhimurium, chenodeoxycholic acid (CDCA) most effectively inhibited the expression of virulence genes and invasion of epithelial cells and interacted with many proteins. Notably, we discovered that CDCA can directly bind and inhibit the function of HilD, an important transcriptional regulator of S. Typhimurium virulence and pathogenesis. Our characterization of bile acid-resistant HilD mutants in vitro and in S. Typhimurium infection models suggests that HilD is one of the key protein targets of anti-infective bile acids. This study highlights the utility of chemical proteomics to identify the direct protein targets of microbiota metabolites for mechanistic studies in bacterial pathogens.
Collapse
Affiliation(s)
- Xinglin Yang
- Department of Immunology and Microbiology, Scripps Research, La Jolla, CA, USA
| | - Kathryn R Stein
- Department of Immunology and Microbiology, Scripps Research, La Jolla, CA, USA
| | - Howard C Hang
- Department of Immunology and Microbiology, Scripps Research, La Jolla, CA, USA.
- Department of Chemistry, Scripps Research, La Jolla, CA, USA.
| |
Collapse
|
11
|
Monteiro KLC, Silva ON, Dos Santos Nascimento IJ, Mendonça Júnior FJB, Aquino PGV, da Silva-Júnior EF, de Aquino TM. Medicinal Chemistry of Inhibitors Targeting Resistant Bacteria. Curr Top Med Chem 2022; 22:1983-2028. [PMID: 35319372 DOI: 10.2174/1568026622666220321124452] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2021] [Revised: 02/01/2022] [Accepted: 02/13/2022] [Indexed: 12/15/2022]
Abstract
The discovery of antibiotics was a revolutionary feat that provided countless health benefits. The identification of penicillin by Alexander Fleming initiated the era of antibiotics, represented by constant discoveries that enabled effective treatments for the different classes of diseases caused by bacteria. However, the indiscriminate use of these drugs allowed the emergence of resistance mechanisms of these microorganisms against the available drugs. In addition, the constant discoveries in the 20th century generated a shortage of new molecules, worrying health agencies and professionals about the appearance of multidrug-resistant strains against available drugs. In this context, the advances of recent years in molecular biology and microbiology have allowed new perspectives in drug design and development, using the findings related to the mechanisms of bacterial resistance to generate new drugs that are not affected by such mechanisms and supply new molecules to be used to treat resistant bacterial infections. Besides, a promising strategy against bacterial resistance is the combination of drugs through adjuvants, providing new expectations in designing new antibiotics and new antimicrobial therapies. Thus, this manuscript will address the main mechanisms of bacterial resistance under the understanding of medicinal chemistry, showing the main active compounds against efflux mechanisms, and also the application of the use of drug delivery systems, and finally, the main potential natural products as adjuvants or with promising activity against resistant strains.
Collapse
Affiliation(s)
- Kadja Luana Chagas Monteiro
- Research Group on Therapeutic Strategies - GPET, Laboratory of Synthesis and Research in Medicinal Chemistry - LSPMED, Institute of Chemistry and Biotechnology, Federal University of Alagoas, 57072-970, Maceió, Alagoas, Brazil
| | - Osmar Nascimento Silva
- Faculty of Pharmacy, University Center of Anápolis, Unievangélica, 75083-515, Anápolis, Goiás, Brazil
| | - Igor José Dos Santos Nascimento
- Research Group on Therapeutic Strategies - GPET, Laboratory of Synthesis and Research in Medicinal Chemistry - LSPMED, Institute of Chemistry and Biotechnology, Federal University of Alagoas, 57072-970, Maceió, Alagoas, Brazil
| | | | | | - Edeildo Ferreira da Silva-Júnior
- Laboratory of Medicinal Chemistry, Institute of Pharmaceutical Sciences, Federal University of Alagoas, 57072-970, Maceió, Alagoas, Brazil
| | - Thiago Mendonça de Aquino
- Research Group on Therapeutic Strategies - GPET, Laboratory of Synthesis and Research in Medicinal Chemistry - LSPMED, Institute of Chemistry and Biotechnology, Federal University of Alagoas, 57072-970, Maceió, Alagoas, Brazil
| |
Collapse
|
12
|
Mitchell MK, Ellermann M. Long Chain Fatty Acids and Virulence Repression in Intestinal Bacterial Pathogens. Front Cell Infect Microbiol 2022; 12:928503. [PMID: 35782143 PMCID: PMC9247172 DOI: 10.3389/fcimb.2022.928503] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Accepted: 05/23/2022] [Indexed: 11/13/2022] Open
Abstract
When bacterial pathogens enter the gut, they encounter a complex milieu of signaling molecules and metabolites produced by host and microbial cells or derived from external sources such as the diet. This metabolomic landscape varies throughout the gut, thus establishing a biogeographical gradient of signals that may be sensed by pathogens and resident bacteria alike. Enteric bacterial pathogens have evolved elaborate mechanisms to appropriately regulate their virulence programs, which involves sensing and responding to many of these gut metabolites to facilitate successful gut colonization. Long chain fatty acids (LCFAs) represent major constituents of the gut metabolome that can impact bacterial functions. LCFAs serve as important nutrient sources for all cellular organisms and can function as signaling molecules that regulate bacterial metabolism, physiology, and behaviors. Moreover, in several enteric pathogens, including Salmonella enterica, Listeria monocytogenes, Vibrio cholerae, and enterohemorrhagic Escherichia coli, LCFA sensing results in the transcriptional repression of virulence through two general mechanisms. First, some LCFAs function as allosteric inhibitors that decrease the DNA binding affinities of transcriptional activators of virulence genes. Second, some LCFAs also modulate the activation of histidine kinase receptors, which alters downstream intracellular signaling networks to repress virulence. This mini-review will summarize recent studies that have investigated the molecular mechanisms by which different LCFA derivatives modulate the virulence of enteric pathogens, while also highlighting important gaps in the field regarding the roles of LCFAs as determinants of infection and disease.
Collapse
|
13
|
Biswas Q, Purohit A, Kumar A, Rakshit D, Maiti D, Das B, Bhadra RK. Genetic and mutational analysis of virulence traits and their modulation in an environmental toxigenic Vibrio cholerae non-O1/non-O139 strain, VCE232. MICROBIOLOGY (READING, ENGLAND) 2022; 168. [PMID: 35113781 DOI: 10.1099/mic.0.001135] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Vibrio cholerae O1 and O139 isolates deploy cholera toxin (CT) and toxin-coregulated pilus (TCP) to cause the diarrhoeal disease cholera. The ctxAB and tcpA genes encoding CT and TCP are part of two acquired genetic elements, the CTX phage and Vibrio pathogenicity island-1 (VPI-1), respectively. ToxR and ToxT proteins are the key regulators of virulence genes of V. cholerae O1 and O139. V. cholerae isolates belonging to serogroups other than O1/O139, called non-O1/non-O139, are usually devoid of virulence-related elements and are non-pathogenic. Here, we have analysed the available whole genome sequence of an environmental toxigenic V. cholerae non-O1/non-O139 strain, VCE232, carrying the CTX phage and VPI-1. Extensive bioinformatics and phylogenetic analyses indicated high similarity of the VCE232 genome sequence with the genome of V. cholerae O1 strains, including organization of the VPI-1 locus, ctxAB, tcpA and toxT genes, and promoters. We established that the VCE232 strain produces an optimal amount of CT at 30 °C under AKI conditions. To investigate the role of ToxT and ToxR in the regulation of virulence factors, we constructed ΔtoxT, ΔtoxR and ΔtoxTΔtoxR deletion mutants of VCE232. Extensive genetic analyses of these mutants indicated that the toxT and toxR genes of VCE232 are crucial for CT and TCP production. However, unlike O1 isolates, the presence of either toxT or toxR gene is sufficient for optimal CT production in VCE232. In addition, the VCE232 ΔtoxR mutant showed differential regulation of the major outer membrane proteins, OmpT and OmpU. This is the first attempt to explore the regulation of expression of major virulence genes and regulators in an environmental toxigenic V. cholerae non-O1/non-O139 strain.
Collapse
Affiliation(s)
- Quoelee Biswas
- Infectious Diseases and Immunology Division, CSIR - Indian Institute of Chemical Biology, Kolkata 700 032, India
| | - Ayushi Purohit
- Translational Health Science and Technology Institute, NCR Biotech Science Cluster, Faridabad 121 001, India
| | - Ashok Kumar
- Translational Health Science and Technology Institute, NCR Biotech Science Cluster, Faridabad 121 001, India
- School of Life Sciences, Manipal Academy of Higher Education, Manipal 576 104, Karnataka, India
| | - Dipayan Rakshit
- Infectious Diseases and Immunology Division, CSIR - Indian Institute of Chemical Biology, Kolkata 700 032, India
| | - Diganta Maiti
- Infectious Diseases and Immunology Division, CSIR - Indian Institute of Chemical Biology, Kolkata 700 032, India
| | - Bhabatosh Das
- Translational Health Science and Technology Institute, NCR Biotech Science Cluster, Faridabad 121 001, India
- School of Life Sciences, Manipal Academy of Higher Education, Manipal 576 104, Karnataka, India
| | - Rupak K Bhadra
- Infectious Diseases and Immunology Division, CSIR - Indian Institute of Chemical Biology, Kolkata 700 032, India
| |
Collapse
|
14
|
Roncarati D, Scarlato V, Vannini A. Targeting of Regulators as a Promising Approach in the Search for Novel Antimicrobial Agents. Microorganisms 2022; 10:microorganisms10010185. [PMID: 35056634 PMCID: PMC8777881 DOI: 10.3390/microorganisms10010185] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Revised: 01/12/2022] [Accepted: 01/12/2022] [Indexed: 02/01/2023] Open
Abstract
Since the discovery of penicillin in the first half of the last century, antibiotics have become the pillars of modern medicine for fighting bacterial infections. However, pathogens resistant to antibiotic treatment have increased in recent decades, and efforts to discover new antibiotics have decreased. As a result, it is becoming increasingly difficult to treat bacterial infections successfully, and we look forward to more significant efforts from both governments and the scientific community to research new antibacterial drugs. This perspective article highlights the high potential of bacterial transcriptional and posttranscriptional regulators as targets for developing new drugs. We highlight some recent advances in the search for new compounds that inhibit their biological activity and, as such, appear very promising for treating bacterial infections.
Collapse
Affiliation(s)
- Davide Roncarati
- Department of Pharmacy and Biotechnology (FaBiT), University of Bologna, 40126 Bologna, Italy
- Correspondence: (D.R.); (V.S.); (A.V.)
| | - Vincenzo Scarlato
- Department of Pharmacy and Biotechnology (FaBiT), University of Bologna, 40126 Bologna, Italy
- Correspondence: (D.R.); (V.S.); (A.V.)
| | - Andrea Vannini
- Department of Experimental, Diagnostic and Specialty Medicine, University of Bologna, 40126 Bologna, Italy
- Correspondence: (D.R.); (V.S.); (A.V.)
| |
Collapse
|
15
|
Streptococcus pyogenes ("Group A Streptococcus"), a Highly Adapted Human Pathogen-Potential Implications of Its Virulence Regulation for Epidemiology and Disease Management. Pathogens 2021; 10:pathogens10060776. [PMID: 34205500 PMCID: PMC8234341 DOI: 10.3390/pathogens10060776] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Revised: 06/02/2021] [Accepted: 06/17/2021] [Indexed: 11/16/2022] Open
Abstract
Streptococcus pyogenes (group A streptococci; GAS) is an exclusively human pathogen. It causes a variety of suppurative and non-suppurative diseases in people of all ages worldwide. Not all can be successfully treated with antibiotics. A licensed vaccine, in spite of its global importance, is not yet available. GAS express an arsenal of virulence factors responsible for pathological immune reactions. The transcription of all these virulence factors is under the control of three types of virulence-related regulators: (i) two-component systems (TCS), (ii) stand-alone regulators, and (iii) non-coding RNAs. This review summarizes major TCS and stand-alone transcriptional regulatory systems, which are directly associated with virulence control. It is suggested that this treasure of knowledge on the genetics of virulence regulation should be better harnessed for new therapies and prevention methods for GAS infections, thereby changing its global epidemiology for the better.
Collapse
|
16
|
Jana SK, Gucchait A, Paul S, Saha T, Acharya S, Hoque KM, Misra AK, Chatterjee BK, Chatterjee T, Chakrabarti P. Virstatin-Conjugated Gold Nanoparticle with Enhanced Antimicrobial Activity against the Vibrio cholerae El Tor Biotype. ACS APPLIED BIO MATERIALS 2021; 4:3089-3100. [DOI: 10.1021/acsabm.0c01483] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Affiliation(s)
- Swapan Kumar Jana
- Department of Biochemistry, Bose Institute, P1/12 CIT Scheme VIIM, Kolkata 700054, India
| | - Arin Gucchait
- Division of Molecular Medicine, Bose Institute, P1/12 CIT Scheme VIIM, Kolkata 700054, India
| | - Susmita Paul
- School of Applied & Interdisciplinary Sciences, Indian Association for the Cultivation of Science, Jadavpur, Kolkata 700032, India
| | - Tultul Saha
- Division of Molecular Pathophysiology, National Institute of Cholera & Enteric Diseases, P-33 CIT Road, Scheme XM, Beliaghata, Kolkata 700010, India
| | - Somobrata Acharya
- School of Applied & Interdisciplinary Sciences, Indian Association for the Cultivation of Science, Jadavpur, Kolkata 700032, India
| | - Kazi Mirajul Hoque
- Division of Molecular Pathophysiology, National Institute of Cholera & Enteric Diseases, P-33 CIT Road, Scheme XM, Beliaghata, Kolkata 700010, India
| | - Anup Kumar Misra
- Division of Molecular Medicine, Bose Institute, P1/12 CIT Scheme VIIM, Kolkata 700054, India
| | - Barun K. Chatterjee
- Department of Physics, Bose Institute, 93/1 A.P.C. Road, Kolkata 700009, India
| | - Tanaya Chatterjee
- Department of Biochemistry, Bose Institute, P1/12 CIT Scheme VIIM, Kolkata 700054, India
| | - Pinak Chakrabarti
- Department of Biochemistry, Bose Institute, P1/12 CIT Scheme VIIM, Kolkata 700054, India
| |
Collapse
|
17
|
Cortés-Avalos D, Martínez-Pérez N, Ortiz-Moncada MA, Juárez-González A, Baños-Vargas AA, Estrada-de Los Santos P, Pérez-Rueda E, Ibarra JA. An update of the unceasingly growing and diverse AraC/XylS family of transcriptional activators. FEMS Microbiol Rev 2021; 45:6219864. [PMID: 33837749 DOI: 10.1093/femsre/fuab020] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Accepted: 03/31/2021] [Indexed: 01/09/2023] Open
Abstract
Transcriptional factors play an important role in gene regulation in all organisms, especially in Bacteria. Here special emphasis is placed in the AraC/XylS family of transcriptional regulators. This is one of the most abundant as many predicted members have been identified and more members are added because more bacterial genomes are sequenced. Given the way more experimental evidence has mounded in the past decades, we decided to update the information about this captivating family of proteins. Using bioinformatics tools on all the data available for experimentally characterized members of this family, we found that many members that display a similar functional classification can be clustered together and in some cases they have a similar regulatory scheme. A proposal for grouping these proteins is also discussed. Additionally, an analysis of surveyed proteins in bacterial genomes is presented. Altogether, the current review presents a panoramic view into this family and we hope it helps to stimulate future research in the field.
Collapse
Affiliation(s)
- Daniel Cortés-Avalos
- Laboratorio de Genética Microbiana, Departamento de Microbiología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Ciudad de México, México
| | - Noemy Martínez-Pérez
- Instituto de Investigaciones en Matemáticas Aplicadas y en Sistemas, Universidad Nacional Autónoma de México, Unidad Académica Yucatán, Mérida, Yucatán, México
| | - Mario A Ortiz-Moncada
- Laboratorio de Genética Microbiana, Departamento de Microbiología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Ciudad de México, México
| | - Aylin Juárez-González
- Laboratorio de Genética Microbiana, Departamento de Microbiología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Ciudad de México, México
| | - Arturo A Baños-Vargas
- Laboratorio de Genética Microbiana, Departamento de Microbiología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Ciudad de México, México
| | - Paulina Estrada-de Los Santos
- Laboratorio de Biotecnología Microbiana, Departamento de Microbiología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Ciudad de México, México
| | - Ernesto Pérez-Rueda
- Instituto de Investigaciones en Matemáticas Aplicadas y en Sistemas, Universidad Nacional Autónoma de México, Unidad Académica Yucatán, Mérida, Yucatán, México.,Facultad de Ciencias, Centro de Genómica y Bioinformática, Universidad Mayor, Santiago, Chile
| | - J Antonio Ibarra
- Laboratorio de Genética Microbiana, Departamento de Microbiología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Ciudad de México, México
| |
Collapse
|
18
|
Das S, Chourashi R, Mukherjee P, Kundu S, Koley H, Dutta M, Mukhopadhyay AK, Okamoto K, Chatterjee NS. Inhibition of growth and virulence of Vibrio cholerae by carvacrol, an essential oil component of Origanum spp. J Appl Microbiol 2021; 131:1147-1161. [PMID: 33544959 DOI: 10.1111/jam.15022] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Revised: 01/14/2021] [Accepted: 01/28/2021] [Indexed: 01/11/2023]
Abstract
AIMS In the age where bacterial resistance to conventional antibiotics is increasing at an alarming rate, the use of the traditional plant, herb extracts or other bioactive constituents is gradually becoming popular as an anti-virulence agent to treat pathogenic diseases. Carvacrol, a major essential oil fraction of Oregano, possesses a wide range of bioactivities. Therefore, we aimed to study the effect of sub-inhibitory concentrations of carvacrol on major virulence traits of Vibrio cholerae. METHODS AND RESULTS We have used in vitro as well as ex vivo models to access the anti-pathogenic role of carvacrol. We found that the sub-inhibitory concentration of carvacrol significantly repressed bacterial mucin penetrating ability. Carvacrol also reduced the adherence and fluid accumulation in the rabbit ileal loop model. Reduction in virulence is associated with the downregulated expression of tcpA, ctxB, hlyA and toxT. Furthermore, carvacrol inhibits flagellar synthesis by downregulating the expression of flrC and most of the class III genes. CONCLUSIONS Carvacrol exhibited anti-virulence activity against V. cholerae, which involved many events including the inhibition of mucin penetration, adhesion, reduced expression of virulence-associated genes culminating in reduced fluid accumulation. SIGNIFICANCE AND IMPACT OF THE STUDY These findings indicate that carvacrol possesses inhibitory activity against V. cholerae pathogenesis and might be considered as a potential bio-active therapeutic alternative to combat cholera.
Collapse
Affiliation(s)
- S Das
- Division of Biochemistry, ICMR-National Institute of Cholera and Enteric Diseases, Kolkata, India
| | - R Chourashi
- Division of Biochemistry, ICMR-National Institute of Cholera and Enteric Diseases, Kolkata, India
| | - P Mukherjee
- Division of Bacteriology, ICMR-National Institute of Cholera and Enteric Diseases, Kolkata, India
| | - S Kundu
- Division of Biochemistry, ICMR-National Institute of Cholera and Enteric Diseases, Kolkata, India
| | - H Koley
- Division of Bacteriology, ICMR-National Institute of Cholera and Enteric Diseases, Kolkata, India
| | - M Dutta
- Division of Electron Microscopy, ICMR-National Institute of Cholera and Enteric Diseases, Kolkata, India
| | - A K Mukhopadhyay
- Division of Bacteriology, ICMR-National Institute of Cholera and Enteric Diseases, Kolkata, India
| | - K Okamoto
- Collaborative Research Center of Okayama University for Infectious Diseases at NICED, Kolkata, India
| | - N S Chatterjee
- Division of Biochemistry, ICMR-National Institute of Cholera and Enteric Diseases, Kolkata, India
| |
Collapse
|
19
|
Judy E, Kishore N. Discrepancies in Thermodynamic Information Obtained from Calorimetry and Spectroscopy in Ligand Binding Reactions: Implications on Correct Analysis in Systems of Biological Importance. BULLETIN OF THE CHEMICAL SOCIETY OF JAPAN 2021. [DOI: 10.1246/bcsj.20200248] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Affiliation(s)
- Eva Judy
- Department of Chemistry, Indian Institute of Technology Bombay, Powai, Mumbai – 400 076, India
| | - Nand Kishore
- Department of Chemistry, Indian Institute of Technology Bombay, Powai, Mumbai – 400 076, India
| |
Collapse
|
20
|
Targeting protein self-association in drug design. Drug Discov Today 2021; 26:1148-1163. [PMID: 33548462 DOI: 10.1016/j.drudis.2021.01.028] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Revised: 07/24/2020] [Accepted: 01/26/2021] [Indexed: 01/05/2023]
Abstract
Protein self-association is a universal phenomenon essential for stability and molecular recognition. Disrupting constitutive homomers constitutes an original and emerging strategy in drug design. Inhibition of homomeric proteins can be achieved through direct complex disruption, subunit intercalation, or by promoting inactive oligomeric states. Targeting self-interaction grants several advantages over active site inhibition because of the stimulation of protein degradation, the enhancement of selectivity, substoichiometric inhibition, and by-pass of compensatory mechanisms. This new landscape in protein inhibition is driven by the development of biophysical and biochemical tools suited for the study of homomeric proteins, such as differential scanning fluorimetry (DSF), native mass spectrometry (MS), Förster resonance energy transfer (FRET) spectroscopy, 2D nuclear magnetic resonance (NMR), and X-ray crystallography. In this review, we discuss the different aspects of this new paradigm in drug design.
Collapse
|
21
|
Sousa FBM, Nolêto IRSG, Chaves LS, Pacheco G, Oliveira AP, Fonseca MMV, Medeiros JVR. A comprehensive review of therapeutic approaches available for the treatment of cholera. J Pharm Pharmacol 2020; 72:1715-1731. [DOI: 10.1111/jphp.13344] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Accepted: 07/04/2020] [Indexed: 12/15/2022]
Abstract
Abstract
Objectives
The oral rehydration solution is the most efficient method to treat cholera; however, it does not interfere in the action mechanism of the main virulence factor produced by Vibrio cholerae, the cholera toxin (CT), and this disease still stands out as a problem for human health worldwide. This review aimed to describe therapeutic alternatives available in the literature, especially those related to the search for molecules acting upon the physiopathology of cholera.
Key findings
New molecules have offered a protection effect against diarrhoea induced by CT or even by infection from V. cholerae. The receptor regulator cystic fibrosis channel transmembrane (CFTR), monosialoganglioside (GM1), enkephalinase, AMP-activated protein kinase (AMPK), inhibitors of expression of virulence factors and activators of ADP-ribosylarginine hydrolase are the main therapeutic targets studied. Many of these molecules or extracts still present unclear action mechanisms.
Conclusions
Knowing therapeutic alternatives and their molecular mechanisms for the treatment of cholera could guide us to develop a new drug that could be used in combination with the rehydration solution.
Collapse
Affiliation(s)
- Francisca B M Sousa
- Laboratory of Pharmacology of Inflammation and Gastrointestinal Disorders (Lafidg), Post-graduation Program in Biotechnology, Federal University of Parnaíba Delta, Parnaíba, Brazil
- Northeast Biotechnology Network (RENORBIO), Federal University of Piauí, Teresina, Brazil
| | - Isabela R S G Nolêto
- Laboratory of Pharmacology of Inflammation and Gastrointestinal Disorders (Lafidg), Post-graduation Program in Biotechnology, Federal University of Parnaíba Delta, Parnaíba, Brazil
- Northeast Biotechnology Network (RENORBIO), Federal University of Piauí, Teresina, Brazil
| | - Leticia S Chaves
- Laboratory of Pharmacology of Inflammation and Gastrointestinal Disorders (Lafidg), Post-graduation Program in Biotechnology, Federal University of Parnaíba Delta, Parnaíba, Brazil
- Post-graduation Program in Biomedical Sciences, Federal University of Piauí, Parnaíba, Brazil
| | - Gabriella Pacheco
- Laboratory of Pharmacology of Inflammation and Gastrointestinal Disorders (Lafidg), Post-graduation Program in Biotechnology, Federal University of Parnaíba Delta, Parnaíba, Brazil
| | - Ana P Oliveira
- Laboratory of Pharmacology of Inflammation and Gastrointestinal Disorders (Lafidg), Post-graduation Program in Biotechnology, Federal University of Parnaíba Delta, Parnaíba, Brazil
- Northeast Biotechnology Network (RENORBIO), Federal University of Piauí, Teresina, Brazil
| | - Mikhail M V Fonseca
- Institute of Higher Education of Vale do Parnaíba (IESVAP), Parnaíba, Brazil
| | - Jand V R Medeiros
- Laboratory of Pharmacology of Inflammation and Gastrointestinal Disorders (Lafidg), Post-graduation Program in Biotechnology, Federal University of Parnaíba Delta, Parnaíba, Brazil
- Northeast Biotechnology Network (RENORBIO), Federal University of Piauí, Teresina, Brazil
| |
Collapse
|
22
|
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
|
23
|
HilD, HilC, and RtsA Form Homodimers and Heterodimers To Regulate Expression of the Salmonella Pathogenicity Island I Type III Secretion System. J Bacteriol 2020; 202:JB.00012-20. [PMID: 32041797 DOI: 10.1128/jb.00012-20] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2020] [Accepted: 02/06/2020] [Indexed: 12/19/2022] Open
Abstract
Salmonella enterica serovar Typhimurium colonizes and invades host intestinal epithelial cells using the type three secretion system (T3SS) encoded on Salmonella pathogenicity island 1 (SPI1). The level of SPI1 T3SS gene expression is controlled by the transcriptional activator HilA, encoded on SPI1. Expression of hilA is positively regulated by three homologous transcriptional regulators, HilD, HilC, and RtsA, belonging to the AraC/XylS family. These regulators also activate the hilD, hilC, and rtsA genes by binding to the same DNA sequences upstream of these promoters, forming a complex feed-forward loop to control SPI1 expression. Despite the apparent redundancy in function, HilD has a unique role in SPI1 regulation because the majority of external regulatory inputs act exclusively through HilD. To better understand SPI1 regulation, the nature of interaction between HilD, HilC, and RtsA has been characterized using biochemical and genetic techniques. Our results showed that HilD, HilC, and RtsA can form heterodimers as well as homodimers in solution. Comparison with other AraC family members identified a putative α-helix in the N-terminal domain, which acts as the dimerization domain. Alanine substitution in this region results in reduced dimerization of HilD and HilC and also affects their ability to activate hilA expression. The dimer interactions of HilD, HilC, and RtsA add another layer of complexity to the SPI1 regulatory circuit, providing a more comprehensive understanding of SPI1 T3SS regulation and Salmonella pathogenesis.IMPORTANCE The SPI1 type three secretion system is a key virulence factor required for Salmonella to both cause gastroenteritis and initiate serious systemic disease. The system responds to numerous environmental signals in the intestine, integrating this information via a complex regulatory network. Here, we show that the primary regulatory proteins in the network function as both homodimers and heterodimers, providing information regarding both regulation of virulence in this important pathogen and general signal integration to control gene expression.
Collapse
|
24
|
Abstract
Progress against tuberculosis (TB) requires faster-acting drugs. Mycobacterium tuberculosis (Mtb) is the leading cause of death by an infectious disease and its treatment is challenging and lengthy. Mtb is remarkably successful, in part, due to its ability to become dormant in response to host immune pressures. The DosRST two-component regulatory system is induced by hypoxia, nitric oxide and carbon monoxide and remodels Mtb physiology to promote nonreplicating persistence (NRP). NRP bacteria are thought to play a role in the long course of TB treatment. Therefore, inhibitors of DosRST-dependent adaptation may function to kill this reservoir of persisters and potentially shorten therapy. This review examines the function of DosRST, newly discovered compounds that inhibit DosRST signaling and considers future development of DosRST inhibitors as adjunct therapies.
Collapse
|
25
|
Wang CH, Hsieh YH, Powers ZM, Kao CY. Defeating Antibiotic-Resistant Bacteria: Exploring Alternative Therapies for a Post-Antibiotic Era. Int J Mol Sci 2020; 21:E1061. [PMID: 32033477 PMCID: PMC7037027 DOI: 10.3390/ijms21031061] [Citation(s) in RCA: 110] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2020] [Revised: 02/04/2020] [Accepted: 02/04/2020] [Indexed: 12/11/2022] Open
Abstract
Antibiotics are one of the greatest medical advances of the 20th century, however, they are quickly becoming useless due to antibiotic resistance that has been augmented by poor antibiotic stewardship and a void in novel antibiotic discovery. Few novel classes of antibiotics have been discovered since 1960, and the pipeline of antibiotics under development is limited. We therefore are heading for a post-antibiotic era in which common infections become untreatable and once again deadly. There is thus an emergent need for both novel classes of antibiotics and novel approaches to treatment, including the repurposing of existing drugs or preclinical compounds and expanded implementation of combination therapies. In this review, we highlight to utilize alternative drug targets/therapies such as combinational therapy, anti-regulator, anti-signal transduction, anti-virulence, anti-toxin, engineered bacteriophages, and microbiome, to defeat antibiotic-resistant bacteria.
Collapse
Affiliation(s)
- Chih-Hung Wang
- Department of Power Mechanical Engineering, National Tsing Hua University, Hsinchu 30013, Taiwan;
| | - Yi-Hsien Hsieh
- Institute of Biochemistry, Microbiology and Immunology, Chung Shan Medical University, Taichung 40201, Taiwan;
| | - Zachary M. Powers
- Department of Medical Microbiology and Immunology, University of Wisconsin-Madison, Madison, WI 53706, USA;
| | - Cheng-Yen Kao
- Institute of Microbiology and Immunology, School of Life Science, National Yang-Ming University, Taipei 11221, Taiwan
| |
Collapse
|
26
|
Cruite JT, Kovacikova G, Clark KA, Woodbrey AK, Skorupski K, Kull FJ. Structural basis for virulence regulation in Vibrio cholerae by unsaturated fatty acid components of bile. Commun Biol 2019; 2:440. [PMID: 31815195 PMCID: PMC6882843 DOI: 10.1038/s42003-019-0686-x] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2019] [Accepted: 11/07/2019] [Indexed: 12/20/2022] Open
Abstract
The AraC/XylS-family transcriptional regulator ToxT is the master virulence activator of Vibrio cholerae, the gram-negative bacterial pathogen that causes the diarrheal disease cholera. Unsaturated fatty acids (UFAs) found in bile inhibit the activity of ToxT. Crystal structures of inhibited ToxT bound to UFA or synthetic inhibitors have been reported, but no structure of ToxT in an active conformation had been determined. Here we present the 2.5 Å structure of ToxT without an inhibitor. The structure suggests release of UFA or inhibitor leads to an increase in flexibility, allowing ToxT to adopt an active conformation that is able to dimerize and bind DNA. Small-angle X-ray scattering was used to validate a structural model of an open ToxT dimer bound to the cholera toxin promoter. The results presented here provide a detailed structural mechanism for virulence gene regulation in V. cholerae by the UFA components of bile and other synthetic ToxT inhibitors.
Collapse
Affiliation(s)
- Justin T. Cruite
- Department of Biochemistry and Cell Biology, Geisel School of Medicine at Dartmouth College, Hanover, NH USA
- Guarini School of Graduate and Advanced Studies, Dartmouth College, Hanover, NH USA
| | - Gabriela Kovacikova
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth College, Hanover, NH USA
| | - Kenzie A. Clark
- Department of Chemistry, Dartmouth College, Hanover, NH USA
- Present Address: Department of Chemistry, Princeton University, Princeton, NJ USA
| | - Anne K. Woodbrey
- Guarini School of Graduate and Advanced Studies, Dartmouth College, Hanover, NH USA
- Department of Chemistry, Dartmouth College, Hanover, NH USA
| | - Karen Skorupski
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth College, Hanover, NH USA
| | - F. Jon Kull
- Department of Biochemistry and Cell Biology, Geisel School of Medicine at Dartmouth College, Hanover, NH USA
- Guarini School of Graduate and Advanced Studies, Dartmouth College, Hanover, NH USA
- Department of Chemistry, Dartmouth College, Hanover, NH USA
| |
Collapse
|
27
|
Patel J, Yin HB, Bauchan G, Mowery J. Inhibition of Escherichia coli O157:H7 and Salmonella enterica virulence factors by benzyl isothiocyanate. Food Microbiol 2019; 86:103303. [PMID: 31703885 DOI: 10.1016/j.fm.2019.103303] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2019] [Revised: 08/03/2019] [Accepted: 08/10/2019] [Indexed: 01/17/2023]
Abstract
Escherichia coli O157:H7 and Salmonella enterica are foodborne pathogens with major public health concern in the U.S. These pathogens utilize several virulence factors to initiate infections in humans. The antimicrobial effect of seven glucosinolate hydrolysis compounds against Salmonella and E. coli O157:H7 was investigated by the disc diffusion assay. Among the tested compounds, benzyl isothiocyanate (BIT), which exerted the highest antimicrobial activity, was evaluated for its anti-virulence properties against these pathogens. The effect of BIT on motility of Salmonella and E. coli O157:H7 and Shiga toxin production by E. coli O157:H7 was determined by the motility assay and ELISA procedure, respectively. Confocal and transmission electron microscopy (TEM) procedures were used to determine bacterial damage at the cellular level. Results revealed that sub-inhibitory concentrations (SICs) of BIT significantly inhibited the motility of both bacteria (P < 0.05). Shiga toxin production by E. coli O157:H7 was decreased by ~32% in the presence of BIT at SICs. TEM results showed the disruption of outer membrane, release of cytoplasmic contents, and cell lysis following BIT treatment. Results suggest that BIT could be potentially used to attenuate Salmonella and E. coli O157:H7 infections by reducing the virulence factors including bacterial motility and Shiga toxin production.
Collapse
Affiliation(s)
- Jitendra Patel
- U.S. Department of Agriculture, Agricultural Research Service, Environmental and Microbial Food Safety Laboratory, Beltsville, MD 20705, USA.
| | - Hsin-Bai Yin
- U.S. Department of Agriculture, Agricultural Research Service, Environmental and Microbial Food Safety Laboratory, Beltsville, MD 20705, USA
| | - Gary Bauchan
- U.S. Department of Agriculture, Agricultural Research Service, SGIL Electron and Confocal Microscopy Unit, Beltsville, MD 20705, USA
| | - Joseph Mowery
- U.S. Department of Agriculture, Agricultural Research Service, SGIL Electron and Confocal Microscopy Unit, Beltsville, MD 20705, USA
| |
Collapse
|
28
|
Massai F, Saleeb M, Doruk T, Elofsson M, Forsberg Å. Development, Optimization, and Validation of a High Throughput Screening Assay for Identification of Tat and Type II Secretion Inhibitors of Pseudomonas aeruginosa. Front Cell Infect Microbiol 2019; 9:250. [PMID: 31355152 PMCID: PMC6635566 DOI: 10.3389/fcimb.2019.00250] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2019] [Accepted: 06/26/2019] [Indexed: 11/13/2022] Open
Abstract
Antibiotics are becoming less effective in treatment of infections caused by multidrug-resistant Pseudomonas aeruginosa. Antimicrobial therapies based on the inhibition of specific virulence-related traits, as opposed to growth inhibitors, constitute an innovative and appealing approach to tackle the threat of P. aeruginosa infections. The twin-arginine translocation (Tat) pathway plays an important role in the pathogenesis of P. aeruginosa, and constitutes a promising target for the development of anti-pseudomonal drugs. In this study we developed and optimized a whole-cell, one-well assay, based on native phospholipase C activity, to identify compounds active against the Tat system. Statistical robustness, sensitivity and consequently suitability for high-throughput screening (HTS) were confirmed by a dry run/pre-screening test scoring a Z′ of 0.82 and a signal-to-noise ratio of 49. Using this assay, we evaluated ca. 40,000 molecules and identified 59 initial hits as possible Tat inhibitors. Since phospholipase C is exported into the periplasm by Tat, and subsequently translocated across the outer membrane by the type II secretion system (T2SS), our assay could also identify T2SS inhibitors. To validate our hits and discriminate between compounds that inhibited either Tat or T2SS, two separate counter assays were developed and optimized. Finally, three Tat inhibitors and one T2SS inhibitor were confirmed by means of dose-response analysis and additional counter and confirming assays. Although none of the identified inhibitors was suitable as a lead compound for drug development, this study validates our assay as a simple, efficient, and HTS compatible method for the identification of Tat and T2SS inhibitors.
Collapse
Affiliation(s)
- Francesco Massai
- Laboratory for Molecular Infection Medicine Sweden, Department of Molecular Biology, Umeå University, Umeå, Sweden.,Department of Molecular Biology, Umeå Centre for Microbial Research, Umeå University, Umeå, Sweden
| | - Michael Saleeb
- Department of Chemistry, Umeå Centre for Microbial Research, Umeå University, Umeå, Sweden
| | - Tugrul Doruk
- Laboratory for Molecular Infection Medicine Sweden, Department of Molecular Biology, Umeå University, Umeå, Sweden.,Department of Molecular Biology, Umeå Centre for Microbial Research, Umeå University, Umeå, Sweden
| | - Mikael Elofsson
- Department of Chemistry, Umeå Centre for Microbial Research, Umeå University, Umeå, Sweden
| | - Åke Forsberg
- Laboratory for Molecular Infection Medicine Sweden, Department of Molecular Biology, Umeå University, Umeå, Sweden.,Department of Molecular Biology, Umeå Centre for Microbial Research, Umeå University, Umeå, Sweden
| |
Collapse
|
29
|
Tsai KC, Hung PP, Cheng CF, Chen C, Tseng TS. Exploring the mode of action of inhibitors targeting the PhoP response regulator of Salmonella enterica through comprehensive pharmacophore approaches. RSC Adv 2019; 9:9308-9312. [PMID: 35517705 PMCID: PMC9062048 DOI: 10.1039/c9ra00620f] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2019] [Accepted: 02/28/2019] [Indexed: 11/23/2022] Open
Abstract
The PhoQ/PhoP two-component system regulates the physiological and virulence functions of Salmonella enterica. However, the mode of action of known PhoP inhibitors is unclear. We systematically constructed a pharmacophore model of inhibitors to probe the interface pharmacophore model of the PhoP dimer, coupling it with Ligplot analysis. We found that these inhibitors bind on the α5-helix, altering the conformation and interfering with PhoP binding on DNA. Comprehensive pharmacophore approaches explore the mode of action of inhibitors targeting PhoP response regulator of Salmonella enterica.![]()
Collapse
Affiliation(s)
- Keng-Chang Tsai
- National Research Institute of Chinese Medicine, Ministry of Health and Welfare Taipei 112 Taiwan.,The PhD Program in Medical Biotechnology, College of Medical Science and Technology, Taipei Medical University Taipei Taiwan
| | - Po-Pin Hung
- Division of Infectious Disease, Department of Internal Medicine, Taipei Tzu Chi Hospital, The Buddhist Tzu Chi Medical Foundation New Taipei City 231 Taiwan
| | - Ching-Feng Cheng
- Department of Pediatrics, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Taipei and Tzu Chi University Hualien Taiwan.,Institute of Biomedical Sciences, Academia Sinica Taipei 115 Taiwan
| | - Chinpan Chen
- Institute of Biomedical Sciences, Academia Sinica Taipei 115 Taiwan
| | - Tien-Sheng Tseng
- Department of Research, Taipei Tzu Chi Hospital, The Buddhist Tzu Chi Medical Foundation New Taipei City 231 Taiwan
| |
Collapse
|
30
|
Small-molecule inhibitor of HlyU attenuates virulence of Vibrio species. Sci Rep 2019; 9:4346. [PMID: 30867441 PMCID: PMC6416295 DOI: 10.1038/s41598-019-39554-y] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2018] [Accepted: 01/10/2019] [Indexed: 02/08/2023] Open
Abstract
Increasing antibiotic resistance has led to the development of new strategies to combat bacterial infection. Anti-virulence strategies that impair virulence of bacterial pathogens are one of the novel approaches with less selective pressure for developing resistance than traditional strategies that impede viability. In this study, a small molecule CM14 [N-(4-oxo-4H-thieno[3,4-c]chromen-3-yl)-3-phenylprop-2-ynamide] that inhibits the activity of HlyU, a transcriptional regulator essential for the virulence of the fulminating human pathogen Vibrio vulnificus, has been identified. Without affecting bacterial growth or triggering the host cell death, CM14 reduces HlyU-dependent expression of virulence genes in V. vulnificus. In addition to the decreased hemolysis of human erythrocytes, CM14 impedes host cell rounding and lysis caused by V. vulnificus. Notably, CM14 significantly enhances survival of mice infected with V. vulnificus by alleviating hepatic and renal dysfunction and systemic inflammation. Biochemical, mass spectrometric, and mutational analyses revealed that CM14 inhibits HlyU from binding to target DNA by covalently modifying Cys30. Remarkably, CM14 decreases the expression of various virulence genes of other Vibrio species and thus attenuates their virulence phenotypes. Together, this molecule could be an anti-virulence agent against HlyU-harboring Vibrio species with a low selective pressure for the emergence of resistance.
Collapse
|
31
|
Peterson KM, Gellings PS. Multiple intraintestinal signals coordinate the regulation of Vibrio cholerae virulence determinants. Pathog Dis 2018; 76:4791527. [PMID: 29315383 DOI: 10.1093/femspd/ftx126] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2017] [Accepted: 12/27/2017] [Indexed: 12/17/2022] Open
Abstract
Vibrio cholerae is a Gram-negative motile bacterium capable of causing fatal pandemic disease in humans via oral ingestion of contaminated water or food. Within the human intestine, the motile vibrios must evade the innate host defense mechanisms, penetrate the mucus layer covering the small intestine, adhere to and multiply on the surface of the microvilli and cause disease via the action of cholera toxin. The explosive diarrhea associated with V. cholerae intestinal colonization leads to dissemination of the vibrios back into the environment to complete this phase of the life cycle. The host phase of the vibrio life cycle is made possible via the concerted action of a signaling cascade that controls the synthesis of V. cholerae colonization determinants. These virulence proteins are coordinately synthesized in response to specific host signals that are still largely undefined. A more complete understanding of the molecular events involved in the V. cholerae recognition of intraintestinal signals and the subsequent transcriptional response will provide important information regarding how pathogenic bacteria establish infection and provide novel methods for treating and/or preventing bacterial infections such as Asiatic cholera. This review will summarize what is currently known in regard to host intraintestinal signals that inform the complex ToxR regulatory cascade in order to coordinate in a spatial and temporal fashion virulence protein synthesis within the human small intestine.
Collapse
Affiliation(s)
- Kenneth M Peterson
- Department of Microbiology and Immunology, Louisiana State University Health Science Center, Shreveport, LA 71130, USA
| | - Patrick S Gellings
- Department of Microbiology and Immunology, Louisiana State University Health Science Center, Shreveport, LA 71130, USA
| |
Collapse
|
32
|
Woodbrey AK, Onyango EO, Kovacikova G, Kull FJ, Gribble GW. A Modified ToxT Inhibitor Reduces Vibrio cholerae Virulence in Vivo. Biochemistry 2018; 57:5609-5615. [PMID: 30160100 DOI: 10.1021/acs.biochem.8b00667] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
We have previously designed and synthesized small-molecule inhibitors that reduce Vibrio cholerae virulence in vitro by targeting the transcription factor ToxT. Here we report the synthesis and biological activity of derivatives of our previous bicyclic, fatty acid-like inhibitors. All of the synthesized derivatives show antivirulence activity in vitro. For the most potent compounds, a concentration of 5 μM completely inhibited ToxT-mediated tcpA expression as measured in the β-galactosidase assay. One indole compound, 3-(1-butyl-1 H-indol-7-yl)propanoic acid (8), was also effective at inhibiting intestinal colonization in the infant mouse. These modified compounds may serve as good candidates for further anti-cholera drug development.
Collapse
Affiliation(s)
- Anne K Woodbrey
- Department of Chemistry , Dartmouth College , Hanover , New Hampshire 03755 , United States
| | - Evans O Onyango
- Department of Chemistry , Dartmouth College , Hanover , New Hampshire 03755 , United States
| | - Gabriela Kovacikova
- Department of Microbiology and Immunology , Geisel School of Medicine at Dartmouth , Hanover , New Hampshire 03755 , United States
| | - F Jon Kull
- Department of Chemistry , Dartmouth College , Hanover , New Hampshire 03755 , United States
| | - Gordon W Gribble
- Department of Chemistry , Dartmouth College , Hanover , New Hampshire 03755 , United States
| |
Collapse
|
33
|
Protein‐protein interactions as antibiotic targets: A medicinal chemistry perspective. Med Res Rev 2018; 40:469-494. [DOI: 10.1002/med.21519] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2017] [Revised: 05/28/2018] [Accepted: 06/03/2018] [Indexed: 12/27/2022]
|
34
|
Abstract
Antibiotics have saved millions of lives over the past decades. However, the accumulation of so many antibiotic resistance genes by some clinically relevant pathogens has begun to lead to untreatable infections worldwide. The current antibiotic resistance crisis will require greater efforts by governments and the scientific community to increase the research and development of new antibacterial drugs with new mechanisms of action. A major challenge is the identification of novel microbial targets, essential for in vivo growth or pathogenicity, whose inhibitors can overcome the currently circulating resistome of human pathogens. In this article, we focus on the potential high value of bacterial transcriptional regulators as targets for the development of new antibiotics, discussing in depth the molecular role of these regulatory proteins in bacterial physiology and pathogenesis. Recent advances in the search for novel compounds that inhibit the biological activity of relevant transcriptional regulators in pathogenic bacteria are reviewed.
Collapse
|
35
|
Nicol M, Alexandre S, Luizet JB, Skogman M, Jouenne T, Salcedo SP, Dé E. Unsaturated Fatty Acids Affect Quorum Sensing Communication System and Inhibit Motility and Biofilm Formation of Acinetobacter baumannii. Int J Mol Sci 2018; 19:ijms19010214. [PMID: 29320462 PMCID: PMC5796163 DOI: 10.3390/ijms19010214] [Citation(s) in RCA: 56] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2017] [Revised: 01/07/2018] [Accepted: 01/08/2018] [Indexed: 02/07/2023] Open
Abstract
The increasing threat of Acinetobacter baumannii as a nosocomial pathogen is mainly due to the occurrence of multidrug-resistant strains that are associated with the real problem of its eradication from hospital wards. The particular ability of this pathogen to form biofilms contributes to its persistence, increases antibiotic resistance, and promotes persistent/device-related infections. We previously demonstrated that virstatin, which is a small organic compound known to decrease virulence of Vibrio cholera via an inhibition of T4-pili expression, displayed very promising activity to prevent A. baumannii biofilm development. Here, we examined the antibiofilm activity of mono-unsaturated chain fatty acids, palmitoleic (PoA), and myristoleic (MoA) acids, presenting similar action on V. cholerae virulence. We demonstrated that PoA and MoA (at 0.02 mg/mL) were able to decrease A. baumannii ATCC 17978 biofilm formation up to 38% and 24%, respectively, presented a biofilm dispersing effect and drastically reduced motility. We highlighted that these fatty acids decreased the expression of the regulator abaR from the LuxIR-type quorum sensing (QS) communication system AbaIR and consequently reduced the N-acyl-homoserine lactone production (AHL). This effect can be countered by addition of exogenous AHLs. Besides, fatty acids may have additional non-targeted effects, independent from QS. Atomic force microscopy experiments probed indeed that PoA and MoA could also act on the initial adhesion process in modifying the material interface properties. Evaluation of fatty acids effect on 22 clinical isolates showed a strain-dependent antibiofilm activity, which was not correlated to hydrophobicity or pellicle formation ability of the tested strains, and suggested a real diversity in cell-to-cell communication systems involved in A. baumannii biofilm formation.
Collapse
Affiliation(s)
- Marion Nicol
- Normandie University, Unirouen, 76000 Rouen, France.
- CNRS, UMR 6270, Polymers, Biopolymers, Surfaces Laboratory, F-76821 Mont-Saint-Aignan, France.
| | - Stéphane Alexandre
- Normandie University, Unirouen, 76000 Rouen, France.
- CNRS, UMR 6270, Polymers, Biopolymers, Surfaces Laboratory, F-76821 Mont-Saint-Aignan, France.
| | - Jean-Baptiste Luizet
- Laboratory of Molecular Microbiology and Structural Biochemistry, University of Lyon, Centre National de la Recherche Scientifique, F-69367 Lyon, France.
| | - Malena Skogman
- Department of Pharmaceutical Biosciences, Faculty of Pharmacy, University of Helsinki, Viikinkaari 5E, FI-00014 Helsinki, Finland.
| | - Thierry Jouenne
- Normandie University, Unirouen, 76000 Rouen, France.
- CNRS, UMR 6270, Polymers, Biopolymers, Surfaces Laboratory, F-76821 Mont-Saint-Aignan, France.
| | - Suzana P Salcedo
- Laboratory of Molecular Microbiology and Structural Biochemistry, University of Lyon, Centre National de la Recherche Scientifique, F-69367 Lyon, France.
| | - Emmanuelle Dé
- Normandie University, Unirouen, 76000 Rouen, France.
- CNRS, UMR 6270, Polymers, Biopolymers, Surfaces Laboratory, F-76821 Mont-Saint-Aignan, France.
| |
Collapse
|
36
|
Sarwar S, Ali A, Pal M, Chakrabarti P. Zinc oxide nanoparticles provide anti-cholera activity by disrupting the interaction of cholera toxin with the human GM1 receptor. J Biol Chem 2017; 292:18303-18311. [PMID: 28882894 PMCID: PMC5672052 DOI: 10.1074/jbc.m117.793240] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2017] [Revised: 09/04/2017] [Indexed: 11/06/2022] Open
Abstract
Vibrio cholerae causes cholera and is the leading cause of diarrhea in developing countries, highlighting the need for the development of new treatment strategies to combat this disease agent. While exploring the possibility of using zinc oxide (ZnO) nanoparticles (NPs) in cholera treatment, we previously found that ZnO NPs reduce fluid accumulation in mouse ileum induced by the cholera toxin (CT) protein. To uncover the mechanism of action of ZnO NPs on CT activity, here we used classical (O395) and El Tor (C6706) V. cholerae biotypes in growth and biochemical assays. We found that a ZnO NP concentration of 10 μg/ml did not affect the growth rates of these two strains, nor did we observe that ZnO NPs reduce the expression levels of CT mRNA and protein. It was observed that ZnO NPs form a complex with CT, appear to disrupt the CT secondary structure, and block its interaction with the GM1 ganglioside receptor in the outer leaflet of the plasma membrane in intestinal (HT-29) cells and thereby reduce CT uptake into the cells. In the range of 2.5-10 μg/ml, ZnO NPs exhibited no cytotoxicity on kidney (HEK293) and HT-29 cells. We conclude that ZnO NPs prevent the first step in the translocation of cholera toxin into intestinal epithelial cells without exerting measurable toxic effects on HEK293 and HT-29 cells.
Collapse
Affiliation(s)
| | - Asif Ali
- Division of Molecular Medicine, Bose Institute, P1/12 CIT Scheme VIIM, Kolkata, India
| | - Mahadeb Pal
- Division of Molecular Medicine, Bose Institute, P1/12 CIT Scheme VIIM, Kolkata, India
| | | |
Collapse
|
37
|
Waack U, Johnson TL, Chedid K, Xi C, Simmons LA, Mobley HLT, Sandkvist M. Targeting the Type II Secretion System: Development, Optimization, and Validation of a High-Throughput Screen for the Identification of Small Molecule Inhibitors. Front Cell Infect Microbiol 2017; 7:380. [PMID: 28894700 PMCID: PMC5581314 DOI: 10.3389/fcimb.2017.00380] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2017] [Accepted: 08/09/2017] [Indexed: 12/26/2022] Open
Abstract
Nosocomial pathogens that develop multidrug resistance present an increasing problem for healthcare facilities. Due to its rapid rise in antibiotic resistance, Acinetobacter baumannii is one of the most concerning gram-negative species. A. baumannii typically infects immune compromised individuals resulting in a variety of outcomes, including pneumonia and bacteremia. Using a murine model for bacteremia, we have previously shown that the type II secretion system (T2SS) contributes to in vivo fitness of A. baumannii. Here, we provide support for a role of the T2SS in protecting A. baumannii from human complement as deletion of the T2SS gene gspD resulted in a 100-fold reduction in surviving cells when incubated with human serum. This effect was abrogated in the absence of Factor B, a component of the alternative pathway of complement activation, indicating that the T2SS protects A. baumannii against the alternative complement pathway. Because inactivation of the T2SS results in loss of secretion of multiple enzymes, reduced in vivo fitness, and increased sensitivity to human complement, the T2SS may be a suitable target for therapeutic intervention. Accordingly, we developed and optimized a whole-cell high-throughput screening (HTS) assay based on secreted lipase activity to identify small molecule inhibitors of the T2SS. We tested the reproducibility of our assay using a 6,400-compound library. With small variation within controls and a dynamic range between positive and negative controls, the assay had a z-factor of 0.65, establishing its suitability for HTS. Our screen identified the lipase inhibitors Orlistat and Ebelactone B demonstrating the specificity of the assay. To eliminate inhibitors of lipase activity and lipase expression, two counter assays were developed and optimized. By implementing these assays, all seven tricyclic antidepressants present in the library were found to be inhibitors of the lipase, highlighting the potential of identifying alternative targets for approved pharmaceuticals. Although no T2SS inhibitor was identified among the compounds that reduced lipase activity by ≥30%, our small proof-of-concept pilot study indicates that the HTS regimen is simple, reproducible, and specific and that it can be used to screen larger libraries for the identification of T2SS inhibitors that may be developed into novel A. baumannii therapeutics.
Collapse
Affiliation(s)
- Ursula Waack
- Department of Microbiology and Immunology, University of Michigan Medical SchoolAnn Arbor, MI, United States
| | - Tanya L Johnson
- Department of Microbiology and Immunology, University of Michigan Medical SchoolAnn Arbor, MI, United States.,Department of Chemistry, Eastern Michigan UniversityYpsilanti, MI, United States
| | - Khalil Chedid
- Department of Microbiology and Immunology, University of Michigan Medical SchoolAnn Arbor, MI, United States
| | - Chuanwu Xi
- Department of Environmental Health Sciences, University of Michigan School of Public HealthAnn Arbor, MI, United States
| | - Lyle A Simmons
- Department of Molecular, Cellular, and Developmental Biology, University of MichiganAnn Arbor, MI, United States
| | - Harry L T Mobley
- Department of Microbiology and Immunology, University of Michigan Medical SchoolAnn Arbor, MI, United States
| | - Maria Sandkvist
- Department of Microbiology and Immunology, University of Michigan Medical SchoolAnn Arbor, MI, United States
| |
Collapse
|
38
|
Wang H, Silva AJ, Benitez JA. 3-Amino 1,8-naphthalimide, a structural analog of the anti-cholera drug virstatin inhibits chemically-biased swimming and swarming motility in vibrios. Microbes Infect 2017. [PMID: 28392408 DOI: 10.1016/j.micinf] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/13/2023]
Abstract
A screen for inhibitors of Vibrio cholerae motility identified the compound 3-amino 1,8-naphthalimide (3-A18NI), a structural analog of the cholera drug virstatin. Similar to virstatin, 3-A18NI diminished cholera toxin production. In contrast, 3-A18NI impeded swimming and/or swarming motility of V. cholerae and V. parahemolyticus suggesting that it could target the chemotaxis pathway shared by the polar and lateral flagellar system of vibrios. 3-A18NI did not inhibit the expression of V. cholerae major flagellin FlaA or the assembly of its polar flagellum. Finally, 3-A18NI enhanced V. cholerae colonization mimicking the phenotype of chemotaxis mutants that exhibit counterclockwise-biased flagellum rotation.
Collapse
Affiliation(s)
- Hongxia Wang
- Morehouse School of Medicine, Department of Microbiology, Biochemistry and Immunology, 720 Westview Dr., SW, Atlanta, GA 30310, USA
| | - Anisia J Silva
- Morehouse School of Medicine, Department of Microbiology, Biochemistry and Immunology, 720 Westview Dr., SW, Atlanta, GA 30310, USA
| | - Jorge A Benitez
- Morehouse School of Medicine, Department of Microbiology, Biochemistry and Immunology, 720 Westview Dr., SW, Atlanta, GA 30310, USA.
| |
Collapse
|
39
|
3-Amino 1,8-naphthalimide, a structural analog of the anti-cholera drug virstatin inhibits chemically-biased swimming and swarming motility in vibrios. Microbes Infect 2017; 19:370-375. [PMID: 28392408 DOI: 10.1016/j.micinf.2017.03.003] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2017] [Revised: 03/24/2017] [Accepted: 03/28/2017] [Indexed: 11/23/2022]
Abstract
A screen for inhibitors of Vibrio cholerae motility identified the compound 3-amino 1,8-naphthalimide (3-A18NI), a structural analog of the cholera drug virstatin. Similar to virstatin, 3-A18NI diminished cholera toxin production. In contrast, 3-A18NI impeded swimming and/or swarming motility of V. cholerae and V. parahemolyticus suggesting that it could target the chemotaxis pathway shared by the polar and lateral flagellar system of vibrios. 3-A18NI did not inhibit the expression of V. cholerae major flagellin FlaA or the assembly of its polar flagellum. Finally, 3-A18NI enhanced V. cholerae colonization mimicking the phenotype of chemotaxis mutants that exhibit counterclockwise-biased flagellum rotation.
Collapse
|
40
|
A new class of inhibitors of the AraC family virulence regulator Vibrio cholerae ToxT. Sci Rep 2017; 7:45011. [PMID: 28332578 PMCID: PMC5362913 DOI: 10.1038/srep45011] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2016] [Accepted: 02/20/2017] [Indexed: 11/28/2022] Open
Abstract
Vibrio cholerae is responsible for the diarrheal disease cholera that infects millions of people worldwide. While vaccines protecting against cholera exist, and oral rehydration therapy is an effective treatment method, the disease will remain a global health threat until long-term solutions such as improved sanitation and access to clean water become widely available. Because of this, there is a pressing need for potent therapeutics that can either mitigate cholera symptoms, or act prophylactically to prevent the virulent effects of a cholera infection. Here we report the design, synthesis, and characterization of a set of compounds that bind and inhibit ToxT, the transcription factor that directly regulates the two primary V. cholerae virulence factors. Using the folded structure of the monounsaturated fatty acid observed in the X-ray structure of ToxT as a template, we designed ten novel compounds that inhibit the virulence cascade to a greater degree than any known inhibitor. Our findings provide a structural and functional basis for the development of viable antivirulence therapeutics that combat cholera and, potentially, other forms of bacterial pathogenic disease.
Collapse
|
41
|
Affiliation(s)
- Megan Garland
- Cancer
Biology Program, ‡Department of Pathology, §Department of Microbiology and Immunology, and ∥Department of
Chemical and Systems Biology, Stanford University School of Medicine, 300 Pasteur Drive, Stanford, California 94305, United States
| | - Sebastian Loscher
- Cancer
Biology Program, ‡Department of Pathology, §Department of Microbiology and Immunology, and ∥Department of
Chemical and Systems Biology, Stanford University School of Medicine, 300 Pasteur Drive, Stanford, California 94305, United States
| | - Matthew Bogyo
- Cancer
Biology Program, ‡Department of Pathology, §Department of Microbiology and Immunology, and ∥Department of
Chemical and Systems Biology, Stanford University School of Medicine, 300 Pasteur Drive, Stanford, California 94305, United States
| |
Collapse
|
42
|
Small Molecules That Sabotage Bacterial Virulence. Trends Pharmacol Sci 2017; 38:339-362. [PMID: 28209403 DOI: 10.1016/j.tips.2017.01.004] [Citation(s) in RCA: 82] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2016] [Revised: 01/01/2017] [Accepted: 01/06/2017] [Indexed: 02/08/2023]
Abstract
The continued rise of antibiotic-resistant bacterial infections has motivated alternative strategies for target discovery and treatment of infections. Antivirulence therapies function through inhibition of in vivo required virulence factors to disarm the pathogen instead of directly targeting viability or growth. This approach to treating bacteria-mediated diseases may have advantages over traditional antibiotics because it targets factors specific for pathogenesis, potentially reducing selection for resistance and limiting collateral damage to the resident microbiota. This review examines vulnerable molecular mechanisms used by bacteria to cause disease and the antivirulence compounds that sabotage these virulence pathways. By expanding the study of antimicrobial targets beyond those that are essential for growth, antivirulence strategies offer new and innovative opportunities to combat infectious diseases.
Collapse
|
43
|
Ribavirin suppresses bacterial virulence by targeting LysR-type transcriptional regulators. Sci Rep 2016; 6:39454. [PMID: 27991578 PMCID: PMC5171790 DOI: 10.1038/srep39454] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2016] [Accepted: 11/23/2016] [Indexed: 12/30/2022] Open
Abstract
Targeting bacterial virulence mechanisms without compromising bacterial growth is a promising strategy to prevent drug resistance. LysR-type transcriptional regulators (LTTRs) possess structural conservation across bacterial species and regulate virulence in numerous pathogens, making them attractive targets for antimicrobial agents. We targeted AphB, a Vibrio cholerae LTTR, which regulates the expression of genes encoding cholera toxin and toxin-co-regulated pilus for inhibitor designing. Since AphB ligand is unknown, we followed a molecular fragment-based approach for ligand designing using FDA-approved drugs and subsequent screen to identify molecules that exhibited high-affinity binding to AphB ligand-binding pocket. Among the identified compounds, ribavirin, an anti-viral drug, antagonized AphB functions. Ribavirin perturbed Vibrio cholerae pathogenesis in animal models. The inhibitory effects of the drug was limited to the bacteria expressing wild type AphB, but not its constitutively active mutant (AphBN100E), which represents the ligand-bound state, suggesting that ribavirin binds to the active site of AphB to exert its inhibitory role and there exists no AphB-independent mechanism of its action. Similarly, ribavirin suppressed the functions of Salmonella Typhi LTTR Hrg, indicating its broad spectrum efficacy. Moreover, ribavirin did not affect the bacterial viability in culture. This study cites an example of drug repurposing for anti-infective therapy.
Collapse
|
44
|
Good JAD, Andersson C, Hansen S, Wall J, Krishnan KS, Begum A, Grundström C, Niemiec MS, Vaitkevicius K, Chorell E, Wittung-Stafshede P, Sauer UH, Sauer-Eriksson AE, Almqvist F, Johansson J. Attenuating Listeria monocytogenes Virulence by Targeting the Regulatory Protein PrfA. Cell Chem Biol 2016; 23:404-14. [PMID: 26991105 PMCID: PMC4802734 DOI: 10.1016/j.chembiol.2016.02.013] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2015] [Revised: 01/27/2016] [Accepted: 02/19/2016] [Indexed: 01/28/2023]
Abstract
The transcriptional activator PrfA, a member of the Crp/Fnr family, controls the expression of some key virulence factors necessary for infection by the human bacterial pathogen Listeria monocytogenes. Phenotypic screening identified ring-fused 2-pyridone molecules that at low micromolar concentrations attenuate L. monocytogenes cellular uptake by reducing the expression of virulence genes. These inhibitors bind the transcriptional regulator PrfA and decrease its affinity for the consensus DNA-binding site. Structural characterization of this interaction revealed that one of the ring-fused 2-pyridones, compound 1, binds at two separate sites on the protein: one within a hydrophobic pocket or tunnel, located between the C- and N-terminal domains of PrfA, and the second in the vicinity of the DNA-binding helix-turn-helix motif. At both sites the compound interacts with residues important for PrfA activation and helix-turn-helix formation. Ring-fused 2-pyridones represent a new class of chemical probes for studying virulence in L. monocytogenes. Inhibitors of L. monocytogenes infectivity reduce virulence gene expression Binding of inhibitor to the PrfA regulator reduces affinity for its DNA motif First crystal structure of a Crp family regulator with an inhibitor Provides rationale for screening with Crp family transcriptional regulators
Collapse
Affiliation(s)
- James A D Good
- Department of Chemistry, Umeå University, 901 87 Umeå, Sweden; Umeå Centre for Microbial Research (UCMR), Umeå University, 901 87 Umeå, Sweden
| | - Christopher Andersson
- Umeå Centre for Microbial Research (UCMR), Umeå University, 901 87 Umeå, Sweden; Department of Molecular Biology, Umeå University, 901 87 Umeå, Sweden; Molecular Infection Medicine, Sweden (MIMS), Umeå University, 901 87 Umeå, Sweden
| | - Sabine Hansen
- Umeå Centre for Microbial Research (UCMR), Umeå University, 901 87 Umeå, Sweden; Department of Molecular Biology, Umeå University, 901 87 Umeå, Sweden; Molecular Infection Medicine, Sweden (MIMS), Umeå University, 901 87 Umeå, Sweden
| | - Jessica Wall
- Umeå Centre for Microbial Research (UCMR), Umeå University, 901 87 Umeå, Sweden; Department of Molecular Biology, Umeå University, 901 87 Umeå, Sweden; Molecular Infection Medicine, Sweden (MIMS), Umeå University, 901 87 Umeå, Sweden
| | - K Syam Krishnan
- Department of Chemistry, Umeå University, 901 87 Umeå, Sweden; Umeå Centre for Microbial Research (UCMR), Umeå University, 901 87 Umeå, Sweden
| | - Afshan Begum
- Department of Chemistry, Umeå University, 901 87 Umeå, Sweden; Umeå Centre for Microbial Research (UCMR), Umeå University, 901 87 Umeå, Sweden
| | - Christin Grundström
- Department of Chemistry, Umeå University, 901 87 Umeå, Sweden; Umeå Centre for Microbial Research (UCMR), Umeå University, 901 87 Umeå, Sweden
| | | | - Karolis Vaitkevicius
- Umeå Centre for Microbial Research (UCMR), Umeå University, 901 87 Umeå, Sweden; Department of Molecular Biology, Umeå University, 901 87 Umeå, Sweden; Molecular Infection Medicine, Sweden (MIMS), Umeå University, 901 87 Umeå, Sweden
| | - Erik Chorell
- Department of Chemistry, Umeå University, 901 87 Umeå, Sweden; Umeå Centre for Microbial Research (UCMR), Umeå University, 901 87 Umeå, Sweden
| | | | - Uwe H Sauer
- Department of Chemistry, Umeå University, 901 87 Umeå, Sweden; Umeå Centre for Microbial Research (UCMR), Umeå University, 901 87 Umeå, Sweden
| | - A Elisabeth Sauer-Eriksson
- Department of Chemistry, Umeå University, 901 87 Umeå, Sweden; Umeå Centre for Microbial Research (UCMR), Umeå University, 901 87 Umeå, Sweden.
| | - Fredrik Almqvist
- Department of Chemistry, Umeå University, 901 87 Umeå, Sweden; Umeå Centre for Microbial Research (UCMR), Umeå University, 901 87 Umeå, Sweden.
| | - Jörgen Johansson
- Umeå Centre for Microbial Research (UCMR), Umeå University, 901 87 Umeå, Sweden; Department of Molecular Biology, Umeå University, 901 87 Umeå, Sweden; Molecular Infection Medicine, Sweden (MIMS), Umeå University, 901 87 Umeå, Sweden.
| |
Collapse
|
45
|
Naidoo N, Pillay M. Bacterial pili, with emphasis on Mycobacterium tuberculosis curli pili: potential biomarkers for point-of care tests and therapeutics. Biomarkers 2016; 22:93-105. [PMID: 27797276 DOI: 10.1080/1354750x.2016.1252960] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
CONTEXT Novel biomarkers are essential for developing rapid diagnostics and therapeutic interventions Objective: This review aimed to highlight biomarker characterisation and assessment of unique bacterial pili. METHODS A PubMed search for bacterial pili, diagnostics, vaccine and therapeutics was performed, with emphasis on the well characterised pili. RESULTS In total, 46 papers were identified and reviewed. CONCLUSION Extensive analyses of pili enabled by advanced nanotechnology and whole genome sequencing provide evidence that they are strong biomarker candidates. Mycobacterium tuberculosis curli pili are emphasised as important epitopes for the development of much needed point-of-care diagnostics and therapeutics.
Collapse
Affiliation(s)
- Natasha Naidoo
- a Medical Microbiology and Infection Control , School of Laboratory Medicine and Medical Sciences, College of Health Science, University of KwaZulu-Natal, Congella , Durban , South Africa
| | - Manormoney Pillay
- a Medical Microbiology and Infection Control , School of Laboratory Medicine and Medical Sciences, College of Health Science, University of KwaZulu-Natal, Congella , Durban , South Africa
| |
Collapse
|
46
|
Oh YT, Kim HY, Kim EJ, Go J, Hwang W, Kim HR, Kim DW, Yoon SS. Selective and Efficient Elimination of Vibrio cholerae with a Chemical Modulator that Targets Glucose Metabolism. Front Cell Infect Microbiol 2016; 6:156. [PMID: 27900286 PMCID: PMC5111416 DOI: 10.3389/fcimb.2016.00156] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2016] [Accepted: 11/02/2016] [Indexed: 12/16/2022] Open
Abstract
Vibrio cholerae, a Gram-negative bacterium, is the causative agent of pandemic cholera. Previous studies have shown that the survival of the seventh pandemic El Tor biotype V. cholerae strain N16961 requires production of acetoin in a glucose-rich environment. The production of acetoin, a neutral fermentation end-product, allows V. cholerae to metabolize glucose without a pH drop, which is mediated by the production of organic acid. This finding suggests that inhibition of acetoin fermentation can result in V. cholerae elimination by causing a pH imbalance under glucose-rich conditions. Here, we developed a simple high-throughput screening method and identified an inducer of medium acidification (iMAC). Of 8364 compounds screened, we identified one chemical, 5-(4-chloro-2-nitrobenzoyl)-6-hydroxy-1,3-dimethylpyrimidine-2,4(1H,3H)-dione, that successfully killed glucose-metabolizing N16961 by inducing acidic stress. When N16961 was grown with abundant glucose in the presence of iMAC, acetoin production was completely suppressed and concomitant accumulation of lactate and acetate was observed. Using a beta-galactosidase activity assay with a single-copy palsD::lacZ reporter fusion, we show that that iMAC likely inhibits acetoin production at the transcriptional level. Thin-layer chromatography revealed that iMAC causes a significantly reduced accumulation of intracellular (p)ppGpp, a bacterial stringent response alarmone known to positively regulate acetoin production. In vivo bacterial colonization and fluid accumulation were also markedly decreased after iMAC treatment. Finally, we demonstrate iMAC-induced bacterial killing for 22 different V. cholerae strains belonging to diverse serotypes. Together, our results suggest that iMAC, acting as a metabolic modulator, has strong potential as a novel antibacterial agent for treatment against cholera.
Collapse
Affiliation(s)
- Young Taek Oh
- Department of Microbiology and Immunology, Yonsei University College of Medicine Seoul, South Korea
| | - Hwa Young Kim
- Department of Microbiology and Immunology, Yonsei University College of MedicineSeoul, South Korea; Brain Korea 21 Project for Medical Science, Yonsei University College of MedicineSeoul, South Korea
| | - Eun Jin Kim
- Department of Pharmacy, College of Pharmacy, Hanyang University Ansan, South Korea
| | - Junhyeok Go
- Department of Microbiology and Immunology, Yonsei University College of MedicineSeoul, South Korea; Brain Korea 21 Project for Medical Science, Yonsei University College of MedicineSeoul, South Korea
| | - Wontae Hwang
- Department of Microbiology and Immunology, Yonsei University College of MedicineSeoul, South Korea; Brain Korea 21 Project for Medical Science, Yonsei University College of MedicineSeoul, South Korea
| | - Hyoung Rae Kim
- Bio and Drug Discovery Division, Korea Research Institute of Chemical Technology Daejeon, South Korea
| | - Dong Wook Kim
- Department of Pharmacy, College of Pharmacy, Hanyang UniversityAnsan, South Korea; Institute of Pharmacological Research, Hanyang UniversityAnsan, South Korea
| | - Sang Sun Yoon
- Department of Microbiology and Immunology, Yonsei University College of MedicineSeoul, South Korea; Brain Korea 21 Project for Medical Science, Yonsei University College of MedicineSeoul, South Korea; Institute for Immunology and Immunological Diseases, Yonsei University College of MedicineSeoul, South Korea
| |
Collapse
|
47
|
Li J, Wehmeyer G, Lovell S, Battaile KP, Egan SM. 1.65 Å resolution structure of the AraC-family transcriptional activator ToxT from Vibrio cholerae. Acta Crystallogr F Struct Biol Commun 2016; 72:726-31. [PMID: 27599865 PMCID: PMC5012214 DOI: 10.1107/s2053230x1601298x] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2016] [Accepted: 08/11/2016] [Indexed: 11/24/2022] Open
Abstract
ToxT is an AraC-family transcriptional activator protein that controls the expression of key virulence factors in Vibrio cholerae, the causative agent of cholera. ToxT directly activates the expression of the genes that encode the toxin-coregulated pilus and cholera toxin, and also positively auto-regulates its own expression from the tcp promoter. The crystal structure of ToxT has previously been solved at 1.9 Å resolution (PDB entry 3gbg). In this study, a crystal structure of ToxT at 1.65 Å resolution with a similar overall structure to the previously determined structure is reported. However, there are distinct differences between the two structures, particularly in the region that extends from Asp101 to Glu110. This region, which can influence ToxT activity but was disordered in the previous structure, can be traced entirely in the current structure.
Collapse
Affiliation(s)
- Jiaqin Li
- Department of Molecular Bioscience, The University of Kansas, 8031 Haworth, 1200 Sunnyside Avenue, Lawrence, KS 66045, USA
| | - Graham Wehmeyer
- Department of Molecular Bioscience, The University of Kansas, 8031 Haworth, 1200 Sunnyside Avenue, Lawrence, KS 66045, USA
| | - Scott Lovell
- Protein Structure Laboratory, Shankel Structural Biology Center, The University of Kansas, 2034 Becker Drive, Lawrence, KS 66047, USA
| | - Kevin P. Battaile
- IMCA-CAT, Hauptman–Woodward Medical Research Institute, 9700 South Cass Avenue, Building 435A, Argonne, IL 60439, USA
| | - Susan M. Egan
- Department of Molecular Bioscience, The University of Kansas, 8031 Haworth, 1200 Sunnyside Avenue, Lawrence, KS 66045, USA
| |
Collapse
|
48
|
Sun YY, Chi H, Sun L. Pseudomonas fluorescens Filamentous Hemagglutinin, an Iron-Regulated Protein, Is an Important Virulence Factor that Modulates Bacterial Pathogenicity. Front Microbiol 2016; 7:1320. [PMID: 27602029 PMCID: PMC4993755 DOI: 10.3389/fmicb.2016.01320] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2016] [Accepted: 08/10/2016] [Indexed: 11/16/2022] Open
Abstract
Pseudomonas fluorescens is a common bacterial pathogen to a wide range of aquaculture animals including various species of fish. In this study, we employed proteomic analysis and identified filamentous hemagglutinin (FHA) as an iron-responsive protein secreted by TSS, a pathogenic P. fluorescens isolate. In vitro study showed that compared to the wild type, the fha mutant TSSfha (i) exhibited a largely similar vegetative growth profile but significantly retarded in the ability of biofilm growth and producing extracellular matrix, (ii) displayed no apparent flagella and motility, (iii) was defective in the attachment to host cells and unable to form self-aggregation, (iv) displayed markedly reduced capacity of hemagglutination and surviving in host serum. In vivo infection analysis revealed that TSSfha was significantly attenuated in the ability of dissemination in fish tissues and inducing host mortality, and that antibody blocking of the natural FHA produced by the wild type TSS impaired the infectivity of the pathogen. Furthermore, when introduced into turbot as a subunit vaccine, recombinant FHA elicited a significant protection against lethal TSS challenge. Taken together, these results indicate for the first time that P. fluorescens FHA is a key virulence factor essential to multiple biological processes associated with pathogenicity.
Collapse
Affiliation(s)
- Yuan-Yuan Sun
- Key Laboratory of Experimental Marine Biology, Institute of Oceanology - Chinese Academy of SciencesQingdao, China; Laboratory for Marine Biology and Biotechnology, Qingdao National Laboratory for Marine Science and TechnologyQingdao, China; University of Chinese Academy of SciencesBeijing, China
| | - Heng Chi
- Key Laboratory of Experimental Marine Biology, Institute of Oceanology - Chinese Academy of SciencesQingdao, China; Laboratory for Marine Biology and Biotechnology, Qingdao National Laboratory for Marine Science and TechnologyQingdao, China
| | - Li Sun
- Key Laboratory of Experimental Marine Biology, Institute of Oceanology - Chinese Academy of SciencesQingdao, China; Laboratory for Marine Biology and Biotechnology, Qingdao National Laboratory for Marine Science and TechnologyQingdao, China
| |
Collapse
|
49
|
Reffuveille F, Nicol M, Dé E, Thébault P. Design of an anti-adhesive surface by a pilicide strategy. Colloids Surf B Biointerfaces 2016; 146:895-901. [PMID: 27469573 DOI: 10.1016/j.colsurfb.2016.07.037] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2016] [Revised: 07/01/2016] [Accepted: 07/19/2016] [Indexed: 01/08/2023]
Abstract
Biofilm formation on surfaces is one of major problems in medical, cosmetic and food industries. Nowadays any efficient treatment is known, as consequence, research of new strategies to inhibit biofilm formation is urgent. Recently, virstatin, which interferes with bacterial type IV pili formation, has demonstrated a capacity to inhibit biofilm formation developed by Acinetobacter baumannii after 24h. In this study, we aim to elaborate anti-adhesive surfaces preventing biofilm development by the covalent immobilization of virstatin on silicon surface. Surfaces were functionalized by self-assembled monolayers of two aminosilanes (11-aminoundecyltrimethoxysilane (AUTMS) and 3-aminopropyltrimethoxysilane (APTMS)). Then, virstatin (2mM) was immobilized on those modified surfaces. We observed an increase in surface hydrophobicity of AUTMS modified substratum leading to an increase of A. baumannii ATCC 17978 adhesion (after 4h). Immobilization of virstatin molecule on APTMS modified surface was efficient to decrease cell attachment by 32.1±5.7% compared to unmodified surface. As virstatin is known to inhibit type IV pili formation in solution, the observed decrease of bacterial adhesion might be due to this pilicide action. We also demonstrated that hydrophobicity of strains plays a role in adhesion according to surface properties. In conclusion, immobilized virstatin succeeded to inhibit bacterial attachment of various Acinetobacter baumannii strains comparing to APTMS modified support.
Collapse
Affiliation(s)
- Fany Reffuveille
- Normandie Univ, UNIROUEN, INSA Rouen, CNRS, PBS, 76000 Rouen, France
| | - Marion Nicol
- Normandie Univ, UNIROUEN, INSA Rouen, CNRS, PBS, 76000 Rouen, France
| | - Emmanuelle Dé
- Normandie Univ, UNIROUEN, INSA Rouen, CNRS, PBS, 76000 Rouen, France
| | - Pascal Thébault
- Normandie Univ, UNIROUEN, INSA Rouen, CNRS, PBS, 76000 Rouen, France.
| |
Collapse
|
50
|
Bolger G, Roy S, Zapol'skii VA, Kaufmann DE, Schnürch M, Mihovilovic MD, Nandy RK, Tegge W. Targeting aphA : a new high-throughput screening assay identifies compounds that reduce prime virulence factors of Vibrio cholerae. J Med Microbiol 2016; 65:678-687. [DOI: 10.1099/jmm.0.000276] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Affiliation(s)
- Galina Bolger
- Department of Chemical Biology, Helmholtz Centre for Infection Research (HZI), Braunschweig, Germany
| | - Sambit Roy
- Division of Bacteriology, National Institute of Cholera and Enteric Diseases (NICED), Kolkata, India
| | - Viktor A. Zapol'skii
- Institute of Organic Chemistry, Technical University of Clausthal, Clausthal-Zellerfeld, Germany
| | - Dieter E. Kaufmann
- Institute of Organic Chemistry, Technical University of Clausthal, Clausthal-Zellerfeld, Germany
| | - Michael Schnürch
- Institute of Applied Synthetic Chemistry, Vienna University of Technology, Vienna, Austria
| | - Marko D. Mihovilovic
- Institute of Applied Synthetic Chemistry, Vienna University of Technology, Vienna, Austria
| | - Ranjan K. Nandy
- Division of Bacteriology, National Institute of Cholera and Enteric Diseases (NICED), Kolkata, India
| | - Werner Tegge
- Department of Chemical Biology, Helmholtz Centre for Infection Research (HZI), Braunschweig, Germany
| |
Collapse
|