1
|
Ngaini Z, Hissam MA, Mortadza NA, Abd Halim AN, Daud AI. In vitro antimicrobial activities, molecular docking and density functional theory (DFT) evaluation of natural product-based vanillin derivatives featuring halogenated azo dyes. Nat Prod Res 2024; 38:3762-3772. [PMID: 37752781 DOI: 10.1080/14786419.2023.2262713] [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: 05/31/2023] [Accepted: 09/17/2023] [Indexed: 09/28/2023]
Abstract
Chemical modification of active scaffolds from natural products has gained interest in pharmaceutical industries. Nevertheless, the metabolites extraction is time-consuming while the lead is frequently mismatched with the receptor. Here, the diazo coupling approach was introduced to generate a series of vanillin derivatives featuring halogenated azo dyes (1a-h). The vanillin derivatives showed effective inhibition of S. aureus (7-9 mm) and E. coli (7-8 mm) compared to the parent vanillin, while 1b had the highest inhibition zone (9 mm) against S. aureus comparable to the reference ampicillin. The presence of N = N, C = O, -OH, -OCH3 and halogens established strategic binding interactions with the receptor. The potential vanillin-azo as an antimicrobial drug was supported by in silico docking with penicillin-binding proteins and DFT (using Gaussian 09) with binding affinity -7.5 kcal/mol and energy gap (Egap) 3.77 eV, respectively. This study represents a significant advancement in drug discovery for effective antibiotics with excellent properties.
Collapse
Affiliation(s)
- Zainab Ngaini
- Faculty of Resource Science and Technology, Universiti Malaysia Sarawak, Kota Samarahan, Sarawak, Malaysia
| | - Mohamad Azmeer Hissam
- Faculty of Resource Science and Technology, Universiti Malaysia Sarawak, Kota Samarahan, Sarawak, Malaysia
| | - Nur Arif Mortadza
- Faculty of Resource Science and Technology, Universiti Malaysia Sarawak, Kota Samarahan, Sarawak, Malaysia
| | - Ainaa Nadiah Abd Halim
- Faculty of Resource Science and Technology, Universiti Malaysia Sarawak, Kota Samarahan, Sarawak, Malaysia
| | - Adibah Izzati Daud
- Faculty of Chemical Engineering and Technology, Universiti Malaysia Perlis, Padang Besar, Perlis, Malaysia
| |
Collapse
|
2
|
Davignon G, Pietrosemoli N, Benaroudj N, Soupé-Gilbert ME, Cagliero J, Turc É, Picardeau M, Guentas L, Goarant C, Thibeaux R. Leptospira interrogans biofilm transcriptome highlights adaption to starvation and general stress while maintaining virulence. NPJ Biofilms Microbiomes 2024; 10:95. [PMID: 39349472 PMCID: PMC11442865 DOI: 10.1038/s41522-024-00570-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2024] [Accepted: 09/15/2024] [Indexed: 10/02/2024] Open
Abstract
Life-threatening Leptospira interrogans navigate a dual existence: surviving in the environment and infecting mammalian hosts. Biofilm formation is presumably an important survival strategy to achieve this process. Understanding the relation between biofilm and virulence might improve our comprehension of leptospirosis epidemiology. Our study focused on elucidating Leptospira's adaptations and regulations involved in such complex microenvironments. To determine the transcriptional profile of Leptospira in biofilm, we compared the transcriptomes in late biofilms and in exponential planktonic cultures. While genes for motility, energy production, and metabolism were downregulated, those governing general stress response, defense against metal stress, and redox homeostasis showed a significant upsurge, hinting at a tailored defensive strategy against stress. Further, despite a reduced metabolic state, biofilm disruption swiftly restored metabolic activity. Crucially, bacteria in late biofilms or resulting from biofilm disruption retained virulence in an animal model. In summary, our study highlights Leptospira's adaptive equilibrium in biofilms: minimizing energy expenditure, potentially aiding in withstanding stresses while maintaining pathogenicity. These insights are important for explaining the survival strategies of Leptospira, revealing that a biofilm lifestyle may confer an advantage in maintaining virulence, an understanding essential for managing leptospirosis across both environmental and mammalian reservoirs.
Collapse
Affiliation(s)
- Grégoire Davignon
- Leptospirosis Research and Expertise Unit, Institut Pasteur de Nouvelle-Calédonie, Institut Pasteur International Network, Nouméa, New Caledonia
- Exact and Applied Sciences Institute (ISEA), University of New Caledonia, BP R4, 98851, Nouméa, New Caledonia
| | - Natalia Pietrosemoli
- Institut Pasteur, Université Paris Cité, Bioinformatics and Biostatistics Hub, F-75015, Paris, France
| | - Nadia Benaroudj
- Biology of Spirochetes, Institut Pasteur, Université Paris Cité, CNRS UMR 6047, F-75015, Paris, France
| | - Marie-Estelle Soupé-Gilbert
- Leptospirosis Research and Expertise Unit, Institut Pasteur de Nouvelle-Calédonie, Institut Pasteur International Network, Nouméa, New Caledonia
| | - Julie Cagliero
- Leptospirosis Research and Expertise Unit, Institut Pasteur de Nouvelle-Calédonie, Institut Pasteur International Network, Nouméa, New Caledonia
| | - Élodie Turc
- Institut Pasteur, Université Paris Cité, Plate-forme Technologique Biomics, F-75015, Paris, France
| | - Mathieu Picardeau
- Biology of Spirochetes, Institut Pasteur, Université Paris Cité, CNRS UMR 6047, F-75015, Paris, France
| | - Linda Guentas
- Exact and Applied Sciences Institute (ISEA), University of New Caledonia, BP R4, 98851, Nouméa, New Caledonia
| | - Cyrille Goarant
- Pacific Community SPC - Public Health Division - B.P. D5, Nouméa, New Caledonia
| | - Roman Thibeaux
- Leptospirosis Research and Expertise Unit, Institut Pasteur de Nouvelle-Calédonie, Institut Pasteur International Network, Nouméa, New Caledonia.
| |
Collapse
|
3
|
Anand A, Verma A, Kaur S, Kathayat P, Manoj RM, Aakanksha A, Turzin JK, Satapathy P, Khatib MN, Gaidhane S, Zahiruddin QS, Kukreti N, Rustagi S, Surana A. An overview of sulbactam-durlobactam approval and implications in advancing therapeutics for hospital-acquired and ventilator-associated pneumonia by acinetobacter baumannii-calcoaceticus complex: A narrative review. Health Sci Rep 2024; 7:e70066. [PMID: 39257909 PMCID: PMC11386240 DOI: 10.1002/hsr2.70066] [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: 01/17/2024] [Revised: 08/26/2024] [Accepted: 08/29/2024] [Indexed: 09/12/2024] Open
Abstract
Purpose Infections caused by Acinetobacter baumannii, particularly those resistant to antibiotics such as carbapenem, have become a global health crisis with a significant mortality rate. Hospital-acquired pneumonia (HAP) and ventilator-associated pneumonia (VAP) resulting from the A. baumannii-calcoaceticus (ABC) complex represent a major clinical challenge. This review aimed to understand the approval process, mechanism of action, therapeutic potential, and future implications of sulbactam-durlobactam therapy (SUL-DUR). Methods PubMed, Web of Science, EMBASE, Clinical trials. gov, ICTRP, and CENTRAL were searched for studies on SUL-DUR for the treatment of hospital-acquired pneumonia and ventilator-associated pneumonia. Also, World Health Organization, U.S. Food and Drug Administration, and Centers for Disease Control and Prevention websites were searched for relevant information. Results SUL-DUR, marketed as Xacduro, is a novel pharmaceutical combination that functions as a narrow-spectrum parenterally administered antibiotic. Sulbactam acts as a β-lactamase inhibitor, whereas durlobactam protects against degradation by A. baumannii enzymes. A phase 1 trial successfully established the safety and tolerability of SUL-DUR in patients with normal and mild renal impairment. A phase 2 trial demonstrated the safety and tolerability of SUL-DUR in a larger population with urinary tract infections. A phase 3 trial showed that SUL-DUR was non-inferior to colistin in terms of mortality in A. baumannii-related VAP, HAP, and bacteremia. Conclusion The combination of sulbactam and durlobactam is a promising treatment option for HAP and VAP caused by ABC complex.
Collapse
Affiliation(s)
- Ayush Anand
- B. P. Koirala Institute of Health Sciences Dharan Nepal
- MediSurg Research Darbhanga India
- Global Consortium of Medical Education and Research Pune India
| | - Amogh Verma
- Rama Medical College Hospital and Research Centre Hapur India
| | - Sarabjeet Kaur
- Global Consortium of Medical Education and Research Pune India
- Government Medical College Patiala India
| | - Priyangi Kathayat
- Global Consortium of Medical Education and Research Pune India
- Smt. NHL Municipal Medical College Ahmedabad India
| | - Rachel M Manoj
- Global Consortium of Medical Education and Research Pune India
- Nicolae Testemițanu State University of Medicine and Pharmacy Chisinau Moldova
| | - Aakanksha Aakanksha
- Global Consortium of Medical Education and Research Pune India
- Tbilisi State Medical University Tbilisi Georgia
| | - Justice K Turzin
- Global Consortium of Medical Education and Research Pune India
- Department of Biomedical Science, College of Health and Allied Sciences University of Cape Coast Cape Coast Ghana
| | - Prakasini Satapathy
- Center for Global Health Research, Saveetha Institute of Medical and Technical Sciences, Saveetha Medical College and Hospital Saveetha University Chennai India
- Medical Laboratories Techniques Department AL-Mustaqbal University Hillah Iraq
| | - Mahalaqua N Khatib
- Division of Evidence Synthesis, Datta Meghe Institute of Higher Education Global Consortium of Public Health and Research Wardha India
| | - Shilpa Gaidhane
- Datta Meghe Institute of Higher Education and Research Jawaharlal Nehru Medical College Wardha India
| | - Quazi S Zahiruddin
- Division of Evidence Synthesis, Datta Meghe Institute of Higher Education South Asia Infant Feeding Research Network (SAIFRN), Global Consortium of Public Health and Research Wardha India
| | - Neelima Kukreti
- School of Pharmacy Graphic Era Hill University Dehradun India
| | - Sarvesh Rustagi
- School of Applied and Life Sciences Uttaranchal University Dehradun India
| | - Arihant Surana
- Department of Internal Medicine St. Vincent Hospital Worcester Massachusetts USA
| |
Collapse
|
4
|
Shahalaei M, Azad AK, Sulaiman WMAW, Derakhshani A, Mofakham EB, Mallandrich M, Kumarasamy V, Subramaniyan V. A review of metallic nanoparticles: present issues and prospects focused on the preparation methods, characterization techniques, and their theranostic applications. Front Chem 2024; 12:1398979. [PMID: 39206442 PMCID: PMC11351095 DOI: 10.3389/fchem.2024.1398979] [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: 03/11/2024] [Accepted: 07/04/2024] [Indexed: 09/04/2024] Open
Abstract
Metallic nanoparticles (MNPs) have garnered significant attention due to their ability to improve the therapeutic index of medications by reducing multidrug resistance and effectively delivering therapeutic agents through active targeting. In addition to drug delivery, MNPs have several medical applications, including in vitro and in vivo diagnostics, and they improve the biocompatibility of materials and nutraceuticals. MNPs have several advantages in drug delivery systems and genetic manipulation, such as improved stability and half-life in circulation, passive or active targeting into the desired target selective tissue, and gene manipulation by delivering genetic materials. The main goal of this review is to provide current information on the present issues and prospects of MNPs in drug and gene delivery systems. The current study focused on MNP preparation methods and their characterization by different techniques, their applications to targeted delivery, non-viral vectors in genetic manipulation, and challenges in clinical trial translation.
Collapse
Affiliation(s)
- Mona Shahalaei
- Biomaterial Group, Nanotechnology and Advanced Materials Department, Materials and Energy Research Center, Karaj, Iran
| | - Abul Kalam Azad
- Department of Pharmaceutical Technology, Faculty of Pharmacy, University College of MAIWP International (UCMI), Kuala Lumpur, Malaysia
| | - Wan Mohd Azizi Wan Sulaiman
- Department of Pharmaceutical Technology, Faculty of Pharmacy, University College of MAIWP International (UCMI), Kuala Lumpur, Malaysia
| | - Atefeh Derakhshani
- Department of Tissue Engineering and Applied Cell Sciences, Faculty of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Elmira Banaee Mofakham
- Biomaterial Group, Nanotechnology and Advanced Materials Department, Materials and Energy Research Center, Karaj, Iran
| | - Mireia Mallandrich
- Department of Pharmacy, Pharmaceutical Technology and Physical-Chemistry, Faculty of Pharmacy and Food Sciences, University of Barcelona, Barcelona, Spain
| | - Vinoth Kumarasamy
- Department of Parasitology and Medical Entomology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| | - Vetriselvan Subramaniyan
- Department of Medical Sciences, School of Medical and Life Sciences, Sunway University, Sunway, Malaysia
| |
Collapse
|
5
|
Kulig K, Kowalik K, Surowiec M, Karnas E, Barczyk-Woznicka O, Zuba-Surma E, Pyza E, Kozik A, Rapala-Kozik M, Karkowska-Kuleta J. Isolation and Characteristics of Extracellular Vesicles Produced by Probiotics: Yeast Saccharomyces boulardii CNCM I-745 and Bacterium Streptococcus salivarius K12. Probiotics Antimicrob Proteins 2024; 16:936-948. [PMID: 37209320 PMCID: PMC11126510 DOI: 10.1007/s12602-023-10085-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/04/2023] [Indexed: 05/22/2023]
Abstract
Numerous probiotic microorganisms have repeatedly been shown to produce nanometer-sized structures named extracellular vesicles (EVs). Recently, it has been suggested that similarly to whole microbial cells, EVs produced by probiotics may also demonstrate health benefits to the host, while their application does not involve the risk of infection caused by live microorganisms. In this work, we isolated EVs from two probiotic species originating from different taxonomic domains - yeast Saccharomyces boulardii CNCM I-745 and bacterium Streptococcus salivarius K12. The diameters of S. boulardii EVs were about 142 nm and for S. salivarius EVs about 123 nm. For S. boulardii EVs, 1641 proteins and for S. salivarius EVs, 466 proteins were identified with a liquid chromatography-coupled tandem mass spectrometry and then functionally classified. In both microbial species, metabolic proteins significantly contributed to the cargo of EVs comprising 25% and 26% of all identified vesicular proteins for fungi and bacteria, respectively. Moreover, enzymes associated with cell wall rearrangement, including enzymatically active glucanases, were also identified in EVs. Furthermore, probiotic EVs were shown to influence host cells and stimulate the production of IL-1β and IL-8 by the human monocytic cell line THP-1, and, at the same time, did not cause any remarkable reduction in the survival rate of Galleria mellonella larvae in this invertebrate model commonly used to evaluate microbial EV toxicity. These observations suggest that the EVs produced by the investigated probiotic microorganisms may be promising structures for future use in pro-health applications.
Collapse
Affiliation(s)
- Kamila Kulig
- Department of Comparative Biochemistry and Bioanalytics, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Kraków, Poland
| | - Katarzyna Kowalik
- Department of Comparative Biochemistry and Bioanalytics, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Kraków, Poland
| | - Magdalena Surowiec
- Department of Comparative Biochemistry and Bioanalytics, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Kraków, Poland
- Doctoral School of Exact and Natural Sciences, Jagiellonian University, Kraków, Poland
| | - Elzbieta Karnas
- Department of Cell Biology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Kraków, Poland
| | - Olga Barczyk-Woznicka
- Department of Cell Biology and Imaging, Institute of Zoology and Biomedical Research, Jagiellonian University, Kraków, Poland
| | - Ewa Zuba-Surma
- Department of Cell Biology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Kraków, Poland
| | - Elzbieta Pyza
- Department of Cell Biology and Imaging, Institute of Zoology and Biomedical Research, Jagiellonian University, Kraków, Poland
| | - Andrzej Kozik
- Department of Analytical Biochemistry, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Kraków, Poland
| | - Maria Rapala-Kozik
- Department of Comparative Biochemistry and Bioanalytics, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Kraków, Poland
| | - Justyna Karkowska-Kuleta
- Department of Comparative Biochemistry and Bioanalytics, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Kraków, Poland.
| |
Collapse
|
6
|
Hsu TW, Fang JM. Advances and prospects of analytic methods for bacterial transglycosylation and inhibitor discovery. Analyst 2024; 149:2204-2222. [PMID: 38517346 DOI: 10.1039/d3an01968c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/23/2024]
Abstract
The cell wall is essential for bacteria to maintain structural rigidity and withstand external osmotic pressure. In bacteria, the cell wall is composed of peptidoglycan. Lipid II is the basic unit for constructing highly cross-linked peptidoglycan scaffolds. Transglycosylase (TGase) is the initiating enzyme in peptidoglycan synthesis that catalyzes the ligation of lipid II moieties into repeating GlcNAc-MurNAc polysaccharides, followed by transpeptidation to generate cross-linked structures. In addition to the transglycosylases in the class-A penicillin-binding proteins (aPBPs), SEDS (shape, elongation, division and sporulation) proteins are also present in most bacteria and play vital roles in cell wall renewal, elongation, and division. In this review, we focus on the latest analytical methods including the use of radioactive labeling, gel electrophoresis, mass spectrometry, fluorescence labeling, probing undecaprenyl pyrophosphate, fluorescence anisotropy, ligand-binding-induced tryptophan fluorescence quenching, and surface plasmon resonance to evaluate TGase activity in cell wall formation. This review also covers the discovery of TGase inhibitors as potential antibacterial agents. We hope that this review will give readers a better understanding of the chemistry and basic research for the development of novel antibiotics.
Collapse
Affiliation(s)
- Tse-Wei Hsu
- Department of Chemistry, National Taiwan University, Taipei 106, Taiwan.
| | - Jim-Min Fang
- Department of Chemistry, National Taiwan University, Taipei 106, Taiwan.
| |
Collapse
|
7
|
Liang Y, Wang J, Liu X, Chen S, He G, Fang X, Yang J, Teng Z, Liu HB. Anti-adhesion multifunctional poly(lactic-co-glycolic acid)/polydimethylsiloxane wound dressing for bacterial infection monitoring and photodynamic antimicrobial therapy. Int J Biol Macromol 2024; 260:129501. [PMID: 38224803 DOI: 10.1016/j.ijbiomac.2024.129501] [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: 08/07/2023] [Revised: 01/06/2024] [Accepted: 01/12/2024] [Indexed: 01/17/2024]
Abstract
Wound infection and adhesion are important factors affecting wound healing. Early detection of pathogen infection and reduction of wound-to-dressing adhesion are critical for improving wound healing. Herein, Ester-J, which can rapidly respond to lipase secreted by bacteria, was designed and synthesized. Then, Ester-J was co-spun with poly(lactic-co-glycolic acid) (PLGA) and polydimethylsiloxane (PDMS) to prepare a PP-EsJ hydrophobic anti-adhesion dressing with a contact angle of 140.7°. When the PP-EsJ membrane came into contact with the bacteria, the loaded Ester-J was hydrolyzed to Tph-TSF-OH, releasing bright cyan-blue fluorescence, thus providing a fluorescence switch for an early warning of infection. The detection limits of PP-EsJ for Pseudomonas aeruginosa and Staphylococcus aureus were 1.0 × 105 and 1.0 × 106 CFU/mL, respectively. Subsequently, Tph-TSF-OH released 1O2 through light irradiation, which rapidly killed P. aeruginosa and S. aureus, and accelerated wound healing. Compared with the control group, enhanced wound closure (up to 99.80 ± 1.10 %) was observed in mice treated with the PP-EsJ membrane. The PP-EsJ membrane not only effectively reduced the risk of external infection but also reduced adhesions to the skin during dressing changes. These characteristics make PP-EsJ membranes potentially useful for clinical treatment.
Collapse
Affiliation(s)
- Yuehui Liang
- School of Chemistry and Chemical Engineering, Guangxi University, Nanning 53004, PR China
| | - Jing Wang
- School of Chemistry and Chemical Engineering, Guangxi University, Nanning 53004, PR China
| | - Xu Liu
- Medical College of Guangxi University, Guangxi University, Nanning 53004, PR China
| | - Shirong Chen
- School of Chemistry and Chemical Engineering, Guangxi University, Nanning 53004, PR China
| | - Guangpeng He
- School of Chemistry and Chemical Engineering, Guangxi University, Nanning 53004, PR China
| | - Xiru Fang
- School of Chemistry and Chemical Engineering, Guangxi University, Nanning 53004, PR China
| | - Jiaying Yang
- School of Chemistry and Chemical Engineering, Guangxi University, Nanning 53004, PR China
| | - Zhongshan Teng
- School of Chemistry and Chemical Engineering, Guangxi University, Nanning 53004, PR China
| | - Hai-Bo Liu
- School of Chemistry and Chemical Engineering, Guangxi University, Nanning 53004, PR China.
| |
Collapse
|
8
|
Huang LD, Gou XY, Yang MJ, Li MJ, Chen SN, Yan J, Liu XX, Sun AH. Peptidoglycan biosynthesis-associated enzymatic kinetic characteristics and β-lactam antibiotic inhibitory effects of different Streptococcus pneumoniae penicillin-binding proteins. Int J Biol Macromol 2024; 254:127784. [PMID: 37949278 DOI: 10.1016/j.ijbiomac.2023.127784] [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: 05/24/2023] [Revised: 09/15/2023] [Accepted: 10/28/2023] [Indexed: 11/12/2023]
Abstract
Penicillin-binding proteins (PBPs) include transpeptidases, carboxypeptidases, and endopeptidases for biosynthesis of peptidoglycans in the cell wall to maintain bacterial morphology and survival in the environment. Streptococcus pneumoniae expresses six PBPs, but their enzymatic kinetic characteristics and inhibitory effects on different β-lactam antibiotics remain poorly understood. In this study, all the six recombinant PBPs of S. pneumoniae displayed transpeptidase activity with different substrate affinities (Km = 1.56-9.11 mM) in a concentration-dependent manner, and rPBP3 showed a greater catalytic efficiency (Kcat = 2.38 s-1) than the other rPBPs (Kcat = 3.20-7.49 × 10-2 s-1). However, only rPBP3 was identified as a carboxypeptidase (Km = 8.57 mM and Kcat = 2.57 s-1). None of the rPBPs exhibited endopeptidase activity. Penicillin and cefotaxime inhibited the transpeptidase and carboxypeptidase activity of all the rPBPs but imipenem did not inhibited the enzymatic activities of rPBP3. Except for the lack of binding of imipenem to rPBP3, penicillin, cefotaxime, and imipenem bound to all the other rPBPs (KD = 3.71-9.35 × 10-4 M). Sublethal concentrations of penicillin, cefotaxime, and imipenem induced a decrease of pneumococcal pbps-mRNA levels (p < 0.05). These results indicated that all six PBPs of S. pneumoniae are transpeptidases, while only PBP3 is a carboxypeptidase. Imipenem has no inhibitory effect on pneumococcal PBP3. The pneumococcal genes for encoding endopeptidases remain to be determined.
Collapse
Affiliation(s)
- Li-Dan Huang
- School of Basic Medical Sciences and Forensic Medicine, Hangzhou Medical College, Hangzhou, Zhejiang 310053, PR China; Yiwu Central Blood Station, Yiwu, Zhejiang 322000, PR China
| | - Xiao-Yu Gou
- School of Basic Medical Sciences and Forensic Medicine, Hangzhou Medical College, Hangzhou, Zhejiang 310053, PR China
| | - Mei-Juan Yang
- School of Basic Medical Sciences and Forensic Medicine, Hangzhou Medical College, Hangzhou, Zhejiang 310053, PR China; The First Hospital of Putian City, Putian, Fujian 351100, PR China
| | - Meng-Jie Li
- School of Basic Medical Sciences and Forensic Medicine, Hangzhou Medical College, Hangzhou, Zhejiang 310053, PR China
| | - Sui-Ning Chen
- School of Basic Medical Sciences and Forensic Medicine, Hangzhou Medical College, Hangzhou, Zhejiang 310053, PR China
| | - Jie Yan
- Zhejiang University School of Medicine, Hangzhou, Zhejiang 310058, PR China
| | - Xiao-Xiang Liu
- School of Basic Medical Sciences and Forensic Medicine, Hangzhou Medical College, Hangzhou, Zhejiang 310053, PR China.
| | - Ai-Hua Sun
- School of Basic Medical Sciences and Forensic Medicine, Hangzhou Medical College, Hangzhou, Zhejiang 310053, PR China.
| |
Collapse
|
9
|
Feng J, Zheng Y, Ma W, Ihsan A, Hao H, Cheng G, Wang X. Multitarget antibacterial drugs: An effective strategy to combat bacterial resistance. Pharmacol Ther 2023; 252:108550. [PMID: 39492518 DOI: 10.1016/j.pharmthera.2023.108550] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Revised: 10/19/2023] [Accepted: 10/23/2023] [Indexed: 11/05/2024]
Abstract
The rise of antibiotic resistance and the decrease in the discovery of new antibiotics have caused a global health crisis. Of particular concern is the fact that despite efforts to develop new antibiotics, drug discovery is unable to keep up with the rapid development of resistance. This ongoing crisis highlights the fact that single-target drugs may not always exhibit satisfactory therapeutic effects and are prone to target mutations and resistance due to the complexity of bacterial mechanisms. Retrospective studies have shown that most successful antibiotics have multiple targets. Compared with single-target drugs, successfully designed multitarget drugs can simultaneously regulate multiple targets to reduce resistance caused by single-target mutations or expression changes. In addition to a lower risk of drug-drug interactions, multitarget drugs show superior pharmacokinetics and higher patient compliance compared with combination therapies. Therefore, to reduce resistance, many efforts have been made to discover and design multitarget drugs with different chemical structures and functions. Although there have been numerous studies on how to develop drugs and slow down the development of drug resistance, the reduction of bacterial resistance by multitarget antibacterial drugs has not received widespread attention and is rarely mentioned in the peer-reviewed literature. This review summarises the development of antibiotic resistance and the mechanisms proposed for its emergence, examines the potential of multitarget drugs as an effective strategy to slow the development of resistance, and discusses the rationale for multitarget drug therapy. We also describe multitarget antibacterial compounds with the potential to reduce drug resistance and the available strategies to develop multitarget drugs.
Collapse
Affiliation(s)
- Jin Feng
- National Reference Laboratory of Veterinary Drug Residues (HZAU) and MAO Key Laboratory for Detection of Veterinary Drug Residues, Huazhong Agricultural University, Wuhan, Hubei 430070, China
| | - Youle Zheng
- MAO Laboratory for Risk Assessment of Quality and Safety of Livestock and Poultry Products, Huazhong Agricultural University, Wuhan, Hubei 430070, China
| | - Wanqing Ma
- MAO Laboratory for Risk Assessment of Quality and Safety of Livestock and Poultry Products, Huazhong Agricultural University, Wuhan, Hubei 430070, China
| | - Awais Ihsan
- Department of Biosciences, COMSATS University Islamabad, Sahiwal Campus, Islamabad 45550, Pakistan
| | - Haihong Hao
- National Reference Laboratory of Veterinary Drug Residues (HZAU) and MAO Key Laboratory for Detection of Veterinary Drug Residues, Huazhong Agricultural University, Wuhan, Hubei 430070, China; MAO Laboratory for Risk Assessment of Quality and Safety of Livestock and Poultry Products, Huazhong Agricultural University, Wuhan, Hubei 430070, China
| | - Guyue Cheng
- National Reference Laboratory of Veterinary Drug Residues (HZAU) and MAO Key Laboratory for Detection of Veterinary Drug Residues, Huazhong Agricultural University, Wuhan, Hubei 430070, China; MAO Laboratory for Risk Assessment of Quality and Safety of Livestock and Poultry Products, Huazhong Agricultural University, Wuhan, Hubei 430070, China
| | - Xu Wang
- National Reference Laboratory of Veterinary Drug Residues (HZAU) and MAO Key Laboratory for Detection of Veterinary Drug Residues, Huazhong Agricultural University, Wuhan, Hubei 430070, China; MAO Laboratory for Risk Assessment of Quality and Safety of Livestock and Poultry Products, Huazhong Agricultural University, Wuhan, Hubei 430070, China.
| |
Collapse
|
10
|
Hahn V. Potential of the enzyme laccase for the synthesis and derivatization of antimicrobial compounds. World J Microbiol Biotechnol 2023; 39:107. [PMID: 36854853 PMCID: PMC9974771 DOI: 10.1007/s11274-023-03539-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2022] [Accepted: 02/01/2023] [Indexed: 03/02/2023]
Abstract
Laccases [E.C. 1.10.3.2, benzenediol:dioxygen oxidoreductase] can oxidize phenolic substances, e.g. di- and polyphenols, hydroxylated biaryls, aminophenols or aryldiamines. This large substrate spectrum is the basis for various reaction possibilities, which include depolymerization and polymerization reactions, but also the coupling of different substance classes. To catalyze these reactions, laccases demand only atmospheric oxygen and no depletive cofactors. The utilization of mild and environmentally friendly reaction conditions such as room temperature, atmospheric pressure, and the avoidance of organic solvents makes the laccase-mediated reaction a valuable tool in green chemistry for the synthesis of biologically active compounds such as antimicrobial substances. In particular, the production of novel antibiotics becomes vital due to the evolution of antibiotic resistances amongst bacteria and fungi. Therefore, laccase-mediated homo- and heteromolecular coupling reactions result in derivatized or newly synthesized antibiotics. The coupling or derivatization of biologically active compounds or its basic structures may allow the development of novel pharmaceuticals, as well as the improvement of efficacy or tolerability of an already applied drug. Furthermore, by the laccase-mediated coupling of two different active substances a synergistic effect may be possible. However, the coupling of compounds that have no described efficacy can lead to biologically active substances by means of laccase. The review summarizes laccase-mediated reactions for the synthesis of antimicrobial compounds valuable for medical purposes. In particular, reactions with two different reaction partners were shown in detail. In addition, studies with in vitro and in vivo experimental data for the confirmation of the antibacterial and/or antifungal efficacy of the products, synthesized with laccase, were of special interest. Analyses of the structure-activity relationship confirm the great potential of the novel compounds. These substances may represent not only a value for pharmaceutical and chemical industry, but also for other industries due to a possible functionalization of surfaces such as wood or textiles.
Collapse
Affiliation(s)
- Veronika Hahn
- Leibniz Institute for Plasma Science and Technology (INP), Felix-Hausdorff-Str. 2, 17489, Greifswald, Germany.
- Institute for Microbiology, University of Greifswald, Felix-Hausdorff-Str. 8, 17489, Greifswald, Germany.
| |
Collapse
|
11
|
clbP Gene, a Potential New Member of the β-Lactamase Family. Int J Mol Sci 2022; 23:ijms232415642. [PMID: 36555283 PMCID: PMC9778894 DOI: 10.3390/ijms232415642] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2022] [Revised: 12/05/2022] [Accepted: 12/07/2022] [Indexed: 12/14/2022] Open
Abstract
The colibactin island (pks) of Escherichia coli formed by 19 genes (55-Kb), encodes non-ribosomal peptide (NRP) and polyketide (PK) synthases, which allow the synthesis of colibactin, a suspected hybrid PK-NRP compound that causes damage to DNA in eukaryotic cells. The clbP, an unusual essential gene, is found in the operon structure with the clbS gene in the pks-encoded machinery. Interestingly, the clbP gene has been annotated as a β-lactamase but no previous study has reported its β-lactamase characteristics. In this study, we (i) investigated the β-lactamase properties of the clbP gene in silico by analysing its phylogenetic relationship with bacterial β-lactamase and peptidase enzymes, (ii) compared its three-dimensional (3D) protein structure with those of bacterial β-lactamase proteins using the Phyr2 database and PyMOL software, and (iii) evaluated in vitro its putative enzymatic activities, including β-lactamase, nuclease, and ribonuclease using protein expression and purification from an E. coli BL21 strain. In this study, we reveal a structural configuration of toxin/antitoxin systems in this island. Thus, similar to the toxin/antitoxin systems, the role of the clbP gene within the pks-island gene group appears as an antitoxin, insofar as it is responsible for the activation of the toxin, which is colibactin. In silico, our analyses revealed that ClbP belonged to the superfamily of β-lactamase, class C. Furthermore, in vitro we were unable to demonstrate its β-lactamase activity, likely due to the fact that the clbP gene requires co-expression with other genes, such as the genes present in the pks-island (19 genes). More research is needed to better understand its actions, particularly with regards to antibiotics, and to discover whether it has any additional functions due to the importance of this gene and its toxicity.
Collapse
|
12
|
Flanders PL, Contreras-Martel C, Brown NW, Shirley JD, Martins A, Nauta KN, Dessen A, Carlson EE, Ambrose EA. Combined Structural Analysis and Molecular Dynamics Reveal Penicillin-Binding Protein Inhibition Mode with β-Lactones. ACS Chem Biol 2022; 17:3110-3120. [PMID: 36173746 PMCID: PMC10057605 DOI: 10.1021/acschembio.2c00503] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
β-Lactam antibiotics comprise one of the most widely used therapeutic classes to combat bacterial infections. This general scaffold has long been known to inhibit bacterial cell wall biosynthesis by inactivating penicillin-binding proteins (PBPs); however, bacterial resistance to β-lactams is now widespread, and new strategies are urgently needed to target PBPs and other proteins involved in bacterial cell wall formation. A key requirement in the identification of strategies to overcome resistance is a deeper understanding of the roles of the PBPs and their associated proteins during cell growth and division, such as can be obtained with the use of selective chemical probes. Probe development has typically depended upon known PBP inhibitors, which have historically been thought to require a negatively charged moiety that mimics the C-terminus of the PBP natural peptidoglycan substrate, d-Ala-d-Ala. However, we have identified a new class of β-lactone-containing molecules that interact with PBPs, often in an isoform-specific manner, and do not incorporate this C-terminal mimetic. Here, we report a series of structural biology experiments and molecular dynamics simulations that we utilized to evaluate specific binding modes of this novel PBP inhibitor class. In this work, we obtained <2 Å resolution X-ray structures of four β-lactone probes bound to PBP1b from Streptococcus pneumoniae. Despite their diverging recognition modes beyond the site of covalent modification, these four probes all efficiently labeled PBP1b, as well as other PBPs from S. pneumoniae. From these structures, we analyzed protein-ligand interactions and characterized the β-lactone-bound active sites using in silico mutagenesis and molecular dynamics. Our approach has clarified the dynamic interaction profile in this series of ligands, expanding the understanding of PBP inhibitor binding.
Collapse
Affiliation(s)
- Parker L Flanders
- Department of Medicinal Chemistry, University of Minnesota, 208 Harvard Street SE, Minneapolis, Minnesota 55454, United States
| | - Carlos Contreras-Martel
- Université Grenoble Alpes, CNRS, CEA, Institut de Biologie Structurale (IBS), F-38044 Grenoble, France
| | - Nathaniel W Brown
- Department of Chemistry, University of Minnesota, 207 Pleasant Street SE, Minneapolis, Minnesota 55455, United States
| | - Joshua D Shirley
- Department of Medicinal Chemistry, University of Minnesota, 208 Harvard Street SE, Minneapolis, Minnesota 55454, United States
| | - Alexandre Martins
- Université Grenoble Alpes, CNRS, CEA, Institut de Biologie Structurale (IBS), F-38044 Grenoble, France
| | - Kelsie N Nauta
- Department of Chemistry, University of Minnesota, 207 Pleasant Street SE, Minneapolis, Minnesota 55455, United States
| | - Andréa Dessen
- Université Grenoble Alpes, CNRS, CEA, Institut de Biologie Structurale (IBS), F-38044 Grenoble, France.,Brazilian Biosciences National Laboratory (LNBio), CNPEM, Campinas 13084-971, São Paulo, Brazil
| | - Erin E Carlson
- Department of Medicinal Chemistry, University of Minnesota, 208 Harvard Street SE, Minneapolis, Minnesota 55454, United States.,Department of Chemistry, University of Minnesota, 207 Pleasant Street SE, Minneapolis, Minnesota 55455, United States.,Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota, 321 Church Street SE, Minneapolis, Minnesota 55454, United States.,Department of Pharmacology, University of Minnesota, 321 Church Street SE, Minneapolis, Minnesota 55454, United States
| | - Elizabeth A Ambrose
- Department of Medicinal Chemistry, University of Minnesota, 208 Harvard Street SE, Minneapolis, Minnesota 55454, United States.,Minnesota Supercomputing Institute for Advanced Computational Research, University of Minnesota, Minneapolis, Minnesota 55455, United States
| |
Collapse
|
13
|
The Mechanism of Bacterial Resistance and Potential Bacteriostatic Strategies. Antibiotics (Basel) 2022; 11:antibiotics11091215. [PMID: 36139994 PMCID: PMC9495013 DOI: 10.3390/antibiotics11091215] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2022] [Revised: 09/04/2022] [Accepted: 09/05/2022] [Indexed: 12/26/2022] Open
Abstract
Bacterial drug resistance is rapidly developing as one of the greatest threats to human health. Bacteria will adopt corresponding strategies to crack the inhibitory effect of antibiotics according to the antibacterial mechanism of antibiotics, involving the mutation of drug target, secreting hydrolase, and discharging antibiotics out of cells through an efflux pump, etc. In recent years, bacteria are found to constantly evolve new resistance mechanisms to antibiotics, including target protective protein, changes in cell morphology, and so on, endowing them with multiple defense systems against antibiotics, leading to the emergence of multi-drug resistant (MDR) bacteria and the unavailability of drugs in clinics. Correspondingly, researchers attempt to uncover the mystery of bacterial resistance to develop more convenient and effective antibacterial strategies. Although traditional antibiotics still play a significant role in the treatment of diseases caused by sensitive pathogenic bacteria, they gradually lose efficacy in the MDR bacteria. Therefore, highly effective antibacterial compounds, such as phage therapy and CRISPER-Cas precision therapy, are gaining an increasing amount of attention, and are considered to be the treatments with the moist potential with regard to resistance against MDR in the future. In this review, nine identified drug resistance mechanisms are summarized, which enhance the retention rate of bacteria under the action of antibiotics and promote the distribution of drug-resistant bacteria (DRB) in the population. Afterwards, three kinds of potential antibacterial methods are introduced, in which new antibacterial compounds exhibit broad application prospects with different action mechanisms, the phage therapy has been successfully applied to infectious diseases caused by super bacteria, and the CRISPER-Cas precision therapy as a new technology can edit drug-resistant genes in pathogenic bacteria at the gene level, with high accuracy and flexibility. These antibacterial methods will provide more options for clinical treatment, and will greatly alleviate the current drug-resistant crisis.
Collapse
|
14
|
Shirley JD, Nauta KM, Carlson EE. Live-Cell Profiling of Penicillin-Binding Protein Inhibitors in Escherichia coli MG1655. ACS Infect Dis 2022; 8:1241-1252. [PMID: 35763562 PMCID: PMC10040144 DOI: 10.1021/acsinfecdis.2c00004] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Penicillin-binding proteins (PBPs) make up an essential class of bacterial enzymes that carry out the final steps of peptidoglycan synthesis and regulate the recycling of this polymeric structure. PBPs are an excellent drug target and have been the most clinically relevant antibacterial target since the 1940s with the introduction of β-lactams. Despite this, a large gap in knowledge remains regarding the individual function and regulation of each PBP homologue in most bacteria. This can be attributed to a lack of chemical tools and methods that enable the study of individual PBPs in an activity-dependent manner and in their native environment. The development of such methods in Gram-negative bacteria has been particularly challenging due to the presence of an outer membrane and numerous resistance mechanisms. To address this, we have developed an optimized live-cell assay for screening inhibitors of the PBPs in Escherichia coli MG1655. We utilized EDTA to permeabilize Gram-negative cells, enabling increased penetration of our readout probe, Bocillin-FL, and subsequent analysis of PBP-inhibition profiles. To identify scaffolds for future development of PBP-selective activity-based probes, we screened ten β-lactams, one diazabicyclooctane, and one monobactam for their PBP-selectivity profiles in E. coli MG1655. These results demonstrate the utility of our assay for the screening of inhibitors in live, non-hypersusceptible Gram-negative organisms.
Collapse
Affiliation(s)
- Joshua D Shirley
- Department of Medicinal Chemistry, University of Minnesota, 208 Harvard Street SE, Minneapolis, Minnesota 55454, United States
| | - Kelsie M Nauta
- Department of Chemistry, University of Minnesota, 207 Pleasant Street SE, Minneapolis, Minnesota 55455, United States
| | - Erin E Carlson
- Department of Medicinal Chemistry, University of Minnesota, 208 Harvard Street SE, Minneapolis, Minnesota 55454, United States.,Department of Chemistry, University of Minnesota, 207 Pleasant Street SE, Minneapolis, Minnesota 55455, United States.,Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota, 321 Church Street SE, Minneapolis, Minnesota 55454, United States.,Department of Pharmacology, University of Minnesota, 321 Church Street SE, Minneapolis, Minnesota 55454, United States
| |
Collapse
|
15
|
Bhat BA, Mir WR, Sheikh BA, Alkanani M, Mir MA. Metabolite fingerprinting of phytoconstituents from Fritillaria cirrhosa D. Don and molecular docking analysis of bioactive peonidin with microbial drug target proteins. Sci Rep 2022; 12:7296. [PMID: 35508512 PMCID: PMC9068770 DOI: 10.1038/s41598-022-10796-7] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Accepted: 04/07/2022] [Indexed: 12/15/2022] Open
Abstract
Fritillaria cirrhosa D. Don (Liliaceae), a valuable and critically endangered medicinal herb of northwest India, including Jammu and Kashmir, grows in temperate to alpine regions of the Himalaya. It is known as the traditional herb for cardiovascular diseases, respiratory diseases, and metabolic disorders. The plant bulbs are precious and are used to cure many other health complications. The current study analysed the phytoconstituents by liquid chromatography-mass spectrometry (LC-MS) of different crude extracts (methanolic, petroleum ether, and ethyl acetate) of F. cirrhosa. The LC-MS analysis from the bulbs of F. cirrhosa yielded 88 bioactive compounds, with the vast majority having therapeutic applications. Further, determination of minimum inhibitory concentrations (MICs) by broth microdilution method of F. cirrhosa against tested bacterial and fungal pathogens showed remarkable results with MICs ranging between 6.25-200 µg/mL and 50-400 µg/mL, respectively. Subsequently, these 88 identified phytocompounds were tested for their bioactivity through ADMET prediction by SwissADME and in silico molecular docking studies. Results revealed that Peonidin might have maximum antibacterial and antifungal activity against various microbial protein drug targets among the phytochemical compounds identified. Furthermore, the highest binding affinity complex was subjected to molecular dynamic simulation (MDS) analysis using Desmond Schrodinger v3.8. The root-mean-square deviation (RMSD) graphs obtained through the molecular dynamic simulations indicated the true bonding interactions, further validated using the root-mean-square fluctuation (RMSF) graphs which provided a better understanding of the amino acids present in the proteins responsible for the molecular motions and fluctuations. To our best knowledge, this is the first description of the phytochemical constituents of the bulbs of F.cirrhosa analyzed through LC-MS, which show pharmacological significance. The in silico molecular docking and molecular dynamics study of peonidin was also performed to confirm its broad-spectrum activities based on the binding interactions with the antibacterial and antifungal target proteins. The present study results will create a way for the invention of herbal medicines for several ailments by using F. cirrhosa plants, which may lead to the development of novel drugs.
Collapse
Affiliation(s)
- Basharat Ahmad Bhat
- Department of Bioresources, School of Biological Sciences, University of Kashmir, Srinagar, 190006, India
| | - Wajahat Rashid Mir
- Department of Bioresources, School of Biological Sciences, University of Kashmir, Srinagar, 190006, India
| | - Bashir Ahmad Sheikh
- Department of Bioresources, School of Biological Sciences, University of Kashmir, Srinagar, 190006, India
| | - Mustafa Alkanani
- College of Applied Medical Sciences, Almaarefa University, Riyadh, 11597, Kingdom of Saudi Arabia
| | - Manzoor Ahmad Mir
- Department of Bioresources, School of Biological Sciences, University of Kashmir, Srinagar, 190006, India.
| |
Collapse
|
16
|
Mir WR, Bhat BA, Almilaibary A, Asdaq SMB, Mir MA. Evaluation of the In Vitro Antimicrobial Activities of Delphinium roylei: An Insight from Molecular Docking and MD-Simulation Studies. Med Chem 2022; 18:1109-1121. [PMID: 35507782 DOI: 10.2174/1573406418666220429093956] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Revised: 03/27/2022] [Accepted: 03/29/2022] [Indexed: 11/22/2022]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE The burden of antimicrobial resistance demands a continued search for new antimicrobial drugs. The synthetic drugs which are used clinically have serious side effects. Natural products or compounds derived from natural sources show diversity in structure and play an essential role in drug discovery and development. OBJECTIVE Delphinium roylei is an important medicinal herb of Kashmir Himalaya, India. Traditionally this medicinal plant treats liver infections, skin problems, and chronic lower back pain. The current study evaluates the antimicrobial potential by various in -vitro and in -silico parameters. METHODS Three extracts and 168 bioactive compounds analysed through LC-MS data, with the vast majority of them having therapeutic applications of D. roylei, have been screened for the antimicrobial activity against bacteria (E. coli, M. luteus, K. pneumoniae, Streptococcus pneumonia, Haemophilus influenzae, Neisseria mucosa) and fungi (Candida albicans, Candida glabrata, Candida Paropsilosis) species through molecular docking using autodock Vina, MD simulation and broth microdilution method for minimum inhibitory concentration (MIC) evaluation. RESULTS The extracts, as well as the compounds analyzed through the LC-MS technique of Delphinium roylie showed significant antimicrobial activity. CONCLUSION Our study established that the leaf extracts of Delphinium roylei exhibit antimicrobial activity and thus confirm its importance in traditional medicine.
Collapse
Affiliation(s)
- Wajahat Rashid Mir
- Department of Bioresources, School of Biological Sciences, University of Kashmir, Srinagar-190006, India
| | - Basharat Ahmad Bhat
- Department of Bioresources, School of Biological Sciences, University of Kashmir, Srinagar-190006, India
| | - Abdullah Almilaibary
- Department of Family and Community Medicine, Albaha University, Albaha-65511, KSA
| | | | - Manzoor Ahmad Mir
- Department of Bioresources, School of Biological Sciences, University of Kashmir, Srinagar-190006, India
| |
Collapse
|
17
|
Kumar S, Mollo A, Kahne D, Ruiz N. The Bacterial Cell Wall: From Lipid II Flipping to Polymerization. Chem Rev 2022; 122:8884-8910. [PMID: 35274942 PMCID: PMC9098691 DOI: 10.1021/acs.chemrev.1c00773] [Citation(s) in RCA: 45] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
The peptidoglycan (PG) cell wall is an extra-cytoplasmic glycopeptide polymeric structure that protects bacteria from osmotic lysis and determines cellular shape. Since the cell wall surrounds the cytoplasmic membrane, bacteria must add new material to the PG matrix during cell elongation and division. The lipid-linked precursor for PG biogenesis, Lipid II, is synthesized in the inner leaflet of the cytoplasmic membrane and is subsequently translocated across the bilayer so that the PG building block can be polymerized and cross-linked by complex multiprotein machines. This review focuses on major discoveries that have significantly changed our understanding of PG biogenesis in the past decade. In particular, we highlight progress made toward understanding the translocation of Lipid II across the cytoplasmic membrane by the MurJ flippase, as well as the recent discovery of a novel class of PG polymerases, the SEDS (shape, elongation, division, and sporulation) glycosyltransferases RodA and FtsW. Since PG biogenesis is an effective target of antibiotics, these recent developments may lead to the discovery of much-needed new classes of antibiotics to fight bacterial resistance.
Collapse
Affiliation(s)
- Sujeet Kumar
- Department of Microbiology, The Ohio State University, Columbus, Ohio 43210, United States
| | - Aurelio Mollo
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, Massachusetts 02138, United States
| | - Daniel Kahne
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, Massachusetts 02138, United States.,Department of Molecular and Cellular Biology, Harvard University, Cambridge, Massachusetts 02138, United States.,Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, Massachusetts 02115, United States
| | - Natividad Ruiz
- Department of Microbiology, The Ohio State University, Columbus, Ohio 43210, United States
| |
Collapse
|
18
|
Looks can be deceiving: Bacterial enzymes work through unanticipated mechanism. Proc Natl Acad Sci U S A 2021; 118:2114568118. [PMID: 34526405 DOI: 10.1073/pnas.2114568118] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/23/2021] [Indexed: 11/18/2022] Open
|
19
|
Li Y, Qiao D, Zhang Y, Hao W, Xi Y, Deng X, Ge X, Xu M. MapZ deficiency leads to defects in the envelope structure and changes stress tolerance of Streptococcus mutans. Mol Oral Microbiol 2021; 36:295-307. [PMID: 34463029 DOI: 10.1111/omi.12352] [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: 06/18/2021] [Revised: 07/20/2021] [Accepted: 07/28/2021] [Indexed: 11/26/2022]
Abstract
Cell division is a central process in bacteria and a prerequisite for pathogenicity. Several proteins are involved in this process to ensure the accurate localization and proper function of the division machinery. In Streptococcus mutans, MapZ marks the division sites and position of the Z-ring to regulate cell division; however, whether MapZ deficiency can impair the cariogenic virulence of S. mutans remains unclear. Here, using a phenotypic assay and RNA-seq, we investigated the role of MapZ in cell envelope maintenance, biofilm formation, and stress tolerance in S. mutans. The results show that MapZ is important for normal cell shape and envelope structure, and its deletion causes abnormal septum structure and a thin cell wall. Subsequently, we found that the absence of MapZ leads to a greater level of cell death within 12 h biofilms, but it does not seem to affect biofilm architecture and accumulation. mapZ deletion also results in a decreased acid and osmotic stress tolerance. Furthermore, RNA-seq data reveal that MapZ deficiency causes changes in the expression levels of genes involved in transport systems, sugar metabolism, nature competence, and bacteriocin synthesis. Interestingly, we found that mapZ mutation renders S. mutans more sensitive to chlorhexidine. Taken together, our study suggests that MapZ plays a role in maintaining cell envelope structure and stress tolerance in S. mutans, showing a potential application as a drug target for caries prevention.
Collapse
Affiliation(s)
- Yongliang Li
- Department of Geriatric Dentistry, Peking University Hospital of Stomatology, Beijing, P. R. China.,National Engineering Laboratory for Digital and Material Technology of Stomatology, NMPA Key Laboratory for Dental Materials, Beijing Laboratory of Biomedical Materials, Peking University Hospital of Stomatology, Beijing, P. R. China
| | - Dan Qiao
- Shanxi Medical University School and Hospital of Stomatology, Taiyuan, Shanxi, P. R. China
| | - Yifei Zhang
- National Engineering Laboratory for Digital and Material Technology of Stomatology, NMPA Key Laboratory for Dental Materials, Beijing Laboratory of Biomedical Materials, Peking University Hospital of Stomatology, Beijing, P. R. China.,Central Laboratory, Peking University School and Hospital of Stomatology, Beijing, P. R. China
| | - Weifeng Hao
- National Engineering Laboratory for Digital and Material Technology of Stomatology, NMPA Key Laboratory for Dental Materials, Beijing Laboratory of Biomedical Materials, Peking University Hospital of Stomatology, Beijing, P. R. China
| | - Yue Xi
- National Engineering Laboratory for Digital and Material Technology of Stomatology, NMPA Key Laboratory for Dental Materials, Beijing Laboratory of Biomedical Materials, Peking University Hospital of Stomatology, Beijing, P. R. China
| | - Xuliang Deng
- Department of Geriatric Dentistry, Peking University Hospital of Stomatology, Beijing, P. R. China.,National Engineering Laboratory for Digital and Material Technology of Stomatology, NMPA Key Laboratory for Dental Materials, Beijing Laboratory of Biomedical Materials, Peking University Hospital of Stomatology, Beijing, P. R. China
| | - Xuejun Ge
- Shanxi Medical University School and Hospital of Stomatology, Taiyuan, Shanxi, P. R. China
| | - Mingming Xu
- Department of Geriatric Dentistry, Peking University Hospital of Stomatology, Beijing, P. R. China.,National Engineering Laboratory for Digital and Material Technology of Stomatology, NMPA Key Laboratory for Dental Materials, Beijing Laboratory of Biomedical Materials, Peking University Hospital of Stomatology, Beijing, P. R. China
| |
Collapse
|
20
|
Bacterial Resistance to Antimicrobial Agents. Antibiotics (Basel) 2021; 10:antibiotics10050593. [PMID: 34067579 PMCID: PMC8157006 DOI: 10.3390/antibiotics10050593] [Citation(s) in RCA: 96] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Revised: 05/12/2021] [Accepted: 05/14/2021] [Indexed: 12/26/2022] Open
Abstract
Bacterial pathogens as causative agents of infection constitute an alarming concern in the public health sector. In particular, bacteria with resistance to multiple antimicrobial agents can confound chemotherapeutic efficacy towards infectious diseases. Multidrug-resistant bacteria harbor various molecular and cellular mechanisms for antimicrobial resistance. These antimicrobial resistance mechanisms include active antimicrobial efflux, reduced drug entry into cells of pathogens, enzymatic metabolism of antimicrobial agents to inactive products, biofilm formation, altered drug targets, and protection of antimicrobial targets. These microbial systems represent suitable focuses for investigation to establish the means for their circumvention and to reestablish therapeutic effectiveness. This review briefly summarizes the various antimicrobial resistance mechanisms that are harbored within infectious bacteria.
Collapse
|
21
|
Lv N, Kong Q, Zhang H, Li J. Discovery of novel Staphylococcus aureus penicillin binding protein 2a inhibitors by multistep virtual screening and biological evaluation. Bioorg Med Chem Lett 2021; 41:128001. [PMID: 33811991 DOI: 10.1016/j.bmcl.2021.128001] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Revised: 03/21/2021] [Accepted: 03/23/2021] [Indexed: 01/27/2023]
Abstract
Penicillin-binding protein 2a (PBP2a) is an essential protein involved in the resistance to β-lactam antibiotics acquired by methicillin-resistant Staphylococcus aureus (MRSA) and is a potential antibacterial target. In the current study, we employed a strategy that combined virtual screening with biological evaluation to discover novel inhibitors of PBP2a. In this investigation, a hybrid virtual screening method, consisting of drug-likeness evaluation (Lipinski's Rule of Five and ADMET) and rigid (LibDock) and semi-flexible (CDOCKER) docking-based virtual screenings, was used for retrieving novel PBP2a inhibitors from commercially available chemical databases. 11 compounds were selected from the final hits and subsequently shifted to experimental studies. Among them, Hit 2, Hit 3, and Hit 10 exhibited excellent anti-MRSA ATCC 33591 activity and weak toxicity in vitro. The affinity of the three compounds to bind to PBP2a was further confirmed by surface plasmon resonance (SPR) experiments and molecular dynamics (MD) simulation. An inter-complex interaction study showed that all hit compounds adapted well to the allosteric site of the PBP2a protein. In addition, Hit 2 (with best binding affinity to PBP2a, KD = 1.29 × 10-7 M) significantly inhibits proliferation of MRSA clinical isolates. Together, the 3 hit compounds, especially Hit 2, may be potential non-β-lactam antibiotics against MRSA and the work will provide clues for the future development of specific compounds that block the interaction of PBP2a with their targets.
Collapse
Affiliation(s)
- Na Lv
- Department of Infectious Diseases, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230032, China; Department of Stomatology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230032, China
| | - Qinxiang Kong
- Department of Infectious Diseases, Chaohu Hospital of Anhui Medical University, Hefei, China
| | - Hui Zhang
- Department of Infectious Diseases, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230032, China
| | - Jiabin Li
- Department of Infectious Diseases, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230032, China; Institute of Bacterial Resistance, Anhui Medical University, Hefei, Anhui 230032, China.
| |
Collapse
|
22
|
Cui J, Zhang H, Mo Z, Yu M, Liang Z. Cell wall thickness and the molecular mechanism of heterogeneous vancomycin-intermediate Staphylococcus aureus. Lett Appl Microbiol 2021; 72:604-609. [PMID: 33539564 PMCID: PMC8248079 DOI: 10.1111/lam.13456] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2020] [Revised: 01/28/2021] [Accepted: 01/28/2021] [Indexed: 12/04/2022]
Abstract
Methicillin‐resistant Staphylococcus aureus (MRSA) with reduced sensitivity to vancomycin (VAN) has caused many clinical cases of VAN treatment failure, but the molecular mechanism underlying the reduced sensitivity to VAN is still unclear. We isolated a heterogeneous VAN‐intermediate Staphylococcus aureus (hVISA), which was also a MRSA strain with reduced sensitivity to VAN. To investigate the molecular mechanism underlying the reduced sensitivity to VAN exhibited by the hVISA strain, we compared the hVISA strain with a VAN‐sensitive MRSA strain, known as the N315 strain. The images captured by transmission electron microscopy showed that the cell wall of the hVISA strain was significantly thicker than that of the N315 strain (36·72 ± 1·04 nm vs 28·15 ± 1·25 nm, P < 0·05), and the results of real‐time quantitative PCR analysis suggested that the expression levels of the cell wall thickness related genes (glmS, vraR/S, sgtB, murZ and PBP4) of the hVISA strain were significantly higher than those of the N315 strain (P < 0·05). In conclusion, this study indicated that the upregulation of the expression of the genes related to cell wall synthesis might be the molecular mechanism underlying the cell wall thickening of the hVISA strain and might be related to its resistance to VAN.
Collapse
Affiliation(s)
- J Cui
- Department of Pulmonary and Critical Care Medicine, Chinese PLA General Hospital, Beijing, China
| | - H Zhang
- Department of Respiratory disease, Beijing Luhe Hospital Affiliated to Capital Medical University, Beijing, China
| | - Z Mo
- Department of Pulmonary and Critical Care Medicine, Chinese PLA General Hospital, Beijing, China
| | - M Yu
- Department of Pulmonary and Critical Care Medicine, Chinese PLA General Hospital, Beijing, China
| | - Z Liang
- Department of Pulmonary and Critical Care Medicine, Chinese PLA General Hospital, Beijing, China
| |
Collapse
|
23
|
Structural analysis of the sensor domain of the β-lactam antibiotic receptor VbrK from Vibrio parahaemolyticus. Biochem Biophys Res Commun 2020; 533:155-161. [DOI: 10.1016/j.bbrc.2020.09.011] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Accepted: 09/06/2020] [Indexed: 01/16/2023]
|