1
|
Moradkasani S, Esmaeili S, Asadi Karam MR, Mostafavi E, Shahbazi B, Salek Farrokhi A, Chiani M, Badmasti F. Development of a multi-epitope vaccine from outer membrane proteins and identification of novel drug targets against Francisella tularensis: an In Silico approach. Front Immunol 2025; 16:1479862. [PMID: 40248715 PMCID: PMC12003292 DOI: 10.3389/fimmu.2025.1479862] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Accepted: 03/17/2025] [Indexed: 04/19/2025] Open
Abstract
Background Francisella tularensis is a category A potential thread agent, making the development of vaccines and countermeasures a high priority. Therefore, identifying new vaccine candidates and novel drug targets is essential for addressing this significant public health concern. Methods This study presents an in silico analysis of two strategies against F. tularensis infection: the development of a multi-epitope vaccine (MEV) and the identification of novel drug targets. Outer membrane proteins (OMPs) were predicted using subcellular localization tools and immunogenicity was evaluated using a reverse vaccinology pipeline. Epitopes from these OMPs were combined to create candidate MEV for prophylactic protection. Concurrently, cytoplasmic proteins were subjected to rigorous analysis to identify potential novel drug targets. Results Of 1,921 proteins, we identified 12 promising protein vaccine candidates from F. tularensis OMPs and proposed a multi-epitope vaccine (MEV) designed using seven immunodominant epitopes derived from four of these OMPs, including two hypothetical proteins (WP_003026145.1 and WP_003029346.1), an OmpA family protein (WP_003020808.1), and PD40 (WP_003021546.1). In addition, we proposed 10 novel drug targets for F. tularensis: Asp-tRNA (Asn)/Glu-tRNA (Gln) amidotransferase subunit GatC (WP_003017413.1), NAD(P)-binding protein (WP_042522581.1), 30S ribosomal protein S16 (WP_003023081.1), Class I SAM-dependent methyltransferase (WP_003022345.1), haloacid dehalogenase (WP_003014157.1), uroporphyrinogen-III synthase (WP_003022220.1), and four hypothetical proteins (WP_003017784.1, WP_003020080.1, WP_003020066.1, and WP_003022350.1). Conclusion This study designed an MEV and proposed novel drug targets to address tularemia, offering broad protection against various F. tularensis strains. MEV, with favorable physicochemical properties, showed strong potential through molecular docking and dynamic simulations. Immune simulations suggest that it may elicit robust responses against pathogens. The identification of novel drug targets can lead to the discovery of new antimicrobial agents. However, further in vitro and in vivo studies are required to validate their efficacy and capability.
Collapse
Affiliation(s)
- Safoura Moradkasani
- WHO Collaborating Centre for Vector-Borne Diseases, Department of Epidemiology and Biostatistics, Research Centre for Emerging and Reemerging Infectious Diseases, Pasteur Institute of Iran, Tehran, Iran
- Student Research Committee, Pasteur Institute of Iran, Tehran, Iran
| | - Saber Esmaeili
- WHO Collaborating Centre for Vector-Borne Diseases, Department of Epidemiology and Biostatistics, Research Centre for Emerging and Reemerging Infectious Diseases, Pasteur Institute of Iran, Tehran, Iran
- Student Research Committee, Pasteur Institute of Iran, Tehran, Iran
- National Reference Laboratory for Plague, Tularemia and Q Fever, Research Centre for Emerging and Reemerging Infectious Diseases, Pasteur Institute of Iran, Akanlu, KabudarAhang, Hamadan, Iran
| | | | - Ehsan Mostafavi
- WHO Collaborating Centre for Vector-Borne Diseases, Department of Epidemiology and Biostatistics, Research Centre for Emerging and Reemerging Infectious Diseases, Pasteur Institute of Iran, Tehran, Iran
- National Reference Laboratory for Plague, Tularemia and Q Fever, Research Centre for Emerging and Reemerging Infectious Diseases, Pasteur Institute of Iran, Akanlu, KabudarAhang, Hamadan, Iran
| | - Behzad Shahbazi
- School of Pharmacy, Semnan University of Medical Sciences, Semnan, Iran
- Nervous System Stem Cells Research Center, Semnan University of Medical Sciences, Semnan, Iran
| | | | - Mohsen Chiani
- Department of Nanobiotechnology, Pasteur Institute of Iran, Tehran, Iran
| | - Farzad Badmasti
- Department of Bacteriology, Pasteur Institute of Iran, Tehran, Iran
| |
Collapse
|
2
|
Gaur V, Kumar N, Vyas A, Chowdhury D, Singh J, Bera S. Identification of potential inhibitors against Escherichia coli Mur D enzyme to combat rising drug resistance: an in-silico approach. J Biomol Struct Dyn 2025; 43:3286-3296. [PMID: 38149858 DOI: 10.1080/07391102.2023.2297007] [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: 05/02/2023] [Accepted: 12/13/2023] [Indexed: 12/28/2023]
Abstract
Indiscriminate use of anti-microbial agents has resulted in the inception, frequency, and spread of antibiotic resistance among targeted bacterial pathogens and the commensal flora. Mur enzymes, playing a crucial role in cell-wall synthesis, are one of the most appropriate targets for developing novel inhibitors against antibiotic-resistant bacterial pathogens. In the present study, in-silico high-throughput virtual (HTVS) and Standard-Precision (SP) screening was carried out with 0.3 million compounds from several small-molecule libraries against the E. coli Mur D enzyme (PDB ID 2UUP). The docked complexes were further subjected to extra-precision (XP) docking calculations, and highest Glide-score compound was further subjected to molecular simulation studies. The top six virtual hits (S1-S6) displayed a glide score (G-score) within the range of -9.013 to -7.126 kcal/mol and compound S1 was found to have the highest stable interactions with the Mur D enzyme (2UUP) of E. coli. The stability of compound S1 with the Mur D (2UUP) complex was validated by a 100-ns molecular dynamics simulation. Binding free energy calculation by the MM-GBSA strategy of the S1-2UUP (Mur D) complex established van der Waals, hydrogen bonding, lipophilic, and Coulomb energy terms as significant favorable contributors for ligand binding. The final lead molecules were subjected to ADMET predictions to study their pharmacokinetic properties and displayed promising results, except for certain modifications required to improve QPlogHERG values. So, the compounds screened against the Mur D enzyme can be further studied as preparatory points for in-vivo studies to develop potential drugs. HIGHLIGHTSE.coli is a common cause of urinary tract infections.E.coli MurD enzyme is a suitable target for drug development.Novel inhibitors against E.coli MurD enzyme were identified.Molecular dynamics studies identified in-silico potential of identified compound.ADMET predictions and Lipinski's rule of five studies showed promising results.
Collapse
Affiliation(s)
- Vinita Gaur
- Department of Microbiology, School of Bioengineering and Biosciences, Lovely Professional University, Punjab, India
| | - Neeraj Kumar
- Department of Pharmaceutical Chemistry, Bhupal Nobles' University, Udaipur, Rajasthan, India
| | - Ashish Vyas
- Department of Microbiology, School of Bioengineering and Biosciences, Lovely Professional University, Punjab, India
| | - Debabrata Chowdhury
- School of Medicine - Infectious Diseases, Stanford University, Stanford, CA, USA
| | - Joginder Singh
- Department of Microbiology, School of Bioengineering and Biosciences, Lovely Professional University, Punjab, India
| | - Surojit Bera
- Department of Microbiology, School of Bioengineering and Biosciences, Lovely Professional University, Punjab, India
| |
Collapse
|
3
|
Shastri T, Binsuwaidan R, Siddiqui AJ, Badraoui R, Jahan S, Alshammari N, Adnan M, Patel M. Quercetin Exhibits Broad-Spectrum Antibiofilm and Antiquorum Sensing Activities Against Gram-Negative Bacteria: In Vitro and In Silico Investigation Targeting Antimicrobial Therapy. THE CANADIAN JOURNAL OF INFECTIOUS DISEASES & MEDICAL MICROBIOLOGY = JOURNAL CANADIEN DES MALADIES INFECTIEUSES ET DE LA MICROBIOLOGIE MEDICALE 2025; 2025:2333207. [PMID: 40196379 PMCID: PMC11972862 DOI: 10.1155/cjid/2333207] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Accepted: 03/11/2025] [Indexed: 04/09/2025]
Abstract
Quercetin (QC), a flavonoid abundant in fruits and vegetables, has garnered attention for its potential therapeutic properties. In this study, we investigated the antibiofilm and antiquorum sensing (QS) activities of QC against Gram-negative bacteria both in vitro and in silico. The findings of this study demonstrate MIC values of 125 μg/mL for Chromobacterium violaceum, 250 μg/mL for Pseudomonas aeruginosa, and 500 μg/mL for Serratia marcescens, indicating its antibacterial potential abilities. QS-mediated production of violacein and prodigiosin was significantly inhibited in a dose-dependent manner at sub-MIC concentrations. Additionally, a dose-dependent reduction in the virulence factors of P. aeruginosa, including production of pyocyanin, pyoverdine, and rhamnolipid, was noted with QC. Biofilm formation decreased by 66.40%, 59.28%, and 63.70% at the highest sub-MIC for C. violaceum, P. aeruginosa, and S. marcescens, respectively. Furthermore, swimming motility and exopolysaccharide (EPS) production were also reduced in the presence of QC. Additionally, molecular docking and molecular dynamics simulations elucidate the binding interactions between QC and key molecular targets (LasI, LasR, PilY1, LasA, PilT, CviR, CviR', PqsR, RhlR, and PigG) involved in biofilm formation and QS pathways. Our results indicated that the antibiofilm and anti-QS sensing activities of QC may be attributed to its ability to interfere with critical signaling molecules and regulatory proteins. Overall, this study highlights QC as a promising natural compound for combating biofilm-associated infections caused by Gram-negative bacteria. The multifaceted antimicrobial mechanisms of QC underscore its potential as a therapeutic agent for the treatment of biofilm-related infections, providing the way for further exploration, and development of QC-based strategies in antimicrobial therapy.
Collapse
Affiliation(s)
- Tanvi Shastri
- Department of Microbiology, Parul Institute of Applied Sciences, Parul University, Waghodia, Vadodara, Gujarat 391760, India
| | - Reem Binsuwaidan
- Department of Pharmaceutical Sciences, College of Pharmacy, Princess Nourah bint Abdulrahman University, P.O. Box 84428, Riyadh 11671, Saudi Arabia
| | - Arif Jamal Siddiqui
- Department of Biology, College of Science, University of Ha'il, P.O. Box 2440, Ha'il, Saudi Arabia
| | - Riadh Badraoui
- Department of Biology, College of Science, University of Ha'il, P.O. Box 2440, Ha'il, Saudi Arabia
| | - Sadaf Jahan
- Department of Medical Laboratory Sciences, College of Applied Medical Sciences, Majmaah University, Majmaah 11952, Saudi Arabia
| | - Nawaf Alshammari
- Department of Biology, College of Science, University of Ha'il, P.O. Box 2440, Ha'il, Saudi Arabia
| | - Mohd Adnan
- Department of Biology, College of Science, University of Ha'il, P.O. Box 2440, Ha'il, Saudi Arabia
| | - Mitesh Patel
- Research and Development Cell (RDC), Parul University, Waghodia, Vadodara, Gujarat 391760, India
- Department of Biotechnology, Parul Institute of Applied Sciences, Parul University, Waghodia, Vadodara, Gujarat 391760, India
| |
Collapse
|
4
|
Ghosh S, Basu S, Anbarasu A, Ramaiah S. A Comprehensive Review of Antimicrobial Agents Against Clinically Important Bacterial Pathogens: Prospects for Phytochemicals. Phytother Res 2025; 39:138-161. [PMID: 39496516 DOI: 10.1002/ptr.8365] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Revised: 09/08/2024] [Accepted: 09/19/2024] [Indexed: 11/06/2024]
Abstract
Antimicrobial resistance (AMR) hinders the effective treatment of a range of bacterial infections, posing a serious threat to public health globally, as it challenges the currently available antimicrobial drugs. Among the various modes of antimicrobial action, antimicrobial agents that act on membranes have the most promising efficacy. However, there are no consolidated reports on the shortcomings of these drugs, existing challenges, or the potential applications of phytochemicals that act on membranes. Therefore, in this review, we have addressed the challenges and focused on various phytochemicals as antimicrobial agents acting on the membranes of clinically important bacterial pathogens. Antibacterial phytochemicals comprise diverse group of agents found in a wide range of plants. These compounds have been found to disrupt cell membranes, inhibit enzymes, interfere with protein synthesis, generate reactive oxygen species, modulate quorum sensing, and inhibit bacterial adhesion, making them promising candidates for the development of novel antibacterial therapies. Recently, polyphenolic compounds have been reported to have proven efficacy against nosocomial multidrug-resistant pathogens. However, more high-quality studies, improved standards, and the adoption of rules and regulations are required to firmly confirm the clinical efficacy of phytochemicals derived from plants. Identifying potential challenges, thrust areas of research, and considering viable approaches is essential for the successful clinical translation of these compounds.
Collapse
Affiliation(s)
- Soumyadip Ghosh
- Medical and Biological Computing Laboratory, School of Biosciences and Technology (SBST), Vellore Institute of Technology (VIT), Vellore, India
- Department of Bio Sciences, SBST, VIT, Vellore, India
| | - Soumya Basu
- Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
- Department of Biotechnology, National Institute of Science and Technology (NIST), Berhampur, India
| | - Anand Anbarasu
- Medical and Biological Computing Laboratory, School of Biosciences and Technology (SBST), Vellore Institute of Technology (VIT), Vellore, India
- Department of Biotechnology, SBST, VIT, Vellore, India
| | - Sudha Ramaiah
- Medical and Biological Computing Laboratory, School of Biosciences and Technology (SBST), Vellore Institute of Technology (VIT), Vellore, India
- Department of Bio Sciences, SBST, VIT, Vellore, India
| |
Collapse
|
5
|
Rai P, Mehrotra S, Prajapati VK. Exploring immunotherapy to control human infectious diseases. ADVANCES IN PROTEIN CHEMISTRY AND STRUCTURAL BIOLOGY 2024; 144:389-429. [PMID: 39978973 DOI: 10.1016/bs.apcsb.2024.10.010] [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: 02/22/2025]
Abstract
Infectious diseases continue to pose significant challenges to global health, especially with the rise of antibiotic resistance and emerging pathogens. Traditional treatments, while effective, are often limited in the face of rapidly evolving pathogens. Immunotherapy, which harnesses and enhances the body's immune response, offers a promising alternative to conventional approaches for the treatment of infectious diseases. By employing use of monoclonal antibodies, vaccines, cytokine therapies, and immune checkpoint inhibitors, immunotherapy has demonstrated considerable potential in overcoming treatment resistance and improving patient outcomes. Key innovations, including the development of mRNA vaccines, use of immune modulators, adoptive cell transfer, and chimeric antigen receptor (CAR)-T cell therapy are paving the way for more targeted pathogen clearance. Further, combining immunotherapy with conventional antibiotic treatment has demonstrated effectiveness against drug-resistant strains, but this chapter explores the evolving field of immunotherapy for the treatment of bacterial, viral, fungal, and parasitic infections. The chapter also explores the recent breakthroughs and ongoing clinical trials in infectious disease immunotherapy.
Collapse
Affiliation(s)
- Praveen Rai
- Department of Biochemistry, University of Delhi South Campus, Benito Juarez Road, Dhaula Kuan, New Delhi, India
| | - Sanjana Mehrotra
- Department of Human Genetics, Guru Nanak Dev University, Amritsar, Punjab, India
| | - Vijay Kumar Prajapati
- Department of Biochemistry, University of Delhi South Campus, Benito Juarez Road, Dhaula Kuan, New Delhi, India.
| |
Collapse
|
6
|
Shweta, Chahal S, Kumar Dhaka R, Rana A, Joshi G, Singh R, Singh S, Singh D, Kumar P, Sindhu J. Multicomponent Synthesis of 2,4,5-Trisubstituted Thiazoles Using a Sustainable Carbonaceous Catalyst and Assessment of Its Herbicidal and Antibacterial Potential. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024; 72:27762-27774. [PMID: 39630023 DOI: 10.1021/acs.jafc.4c05293] [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: 12/19/2024]
Abstract
Herein, a novel, biocatalyzed, and on-water microwave-assisted multicomponent methodology have been developed for the synthesis of trisubstituted thiazoles (4a-4v). The reaction was catalyzed using a sulfonated peanut shell residue-derived carbonaceous catalyst (SPWB). The developed catalyst was characterized using Fourier transform infrared (FTIR), a Brunauer-Emmett-Teller (BET) surface area analyzer, a field emission scanning electron microscope (FE-SEM), energy-dispersive X-ray (EDX), and a particle size analyzer (PSA). The acidic sites have been established using acid-base back-titration methods. The molecular structures of all the synthesized compounds were validated using FT-IR, 1H NMR, 13C NMR, elemental, and HRMS analyses. Herbicidal potential was evaluated by using Raphanus sativus L. as a model. Furthermore, the antibacterial potential of thiazoles was evaluated against Staphylococcus aureus, Bacillus subtilis, Xanthomonas campestris, Escherichia coli, Micrococcus luteus, and Pseudomonas aeruginosa bacterial strains. The compound 4r displayed improved seed growth inhibition in Raphanus sativus L. versus a commercially available herbicide, i.e., pendimethalin. The antibacterial activity was promising against bacterial strains (MIC: 4-64 μg/mL). The compound 4r was the most potent against P. aeruginosa and S. aureus (MIC: 0.0076 μM) versus standard drug streptomycin (MIC: 0.0138 μM). Moreover, in silico studies performed with the most effective compound 4r against P. aeruginosa revealed its potential binding mode within the protein binding pocket. The biological data revealed compound 4r as a potential candidate for the development of potent herbicidal and antibacterial agents. In a nutshell, this study offers peanut shell biowaste to be a sustainable biomass for heterogeneous acid catalyst preparation and its application in the multicomponent synthesis of bioactive thiazoles, accommodating the concept of sustainable development goals and circular bioeconomy.
Collapse
Affiliation(s)
- Shweta
- Department of Chemistry, COBS&H, CCS Haryana Agricultural University, Hisar 125004, India
| | - Sandhya Chahal
- Department of Chemistry, COBS&H, CCS Haryana Agricultural University, Hisar 125004, India
| | - Rahul Kumar Dhaka
- Department of Chemistry, COBS&H, CCS Haryana Agricultural University, Hisar 125004, India
| | - Anuj Rana
- Department of Microbiology, COBS&H, CCS Haryana Agricultural University, Hisar 125004, India
| | - Gaurav Joshi
- Department of Pharmaceutical Sciences, Hemvati Nandan Bahuguna Garhwal University (Central University), Srinagar, Dist. Garhwal (Uttarakhand 246174, India
| | - Rajvir Singh
- Department of Chemistry, COBS&H, CCS Haryana Agricultural University, Hisar 125004, India
| | - Snigdha Singh
- Department of Chemistry, University of Delhi, Delhi 110007, India
| | - Devender Singh
- Department of Chemistry, Maharshi Dayanand University, Rohtak 124001, India
| | - Parvin Kumar
- Department of Chemistry, Kurukshetra University, Kurukshetra 136119, India
| | - Jayant Sindhu
- Department of Chemistry, COBS&H, CCS Haryana Agricultural University, Hisar 125004, India
| |
Collapse
|
7
|
Obong’o BO, Ogutu FO, Hurley SK, Okiko GM, Mahony J. Exploring the Microbial Ecology of Water in Sub-Saharan Africa and the Potential of Bacteriophages in Water Quality Monitoring and Treatment to Improve Its Safety. Viruses 2024; 16:1897. [PMID: 39772204 PMCID: PMC11680409 DOI: 10.3390/v16121897] [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: 11/06/2024] [Revised: 12/04/2024] [Accepted: 12/06/2024] [Indexed: 01/04/2025] Open
Abstract
Access to safe water and food is a critical issue in sub-Saharan Africa, where microbial contamination poses significant health risks. Conventional water treatment and food preservation methods have limitations in addressing water safety, particularly for antibiotic-resistant bacteria and other pathogenic microorganisms. This review explores the potential application of bacteriophages as an innovative solution for water treatment and food safety in the region. Bacteriophages specifically infect bacteria and offer a targeted approach to reducing bacterial load, including multidrug-resistant strains, without the drawbacks of chemical disinfectants. This review also highlights the advantages of phage bioremediation, including its specificity, adaptability, and minimal environmental impact. It also discusses various case studies demonstrating its efficacy in different water systems. Additionally, we underscore the need for further research and the development of region-specific phage applications to improve water quality and public health outcomes in sub-Saharan Africa. By integrating bacteriophage strategies into water treatment and food production, the region can address critical microbial threats, mitigate the spread of antimicrobial resistance, and advance global efforts toward ensuring safe water for all.
Collapse
Affiliation(s)
- Boniface Oure Obong’o
- Food Technology Division, Kenya Industrial Research and Development Institute (KIRDI), Nairobi P.O. Box 30650-00100, Kenya; (B.O.O.); (G.M.O.)
| | - Fredrick Onyango Ogutu
- Food Technology Division, Kenya Industrial Research and Development Institute (KIRDI), Nairobi P.O. Box 30650-00100, Kenya; (B.O.O.); (G.M.O.)
| | - Shauna Kathleen Hurley
- APC Microbiome Ireland, School of Microbiology, University College Cork, College Road, T12 K8AF Cork, Ireland;
| | - Gertrude Maisiba Okiko
- Food Technology Division, Kenya Industrial Research and Development Institute (KIRDI), Nairobi P.O. Box 30650-00100, Kenya; (B.O.O.); (G.M.O.)
| | - Jennifer Mahony
- APC Microbiome Ireland, School of Microbiology, University College Cork, College Road, T12 K8AF Cork, Ireland;
| |
Collapse
|
8
|
Nazeer RR, Askenasy I, Swain JEV, Welch M. Contribution of the infection ecosystem and biogeography to antibiotic failure in vivo. NPJ ANTIMICROBIALS AND RESISTANCE 2024; 2:45. [PMID: 39649078 PMCID: PMC11618093 DOI: 10.1038/s44259-024-00063-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Accepted: 11/11/2024] [Indexed: 12/10/2024]
Abstract
The acquisition of antibiotic resistance in bacteria, though a deeply concerning international issue, is reasonably well-understood at a mechanistic level. Less well-understood is why bacteria that are sensitive in vitro to well-established and widely-used antibiotics sometimes fail to respond to these agents in vivo. This is a particularly common problem in chronic, polymicrobial infection scenarios. Here, we discuss this in vitro-in vivo disconnect from the perspective of the bacterium, focusing in particular on how infection micro/macro-environment, biogeography, and the presence of co-habiting species affect the response to antibiotics. Using selected exemplars, we also consider interventions that might improve treatment outcomes, as well as ecologically 'eubiotic' approaches that have less of an impact on the patient's commensal microflora. In our view, the accrued data strongly suggest that we need a more comprehensive understanding of the in situ microbiology at infection sites.
Collapse
Affiliation(s)
| | - Isabel Askenasy
- Department of Biochemistry, University of Cambridge, Cambridge, UK
| | | | - Martin Welch
- Department of Biochemistry, University of Cambridge, Cambridge, UK
| |
Collapse
|
9
|
Kamboyi HK, Paudel A, Shawa M, Sugawara M, Zorigt T, Chizimu JY, Kitao T, Furuta Y, Hang'ombe BM, Munyeme M, Higashi H. EsxA, a type VII secretion system-dependent effector, reveals a novel function in the sporulation of Bacillus cereus ATCC14579. BMC Microbiol 2024; 24:351. [PMID: 39289639 PMCID: PMC11406982 DOI: 10.1186/s12866-024-03492-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Accepted: 09/03/2024] [Indexed: 09/19/2024] Open
Abstract
BACKGROUND Bacillus cereus is a Gram-positive, spore-forming bacterium that produces a spectrum of effectors integral to bacterial niche adaptation and the development of various infections. Among those is EsxA, whose secretion depends on the EssC component of the type VII secretion system (T7SS). EsxA's roles within the bacterial cell are poorly understood, although postulations indicate that it may be involved in sporulation. However, the T7SS repertoire in B. cereus has not been reported, and its functions are unestablished. METHODS We used the type strain, B. cereus ATCC14579, to generate ΔessC mutant through homologous recombination using the homing endonuclease I-SceI mediated markerless gene replacement. Comparatively, we analyzed the culture supernatant of type strain and the ΔessC mutant through Liquid chromatography-tandem mass spectrometry (LC-MS/MS). We further generated T7SSb-specific gene mutations to explore the housekeeping roles of the T7SSb-dependent effectors. The sporulation process of B. cereus ATCC14579 and its mutants was observed microscopically through the classic Schaeffer-Fulton staining method. The spore viability of each strain in this study was established by enumerating the colony-forming units on LB agar. RESULTS Through LC-MS/MS, we identified a pair of nearly identical (94%) effector proteins named EsxA belonging to the sagEsxA-like subfamily of the WXG100 protein superfamily in the culture supernatant of the wild type and none in the ΔessC mutant. Homology analysis of the T7SSb gene cluster among B. cereus strains revealed diversity from the 3' end of essC, encoding additional substrates. Deletions in esxA1 and esxA2 neither altered cellular morphology nor growth rate, but the ΔesxA1ΔesxA2 deletion resulted in significantly fewer viable spores and an overall slower sporulation process. Within 24 h culture, more than 80% of wild-type cells formed endospores compared to less than 5% in the ΔesxA1ΔesxA2 mutant. The maximum spore ratios for the wild type and ΔesxA1ΔesxA2 were 0.96 and 0.72, respectively. Altogether, these results indicated that EsxA1 and EsxA2 work cooperatively and are required for sporulation in B. cereus ATCC14567. CONCLUSION B. cereus ATCC14579 possesses two nearly identical T7SSb-dependent effectors belonging to the sagEsxA-like proteins. Simultaneous deletion of genes encoding these effectors significantly delayed and reduced sporulation, a novel finding for EsxA.
Collapse
Affiliation(s)
- Harvey K Kamboyi
- Division of Infection and Immunity, International Institute for Zoonosis Control, Hokkaido University, Sapporo, Japan
| | - Atmika Paudel
- Division of Infection and Immunity, International Institute for Zoonosis Control, Hokkaido University, Sapporo, Japan
- GenEndeavor LLC, 26219 Eden Landing Rd, Hayward, CA, 94545, USA
| | - Misheck Shawa
- Division of Infection and Immunity, International Institute for Zoonosis Control, Hokkaido University, Sapporo, Japan
- Hokudai Center for Zoonosis Control in Zambia, University of Zambia, Lusaka, Zambia
| | - Misa Sugawara
- Division of Infection and Immunity, International Institute for Zoonosis Control, Hokkaido University, Sapporo, Japan
| | - Tuvshinzaya Zorigt
- Division of Infection and Immunity, International Institute for Zoonosis Control, Hokkaido University, Sapporo, Japan
| | - Joseph Y Chizimu
- International Collaboration Unit, International Institute for Zoonosis Control, Hokkaido University, Sapporo, Japan
- Zambia National Public Health Institute, Ministry of Health, Lusaka, Zambia
| | - Tomoe Kitao
- Division of Infection and Immunity, International Institute for Zoonosis Control, Hokkaido University, Sapporo, Japan
| | - Yoshikazu Furuta
- Division of Infection and Immunity, International Institute for Zoonosis Control, Hokkaido University, Sapporo, Japan
| | - Bernard M Hang'ombe
- Microbiology Unit, Paraclinical Studies, School of Veterinary Medicine, University of Zambia, Lusaka, Zambia
| | - Musso Munyeme
- Public Health Unit, Disease Control Studies, School of Veterinary Medicine, University of Zambia, Lusaka, Zambia
| | - Hideaki Higashi
- Division of Infection and Immunity, International Institute for Zoonosis Control, Hokkaido University, Sapporo, Japan.
| |
Collapse
|
10
|
Gong W, Wang ML, Liu Y, Yu DG, Bligh SWA. Shell Distribution of Vitamin K3 within Reinforced Electrospun Nanofibers for Improved Photo-Antibacterial Performance. Int J Mol Sci 2024; 25:9556. [PMID: 39273503 PMCID: PMC11394794 DOI: 10.3390/ijms25179556] [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: 08/05/2024] [Revised: 08/29/2024] [Accepted: 08/30/2024] [Indexed: 09/15/2024] Open
Abstract
Personal protective equipment (PPE) has attracted more attention since the outbreak of the epidemic in 2019. Advanced nano techniques, such as electrospinning, can provide new routes for developing novel PPE. However, electrospun antibacterial PPE is not easily obtained. Fibers loaded with photosensitizers prepared using single-fluid electrospinning have a relatively low utilization rate due to the influence of embedding and their inadequate mechanical properties. For this study, monolithic nanofibers and core-shell nanofibers were prepared and compared. Monolithic F1 fibers comprising polyethylene oxide (PEO), poly(vinyl alcohol-co-ethylene) (PVA-co-PE), and the photo-antibacterial agent vitamin K3 (VK3) were created using a single-fluid blending process. Core-shell F2 nanofibers were prepared using coaxial electrospinning, in which the extensible material PEO was set as the core section, and a composite consisting of PEO, PVA-co-PE, and VK3 was set as the shell section. Both F1 and F2 fibers with the designed structural properties had an average diameter of approximately 1.0 μm, as determined using scanning electron microscopy and transmission electron microscopy. VK3 was amorphously dispersed within the polymeric matrices of F1 and F2 fibers in a compatible manner, as revealed using X-ray diffraction and Fourier transform infrared spectroscopy. Monolithic F1 fibers had a higher tensile strength of 2.917 ± 0.091 MPa, whereas the core-shell F2 fibers had a longer elongation with a break rate of 194.567 ± 0.091%. Photoreaction tests showed that, with their adjustment, core-shell F2 nanofibers could produce 0.222 μmol/L ·OH upon illumination. F2 fibers had slightly better antibacterial performance than F1 fibers, with inhibition zones of 1.361 ± 0.012 cm and 1.296 ± 0.022 cm for E. coli and S. aureus, respectively, but with less VK3. The intentional tailoring of the components and compositions of the core-shell nanostructures can improve the process-structure-performance relationship of electrospun nanofibers for potential sunlight-activated antibacterial PPE.
Collapse
Affiliation(s)
- Wenjian Gong
- School of Materials and Chemistry, University of Shanghai for Science and Technology, Shanghai 200093, China
| | - Meng-Long Wang
- School of Materials and Chemistry, University of Shanghai for Science and Technology, Shanghai 200093, China
- School of Health Sciences, Saint Francis University, Hong Kong 999077, China
| | - Yanan Liu
- School of Materials and Chemistry, University of Shanghai for Science and Technology, Shanghai 200093, China
| | - Deng-Guang Yu
- School of Materials and Chemistry, University of Shanghai for Science and Technology, Shanghai 200093, China
| | - Sim Wan Annie Bligh
- School of Health Sciences, Saint Francis University, Hong Kong 999077, China
| |
Collapse
|
11
|
Avci FG. Unraveling bacterial stress responses: implications for next-generation antimicrobial solutions. World J Microbiol Biotechnol 2024; 40:285. [PMID: 39073503 PMCID: PMC11286680 DOI: 10.1007/s11274-024-04090-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2024] [Accepted: 07/18/2024] [Indexed: 07/30/2024]
Abstract
The accelerated spread of antimicrobial-resistant bacteria has caused a serious health problem and rendered antimicrobial treatments ineffective. Innovative approaches are crucial to overcome the health threat posed by resistant pathogens and prevent the emergence of untreatable infections. Triggering stress responses in bacteria can diminish susceptibility to various antimicrobials by inducing resistance mechanisms. Therefore, a thorough understanding of stress response control, especially in relation to antimicrobial resistance, offers valuable perspectives for innovative and efficient therapeutic approaches to combat antimicrobial resistance. The aim of this study was to evaluate the stress responses of 8 different bacteria by analyzing reporter metabolites, around which significant alterations were observed, using a pathway-driven computational approach. For this purpose, the transcriptomic data that the bacterial pathogens were grown under 11 different stress conditions mimicking the human host environments were integrated with the genome-scale metabolic models of 8 pathogenic species (Enterococcus faecalis OG1R, Escherichia coli EPEC O127:H6 E2348/69, Escherichia coli ETEC H10407, Escherichia coli UPEC 536, Klebsiella pneumoniae MGH 78578, Pseudomonas aeruginosa PAO1, Staphylococcus aureus MRSA252, and Staphylococcus aureus MSSA476). The resulting reporter metabolites were enriched in multiple metabolic pathways, with cofactor biosynthesis being the most important. The results of this study will serve as a guide for the development of antimicrobial agents as they provide a first insight into potential drug targets.
Collapse
Affiliation(s)
- Fatma Gizem Avci
- Department of Bioengineering, Faculty of Engineering and Natural Sciences, Üsküdar University, Istanbul, Türkiye.
- Genetics of Prokaryotes, Faculty of Biology and Center for Biotechnology (CeBiTec), Bielefeld University, Bielefeld, Germany.
| |
Collapse
|
12
|
Rathod S, Dey S, Pawar S, Dhavale R, Choudhari P, Rajakumara E, Mahuli D, Bhagwat D, Tamboli Y, Sankpal P, Mali S, More H. Identification of potential biogenic chalcones against antibiotic resistant efflux pump (AcrB) via computational study. J Biomol Struct Dyn 2024; 42:5178-5196. [PMID: 37340697 DOI: 10.1080/07391102.2023.2225099] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Accepted: 06/09/2023] [Indexed: 06/22/2023]
Abstract
The cases of bacterial multidrug resistance are increasing every year and becoming a serious concern for human health. Multidrug efflux pumps are key players in the formation of antibiotic resistance, which transfer out a broad spectrum of drugs from the cell and convey resistance to the host. Efflux pumps have significantly reduced the efficacy of the previously available antibiotic armory, thereby increasing the frequency of therapeutic failures. In gram-negative bacteria, the AcrAB-TolC efflux pump is the principal transporter of the substrate and plays a major role in the formation of antibiotic resistance. In the current work, advanced computer-aided drug discovery approaches were utilized to find hit molecules from the library of biogenic chalcones against the bacterial AcrB efflux pump. The results of the performed computational studies via molecular docking, drug-likeness prediction, pharmacokinetic profiling, pharmacophore mapping, density functional theory, and molecular dynamics simulation study provided ZINC000004695648, ZINC000014762506, ZINC000014762510, ZINC000095099506, and ZINC000085510993 as stable hit molecules against the AcrB efflux pumps. Identified hits could successfully act against AcrB efflux pumps after optimization as lead molecules.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Sanket Rathod
- Department of Pharmaceutical Chemistry, Bharati Vidyapeeth College of Pharmacy, Kolhapur, MS, India
| | - Sreenath Dey
- Department of Biotechnology, Indian Institute of Technology, Hyderabad, Kandi, Sangareddy, Telangana, India
| | - Swaranjali Pawar
- Department of Pharmaceutical Chemistry, Bharati Vidyapeeth College of Pharmacy, Kolhapur, MS, India
| | - Rakesh Dhavale
- Department of Pharmaceutics, Bharati Vidyapeeth College of Pharmacy, Kolhapur, MS, India
| | - Prafulla Choudhari
- Department of Pharmaceutical Chemistry, Bharati Vidyapeeth College of Pharmacy, Kolhapur, MS, India
| | - Eerappa Rajakumara
- Department of Biotechnology, Indian Institute of Technology, Hyderabad, Kandi, Sangareddy, Telangana, India
| | - Deepak Mahuli
- Department of Pharmacology, Bharati Vidyapeeth College of Pharmacy, Kolhapur, MS, India
| | - Durgacharan Bhagwat
- Department of Pharmaceutics, Bharati Vidyapeeth College of Pharmacy, Kolhapur, MS, India
| | - Yasinalli Tamboli
- King Abdullah International Medical Research Center (KAIMRC), King Saud Bin Abdulaziz University for Health Sciences, Ministry of National Guard-Health Affairs, Riyadh, Saudi Arabia
| | - Poournima Sankpal
- Department of Pharmaceutical Chemistry, Ashokrao Mane College of Pharmacy, Kolhapur, MS, India
| | - Sachin Mali
- Department of Pharmaceutics, Y. D. Mane College of Pharmacy, Kagal, MS, India Kolhapur
| | - Harinath More
- Department of Pharmaceutical Chemistry, Bharati Vidyapeeth College of Pharmacy, Kolhapur, MS, India
| |
Collapse
|
13
|
Naithani K, Das A, Ushare M, Nath S, Biswas R, Kundu A, Ahmed KT, Mohan U, Bhowmik S. Design, synthesis, and evaluation of 1,4-benzothiazine-3-one containing bisamide derivatives as dual inhibitors of Staphylococcus aureus with plausible application in a urinary catheter. Front Chem 2024; 12:1420593. [PMID: 38988728 PMCID: PMC11233542 DOI: 10.3389/fchem.2024.1420593] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2024] [Accepted: 06/03/2024] [Indexed: 07/12/2024] Open
Abstract
In this study, 1,4-benzothiazine-based bisamide derivatives, a new class of antibacterial agents targeting bacterial peptide deformylase (PDF), were designed and synthesized to combat Staphylococcus aureus infection. Molecular modeling of the designed molecules showed better docking scores compared to the natural product actinonin. Bioactivity assessment identified two derivatives with promising antibacterial activity in vitro. The stability of the most active molecule, 8bE, was assessed using molecular dynamics (MD) simulation. Significantly, compound 8bE could also inhibit the S. aureus biofilm at low concentrations. Furthermore, the capability of the synthesized molecule to inhibit S. aureus biofilm formation on medical devices like urinary catheters is also demonstrated.
Collapse
Affiliation(s)
- Kaushal Naithani
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research, Kolkata, West Bengal, India
| | - Arka Das
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research, Kolkata, West Bengal, India
| | - Mamta Ushare
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research, Kolkata, West Bengal, India
| | - Subham Nath
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research, Kolkata, West Bengal, India
- Microbiology Division, Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research, Kolkata, West Bengal, India
| | - Rashmita Biswas
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research, Kolkata, West Bengal, India
- Microbiology Division, Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research, Kolkata, West Bengal, India
| | - Anirban Kundu
- Department of Natural Product, National Institute of Pharmaceutical Education and Research, Kolkata, West Bengal, India
| | - Kazi Tawsif Ahmed
- Department of Botany, Visva Bharati University, Santiniketan, West Bengal, India
| | - Utpal Mohan
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research, Kolkata, West Bengal, India
- Microbiology Division, Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research, Kolkata, West Bengal, India
| | - Subhendu Bhowmik
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research, Kolkata, West Bengal, India
| |
Collapse
|
14
|
Mhaidat I, Banidomi S, Wedian F, Badarneh R, Tashtoush H, Almomani W, Al-Mazaideh GM, Alharbi NS, Thiruvengadam M. Antioxidant and antibacterial activities of 5-mercapto(substitutedthio)-4-substituted-1,2,4-triazol based on nalidixic acid: A comprehensive study on its synthesis, characterization, and In silico evaluation. Heliyon 2024; 10:e28204. [PMID: 38571635 PMCID: PMC10987910 DOI: 10.1016/j.heliyon.2024.e28204] [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: 12/28/2023] [Revised: 03/13/2024] [Accepted: 03/13/2024] [Indexed: 04/05/2024] Open
Abstract
This study introduces a series of novel Alkyl thio-1,2,4-triazole (4a-p) and mercapto-1,2,4-triazole (3a-d) compounds derived from nalidixic acid. The synthesis was streamlined, involving interactions between nalidixic acid hydrazide and various isothiocyanates to yield cyclic and alkyl(aryl) sulfide compounds, characterized using 1H NMR, 13C NMR, IR, and elemental analysis. Antioxidant capabilities were quantified through DPPH and ABTS assays, highlighting significant potential, especially for compound 3d, which demonstrated an ABTS IC50 value of 0.397 μM, on par with ascorbic acid (IC50 = 0.87 μM). Antibacterial efficacy was established through MIC assessments against a broad spectrum of Gram-positive and Gram-negative bacteria, including Candida albicans. Compounds 3b, 4e, 4h, 4j, 4i, 4m, and 4o showed broad-spectrum activity, with 4k and 4m exhibiting pronounced potency against E. coli. Molecular docking studies validated the antibacterial potential, with compounds 4f and 4h showing high binding affinities (docking scores of -9.8 and -9.6 kcal/mol, respectively), indicating robust interactions with the bacterial enzyme targets. These scores underscore the compounds' mechanistic basis for their antibacterial action and support their therapeutic promise. Furthermore, compounds 3b, 4i, and 4m, identified through drug-likeness and toxicity predictions, were highlighted for their favorable profiles, suggesting their suitability for oral antibiotic therapies. This comprehensive study, blending synthetic, in vitro, and in silico approaches, emphasizes the triazole derivatives' potential as future candidates for antibiotic and antioxidant applications, particularly spotlighting compounds 3b, 4i, and 4m due to their promising efficacy and safety profiles.
Collapse
Affiliation(s)
- Ibrahim Mhaidat
- Department of Chemistry, Faculty of Sciences, Yarmouk University, Irbid, 21163, Jordan
| | - Sojoud Banidomi
- Department of Chemistry, Faculty of Sciences, Yarmouk University, Irbid, 21163, Jordan
| | - Fadel Wedian
- Department of Chemistry, Faculty of Sciences, Yarmouk University, Irbid, 21163, Jordan
| | - Rahaf Badarneh
- Department of Chemistry, Faculty of Sciences, Yarmouk University, Irbid, 21163, Jordan
| | - Hasan Tashtoush
- Department of Chemistry, Faculty of Sciences, Yarmouk University, Irbid, 21163, Jordan
| | - Waleed Almomani
- Department of Basic Medical Sciences, Faculty of Medicine, Yarmouk University, Irbid, 21163, Jordan
| | - Ghassab M. Al-Mazaideh
- Department of Chemistry and Chemical Technology, Tafila Technical University, Tafila, Jordan
| | - Naiyf S. Alharbi
- Department of Botany and Microbiology, College of Science, King Saud University, P. O. Box 2455, Riyadh, 11451, Saudi Arabia
| | - Muthu Thiruvengadam
- Department of Crop Science, College of Sanghuh Life Sciences, Konkuk University, Seoul, 05029, South Korea
| |
Collapse
|
15
|
Orozco-Ugarriza ME, Arrieta Caldera NP, Olivo-Martínez Y. Antimicrobial phytoconstituents from Azadirachta indica (neem) with potential inhibitor against FtsZ protein of Pseudomonas aeruginosa. Nat Prod Res 2024:1-6. [PMID: 38516734 DOI: 10.1080/14786419.2024.2332946] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2023] [Accepted: 03/11/2024] [Indexed: 03/23/2024]
Abstract
Pseudomonas aeruginosa is a well-known pathogen for its rapid development of multi-drug antibiotic resistance. This pathogen is responsible for numerous human diseases, particularly affecting immunocompromised and elderly patients. Hence, discovering novel therapeutics has become necessary in the fight against antimicrobial resistance. This study is focused on evaluating the potential inhibitory activity of eleven phytocompounds from Azadirachta indica against the nucleotide-binding site of the FtsZ protein of P. aeruginosa through a cheminformatics approach. FtsZ is an indispensable and highly conserved protein in prokaryotic cell division. Docking studies revealed favourable binding energies (ΔG= - 8.3 to - 5.4 kcal/mol) for all selected phytoconstituents. Finally, we selected Nimbiol (CID 11119228), as a lead compound, exhibiting a binding energy (ΔG= -7.8 kcal/mol) for the target. Based on our findings, Nimbiol shows potential as an anti-FtsZ compound, making it a promising candidate for further in vitro and in vivo investigations to assess its antimicrobial activity.
Collapse
Affiliation(s)
- Mauricio E Orozco-Ugarriza
- Grupo de investigación en Microbiología y Ambiente (GIMA), Universidad de San Buenaventura, Cartagena, Colombia
- Grupo de investigación traslacional en Biomedicina y Biotecnología (GITB&B), Corporación para el desarrollo de la Investigación en Biomedicina & Biotecnología, Cartagena, Colombia
| | | | - Yenifer Olivo-Martínez
- Grupo de investigación en Microbiología y Ambiente (GIMA), Universidad de San Buenaventura, Cartagena, Colombia
- Biochemistry and Diseases Research Group, Facultad de Medicina, Universidad de Cartagena, Cartagena, Colombia
| |
Collapse
|
16
|
Matotoka MM, Masoko P. Evaluation of the Antioxidant, Cytotoxicity, Antibacterial, Anti-Motility, and Anti-Biofilm Effects of Myrothamnus flabellifolius Welw. Leaves and Stem Defatted Subfractions. PLANTS (BASEL, SWITZERLAND) 2024; 13:847. [PMID: 38592866 PMCID: PMC10974473 DOI: 10.3390/plants13060847] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/19/2024] [Revised: 03/11/2024] [Accepted: 03/12/2024] [Indexed: 04/11/2024]
Abstract
The formation of biofilms underscores the challenge of treating bacterial infections. The study aimed to assess the antioxidant, cytotoxicity, antibacterial, anti-motility, and anti-biofilm effects of defatted fractions from Myrothamnus flabellifolius (resurrection plant). Antioxidant activity was assessed using DPPH radical scavenging and hydrogen peroxide assays. Cytotoxicity was screened using a brine shrimp lethality assay. Antibacterial activity was determined using the micro-dilution and growth curve assays. Antibiofilm potential was screened using the crystal violet and tetrazolium reduction assay. Liquid-liquid extraction of crude extracts concentrated polyphenols in the ethyl acetate and n-butanol fractions. Subsequently, these fractions had notable antioxidant activity and demonstrated broad-spectrum antibacterial activity against selected Gram-negative and Gram-positive bacteria and Mycobacterium smegmatis (MIC values < 630 μg/mL). Growth curves showed that the bacteriostatic inhibition by the ethyl acetate fractions was through the extension of the lag phase and/or suppression of the growth rate. The sub-inhibitory concentrations of the ethyl acetate fractions inhibited the swarming motility of Pseudomonas aeruginosa and Klebsiella pneumoniae by 100% and eradicated more than 50% of P. aeruginosa biofilm biomass. The polyphenolic content of M. flabellifolius plays an important role in its antibacterial, anti-motility, and antibiofilm activity, thus offering an additional strategy to treat biofilm-associated infections.
Collapse
Affiliation(s)
| | - Peter Masoko
- Faculty of Science and Agriculture, Department of Biochemistry, Microbiology and Biotechnology, University of Limpopo, Private Bag X1106, Sovenga 0727, South Africa;
| |
Collapse
|
17
|
Wang YP, Jiang TT, Sun J, Han Y, Yan WF, Wang YC, Lu J, Jin J, Liu YF, Li Q. Synthesis, structure, theoretical calculation and antibacterial property of two novel Zn(II)/Ni(II) compounds based on 3, 5-dichlorosalicylaldehyde thiocarbamide ligand. Bioorg Chem 2024; 144:107140. [PMID: 38245950 DOI: 10.1016/j.bioorg.2024.107140] [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: 10/16/2023] [Revised: 12/16/2023] [Accepted: 01/15/2024] [Indexed: 01/23/2024]
Abstract
Two new compounds namely [Zn(L1)phen]31 and Ni(L1)phen(MeOH) 2 (L1 = 3, 5-dichlorosalicylaldehyde thiosemicarbazone) were synthesized by the slow evaporation method at room temperature. The structure of ligand L1 was determined using 1H NMR and 13C NMR spectra. X-ray single crystal diffraction analysis revealed that compounds 1-2 can form 3D supramolecular network structures through π···π stacking and hydrogen bonding interactions. The DFT calculation shows that the coordination of ligand and metal is in good agreement with the experimental results. Hirshfeld surface analysis revealed that H…H and Cl…H interactions were the predominant interactions in compounds 1-2. Energy framework analysis indicated that dispersion energy played a dominant role in the energy composition of compounds 1-2. The inhibitory effects of compounds 1-2 against Escherichia coli (E. coli) and Methicillin-resistant Staphylococcus aureus (MRSA) were tested using the paper disk diffusion method (1: E. coli: 18 mm, MRSA: 17 mm, 2: E. coli: 15 mm, MRSA: 16 mm). Ion releasing experiments were conducted to assess the ion release capacity of compounds 1-2 (Zn2+, 4 days, 38.33 µg/mL; Ni2+, 4 days, 29.12 µg/mL). Molecular docking demonstrated the interaction modes of compounds 1-2 with UDP-N-acetylenolpyruvoylglucosamine reductase (MurB) and dihydrofolate reductase (DHFR) in bacteria, involving hydrophobic, stacking, hydrogen bonding and halogen bonding interactions. The generation of reactive oxygen species (ROS) in bacteria under the presence of compounds 1-2 were evaluated using a fluorescent dye known as dichlorodihydrofluorescein diacetate (DCFH-DA). Potential antibacterial mechanisms of compounds 1-2 were proposed.
Collapse
Affiliation(s)
- Yuan-Peng Wang
- School of Chemistry and Materials Science, Ludong University, Yantai, Shandong 264025, China
| | - Ting-Ting Jiang
- School of Life Science, Ludong University, Yantai, Shandong 264025, China
| | - Jie Sun
- School of Life Science, Ludong University, Yantai, Shandong 264025, China
| | - Yu Han
- School of Chemistry and Materials Science, Ludong University, Yantai, Shandong 264025, China
| | - Wen-Fu Yan
- College of Chemistry and State Key Laboratory of Inorganic Synthesis and Preparative Chemistry, Jilin University, Changchun, Jilin 130023, China
| | - Yu-Chang Wang
- Yantai Valiant Fine Chemicals Co., Ltd, Yantai, Shandong 264006, China
| | - Jing Lu
- Shandong Provincial Key Laboratory of Chemical Energy Storage and Novel Cell Technology, School of Chemistry and Chemical Engineering, Liaocheng University, Liaocheng, Shandong 252059, China
| | - Juan Jin
- School of Chemistry and Materials Science, Ludong University, Yantai, Shandong 264025, China; College of Chemistry and State Key Laboratory of Inorganic Synthesis and Preparative Chemistry, Jilin University, Changchun, Jilin 130023, China.
| | - Yong-Feng Liu
- School of Chemistry and Materials Science, Ludong University, Yantai, Shandong 264025, China
| | - Qing Li
- School of Life Science, Ludong University, Yantai, Shandong 264025, China.
| |
Collapse
|
18
|
Ergüden B, Lüleci HB, Ünver Y. Benzothiophene Schiff Bases Disrupt Cytoplasmic Membrane Integrity of Gram-Positive and -Negative Bacteria Cells. RUSSIAN JOURNAL OF BIOORGANIC CHEMISTRY 2024; 50:128-137. [DOI: 10.1134/s1068162024010096] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Revised: 08/09/2023] [Accepted: 08/10/2023] [Indexed: 07/26/2024]
|
19
|
Ramírez-Trinidad Á, Martínez-Solano E, Tovar-Roman CE, García-Guerrero M, Rivera-Chávez JA, Hernández-Vázquez E. Synthesis, antibiofilm activity and molecular docking of N-acylhomoserine lactones containing cinammic moieties. Bioorg Med Chem Lett 2024; 98:129592. [PMID: 38101651 DOI: 10.1016/j.bmcl.2023.129592] [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: 10/10/2023] [Revised: 12/03/2023] [Accepted: 12/10/2023] [Indexed: 12/17/2023]
Abstract
We prepared a series of cinnamoyl-containing furanones by an affordable and short synthesis. The nineteen compounds hold a variety of substituents including electron-donating, electron-withdrawing, bulky and meta-substituted phenyls, as well as heterocyclic rings. Compounds showed antibiofilm activity in S. aureus, K. pneumoniae and, more pronounced, against P. aeruginosa. The disruption of quorum sensing (QS) was tested using the violacein test and molecular docking predicted the antagonism of LasR as a plausible mechanism of action. The trimethoxylated and diene derivatives showed the best antibiofilm and anti-QS properties, thus becoming candidates for further modifications.
Collapse
Affiliation(s)
- Ángel Ramírez-Trinidad
- Department of Organic Chemistry, Chemistry Institute, UNAM. Circuito exterior S.N., Ciudad Universitaria, Coyoacán, México, DF 04510, Mexico
| | - Ernesto Martínez-Solano
- Department of Organic Chemistry, Chemistry Institute, UNAM. Circuito exterior S.N., Ciudad Universitaria, Coyoacán, México, DF 04510, Mexico
| | - César E Tovar-Roman
- Department of Organic Chemistry, Chemistry Institute, UNAM. Circuito exterior S.N., Ciudad Universitaria, Coyoacán, México, DF 04510, Mexico
| | - Mariana García-Guerrero
- Department of Natural Products, Chemistry Institute, UNAM. Circuito exterior S.N., Ciudad Universitaria, Coyoacán, México, DF 04510, Mexico
| | - José A Rivera-Chávez
- Department of Natural Products, Chemistry Institute, UNAM. Circuito exterior S.N., Ciudad Universitaria, Coyoacán, México, DF 04510, Mexico
| | - Eduardo Hernández-Vázquez
- Department of Organic Chemistry, Chemistry Institute, UNAM. Circuito exterior S.N., Ciudad Universitaria, Coyoacán, México, DF 04510, Mexico.
| |
Collapse
|
20
|
Zorman M, Hrast Rambaher M, Kokot M, Minovski N, Anderluh M. The overview of development of novel bacterial topoisomerase inhibitors effective against multidrug-resistant bacteria in an academic environment: From early hits to in vivo active antibacterials. Eur J Pharm Sci 2024; 192:106632. [PMID: 37949194 DOI: 10.1016/j.ejps.2023.106632] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2023] [Revised: 09/29/2023] [Accepted: 11/06/2023] [Indexed: 11/12/2023]
Abstract
Antimicrobial resistance caused by the excessive and inappropriate use of antibacterial drugs is a global health concern. Currently, we are walking a fine line between the fact that most bacterial infections can still be cured with the antibiotics known so far, and the emergence of infections with bacteria resistant to several drugs at the same time, against which we no longer have an effective drug. Therefore, new antibacterial drugs are urgently needed to curb the hard-to-treat infections. Our group has developed new antibacterials from the class of novel bacterial topoisomerase inhibitors (NBTIs) that exhibit broad-spectrum antibacterial activity. This article reviews our efforts in developing highly potent NBTIs over the past decade. Following the discovery of an initial hit with potent enzyme inhibitory and broad-spectrum antibacterial activity, an extensive hit-to-lead campaign was conducted with the goal of optimizing physicochemical properties, reducing hERG inhibition, and maintaining antibacterial activity against both Gram-positive and Gram-negative bacteria, with a focus on methicillin-resistant Staphylococcus aureus (MRSA). This optimization strategy resulted in an amide-containing, focused NBTI library with compounds exhibiting potent antibacterial activity against Gram-positive bacteria, reduced hERG inhibition, no cardiotoxicity in in vivo zebrafish model, and favorable in vivo efficacy in a neutropenic murine thigh infection model for MRSA infections.
Collapse
Affiliation(s)
- Maša Zorman
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Ljubljana, Aškerčeva Cesta 7, Ljubljana 1000, Slovenia; Theory Department, Laboratory for Cheminformatics, National Institute of Chemistry, Hajdrihova 19, Ljubljana 1001, Slovenia
| | - Martina Hrast Rambaher
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Ljubljana, Aškerčeva Cesta 7, Ljubljana 1000, Slovenia
| | - Maja Kokot
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Ljubljana, Aškerčeva Cesta 7, Ljubljana 1000, Slovenia; Theory Department, Laboratory for Cheminformatics, National Institute of Chemistry, Hajdrihova 19, Ljubljana 1001, Slovenia
| | - Nikola Minovski
- Theory Department, Laboratory for Cheminformatics, National Institute of Chemistry, Hajdrihova 19, Ljubljana 1001, Slovenia.
| | - Marko Anderluh
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Ljubljana, Aškerčeva Cesta 7, Ljubljana 1000, Slovenia.
| |
Collapse
|
21
|
Thakur A, Ganesan R, Ray Dutta J. Antimicrobial Peptide-Based Nanomaterials in Combating Multidrug-Resistant Bacteria. NANOTECHNOLOGY BASED STRATEGIES FOR COMBATING ANTIMICROBIAL RESISTANCE 2024:177-201. [DOI: 10.1007/978-981-97-2023-1_7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
22
|
Zhang Y, Chung WK, Moon SH, Lee JG, Om AS. Comparison of Antibacterial Activities of Korean Pine ( Pinus densiflora) Needle Steam Distillation Extract on Escherichia coli and Staphylococcus aureus Focusing on Membrane Fluidity and Genes Involved in Membrane Lipids and Stress. Molecules 2023; 29:165. [PMID: 38202748 PMCID: PMC10779765 DOI: 10.3390/molecules29010165] [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: 11/10/2023] [Revised: 12/22/2023] [Accepted: 12/23/2023] [Indexed: 01/12/2024] Open
Abstract
The antibacterial activity and mechanism of Pinus densiflora extracts against Escherichia coli and Staphylococcus aureus were investigated. The growth inhibition tests of paper diffusion and optical density exhibited that the extracts have potent antibacterial potentials against foodborne pathogens. The measurement of membrane fluidity by fluorescence polarization has indicated that one of the antibacterial mechanisms involves the disruption of membrane integrity resulting in an increase in the membrane fluidity in both of E. coli and S. aureus. The alteration of fatty acid composition was accompanied by the disturbance of membranes thus shifting the proportion of saturated verses unsaturated fatty acids or trans fatty acids from 1.27:1 to 1.35:1 in E. coli and 1.47:1 to 2.31:1 in S. aureus, most likely to compensate for the increased membrane fluidity by means of a higher proportion of saturated fatty acids which is known to render rigidity in membranes. Realtime q-PCR (polymerase chain reaction) analysis of fatty acid synthetic genes and bacterial stress genes revealed that there was minimal influence of P. densiflora extracts on fatty acid genes except for fab I and the stress rpos in E. coli, and relatively greater impact on fatty acid genes and the stress sigB in S. aureus.
Collapse
Affiliation(s)
| | | | | | | | - Ae-Son Om
- Department of Food and Nutrition, Hanyang University, Seoul 04736, Republic of Korea; (Y.Z.); (W.-K.C.); (S.-H.M.); (J.-G.L.)
| |
Collapse
|
23
|
Becker R, Pederick JL, Dawes EG, Bruning JB, Abell AD. Structure-guided design and synthesis of ATP-competitive N-acyl-substituted sulfamide d-alanine-d-alanine ligase inhibitors. Bioorg Med Chem 2023; 96:117509. [PMID: 37948922 DOI: 10.1016/j.bmc.2023.117509] [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: 08/29/2023] [Revised: 10/10/2023] [Accepted: 10/24/2023] [Indexed: 11/12/2023]
Abstract
d-Alanine-d-alanine ligase (Ddl) catalyses the ATP-dependent formation of d-Ala-d-Ala, a critical component in bacterial cell wall biosynthesis and is a validated target for new antimicrobial agents. Here, we describe the structure-guided design, synthesis, and evaluation of ATP-competitive N-acyl-substituted sulfamides 27-36, 42, 46, 47 as inhibitors of Staphylococcus aureus Ddl (SaDdl). A crystal structure of SaDdl complexed with ATP and d-Ala-d-Ala (PDB: 7U9K) identified ATP-mimetic 8 as an initial scaffold for further inhibitor design. Evaluation of 8 in SaDdl enzyme inhibition assays revealed the ability to reduce enzyme activity to 72 ± 8 % (IC50 = 1.6 mM). The sulfamide linker of 8 was extended with 2-(4-methoxyphenyl)ethanol to give 29, to investigate further interactions with the d-Ala pocket of SaDdl, as predicted by molecular docking. This compound reduced enzyme activity to 89 ± 1 %, with replacement of the 4-methoxyphenyl group in 29 with alternative phenyl substituents (27, 28, 31-33, 35, 36) failing to significantly improve on this (80-89 % remaining enzyme activity). Exchanging these phenyl substituents with selected heterocycles (42, 46, 47) did improve activity, with the most active compound (42) reducing SaDdl activity to 70 ± 1 % (IC50 = 1.7 mM), which compares favourably to the FDA-approved inhibitor d-cycloserine (DCS) (IC50 = 0.1 mM). To the best of our knowledge, this is the first reported study of bisubstrate SaDdl inhibitors.
Collapse
Affiliation(s)
- Rouven Becker
- Department of Chemistry, School of Physics, Chemistry and Earth Sciences, University of Adelaide, Adelaide, South Australia 5005, Australia; Institute for Photonics and Advanced Sensing, (IPAS), School of Biological Sciences, The University of Adelaide, Adelaide, South Australia 5005, Australia; Centre for Nanoscale BioPhotonics (CNBP), University of Adelaide, Adelaide, South Australia 5005, Australia
| | - Jordan L Pederick
- Institute for Photonics and Advanced Sensing, (IPAS), School of Biological Sciences, The University of Adelaide, Adelaide, South Australia 5005, Australia
| | - Edward G Dawes
- Department of Chemistry, School of Physics, Chemistry and Earth Sciences, University of Adelaide, Adelaide, South Australia 5005, Australia; Centre for Nanoscale BioPhotonics (CNBP), University of Adelaide, Adelaide, South Australia 5005, Australia
| | - John B Bruning
- Institute for Photonics and Advanced Sensing, (IPAS), School of Biological Sciences, The University of Adelaide, Adelaide, South Australia 5005, Australia
| | - Andrew D Abell
- Department of Chemistry, School of Physics, Chemistry and Earth Sciences, University of Adelaide, Adelaide, South Australia 5005, Australia; Institute for Photonics and Advanced Sensing, (IPAS), School of Biological Sciences, The University of Adelaide, Adelaide, South Australia 5005, Australia; Centre for Nanoscale BioPhotonics (CNBP), University of Adelaide, Adelaide, South Australia 5005, Australia.
| |
Collapse
|
24
|
Hervin V, Roy V, Agrofoglio LA. Antibiotics and Antibiotic Resistance-Mur Ligases as an Antibacterial Target. Molecules 2023; 28:8076. [PMID: 38138566 PMCID: PMC10745416 DOI: 10.3390/molecules28248076] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2023] [Revised: 11/09/2023] [Accepted: 12/11/2023] [Indexed: 12/24/2023] Open
Abstract
The emergence of Multidrug Resistance (MDR) strains of bacteria has accelerated the search for new antibacterials. The specific bacterial peptidoglycan biosynthetic pathway represents opportunities for the development of novel antibacterial agents. Among the enzymes involved, Mur ligases, described herein, and especially the amide ligases MurC-F are key targets for the discovery of multi-inhibitors, as they share common active sites and structural features.
Collapse
Affiliation(s)
| | - Vincent Roy
- ICOA UMR CNRS 7311, Université d’Orléans et CNRS, Rue de Chartres, 45067 Orléans, France;
| | - Luigi A. Agrofoglio
- ICOA UMR CNRS 7311, Université d’Orléans et CNRS, Rue de Chartres, 45067 Orléans, France;
| |
Collapse
|
25
|
Kumar S, Kumar S, Mir MA, Vishnoi VK, Pandey A, Pandey A. Bioefficacy of Sida cordifolia L. phytoextract against foodborne bacteria: optimization and bioactive compound analysis. Future Microbiol 2023; 18:1235-1249. [PMID: 37750761 DOI: 10.2217/fmb-2023-0064] [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: 03/17/2023] [Accepted: 08/03/2023] [Indexed: 09/27/2023] Open
Abstract
Aim: To elucidate the antibacterial activity of Sida cordifolia L. phytoextract, evaluate its polyphenol profile and optimize conditions against certain common foodborne bacteria. Methods: After polarity-based sequential extraction, S. cordifolia phytoextracts were tested for antibacterial potential against antibiotic-resistant bacteria. Box-Behnken design was used to optimize several process parameters and ultra-performance liquid chromatography confirmed the phenolic composition of the best possible outcome. Results: Agar well diffusion and MIC/MBC assay confirmed a strong bactericidal effect of ethanolic (SC04-ET) extract against ampicillin and colistin-resistant Escherichia coli, Listeria monocytogenes and Staphylococcus aureus. The direct interactive effect of optimized conditions showed maximum antibacterial performance and ultra-performance liquid chromatography revealed a high amount of phenolic compounds. Conclusion: The results confirmed that ethanolic extract of S. cordifolia has potent bactericidal action against foodborne bacteria.
Collapse
Affiliation(s)
- Sachin Kumar
- Department of Botany & Microbiology, Gurukula Kangri (Deemed to be University), Haridwar, India
| | - Sandeep Kumar
- Department of Botany & Microbiology, Gurukula Kangri (Deemed to be University), Haridwar, India
| | - M Amin Mir
- Department of Chemistry, Prince Mohammad Bin Fahd University, Al-Khobar, Saudi Arabia
| | - Vineet Kumar Vishnoi
- Department of Botany & Microbiology, Gurukula Kangri (Deemed to be University), Haridwar, India
| | - Ashutosh Pandey
- National Institute of Plant Genome Research, Aruna Asaf Ali Marg, New Delhi, India
| | - Akanksha Pandey
- Department of Botany & Microbiology, Gurukula Kangri (Deemed to be University), Haridwar, India
| |
Collapse
|
26
|
Costa Dos Santos D, Silva Macêdo N, de Sousa Silveira Z, Silva Pereira RL, Moura Araújo I, Justino Araújo AC, Alves Gonçalves S, da Silveira Regueira Neto M, de Queiroz Balbino V, Torres de Carvalho A, Oliveira de Veras B, Bezerra da Cunha FA, Melo Coutinho HD, Vieira Brito S. Antibacterial and Toxic Activity of Geopropolis Extracts from Melipona subnitida (Ducke, 1910) (Hymenoptera: Apidae) and Scaptotrigona depilis (Moure, 1942) (Hymenoptera: Apidae). Chem Biodivers 2023; 20:e202300931. [PMID: 37776535 DOI: 10.1002/cbdv.202300931] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2023] [Revised: 09/20/2023] [Accepted: 09/24/2023] [Indexed: 10/02/2023]
Abstract
Bacteria are associated with many infections that affect humans and present antibiotic resistance mechanisms, causing problems in health organisations and increased mortality rates. Therefore, it is necessary to find new antibacterial agents that can be used in the treatment of these microorganisms. Geopropolis is a natural product from stingless bees, formed by a mixture of plant resins, salivary secretions, wax and soil particles, the chemical composition of this natural product is diverse. Thus, this study aimed to evaluate antibacterial activity, antibiotic modulation and the toxicity of geopropolis extracts from the stingless bees, Melipona subnitida (Ducke, 1910) and Scaptotrigona depilis (Moure, 1942) against standard and multi-resistant Staphylococcus aureus, Escherichia coli and Pseudomonas aeruginosa bacteria. Geopropolis samples were collected in a meliponary located in Camaragibe, Pernambuco, Brazil. To determine the Minimum Inhibitory Concentration (MIC) and antibiotic modulation we performed broth microdilution tests. Mortality tests were used to verify extract toxicity in the model Drosophila melanogaster. The microbiological tests showing that the M. subnitida extracts had better inhibitory effects compared to S. depilis, presenting direct antibacterial activity against standard and multi-resistant strains. The extracts potentialized antibiotic effects, suggesting possible synergy and did not present toxicity in the model used. The information obtained in this study highlights extracts as promising antibacterial agents and is the first study to evaluate bacterial activity in these extracts, in addition to verifying their modulating effects and determining toxicity in the model used.
Collapse
Affiliation(s)
- Danilo Costa Dos Santos
- Programa de Pós-Graduação em Ciências Ambientais, Centro de Ciências de Chapadinha, Universidade Federal do Maranhão, BR 222, Km 04, S/N, Boa Vista, CEP 65500-000, Chapadinha, Maranhão, Brasil
| | - Nair Silva Macêdo
- Semiarid Bioprospecting Laboratory (LABSEMA), Regional University of Cariri-URCA, Crato, Ceará, Brazil
| | - Zildene de Sousa Silveira
- Semiarid Bioprospecting Laboratory (LABSEMA), Regional University of Cariri-URCA, Crato, Ceará, Brazil
| | - Raimundo Luiz Silva Pereira
- Laboratory of Microbiology and Molecular Biology (LMBM), Regional University of Cariri-URCA, Crato, Ceará, Brazil
| | - Isaac Moura Araújo
- Laboratory of Microbiology and Molecular Biology (LMBM), Regional University of Cariri-URCA, Crato, Ceará, Brazil
| | - Ana Carolina Justino Araújo
- Laboratory of Microbiology and Molecular Biology (LMBM), Regional University of Cariri-URCA, Crato, Ceará, Brazil
| | - Sheila Alves Gonçalves
- Laboratory of Microbiology and Molecular Biology (LMBM), Regional University of Cariri-URCA, Crato, Ceará, Brazil
| | | | | | - Airton Torres de Carvalho
- Department of Biosciences, Center of Biological and Health Sciences, Federal Rural, University of the Semi-Arid, Mossoró, RN, Brazil
| | - Bruno Oliveira de Veras
- Department of Biochemistry, Federal University of Pernambuco, 50670-420, Recife, Pernambuco, Brazil
| | | | | | - Samuel Vieira Brito
- Programa de Pós-Graduação em Ciências Ambientais, Centro de Ciências de Chapadinha, Universidade Federal do Maranhão, BR 222, Km 04, S/N, Boa Vista, CEP 65500-000, Chapadinha, Maranhão, Brasil
| |
Collapse
|
27
|
Hou J, Xianyu Y. Tailoring the Surface and Composition of Nanozymes for Enhanced Bacterial Binding and Antibacterial Activity. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2023; 19:e2302640. [PMID: 37322391 DOI: 10.1002/smll.202302640] [Citation(s) in RCA: 35] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Revised: 05/17/2023] [Indexed: 06/17/2023]
Abstract
With the advantages of diverse structures, tunable enzymatic activity, and high stability, nanozymes are widely used in medicine, chemistry, food, environment, and other fields. As an alternative to traditional antibiotics, nanozymes attract more and more attention from the scientific researchers in recent years. Developing nanozymes-based antibacterial materials opens up a new avenue for the bacterial disinfection and sterilization. In this review, the classification of nanozymes and their antibacterial mechanisms are discussed. The surface and composition of nanozymes are critical for the antibacterial efficacy, which can be tailored to enhance both the bacterial binding and the antibacterial activity. On the one hand, the surface modification of nanozymes enables binding and targeting of bacteria that improves the antibacterial performance of nanozymes including the biochemical recognition, the surface charge, and the surface topography. On the other hand, the composition of nanozymes can be modulated to achieve enhanced antibacterial performance including the single nanozyme-mediated synergistic and multiple nanozymes-mediated cascade catalytic antibacterial applications. In addition, the current challenges and future prospects of tailoring nanozymes for antibacterial applications are discussed. This review can provide insights into the design of future nanozymes-based materials for the antibacterial treatments.
Collapse
Affiliation(s)
- Jinjie Hou
- State Key Laboratory of Fluid Power and Mechatronic Systems, College of Biosystems Engineering and Food Science, Zhejiang University, Hangzhou, 310058, P. R. China
| | - Yunlei Xianyu
- State Key Laboratory of Fluid Power and Mechatronic Systems, College of Biosystems Engineering and Food Science, Zhejiang University, Hangzhou, 310058, P. R. China
- Key Laboratory of Precision Medicine in Diagnosis and Monitoring Research of Zhejiang Province, Sir Run Run Shaw Hospital, Hangzhou, 310016, P. R. China
- Ningbo Research Institute, Zhejiang University, Ningbo, 315100, P. R. China
| |
Collapse
|
28
|
Kim D, Park KW, Park JT, Choi I. Photoactive MOF-Derived Bimetallic Silver and Cobalt Nanocomposite with Enhanced Antibacterial Activity. ACS APPLIED MATERIALS & INTERFACES 2023; 15:22903-22914. [PMID: 36996415 DOI: 10.1021/acsami.3c01529] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/19/2023]
Abstract
Conventional antibiotic-based treatment of bacterial infections remains one of the most difficult challenges in medicine because of the threat of multidrug resistance caused by indiscriminate abuse. To solve these problems, it is essential to develop an effective antibacterial agent that can be used at a small dose while minimizing the occurrence of multiple resistance. Metal-organic frameworks (MOFs), which are hyper-porous hybrid materials containing metal ions linked by organic ligands, have recently attracted attention because of their strong antibacterial activity through metal-ion release, unlike conventional antibiotics. In this study, we developed a photoactive MOF-derived cobalt-silver bimetallic nanocomposite (Ag@CoMOF) by simply depositing silver nanoparticles on a cobalt-based MOF through nanoscale galvanic replacement. The nanocomposite structure continuously releases antibacterial metal ions (i.e., Ag and Co ions) in the aqueous phase and exhibits a strong photothermal conversion effect of Ag nanoparticles, accompanied by a rapid temperature increase of 25-80 °C under near-infrared (NIR) irradiation. Using this MOF-based bimetallic nanocomposite, superior antibacterial activities were achieved by 22.1-fold for Escherichia coli and 18.3-fold for Bacillus subtilis enhanced inhibition of bacterial growth in a liquid culture environment compared with the generally used chemical antibiotics. In addition, we confirmed the synergistic enhancement of the antibacterial ability of the bimetallic nanocomposite induced by NIR-triggered photothermal heating and bacterial membrane disruption even when using a small amount of the nanocomposites. We envision that this novel antibacterial agent using MOF-based nanostructures will replace traditional antibiotics to circumvent multidrug resistance and present a new approach to antibiotic development.
Collapse
Affiliation(s)
- Doyun Kim
- Department of Life Science, University of Seoul, 163, Seoulsiripdae-ro, Dongdaemun-gu, Seoul 02504, Republic of Korea
| | - Kun Woo Park
- Department of Chemical Engineering, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul 05029, Republic of Korea
| | - Jung Tae Park
- Department of Chemical Engineering, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul 05029, Republic of Korea
| | - Inhee Choi
- Department of Life Science, University of Seoul, 163, Seoulsiripdae-ro, Dongdaemun-gu, Seoul 02504, Republic of Korea
- Department of Applied Chemistry, University of Seoul, 163, Seoulsiripdae-ro, Dongdaemun-gu, Seoul 02504, Republic of Korea
| |
Collapse
|
29
|
Recent approaches in the drug research and development of novel antimalarial drugs with new targets. ACTA PHARMACEUTICA (ZAGREB, CROATIA) 2023; 73:1-27. [PMID: 36692468 DOI: 10.2478/acph-2023-0001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 09/16/2022] [Indexed: 01/25/2023]
Abstract
Malaria is a serious worldwide medical issue that results in substantial annual death and morbidity. The availability of treatment alternatives is limited, and the rise of resistant parasite types has posed a significant challenge to malaria treatment. To prevent a public health disaster, novel antimalarial agents with single-dosage therapies, extensive curative capability, and new mechanisms are urgently needed. There are several approaches to developing antimalarial drugs, ranging from alterations of current drugs to the creation of new compounds with specific targeting abilities. The availability of multiple genomic techniques, as well as recent advancements in parasite biology, provides a varied collection of possible targets for the development of novel treatments. A number of promising pharmacological interference targets have been uncovered in modern times. As a result, our review concentrates on the most current scientific and technical progress in the innovation of new antimalarial medications. The protein kinases, choline transport inhibitors, dihydroorotate dehydrogenase inhibitors, isoprenoid biosynthesis inhibitors, and enzymes involved in the metabolism of lipids and replication of deoxyribonucleic acid, are among the most fascinating antimalarial target proteins presently being investigated. The new cellular targets and drugs which can inhibit malaria and their development techniques are summarised in this study.
Collapse
|
30
|
Zaki BM, Fahmy NA, Aziz RK, Samir R, El-Shibiny A. Characterization and comprehensive genome analysis of novel bacteriophage, vB_Kpn_ZCKp20p, with lytic and anti-biofilm potential against clinical multidrug-resistant Klebsiella pneumoniae. Front Cell Infect Microbiol 2023; 13:1077995. [PMID: 36756618 PMCID: PMC9901506 DOI: 10.3389/fcimb.2023.1077995] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2022] [Accepted: 01/02/2023] [Indexed: 01/24/2023] Open
Abstract
Introduction The rise of infections by antibiotic-resistant bacterial pathogens is alarming. Among these, Klebsiella pneumoniae is a leading cause of death by hospital-acquired infections, and its multidrug-resistant strains are flagged as a global threat to human health, which necessitates finding novel antibiotics or alternative therapies. One promising therapeutic alternative is the use of virulent bacteriophages, which specifically target bacteria and coevolve with them to overcome potential resistance. Here, we aimed to discover specific bacteriophages with therapeutic potential against multiresistant K. pneumoniae clinical isolates. Methods and Results Out of six bacteriophages that we isolated from urban and medical sewage, phage vB_Kpn_ZCKp20p had the broadest host range and was thus characterized in detail. Transmission electron microscopy suggests vB_Kpn_ZCKp20p to be a tailed phage of the siphoviral morphotype. In vitro evaluation indicated a high lytic efficiency (30 min latent period and burst size of ∼100 PFU/cell), and extended stability at temperatures up to 70°C and a wide range of (2-12) pH. Additionally, phage vB_Kpn_ZCKp20p possesses antibiofilm activity that was evaluated by the crystal violet assay and was not cytotoxic to human skin fibroblasts. The whole genome was sequenced and annotated, uncovering one tRNA gene and 33 genes encoding proteins with assigned functions out of 85 predicted genes. Furthermore, comparative genomics and phylogenetic analysis suggest that vB_Kpn_ZCKp20p most likely represents a new species, but belongs to the same genus as Klebsiella phages ZCKP8 and 6691. Comprehensive genomic and bioinformatics analyses substantiate the safety of the phage and its strictly lytic lifestyle. Conclusion Phage vB_Kpn_ZCKp20p is a novel phage with potential to be used against biofilm-forming K. pneumoniae and could be a promising source for antibacterial and antibiofilm products, which will be individually studied experimentally in future studies.
Collapse
Affiliation(s)
- Bishoy Maher Zaki
- Department of Microbiology and Immunology, Faculty of Pharmacy, October University for Modern Sciences and Arts (MSA), 6th of October, Giza, Egypt
- Center for Microbiology and Phage Therapy, Biomedical Sciences, Zewail City of Science and Technology, Giza, Egypt
| | - Nada A. Fahmy
- Center for Microbiology and Phage Therapy, Biomedical Sciences, Zewail City of Science and Technology, Giza, Egypt
| | - Ramy Karam Aziz
- Department of Microbiology and Immunology, Faculty of Pharmacy, Cairo University, Cairo, Egypt
- Center for Genome and Microbiome Research, Cairo University, Cairo, Egypt
- Microbiology and Immunology Research Program, Children’s Cancer Hospital Egypt, Cairo, Egypt
| | - Reham Samir
- Department of Microbiology and Immunology, Faculty of Pharmacy, Cairo University, Cairo, Egypt
- Center for Genome and Microbiome Research, Cairo University, Cairo, Egypt
| | - Ayman El-Shibiny
- Center for Microbiology and Phage Therapy, Biomedical Sciences, Zewail City of Science and Technology, Giza, Egypt
- Faculty of Environmental Agricultural Sciences, Arish University, Arish, Egypt
| |
Collapse
|
31
|
Bedendi G, De Moura Torquato LD, Webb S, Cadoux C, Kulkarni A, Sahin S, Maroni P, Milton RD, Grattieri M. Enzymatic and Microbial Electrochemistry: Approaches and Methods. ACS MEASUREMENT SCIENCE AU 2022; 2:517-541. [PMID: 36573075 PMCID: PMC9783092 DOI: 10.1021/acsmeasuresciau.2c00042] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Revised: 08/12/2022] [Accepted: 08/15/2022] [Indexed: 06/17/2023]
Abstract
The coupling of enzymes and/or intact bacteria with electrodes has been vastly investigated due to the wide range of existing applications. These span from biomedical and biosensing to energy production purposes and bioelectrosynthesis, whether for theoretical research or pure applied industrial processes. Both enzymes and bacteria offer a potential biotechnological alternative to noble/rare metal-dependent catalytic processes. However, when developing these biohybrid electrochemical systems, it is of the utmost importance to investigate how the approaches utilized to couple biocatalysts and electrodes influence the resulting bioelectrocatalytic response. Accordingly, this tutorial review starts by recalling some basic principles and applications of bioelectrochemistry, presenting the electrode and/or biocatalyst modifications that facilitate the interaction between the biotic and abiotic components of bioelectrochemical systems. Focus is then directed toward the methods used to evaluate the effectiveness of enzyme/bacteria-electrode interaction and the insights that they provide. The basic concepts of electrochemical methods widely employed in enzymatic and microbial electrochemistry, such as amperometry and voltammetry, are initially presented to later focus on various complementary methods such as spectroelectrochemistry, fluorescence spectroscopy and microscopy, and surface analytical/characterization techniques such as quartz crystal microbalance and atomic force microscopy. The tutorial review is thus aimed at students and graduate students approaching the field of enzymatic and microbial electrochemistry, while also providing a critical and up-to-date reference for senior researchers working in the field.
Collapse
Affiliation(s)
- Giada Bedendi
- Department
of Inorganic and Analytical Chemistry, Faculty of Sciences, University of Geneva, Quai Ernest-Ansermet 30, 1211 Geneva 4, Switzerland
| | | | - Sophie Webb
- Department
of Inorganic and Analytical Chemistry, Faculty of Sciences, University of Geneva, Quai Ernest-Ansermet 30, 1211 Geneva 4, Switzerland
- National
Centre of Competence in Research (NCCR) Catalysis, University of Geneva, Quai Ernest-Ansermet 30, 1211 Geneva 4, Switzerland
| | - Cécile Cadoux
- Department
of Inorganic and Analytical Chemistry, Faculty of Sciences, University of Geneva, Quai Ernest-Ansermet 30, 1211 Geneva 4, Switzerland
- National
Centre of Competence in Research (NCCR) Catalysis, University of Geneva, Quai Ernest-Ansermet 30, 1211 Geneva 4, Switzerland
| | - Amogh Kulkarni
- Department
of Inorganic and Analytical Chemistry, Faculty of Sciences, University of Geneva, Quai Ernest-Ansermet 30, 1211 Geneva 4, Switzerland
| | - Selmihan Sahin
- Department
of Inorganic and Analytical Chemistry, Faculty of Sciences, University of Geneva, Quai Ernest-Ansermet 30, 1211 Geneva 4, Switzerland
| | - Plinio Maroni
- Department
of Inorganic and Analytical Chemistry, Faculty of Sciences, University of Geneva, Quai Ernest-Ansermet 30, 1211 Geneva 4, Switzerland
| | - Ross D. Milton
- Department
of Inorganic and Analytical Chemistry, Faculty of Sciences, University of Geneva, Quai Ernest-Ansermet 30, 1211 Geneva 4, Switzerland
- National
Centre of Competence in Research (NCCR) Catalysis, University of Geneva, Quai Ernest-Ansermet 30, 1211 Geneva 4, Switzerland
| | - Matteo Grattieri
- Dipartimento
di Chimica, Università degli Studi
di Bari “Aldo Moro”, via E. Orabona 4, Bari 70125, Italy
- IPCF-CNR
Istituto per i Processi Chimico Fisici, Consiglio Nazionale delle Ricerche, via E. Orabona 4, Bari 70125, Italy
| |
Collapse
|
32
|
Yadi M, Azizi M, Dianat-Moghadam H, Akbarzadeh A, Abyadeh M, Milani M. Antibacterial activity of green gold and silver nanoparticles using ginger root extract. Bioprocess Biosyst Eng 2022; 45:1905-1917. [PMID: 36269380 DOI: 10.1007/s00449-022-02780-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Accepted: 08/28/2022] [Indexed: 11/27/2022]
Abstract
Recent studies demonstrated that the speed of synthesis, biocompatibility, and antimicrobial activity of gold (Au) and silver (Ag) metals is enhanced when biosynthesized in nano-sized particles. In the present study, Au- and Ag-based nanoparticles (NPs) were synthesized via a biological process using aqueous Ginger root extract and characterized by various spectroscopic methods. The NPs have hexagonal and spherical shapes. The average particle size for Au and Ag NPs was 20 and 15 nm, respectively. The dynamic light scattering (DLS) technique has shown that the zeta potential values of synthesized NPs were 4.8 and - 7.11 mv, respectively. Gas chromatography-mass spectrometry (GC-MS) analysis of Ginger root extract revealed 25 compounds. The synthesized NPs showed significant activity against Staphylococcus aureus and Escherichia (E). coli in vitro, with IC50 and IC90 values for Au and Ag NPs, respectively, noted to be 7.5 and 7.3 µg/ml and 15 and 15.2 µg/ml for both bacterial strains. The protein leakage level was tremendous and morphological changes occurred in bacteria treated with biosynthesized NPs. These results suggest that the biosynthesized metallic NPs have the suitable potential for application as antibacterial agents with enhanced activities.
Collapse
Affiliation(s)
- Morteza Yadi
- Department of Medical Nanotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mehdi Azizi
- Department of Tissue Engineering and Biomaterials, School of Advanced Medical Sciences and Technologies, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Hassan Dianat-Moghadam
- Department of Genetics and Molecular Biology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran.
| | - Abolfazl Akbarzadeh
- Department of Medical Nanotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Morteza Abyadeh
- Infectious Diseases Research Center, Gonabad University of Medical Sciences, Gonabad, Iran
| | - Morteza Milani
- Department of Medical Nanotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
33
|
Chamachar MM, Fazeli MR, Salimi M, Samadi N. Growth promoting activity, anti-biofilm effect, and down regulation of papC and rcsA genes expression by Medicago sativa (alfalfa) extract. FOOD BIOSCI 2022. [DOI: 10.1016/j.fbio.2022.102182] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
|
34
|
Rütten A, Kirchner T, Musiol-Kroll EM. Overview on Strategies and Assays for Antibiotic Discovery. Pharmaceuticals (Basel) 2022; 15:1302. [PMID: 36297414 PMCID: PMC9607151 DOI: 10.3390/ph15101302] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Revised: 10/12/2022] [Accepted: 10/14/2022] [Indexed: 11/23/2022] Open
Abstract
The increase in antibiotic resistance poses a major threat to global health. Actinomycetes, the Gram-positive bacteria of the order Actinomycetales, are fertile producers of bioactive secondary metabolites, including antibiotics. Nearly two-thirds of antibiotics that are used for the treatment of bacterial infections were originally isolated from actinomycetes strains belonging to the genus Streptomyces. This emphasizes the importance of actinomycetes in antibiotic discovery. However, the identification of a new antimicrobial compound and the exploration of its mode of action are very challenging tasks. Therefore, different approaches that enable the "detection" of an antibiotic and the characterization of the mechanisms leading to the biological activity are indispensable. Beyond bioinformatics tools facilitating the identification of biosynthetic gene clusters (BGCs), whole cell-screenings-in which cells are exposed to actinomycete-derived compounds-are a common strategy applied at the very early stage in antibiotic drug development. More recently, target-based approaches have been established. In this case, the drug candidates were tested for interactions with usually validated targets. This review focuses on the bioactivity-based screening methods and provides the readers with an overview on the most relevant assays for the identification of antibiotic activity and investigation of mechanisms of action. Moreover, the article includes examples of the successful application of these methods and suggestions for improvement.
Collapse
Affiliation(s)
- Anika Rütten
- Interfaculty Institute of Microbiology and Infection Medicine (IMIT), Microbiology/Biotechnology, University of Tübingen, Auf der Morgenstelle 28, 72076 Tübingen, Germany
- Cluster of Excellence ‘Controlling Microbes to Fight Infections’ (CMFI), University of Tübingen, Auf der Morgenstelle 28, 72076 Tübingen, Germany
| | - Teresa Kirchner
- Interfaculty Institute of Microbiology and Infection Medicine (IMIT), Microbiology/Biotechnology, University of Tübingen, Auf der Morgenstelle 28, 72076 Tübingen, Germany
- Cluster of Excellence ‘Controlling Microbes to Fight Infections’ (CMFI), University of Tübingen, Auf der Morgenstelle 28, 72076 Tübingen, Germany
| | - Ewa Maria Musiol-Kroll
- Interfaculty Institute of Microbiology and Infection Medicine (IMIT), Microbiology/Biotechnology, University of Tübingen, Auf der Morgenstelle 28, 72076 Tübingen, Germany
- Cluster of Excellence ‘Controlling Microbes to Fight Infections’ (CMFI), University of Tübingen, Auf der Morgenstelle 28, 72076 Tübingen, Germany
| |
Collapse
|
35
|
Akbarizare M. Photodynamic Inactivation Property of Saffron (Crocus sativus) as a Natural Photosensitizer in Combination with Blue Light in Microbial Strains. IRANIAN JOURNAL OF MEDICAL MICROBIOLOGY 2022. [DOI: 10.30699/ijmm.16.6.587] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
|
36
|
Yoneyama T, Elshamy AI, Yamada J, El-Kashak WA, Kasai Y, Imagawa H, Ban S, Noji M, Umeyama A. Antimicrobial metabolite of Cordyceps tenuipes targeting MurE ligase and histidine kinase via in silico study. Appl Microbiol Biotechnol 2022; 106:6483-6491. [DOI: 10.1007/s00253-022-12176-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Revised: 08/10/2022] [Accepted: 09/07/2022] [Indexed: 11/02/2022]
|
37
|
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
|
38
|
Clostridioides difficile Modifies its Aromatic Compound Metabolism in Response to Amidochelocardin-Induced Membrane Stress. mSphere 2022; 7:e0030222. [PMID: 35993700 PMCID: PMC9599328 DOI: 10.1128/msphere.00302-22] [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] [Indexed: 11/20/2022] Open
Abstract
Amidochelocardin is a broad-spectrum antibiotic with activity against many Gram-positive and Gram-negative bacteria. According to recent data, the antibiotic effect of this atypical tetracycline is directed against the cytoplasmic membrane, which is associated with the dissipation of the membrane potential. Here, we investigated the effect of amidochelocardin on the proteome of Clostridioides difficile to gain insight into the membrane stress physiology of this important anaerobic pathogen. For the first time, the membrane-directed action of amidochelocardin was confirmed in an anaerobic pathogen. More importantly, our results revealed that aromatic compounds potentially play an important role in C. difficile upon dissipation of its membrane potential. More precisely, a simultaneously increased production of enzymes required for the synthesis of chorismate and two putative phenazine biosynthesis proteins point to the production of a hitherto unknown compound in response to membrane depolarization. Finally, increased levels of the ClnAB efflux system and its transcriptional regulator ClnR were found, which were previously found in response to cationic antimicrobial peptides like LL-37. Therefore, our data provide a starting point for a more detailed understanding of C. difficile's way to counteract membrane-active compounds. IMPORTANCE C. difficile is an important anaerobe pathogen causing mild to severe infections of the gastrointestinal tract. To avoid relapse of the infection following antibiotic therapy, antibiotics are needed that efficiently eradicate C. difficile from the intestinal tract. Since C. difficile was shown to be substantially sensitive to membrane-active antibiotics, it has been proposed that membrane-active antibiotics might be promising for the therapy of C. difficile infections. Therefore, we studied the response of C. difficile to amidochelocardin, a membrane-active antibiotic dissipating the membrane potential. Interestingly, C. difficile's response to amidochelocardin indicates a role of aromatic metabolites in mediating stress caused by dissipation of the membrane potential.
Collapse
|
39
|
In Silico and In Vitro Assessment of Antimicrobial and Antibiofilm Activity of Some 1,3-Oxazole-Based Compounds and Their Isosteric Analogues. APPLIED SCIENCES-BASEL 2022. [DOI: 10.3390/app12115571] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
In this paper, we report on the antimicrobial activity assessment of 49 compounds previously synthesized as derivatives of alanine or phenylalanine that incorporate a 4-(4-X-phenylsulfonyl)phenyl fragment (X = H, Cl, or Br), namely 21 acyclic compounds (6 × N-acyl-α-amino acids, 1 × N-acyl-α-amino acid ester, and 14 × N-acyl-α-amino ketones) and 28 pentatomic heterocycles from the oxazole-based compound class (6 × 4H-1,3-oxazol-5-ones, 16 × 5-aryl-1,3-oxazoles, and 6 × ethyl 1,3-oxazol-5-yl carbonates). Both in silico and in vitro qualitative and quantitative assays were used to investigate the antimicrobial potential of these derivatives against planktonic and biofilm-embedded microbial strains. Some of the tested compounds showed promising antimicrobial and antibiofilm activity depending on their chemical scaffold and lipophilic character.
Collapse
|
40
|
Bryan EJ, Sagong HY, Parhi AK, Grier MC, Roberge JY, LaVoie EJ, Pilch DS. TXH11106: A Third-Generation MreB Inhibitor with Enhanced Activity against a Broad Range of Gram-Negative Bacterial Pathogens. Antibiotics (Basel) 2022; 11:antibiotics11050693. [PMID: 35625337 PMCID: PMC9137614 DOI: 10.3390/antibiotics11050693] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Revised: 05/17/2022] [Accepted: 05/18/2022] [Indexed: 02/04/2023] Open
Abstract
The emergence of multi-drug-resistant Gram-negative pathogens highlights an urgent clinical need to explore and develop new antibiotics with novel antibacterial targets. MreB is a promising antibacterial target that functions as an essential elongasome protein in most Gram-negative bacterial rods. Here, we describe a third-generation MreB inhibitor (TXH11106) with enhanced bactericidal activity versus the Gram-negative pathogens Escherichia coli, Klebsiella pneumoniae, Acinetobacter baumannii, and Pseudomonas aeruginosa compared to the first- and second-generation compounds A22 and CBR-4830, respectively. Large inocula of these four pathogens are associated with a low frequency of resistance (FOR) to TXH11106. The enhanced bactericidal activity of TXH11106 relative to A22 and CBR-4830 correlates with a correspondingly enhanced capacity to inhibit E. coli MreB ATPase activity via a noncompetitive mechanism. Morphological changes induced by TXH11106 in E. coli, K. pneumoniae, A. baumannii, and P. aeruginosa provide further evidence supporting MreB as the bactericidal target of the compound. Taken together, our results highlight the potential of TXH11106 as an MreB inhibitor with activity against a broad spectrum of Gram-negative bacterial pathogens of acute clinical importance.
Collapse
Affiliation(s)
- Eric J. Bryan
- Department of Pharmacology, Rutgers Robert Wood Johnson Medical School, Piscataway, NJ 08854, USA;
| | - Hye Yeon Sagong
- TAXIS Pharmaceuticals, Inc., Monmouth Junction, NJ 08852, USA; (H.Y.S.); (A.K.P.)
| | - Ajit K. Parhi
- TAXIS Pharmaceuticals, Inc., Monmouth Junction, NJ 08852, USA; (H.Y.S.); (A.K.P.)
| | - Mark C. Grier
- Department of Molecular Design and Synthesis, Rutgers University Biomedical Research Innovation Cores, Piscataway, NJ 08854, USA; (M.C.G.); (J.Y.R.)
| | - Jacques Y. Roberge
- Department of Molecular Design and Synthesis, Rutgers University Biomedical Research Innovation Cores, Piscataway, NJ 08854, USA; (M.C.G.); (J.Y.R.)
| | - Edmond J. LaVoie
- Department of Medicinal Chemistry, Ernest Mario School of Pharmacy, Rutgers—The State University of New Jersey, Piscataway, NJ 08854, USA;
| | - Daniel S. Pilch
- Department of Pharmacology, Rutgers Robert Wood Johnson Medical School, Piscataway, NJ 08854, USA;
- Correspondence:
| |
Collapse
|
41
|
Jhang J, Cheng C, Huang C, Chen Y, Lin J, Lou C. Application of polyhexamethylene guanidine hydrochloride to polylactic acid/polyphenylene block copolymer antibacterial composite membranes: Manufacturing technique and property evaluations. J Appl Polym Sci 2022. [DOI: 10.1002/app.52504] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Affiliation(s)
- Jia‐Ci Jhang
- Department of Textiles and Clothing Fu Jen Catholic University New Taipei Taiwan
| | - Chiao‐Chi Cheng
- Laboratory of Fiber Application and Manufacturing, Department of Fiber and Composite Materials Feng Chia University Taichung Taiwan
| | - Chen‐Hung Huang
- Department of Aerospace and Systems Engineering Feng Chia University Taichung City Taiwan
| | - Yueh‐Sheng Chen
- School of Chinese Medicine China Medical University Taichung Taiwan
| | - Jia‐Horng Lin
- Laboratory of Fiber Application and Manufacturing, Department of Fiber and Composite Materials Feng Chia University Taichung Taiwan
- School of Chinese Medicine China Medical University Taichung Taiwan
- College of Textile and Clothing Qingdao University Qingdao Shangdong China
- Fujian Key Laboratory of Novel Functional Fibers and Materials Minjiang University Fuzhou China
- Advanced Healthcare and Protective Technology Research Center Qingdao University Qingdao Shangdong China
| | - Ching‐Wen Lou
- College of Textile and Clothing Qingdao University Qingdao Shangdong China
- Fujian Key Laboratory of Novel Functional Fibers and Materials Minjiang University Fuzhou China
- Advanced Healthcare and Protective Technology Research Center Qingdao University Qingdao Shangdong China
- Feng Chia University Taichung Taiwan
- Innovation Platform of Intelligent and Energy‐Saving Textiles, School of Textiles Tianjin Polytechnic University Tianjin China
| |
Collapse
|
42
|
Gurnani M, Chauhan A, Ranjan A, Tuli HS, Alkhanani MF, Haque S, Dhama K, Lal R, Jindal T. Filamentous Thermosensitive Mutant Z: An Appealing Target for Emerging Pathogens and a Trek on Its Natural Inhibitors. BIOLOGY 2022; 11:624. [PMID: 35625352 PMCID: PMC9138142 DOI: 10.3390/biology11050624] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Revised: 03/18/2022] [Accepted: 04/01/2022] [Indexed: 12/14/2022]
Abstract
Antibiotic resistance is a major emerging issue in the health care sector, as highlighted by the WHO. Filamentous Thermosensitive mutant Z (Fts-Z) is gaining significant attention in the scientific community as a potential anti-bacterial target for fighting antibiotic resistance among several pathogenic bacteria. The Fts-Z plays a key role in bacterial cell division by allowing Z ring formation. Several in vitro and in silico experiments have demonstrated that inhibition of Fts-Z can lead to filamentous growth of the cells, and finally, cell death occurs. Many natural compounds that have successfully inhibited Fts-Z are also studied. This review article intended to highlight the structural-functional aspect of Fts-Z that leads to Z-ring formation and its contribution to the biochemistry and physiology of cells. The current trend of natural inhibitors of Fts-Z protein is also covered.
Collapse
Affiliation(s)
- Manisha Gurnani
- Amity Institute of Environmental Science, Amity University, Noida 201301, India;
| | - Abhishek Chauhan
- Amity Institute of Environmental Toxicology, Safety and Management, Amity University, Noida 201303, India;
| | - Anuj Ranjan
- Academy of Biology and Biotechnology, Southern Federal University, 344006 Rostov-on-Don, Russia
| | - Hardeep Singh Tuli
- Department of Biotechnology, Maharishi Markandeshwar (Deemed to be University), Ambala 133207, India;
| | - Mustfa F. Alkhanani
- Emergency Service Department, College of Applied Sciences, AlMaarefa University, Riyadh 11597, Saudi Arabia;
| | - Shafiul Haque
- Research and Scientific Studies Unit, College of Nursing and Allied Health Sciences, Jazan University, Jazan 45142, Saudi Arabia;
- Faculty of Medicine, Görükle Campus, Bursa Uludağ University, Nilüfer, Bursa 16059, Turkey
| | - Kuldeep Dhama
- Division of Pathology, ICAR—Indian Veterinary Research Institute, Bareilly 243122, India;
| | - Rup Lal
- Department of Zoology, University of Delhi, Delhi 110021, India;
| | - Tanu Jindal
- Amity Institute of Environmental Toxicology, Safety and Management, Amity University, Noida 201303, India;
| |
Collapse
|
43
|
Mechanisms Underlying Synergistic Killing of Polymyxin B in Combination with Cannabidiol against Acinetobacter baumannii: A Metabolomic Study. Pharmaceutics 2022; 14:pharmaceutics14040786. [PMID: 35456620 PMCID: PMC9025570 DOI: 10.3390/pharmaceutics14040786] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Revised: 03/30/2022] [Accepted: 03/31/2022] [Indexed: 12/04/2022] Open
Abstract
Polymyxins have resurged as the last-resort antibiotics against multidrug-resistant Acinetobacter baumannii. As reports of polymyxin resistance in A. baumannii with monotherapy have become increasingly common, combination therapy is usually the only remaining treatment option. A novel and effective strategy is to combine polymyxins with non-antibiotic drugs. This study aimed to investigate, using untargeted metabolomics, the mechanisms of antibacterial killing synergy of the combination of polymyxin B with a synthetic cannabidiol against A. baumannii ATCC 19606. The antibacterial synergy of the combination against a panel of Gram-negative pathogens (Acinetobacter baumannii, Klebsiella pneumoniae and Pseudomonas aeruginosa) was also explored using checkerboard and static time-kill assays. The polymyxin B–cannabidiol combination showed synergistic antibacterial activity in checkerboard and static time-kill assays against both polymyxin-susceptible and polymyxin-resistant isolates. The metabolomics study at 1 h demonstrated that polymyxin B monotherapy and the combination (to the greatest extent) significantly perturbed the complex interrelated metabolic pathways involved in the bacterial cell envelope biogenesis (amino sugar and nucleotide sugar metabolism, peptidoglycan, and lipopolysaccharide (LPS) biosynthesis), nucleotides (purine and pyrimidine metabolism) and peptide metabolism; notably, these pathways are key regulators of bacterial DNA and RNA biosynthesis. Intriguingly, the combination caused a major perturbation in bacterial membrane lipids (glycerophospholipids and fatty acids) compared to very minimal changes induced by monotherapies. At 4 h, polymyxin B–cannabidiol induced more pronounced effects on the abovementioned pathways compared to the minimal impact of monotherapies. This metabolomics study for the first time showed that in disorganization of the bacterial envelope formation, the DNA and RNA biosynthetic pathways were the most likely molecular mechanisms for the synergy of the combination. The study suggests the possibility of cannabidiol repositioning, in combination with polymyxins, for treatment of MDR polymyxin-resistant Gram-negative infections.
Collapse
|
44
|
Cebeci YU, Ceylan Ş, Karaoğlu ŞA. Conventional and microwave irradiated synthesis, biological activity evaluation of highly substituted indole-triazole hybrids. J Mol Struct 2022. [DOI: 10.1016/j.molstruc.2021.131799] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
|
45
|
Noureldin NA, Richards J, Kothayer H, Baraka MM, Eladl SM, Wootton M, Simons C. Phenylalanyl tRNA synthetase (PheRS) substrate mimics: design, synthesis, molecular dynamics and antimicrobial evaluation. RSC Adv 2022; 12:2511-2524. [PMID: 35425259 PMCID: PMC8979089 DOI: 10.1039/d1ra06439h] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Accepted: 01/05/2022] [Indexed: 11/21/2022] Open
Abstract
Nineteen novel compounds were designed to mimic Phe-AMP, as a new hope to find novel antibacterial agents and combat the antibiotic resistance. E. faecalis PheS homology model was constructed to study the mimics–enzyme interactions in more detail.
Collapse
Affiliation(s)
- Nada A. Noureldin
- School of Pharmacy and Pharmaceutical Sciences, Cardiff University, Cardiff CF10 3NB, UK
- Department of Medicinal Chemistry, Faculty of Pharmacy, Zagazig University, Zagazig P. C., 44519, Egypt
| | - Jennifer Richards
- Specialist Antimicrobial Chemotherapy Unit, University Hospital of Wales, Heath Park, Cardiff CF14 4XW, UK
| | - Hend Kothayer
- Department of Medicinal Chemistry, Faculty of Pharmacy, Zagazig University, Zagazig P. C., 44519, Egypt
| | - Mohammed M. Baraka
- Department of Medicinal Chemistry, Faculty of Pharmacy, Zagazig University, Zagazig P. C., 44519, Egypt
| | - Sobhy M. Eladl
- Department of Medicinal Chemistry, Faculty of Pharmacy, Zagazig University, Zagazig P. C., 44519, Egypt
| | - Mandy Wootton
- Specialist Antimicrobial Chemotherapy Unit, University Hospital of Wales, Heath Park, Cardiff CF14 4XW, UK
| | - Claire Simons
- School of Pharmacy and Pharmaceutical Sciences, Cardiff University, Cardiff CF10 3NB, UK
| |
Collapse
|
46
|
Khwaza V, Oyedeji OO, Aderibigbe BA, Morifi E, Fonkui YT, Ndinteh DT, Nell M, Steenkamp V. Design of Oleanolic Acid-based Hybrid Compounds as Potential Pharmaceutical Scaffolds. LETT DRUG DES DISCOV 2022. [DOI: 10.2174/1570180818666210604112451] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Background:
Infectious diseases, as well as cancer, are the leading causes of death
worldwide. Drug resistance usually results in their treatment requiring a combination of two or more
drugs.
Objective:
Oleanolic-based hybrid compounds were prepared via esterification and characterized
using FTIR, NMR and LC-MS. In vitro antibacterial and in vitro cytotoxicity studies were performed.
Method:
Oleanolic acid was hybridized with selected known pharmaceutical scaffolds via the carboxylic
acid functionality in order to develop therapeutics with increased biological activity. Antibacterial
activity was determined using the micro-dilution assay against selected Gram-positive and
Gram-negative bacteria and cytotoxicity using the sulforhodamine B assay.
Results:
Compound 8 displayed potent antibacterial effect against five strains of bacteria, such as
Bacillus subtilis, Staphylococcus aureus, Proteus vulgaris, Klebsiella oxytoca, and Escherichia coli,
with MIC values of 1.25, 0.078, 0.078, 1.25, 1.25 mg/mL when compared to the control, oleanolic
acid (MIC = 2.5 mg/mL). Furthermore, in vitro cytotoxicity, as determined using the SRB assay,
against selected cancer cells revealed that compound 7 was the most cytotoxic on MDA, DU145, and
MCF-7 cell lines with IC50 values of 69.87 ± 1.04, 73.2 ± 1.08, and 85.27 ± 1.02 μg/mL, respectively,
compared to oleanolic acid with an IC50 > 200 μg/mL.
Conclusion:
Hybridization of oleanolic acid was successful, and further development of these potential
antibacterial compounds with reduced cytotoxicity is therefore warranted.
Collapse
Affiliation(s)
- Vuyolwethu Khwaza
- Department of Chemistry, Faculty of Science and Agriculture, University of Fort Hare, Alice Campus, Alice, Eastern
Cape, South Africa
| | - Opeoluwa Oyehan Oyedeji
- Department of Chemistry, Faculty of Science and Agriculture, University of Fort Hare, Alice Campus, Alice, Eastern
Cape, South Africa
| | - Blessing Atim Aderibigbe
- Department of Chemistry, Faculty of Science and Agriculture, University of Fort Hare, Alice Campus, Alice, Eastern
Cape, South Africa
| | - Eric Morifi
- School of Chemistry, Mass Spectrometry division, University of the Witwatersrand, Johannesburg
Private Bag X3, WITS, 2050, South Africa
| | - Youmbi Thierry Fonkui
- Department of Biotechnology and Food Technology, Faculty of Science,
University of Johannesburg, Doornfontein Campus, Johannesburg, South Africa
| | - Derek Tantoh Ndinteh
- Department of Applied Chemistry,
Faculty of Science, University of Johannesburg, Doornfontein Campus, Johannesburg, South Africa
| | - Margo Nell
- Department of
Pharmacology, Faculty of Health Sciences, University of Pretoria, South Africa
| | - Vanessa Steenkamp
- Department of
Pharmacology, Faculty of Health Sciences, University of Pretoria, South Africa
| |
Collapse
|
47
|
Ali A, Ahmad S, de Albuquerque PMM, Kamil A, Alshammari FA, Alouffi A, da Silva Vaz I. Prediction of Novel Drug Targets and Vaccine Candidates against Human Lice (Insecta), Acari (Arachnida), and Their Associated Pathogens. Vaccines (Basel) 2021; 10:8. [PMID: 35062669 PMCID: PMC8778234 DOI: 10.3390/vaccines10010008] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Revised: 12/14/2021] [Accepted: 12/20/2021] [Indexed: 01/12/2023] Open
Abstract
The emergence of drug-resistant lice, acari, and their associated pathogens (APs) is associated with economic losses; thus, it is essential to find new appropriate therapeutic approaches. In the present study, a subtractive proteomics approach was used to predict suitable therapeutics against these vectors and their infectious agents. We found 9701 proteins in the lice (Pediculus humanus var. corporis) and acari (Ixodes scapularis, Leptotrombidium deliense), and 4822 proteins in the proteomes of their APs (Babesia microti, Borreliella mayonii, Borrelia miyamotoi, Borrelia recurrentis, Rickettsia prowazekii, Orientia tsutsugamushi str. Boryong) that were non-homologous to host proteins. Among these non-homologous proteins, 365 proteins of lice and acari, and 630 proteins of APs, were predicted as essential proteins. Twelve unique essential proteins were predicted to be involved in four unique metabolic pathways of lice and acari, and 103 unique proteins were found to be involved in 75 unique metabolic pathways of APs. The sub cellular localization analysis of 115 unique essential proteins of lice and acari and their APs revealed that 61 proteins were cytoplasmic, 42 as membrane-bound proteins and 12 proteins with multiple localization. The druggability analysis of the identified 73 cytoplasmic and multiple localization essential proteins revealed 22 druggable targets and 51 novel drug targets that participate in unique pathways of lice and acari and their APs. Further, the predicted 42 membrane bound proteins could be potential vaccine candidates. Screening of useful inhibitors against these novel targets may result in finding novel compounds efficient for the control of these parasites.
Collapse
Affiliation(s)
- Abid Ali
- Department of Zoology, Abdul Wali Khan University Mardan, Mardan 23200, Pakistan;
- Centro de Biotecnologia, Universidade Federal do Rio Grande do Sul, Porto Alegre 91501-970, Brazil; (S.A.); (P.M.M.d.A.)
| | - Shabir Ahmad
- Centro de Biotecnologia, Universidade Federal do Rio Grande do Sul, Porto Alegre 91501-970, Brazil; (S.A.); (P.M.M.d.A.)
| | | | - Atif Kamil
- Department of Biotechnology, Abdul Wali Khan University Mardan, Mardan 23200, Pakistan;
| | - Fahdah Ayed Alshammari
- College of Sciences and Literature Microbiology, Nothern Border University, Rafha 76413, Saudi Arabia;
| | - Abdulaziz Alouffi
- King Abdulaziz City for Science and Technology, Riyadh 12354, Saudi Arabia;
- Vaccines Research for Infectious Diseases, King Saud University, Riyadh 11495, Saudi Arabia
- Veterinary Laboratories and Vaccines Center, Ministry of Environment Water & Agriculture, Riyadh 11195, Saudi Arabia
| | - Itabajara da Silva Vaz
- Centro de Biotecnologia, Universidade Federal do Rio Grande do Sul, Porto Alegre 91501-970, Brazil; (S.A.); (P.M.M.d.A.)
| |
Collapse
|
48
|
Hematian A, Goudarzi H, Ghalavand Z, Goudarzi M, Shafieian M, Hashemi A, Ghafourian S. The relationship between phoH and colistin-heteroresistant in clinical isolates of Acinetobacter baumannii. GENE REPORTS 2021. [DOI: 10.1016/j.genrep.2021.101356] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
49
|
Predicting drug targets by homology modelling of Pseudomonas aeruginosa proteins of unknown function. PLoS One 2021; 16:e0258385. [PMID: 34648550 PMCID: PMC8516228 DOI: 10.1371/journal.pone.0258385] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Accepted: 09/24/2021] [Indexed: 11/19/2022] Open
Abstract
The efficacy of antibiotics to treat bacterial infections declines rapidly due to antibiotic resistance. This problem has stimulated the development of novel antibiotics, but most attempts have failed. Consequently, the idea of mining uncharacterized genes of pathogens to identify potential targets for entirely new classes of antibiotics was proposed. Without knowing the biochemical function of a protein, it is difficult to validate its potential for drug targeting; therefore, the functional characterization of bacterial proteins of unknown function must be accelerated. Here, we present a paradigm for comprehensively predicting the biochemical functions of a large set of proteins encoded by hypothetical genes in human pathogens to identify candidate drug targets. A high-throughput approach based on homology modelling with ten templates per target protein was applied to the set of 2103 P. aeruginosa proteins encoded by hypothetical genes. The >21000 homology modelling results obtained and available biological and biochemical information about several thousand templates were scrutinized to predict the function of reliably modelled proteins of unknown function. This approach resulted in assigning one or often multiple putative functions to hundreds of enzymes, ligand-binding proteins and transporters. New biochemical functions were predicted for 41 proteins whose essential or virulence-related roles in P. aeruginosa were already experimentally demonstrated. Eleven of them were shortlisted as promising drug targets that participate in essential pathways (maintaining genome and cell wall integrity), virulence-related processes (adhesion, cell motility, host recognition) or antibiotic resistance, which are general drug targets. These proteins are conserved in other WHO priority pathogens but not in humans; therefore, they represent high-potential targets for preclinical studies. These and many more biochemical functions assigned to uncharacterized proteins of P. aeruginosa, made available as PaPUF database, may guide the design of experimental screening of inhibitors, which is a crucial step towards the validation of the highest-potential targets for the development of novel drugs against P. aeruginosa and other high-priority pathogens.
Collapse
|
50
|
GC-MS Analysis and Antimicrobial Activity of an Iranian Traditional Medicinal Smoke (Anbarnasara). JOURNAL OF MEDICAL MICROBIOLOGY AND INFECTIOUS DISEASES 2021. [DOI: 10.52547/jommid.9.3.148] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
|