1
|
Butman HS, Stefaniak MA, Walsh DJ, Gondil VS, Young M, Crow AH, Nemeth AM, Melander RJ, Dunman PM, Melander C. Phenyl urea based adjuvants for β-lactam antibiotics against methicillin resistant Staphylococcus aureus. Bioorg Med Chem Lett 2025; 121:130164. [PMID: 40043820 DOI: 10.1016/j.bmcl.2025.130164] [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: 11/20/2024] [Revised: 02/04/2025] [Accepted: 03/01/2025] [Indexed: 03/10/2025]
Abstract
Penicillin binding protein 4 (PBP4) is essential for Staphylococcus aureus cortical bone osteocyte lacuno-canalicular network (OLCN) invasion, which causes osteomyelitis and serves as a bacterial niche for recurring bone infection. Moreover, PBP4 is also a key determinant of S. aureus resistance to fifth-generation cephalosporins (ceftobiprole and ceftaroline). From these perspectives, the development of S. aureus PBP4 inhibitors may represent dual functional therapeutics that prevent osteomyelitis, and reverse PBP4-mediated β-lactam resistance. A high-throughput screen for small molecules that inhibit S. aureus PBP4 function identified compound 1. We recently described a preliminary structure activity relationship (SAR) study on 1, identifying several compounds with increased PBP4 inhibitory activity, some of which also inhibit PBP2a. Herein, we expand our exploration of phenyl ureas as antibiotic adjuvants, investigating their activity with penicillins and additional cephalosporins against PBP2a-mediated methicillin-resistant S. aureus (MRSA). We screened the previously reported pilot library, and prepared an additional series of phenyl ureas based on compound 1. Lead compounds potentiate multiple β-lactam antibiotics, lowering minimum inhibitory concentrations (MICs) below susceptibility breakpoints, with up to 64-fold reductions in MIC.
Collapse
Affiliation(s)
- Hailey S Butman
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, IN, 46556, United States
| | - Monica A Stefaniak
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, IN, 46556, United States
| | - Danica J Walsh
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, IN, 46556, United States
| | - Vijay S Gondil
- Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, New York 14642, United States
| | - Mikaeel Young
- Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, New York 14642, United States
| | - Andrew H Crow
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, IN, 46556, United States
| | - Ansley M Nemeth
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, IN, 46556, United States
| | - Roberta J Melander
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, IN, 46556, United States
| | - Paul M Dunman
- Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, New York 14642, United States.
| | - Christian Melander
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, IN, 46556, United States.
| |
Collapse
|
2
|
Rajput P, Nahar KS, Rahman KM. Evaluation of Antibiotic Resistance Mechanisms in Gram-Positive Bacteria. Antibiotics (Basel) 2024; 13:1197. [PMID: 39766587 PMCID: PMC11672434 DOI: 10.3390/antibiotics13121197] [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: 10/24/2024] [Revised: 11/30/2024] [Accepted: 12/05/2024] [Indexed: 01/11/2025] Open
Abstract
The prevalence of resistance in Gram-positive bacterial infections is rapidly rising, presenting a pressing global challenge for both healthcare systems and economies. The WHO categorizes these bacteria into critical, high, and medium priority groups based on the urgency for developing new antibiotics. While the first priority pathogen list was issued in 2017, the 2024 list remains largely unchanged. Despite six years having passed, the progress that has been made in developing novel treatment approaches remains insufficient, allowing antimicrobial resistance to persist and worsen on a global scale. Various strategies have been implemented to address this growing threat by targeting specific resistance mechanisms. This review evaluates antimicrobial resistance (AMR) in Gram-positive bacteria, highlighting its critical impact on global health due to the rise of multidrug-resistant pathogens. It focuses on the unique cell wall structure of Gram-positive bacteria, which influences their identification and susceptibility to antibiotics. The review explores the mechanisms of AMR, including enzymatic inactivation, modification of drug targets, limiting drug uptake, and increased drug efflux. It also examines the resistance strategies employed by high-priority Gram-positive pathogens such as Staphylococcus aureus, Streptococcus pneumoniae, and Enterococcus faecium, as identified in the WHO's 2024 priority list.
Collapse
Affiliation(s)
- Pratiksing Rajput
- Institute of Pharmaceutical Science, King’s College London, 150 Stamford Street, London SE1 9NH, UK;
| | - Kazi S. Nahar
- Department of Natural Sciences, Faculty of Science & Technology, Middlesex University, The Burroughs, Hendon, London NW4 4BT, UK;
| | - Khondaker Miraz Rahman
- Institute of Pharmaceutical Science, King’s College London, 150 Stamford Street, London SE1 9NH, UK;
| |
Collapse
|
3
|
Ledger EVK, Massey RC. PBP4 is required for serum-induced cell wall thickening and antibiotic tolerance in Staphylococcus aureus. Antimicrob Agents Chemother 2024; 68:e0096124. [PMID: 39431816 PMCID: PMC11539222 DOI: 10.1128/aac.00961-24] [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: 06/27/2024] [Accepted: 09/21/2024] [Indexed: 10/22/2024] Open
Abstract
The bacterial pathogen Staphylococcus aureus responds to the host environment by synthesizing a thick peptidoglycan cell wall, which protects the bacterium from membrane-targeting antimicrobials and the immune response. However, the proteins required for this response were previously unknown. Here, we demonstrate by three independent approaches that the penicillin-binding protein PBP4 is crucial for serum-induced cell wall thickening. First, mutants lacking various non-essential cell wall synthesis enzymes were tested, revealing that a mutant lacking pbp4 was unable to generate a thick cell wall in serum. This resulted in reduced serum-induced tolerance of the pbp4 mutant toward the last resort antibiotic daptomycin relative to wild-type cells. Second, we found that serum-induced cell wall thickening occurred in each of a panel of 134 clinical bacteremia isolates, except for one strain with a naturally occurring mutation that results in an S140R substitution in the active site of PBP4. Finally, inhibition of PBP4 with cefoxitin prevented serum-induced cell wall thickening and the resulting antibiotic tolerance in the USA300 strain and clinical MRSA isolates. Together, this provides a rationale for combining daptomycin with cefoxitin, a PBP4 inhibitor, to potentially improve treatment outcomes for patients with invasive MRSA infections.
Collapse
Affiliation(s)
- Elizabeth V. K. Ledger
- School of Microbiology, University College Cork, Cork, Ireland
- APC Microbiome Ireland, University College Cork, Cork, Ireland
- Centre for Bacterial Resistance Biology, Imperial College London, London, United Kingdom
| | - Ruth C. Massey
- School of Microbiology, University College Cork, Cork, Ireland
- APC Microbiome Ireland, University College Cork, Cork, Ireland
- School of Cellular and Molecular Medicine, University of Bristol, Bristol, United Kingdom
| |
Collapse
|
4
|
Brdová D, Ruml T, Viktorová J. Mechanism of staphylococcal resistance to clinically relevant antibiotics. Drug Resist Updat 2024; 77:101147. [PMID: 39236354 DOI: 10.1016/j.drup.2024.101147] [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: 04/16/2024] [Revised: 08/17/2024] [Accepted: 08/26/2024] [Indexed: 09/07/2024]
Abstract
Staphylococcus aureus, a notorious pathogen with versatile virulence, poses a significant challenge to current antibiotic treatments due to its ability to develop resistance mechanisms against a variety of clinically relevant antibiotics. In this comprehensive review, we carefully dissect the resistance mechanisms employed by S. aureus against various antibiotics commonly used in clinical settings. The article navigates through intricate molecular pathways, elucidating the mechanisms by which S. aureus evades the therapeutic efficacy of antibiotics, such as β-lactams, vancomycin, daptomycin, linezolid, etc. Each antibiotic is scrutinised for its mechanism of action, impact on bacterial physiology, and the corresponding resistance strategies adopted by S. aureus. By synthesising the knowledge surrounding these resistance mechanisms, this review aims to serve as a comprehensive resource that provides a foundation for the development of innovative therapeutic strategies and alternative treatments for S. aureus infections. Understanding the evolving landscape of antibiotic resistance is imperative for devising effective countermeasures in the battle against this formidable pathogen.
Collapse
Affiliation(s)
- Daniela Brdová
- Department of Biochemistry and Microbiology, University of Chemistry and Technology, Technicka 3, Prague 16628, Czech Republic.
| | - Tomáš Ruml
- Department of Biochemistry and Microbiology, University of Chemistry and Technology, Technicka 3, Prague 16628, Czech Republic.
| | - Jitka Viktorová
- Department of Biochemistry and Microbiology, University of Chemistry and Technology, Technicka 3, Prague 16628, Czech Republic.
| |
Collapse
|
5
|
Sacco MD, Hammond LR, Noor RE, Bhattacharya D, McKnight LJ, Madsen JJ, Zhang X, Butler SG, Kemp MT, Jaskolka-Brown AC, Khan SJ, Gelis I, Eswara P, Chen Y. Staphylococcus aureus FtsZ and PBP4 bind to the conformationally dynamic N-terminal domain of GpsB. eLife 2024; 13:e85579. [PMID: 38639993 PMCID: PMC11062636 DOI: 10.7554/elife.85579] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Accepted: 04/15/2024] [Indexed: 04/20/2024] Open
Abstract
In the Firmicutes phylum, GpsB is a membrane associated protein that coordinates peptidoglycan synthesis with cell growth and division. Although GpsB has been studied in several bacteria, the structure, function, and interactome of Staphylococcus aureus GpsB is largely uncharacterized. To address this knowledge gap, we solved the crystal structure of the N-terminal domain of S. aureus GpsB, which adopts an atypical, asymmetric dimer, and demonstrates major conformational flexibility that can be mapped to a hinge region formed by a three-residue insertion exclusive to Staphylococci. When this three-residue insertion is excised, its thermal stability increases, and the mutant no longer produces a previously reported lethal phenotype when overexpressed in Bacillus subtilis. In S. aureus, we show that these hinge mutants are less functional and speculate that the conformational flexibility imparted by the hinge region may serve as a dynamic switch to fine-tune the function of the GpsB complex and/or to promote interaction with its various partners. Furthermore, we provide the first biochemical, biophysical, and crystallographic evidence that the N-terminal domain of GpsB binds not only PBP4, but also FtsZ, through a conserved recognition motif located on their C-termini, thus coupling peptidoglycan synthesis to cell division. Taken together, the unique structure of S. aureus GpsB and its direct interaction with FtsZ/PBP4 provide deeper insight into the central role of GpsB in S. aureus cell division.
Collapse
Affiliation(s)
- Michael D Sacco
- Department of Molecular Medicine, Morsani College of Medicine, University of South FloridaTampaUnited States
| | - Lauren R Hammond
- Department of Molecular Biosciences, University of South FloridaTampaUnited States
| | - Radwan E Noor
- Department of Chemistry, University of South FloridaTampaUnited States
| | | | - Lily J McKnight
- Department of Molecular Biosciences, University of South FloridaTampaUnited States
| | - Jesper J Madsen
- Department of Molecular Medicine, Morsani College of Medicine, University of South FloridaTampaUnited States
- Global and Planetary Health, College of Public Health, University of South FloridaTampaUnited States
| | - Xiujun Zhang
- Department of Molecular Medicine, Morsani College of Medicine, University of South FloridaTampaUnited States
| | - Shane G Butler
- Department of Molecular Medicine, Morsani College of Medicine, University of South FloridaTampaUnited States
| | - M Trent Kemp
- Department of Molecular Medicine, Morsani College of Medicine, University of South FloridaTampaUnited States
| | - Aiden C Jaskolka-Brown
- Department of Molecular Medicine, Morsani College of Medicine, University of South FloridaTampaUnited States
| | - Sebastian J Khan
- Department of Molecular Biosciences, University of South FloridaTampaUnited States
| | - Ioannis Gelis
- Department of Chemistry, University of South FloridaTampaUnited States
| | - Prahathees Eswara
- Department of Molecular Biosciences, University of South FloridaTampaUnited States
| | - Yu Chen
- Department of Molecular Medicine, Morsani College of Medicine, University of South FloridaTampaUnited States
| |
Collapse
|
6
|
Beytür S, Essiz S, Özuğur Uysal B. Investigation of Structural and Antibacterial Properties of WS 2-Doped ZnO Nanoparticles. ACS OMEGA 2024; 9:4037-4049. [PMID: 38284036 PMCID: PMC10809239 DOI: 10.1021/acsomega.3c09041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Revised: 11/28/2023] [Accepted: 12/21/2023] [Indexed: 01/30/2024]
Abstract
ZnO nanoparticles, well-known for their structural, optical, and antibacterial properties, are widely applied in diverse fields. The doping of different materials to ZnO, such as metals or metal oxides, is known to ameliorate its properties. Here, nanofilms composed of ZnO doped with WS2 at 5, 15, and 25% ratios are synthesized, and their properties are investigated. Supported by molecular docking analyses, the enhancement of the bactericidal properties after the addition of WS2 at different ratios is highlighted and supported by the inhibitory interaction of residues playing a crucial role in the bacterial survival through the targeting of proteins of interest.
Collapse
Affiliation(s)
- Sercan Beytür
- Faculty of Engineering and
Natural Sciences, Kadir Has University, Cibali, Fatih, Istanbul 34083, Turkey
| | - Sebnem Essiz
- Faculty of Engineering and
Natural Sciences, Kadir Has University, Cibali, Fatih, Istanbul 34083, Turkey
| | - Bengü Özuğur Uysal
- Faculty of Engineering and
Natural Sciences, Kadir Has University, Cibali, Fatih, Istanbul 34083, Turkey
| |
Collapse
|
7
|
Hu J, Han X, Ma X, Chen X, Zhou Z, Peng P, Yu Z, Hou Y, Han P, Pang L, Yang Y, Xu J, Wu W. Comparative proteomic analysis of vancomycin-sensitive and vancomycin-intermediate resistant Staphylococcus aureus. Eur J Clin Microbiol Infect Dis 2024; 43:139-153. [PMID: 37985551 DOI: 10.1007/s10096-023-04709-3] [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: 05/17/2023] [Accepted: 11/09/2023] [Indexed: 11/22/2023]
Abstract
PURPOSE The extensive use of vancomycin has led to the development of Staphylococcus aureus strains with varying degrees of resistance to vancomycin. The present study aimed to explore the molecular causes of vancomycin resistance by conducting a proteomics analysis of subcellular fractions isolated from vancomycin-intermediate resistant S. aureus (VISA) and vancomycin-sensitive S. aureus (VSSA) strains. METHODS We conducted proteomics analysis of subcellular fractions isolated from 2 isogenic S. aureus strains: strain 11 (VSSA) and strain 11Y (VISA). We used an integrated quantitative proteomics approach assisted by bioinformatics analysis, and comprehensively investigated the proteome profile. Intensive bioinformatics analysis, including protein annotation, functional classification, functional enrichment, and functional enrichment-based cluster analysis, was used to annotate quantifiable targets. RESULTS We identified 128 upregulated proteins and 21 downregulated proteins in strain 11Y as compared to strain 11. The largest group of differentially expressed proteins was composed of enzymatic proteins associated with metabolic and catalytic activity, which accounted for 32.1% and 50% of the total proteins, respectively. Some proteins were indispensable parts of the regulatory networks of S. aureus that were altered with vancomycin treatment, and these proteins were related to cell wall metabolism, cell adhesion, proteolysis, and pressure response. CONCLUSION Our proteomics study revealed regulatory proteins associated with vancomycin resistance in S. aureus. Some of these proteins were involved in the regulation of cell metabolism and function, which provides potential targets for the development of strategies to manage vancomycin resistance in S. aureus.
Collapse
Affiliation(s)
- Jian Hu
- Department of Laboratory Medicine, Yixing Hospital of Traditional Chinese Medicine, Yixing, No. 128 East Yangquan Road, Yicheng Subdistrict, Yixing, 214200, Jiangsu, People's Republic of China
| | - Xinjun Han
- Department of Medical Microbiology and Parasitology, College of Basic Medical Sciences, China Medical University, Shenyang, People's Republic of China
| | - Xiaoxue Ma
- Department of Medical Microbiology and Parasitology, College of Basic Medical Sciences, China Medical University, Shenyang, People's Republic of China
| | - Xutao Chen
- Department of Laboratory Medicine, Yixing Hospital of Traditional Chinese Medicine, Yixing, No. 128 East Yangquan Road, Yicheng Subdistrict, Yixing, 214200, Jiangsu, People's Republic of China
| | - Zhenping Zhou
- Department of Laboratory Medicine, Yixing Hospital of Traditional Chinese Medicine, Yixing, No. 128 East Yangquan Road, Yicheng Subdistrict, Yixing, 214200, Jiangsu, People's Republic of China
| | - Peilan Peng
- Department of Medical Microbiology and Parasitology, College of Basic Medical Sciences, China Medical University, Shenyang, People's Republic of China
| | - Zhao Yu
- Department of Medical Microbiology and Parasitology, College of Basic Medical Sciences, China Medical University, Shenyang, People's Republic of China
| | - Yongzhi Hou
- Department of Medical Microbiology and Parasitology, College of Basic Medical Sciences, China Medical University, Shenyang, People's Republic of China
| | - Peiru Han
- Department of Medical Microbiology and Parasitology, College of Basic Medical Sciences, China Medical University, Shenyang, People's Republic of China
| | - Long Pang
- Department of Medical Microbiology and Parasitology, College of Basic Medical Sciences, China Medical University, Shenyang, People's Republic of China
| | - Yali Yang
- Department of Medical Microbiology and Parasitology, College of Basic Medical Sciences, China Medical University, Shenyang, People's Republic of China
| | - Jia Xu
- Department of Medical Microbiology, Key Laboratory of Environmental Pollution and Microecology of Liaoning Province, Shenyang Medical College, Shenyang, 110034, People's Republic of China.
| | - Wenhui Wu
- Department of Laboratory Medicine, Yixing Hospital of Traditional Chinese Medicine, Yixing, No. 128 East Yangquan Road, Yicheng Subdistrict, Yixing, 214200, Jiangsu, People's Republic of China.
| |
Collapse
|
8
|
Lade H, Kim JS. Molecular Determinants of β-Lactam Resistance in Methicillin-Resistant Staphylococcus aureus (MRSA): An Updated Review. Antibiotics (Basel) 2023; 12:1362. [PMID: 37760659 PMCID: PMC10525618 DOI: 10.3390/antibiotics12091362] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Revised: 08/21/2023] [Accepted: 08/24/2023] [Indexed: 09/29/2023] Open
Abstract
The development of antibiotic resistance in Staphylococcus aureus, particularly in methicillin-resistant S. aureus (MRSA), has become a significant health concern worldwide. The acquired mecA gene encodes penicillin-binding protein 2a (PBP2a), which takes over the activities of endogenous PBPs and, due to its low affinity for β-lactam antibiotics, is the main determinant of MRSA. In addition to PBP2a, other genetic factors that regulate cell wall synthesis, cell signaling pathways, and metabolism are required to develop high-level β-lactam resistance in MRSA. Although several genetic factors that modulate β-lactam resistance have been identified, it remains unclear how they alter PBP2a expression and affect antibiotic resistance. This review describes the molecular determinants of β-lactam resistance in MRSA, with a focus on recent developments in our understanding of the role of mecA-encoded PBP2a and on other genetic factors that modulate the level of β-lactam resistance. Understanding the molecular determinants of β-lactam resistance can aid in developing novel strategies to combat MRSA.
Collapse
Affiliation(s)
| | - Jae-Seok Kim
- Department of Laboratory Medicine, Hallym University College of Medicine, Kangdong Sacred Heart Hospital, Seoul 05355, Republic of Korea;
| |
Collapse
|
9
|
Diessner EM, Takahashi GR, Martin RW, Butts CT. Comparative Modeling and Analysis of Extremophilic D-Ala-D-Ala Carboxypeptidases. Biomolecules 2023; 13:328. [PMID: 36830697 PMCID: PMC9953012 DOI: 10.3390/biom13020328] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2022] [Revised: 01/21/2023] [Accepted: 02/02/2023] [Indexed: 02/11/2023] Open
Abstract
Understanding the molecular adaptations of organisms to extreme environments requires a comparative analysis of protein structure, function, and dynamics across species found in different environmental conditions. Computational studies can be particularly useful in this pursuit, allowing exploratory studies of large numbers of proteins under different thermal and chemical conditions that would be infeasible to carry out experimentally. Here, we perform such a study of the MEROPS family S11, S12, and S13 proteases from psychophilic, mesophilic, and thermophilic bacteria. Using a combination of protein structure prediction, atomistic molecular dynamics, and trajectory analysis, we examine both conserved features and trends across thermal groups. Our findings suggest a number of hypotheses for experimental investigation.
Collapse
Affiliation(s)
| | - Gemma R. Takahashi
- Department of Molecular Biology and Biochemistry, University of California, Irvine, CA 92697, USA
| | - Rachel W. Martin
- Department of Chemistry, University of California, Irvine, CA 92697, USA
- Department of Molecular Biology and Biochemistry, University of California, Irvine, CA 92697, USA
| | - Carter T. Butts
- Departments of Sociology, Statistics, Electrical Engineering and Computer Science, University of California, Irvine, CA 92697, USA
| |
Collapse
|
10
|
Sharma S, Sharma BK, Jain S, Gulyani P. A Combined QSAR and Molecular Docking Approach for Identifying
Pyrimidine Derivatives as Penicillin Binding Protein Inhibitors. LETT DRUG DES DISCOV 2022. [DOI: 10.2174/1570180819666220427101322] [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:
Antimicrobial resistance has been rising continuously in the past few years due
to the overuse and exploitation of existing antimicrobials. This has motivated the search for a novel scaffold
that has the capability of rapid antimicrobial action. The hybridized pyrimidines have attracted us due
to their widespread biological activities, such as anti-bacterial and antifungal activities.
Objective:
The present study incorporates a series of pyrimidine-based antimicrobial agents for the 2D
quantitative structure-activity relationship analysis (2D QSAR) and docking analysis.
Methods:
The exploration of the chemical structures in combination with the biological activity in CPMLR led to the detection of six descriptors (Constitutional descriptors, Topological descriptors, Modified Burden Eigenvalues and 2D autocorrelations) for modeling the activity. The resulted QSAR model has been validated using combinatorial protocol in multiple linear regression (CP-MLR) and partial least squares (PLS) analysis.
Methods:
The exploration of the chemical structures in combination with the biological activity in
CPMLR led to the detection of six descriptors (Constitutional descriptors, Topological descriptors, Modified
Burden Eigenvalues and 2D autocorrelations) for modeling the activity. The resulted QSAR model
has been validated using a combinatorial protocol in multiple linear regression (CP-MLR) and partial
least squares (PLS) analysis.
Results:
The best QSAR model displays the r2
t
value of 0.594, Q2
LOO value of 0.779, Q2
L5O value of
0.767. Further docking study was executed using Autodock Vina against Penicillin-binding protein
(PBP2a).
Conclusion:
From the results, Compounds 4, 11and 24 were found to possess a good binding affinity
towards PBP2a.
Collapse
Affiliation(s)
- Smriti Sharma
- Amity Institute of Pharmacy, Amity University, Sector-125, Noida-201313, India
| | - Brij K. Sharma
- Department of Chemistry, Government
College, Bundi-323 001, Rajasthan, India
| | - Surabhi Jain
- Faculty of Pharmacy, B. Pharmacy College Rampura-kakanpur, (Gujarat
Technological University), Panchmahals, Gujarat, India
| | - Puja Gulyani
- Amity Institute of Pharmacy, Amity University, Sector-125, Noida-201313, India
| |
Collapse
|
11
|
Ibrayev MK, Nurkenov OA, Rakhimberlinova ZB, Takibayeva AT, Palamarchuk IV, Turdybekov DM, Kelmyalene AA, Kulakov IV. Synthesis, Structure and Molecular Docking of New 4,5-Dihydrothiazole Derivatives Based on 3,5-Dimethylpyrazole and Cytisine and Salsoline Alkaloids. Molecules 2022; 27:7598. [PMID: 36364423 PMCID: PMC9655236 DOI: 10.3390/molecules27217598] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2022] [Revised: 10/23/2022] [Accepted: 10/27/2022] [Indexed: 10/10/2023] Open
Abstract
The interaction results of 1,2-dibromo-3-isothiocyanatopropane with some pyrazoles as well as cytisine and salsoline alkaloids were presented in this paper. It was shown that the reaction resulted in one one-step and rather mild method for the preparation of the corresponding 1,3-thiazoline bromomethyl derivatives. The yield of this reaction was affected by the presence of a base and an order in which reagents were added. Molecular docking of the synthesized 1,3-thiazoline derivatives for putative antibacterial activity was carried out using the penicillin-binding target protein (PBP4) of the bacteria E. coli "Homo sapiens" and S. aureus "Homo sapiens" as an example. Molecular docking demonstrated that the compounds had insignificant binding energies at the level of selected reference drugs (Cephalotin and Chloramphenicol). The presence of natural alkaloids in the structure of thiazoline derivatives somewhat increased the affinity of these substrates for target proteins selected.
Collapse
Affiliation(s)
- Marat K. Ibrayev
- Department of Chemistry and Chemical Technology, Abylkas Saginov Karaganda Technical University, Ave. Nursultan Nazarbayev, 56, Karaganda 100027, Kazakhstan
- Faculty of Chemistry, Karaganda Buketov University, st. University 28, Karaganda 100024, Kazakhstan
| | - Oralgazy A. Nurkenov
- Department of Chemistry and Chemical Technology, Abylkas Saginov Karaganda Technical University, Ave. Nursultan Nazarbayev, 56, Karaganda 100027, Kazakhstan
- Institute of Organic Synthesis and Coal Chemistry of Republic of Kazakhstan, Alikhanova 1, Karaganda 100008, Kazakhstan
| | - Zhanar B. Rakhimberlinova
- Department of Chemistry and Chemical Technology, Abylkas Saginov Karaganda Technical University, Ave. Nursultan Nazarbayev, 56, Karaganda 100027, Kazakhstan
| | - Altynaray T. Takibayeva
- Department of Chemistry and Chemical Technology, Abylkas Saginov Karaganda Technical University, Ave. Nursultan Nazarbayev, 56, Karaganda 100027, Kazakhstan
| | - Irina V. Palamarchuk
- Institute of Chemistry, Tyumen State University, 15a Perekopskaya St., Tyumen 625003, Russia
| | - Dastan M. Turdybekov
- Department of Chemistry and Chemical Technology, Abylkas Saginov Karaganda Technical University, Ave. Nursultan Nazarbayev, 56, Karaganda 100027, Kazakhstan
| | - Assel A. Kelmyalene
- Department of Chemistry and Chemical Technology, Abylkas Saginov Karaganda Technical University, Ave. Nursultan Nazarbayev, 56, Karaganda 100027, Kazakhstan
| | - Ivan V. Kulakov
- Institute of Chemistry, Tyumen State University, 15a Perekopskaya St., Tyumen 625003, Russia
| |
Collapse
|
12
|
Divergent Effects of Peptidoglycan Carboxypeptidase DacA on Intrinsic β-Lactam and Vancomycin Resistance. Microbiol Spectr 2022; 10:e0173422. [PMID: 35758683 PMCID: PMC9430164 DOI: 10.1128/spectrum.01734-22] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
Vancomycin and β-lactams are clinically important antibiotics that inhibit the formation of peptidoglycan cross-links, but their binding targets are different. The binding target of vancomycin is d-alanine-d-alanine (d-Ala-d-Ala), whereas that of β-lactam is penicillin-binding proteins (PBPs). In this study, we revealed the divergent effects of peptidoglycan (PG) carboxypeptidase DacA on vancomycin and β-lactam resistance in Escherichia coli and Bacillus subtilis. The deletion of DacA induced sensitivity to most β-lactams, whereas it induced strong resistance toward vancomycin. Notably, both phenotypes did not have a strong association with ld-transpeptidases, which are necessary for the formation of PG 3-3 cross-links and covalent bonds between PG and an Lpp outer membrane (OM) lipoprotein. Vancomycin resistance was induced by an increased amount of decoy d-Ala-d-Ala residues within PG, whereas β-lactam sensitivity was associated with physical interactions between DacA and PBPs. The presence of an OM permeability barrier strongly strengthened vancomycin resistance, but it significantly weakened β-lactam sensitivity. Collectively, our results revealed two distinct functions of DacA, which involved inverse modulation of bacterial resistance to clinically important antibiotics, β-lactams and vancomycin, and presented evidence for a link between DacA and PBPs. IMPORTANCE Bacterial PG hydrolases play important roles in various aspects of bacterial physiology, including cytokinesis, PG synthesis, quality control of PG, PG recycling, and stress adaptation. Of all the PG hydrolases, the role of PG carboxypeptidases is poorly understood, especially regarding their impacts on antibiotic resistance. We have revealed two distinct functions of PG carboxypeptidase DacA with respect to antibiotic resistance. The deletion of DacA led to sensitivity to most β-lactams, while it caused strong resistance to vancomycin. Our study provides novel insights into the roles of PG carboxypeptidases in the regulation of antibiotic resistance and a potential clue for the development of a drug to improve the clinical efficacy of β-lactam antibiotics.
Collapse
|
13
|
Altayb HN, Yassin NF, Hosawi S, Kazmi I. In-vitro and in-silico antibacterial activity of Azadirachta indica (Neem), methanolic extract, and identification of Beta.d-Mannofuranoside as a promising antibacterial agent. BMC PLANT BIOLOGY 2022; 22:262. [PMID: 35610569 PMCID: PMC9131563 DOI: 10.1186/s12870-022-03650-5] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Accepted: 05/12/2022] [Indexed: 06/15/2023]
Abstract
BACKGROUND Antimicrobial resistance became the leading cause of death globally, resulting in an urgent need for the discovery of new, safe, and efficient antibacterial agents. Compounds derived from plants can provide an essential source of new types of antibiotics. A. indica (neem) plant is rich in antimicrobial phytoconstituents. Here, we used the sensitive and reliable gas chromatography-mass spectrometry (GC-MS) approach, for the quantitative and quantitative determination of bioactive constituents in methanolic extract of neem leaves grown in Sudan. Subsequently, antibacterial activity, pharmacokinetic and toxicological properties were utilized using in silico tools. RESULTS The methanolic extract of neem leaves was found to have antibacterial activity against all pathogenic and reference strains. The lowest concentration reported with bacterial activity was 3.125%, which showed zones of inhibition of more than 10 mm on P. aeruginosa, K. pneumoniae, Citrobacter spp., and E. coli, and 8 mm on Proteus spp., E. faecalis, S. epidermidis, and the pathogenic S. aureus. GC-MS analysis revealed the presence of 30 chemical compounds, including fatty acids (11), hydrocarbons (9), pyridine derivatives (2), aldehydes (2), phenol group (1), aromatic substances (1), coumarins (1), and monoterpenes (1). In silico and in vitro tools revealed that.beta.d-Mannofuranoside, O-geranyl was the most active compound on different bacterial proteins. It showed the best docking energy (-8 kcal/mol) and best stability with different bacterial essential proteins during molecular dynamic (MD) simulation. It also had a good minimum inhibitory concentration (MIC) (32 μg/ml and 64 μg/ml) against S. aureus (ATCC 25,923) and E. coli (ATCC 25,922) respectively. CONCLUSION The methanolic extract of A. indica leaves possessed strong antibacterial activity against different types of bacteria. Beta.d-Mannofuranoside, O-geranyl was the most active compound and it passed 5 rules of drug-likeness properties. It could therefore be further processed for animal testing and clinical trials for its possible use as an antibacterial agent with commercial values.
Collapse
Affiliation(s)
- Hisham N Altayb
- Biochemistry Department, Faculty of Science, King Abdulaziz University, Jeddah 21589, Saudi Arabia.
- Centre for Artificial Intelligence in Precision Medicine, King Abdulaziz University, Jeddah, 21589, Saudi Arabia.
| | - Nijood F Yassin
- Department Microbiology, College of Medical Laboratory Sciences, Sudan University of Science and Technology, Khartoum, Sudan
| | - Salman Hosawi
- Biochemistry Department, Faculty of Science, King Abdulaziz University, Jeddah 21589, Saudi Arabia
- Centre for Artificial Intelligence in Precision Medicine, King Abdulaziz University, Jeddah, 21589, Saudi Arabia
| | - Imran Kazmi
- Biochemistry Department, Faculty of Science, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| |
Collapse
|
14
|
Staphylococcus pseudintermedius's PBP4 Is Directly Associated with the Dissociated Oxacillin and Cefoxitin Phenotype. Antibiotics (Basel) 2021; 10:antibiotics10111299. [PMID: 34827237 PMCID: PMC8614965 DOI: 10.3390/antibiotics10111299] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Revised: 09/23/2021] [Accepted: 10/23/2021] [Indexed: 11/25/2022] Open
Abstract
Staphylococcus pseudintermedius is an important pathogen responsible for infections in dogs and in humans. The emergence and dissemination of methicillin-resistant S. pseudintermedius (MRSP) and the multidrug resistance frequently seen in this species make difficult the treatment of these pathogens. The cefoxitin disk is widely used as a marker of methicillin resistance mediated by the mecA gene in Staphylococcus aureus and other staphylococcal species; however, it is not useful to detect β-lactam resistance of MRSP in clinical microbiology laboratories. The purpose of this study was to elucidate the molecular bases of the dissociated phenotype between oxacillin and cefoxitin antibiotics. By using a combinatorial approach that included the Penicillin-Binding Proteins’ (PBP) profile, their affinity for different β-lactam antibiotics and the analyses of PBPs’ sequence, we provide evidence that PBP4 showed still affinity for its target cefoxitin, impairing its phenotypic resistant detection in MRSP. Together, these findings provide evidence that S. pseudintermedius PBP4 is directly associated with the dissociated oxacillin and cefoxitin phenotype.
Collapse
|
15
|
Martínez-Caballero S, Mahasenan KV, Kim C, Molina R, Feltzer R, Lee M, Bouley R, Hesek D, Fisher JF, Muñoz IG, Chang M, Mobashery S, Hermoso JA. Integrative structural biology of the penicillin-binding protein-1 from Staphylococcus aureus, an essential component of the divisome machinery. Comput Struct Biotechnol J 2021; 19:5392-5405. [PMID: 34667534 PMCID: PMC8493512 DOI: 10.1016/j.csbj.2021.09.018] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Revised: 09/13/2021] [Accepted: 09/15/2021] [Indexed: 12/18/2022] Open
Abstract
The penicillin-binding proteins are the enzyme catalysts of the critical transpeptidation crosslinking polymerization reaction of bacterial peptidoglycan synthesis and the molecular targets of the penicillin antibiotics. Here, we report a combined crystallographic, small-angle X-ray scattering (SAXS) in-solution structure, computational and biophysical analysis of PBP1 of Staphylococcus aureus (saPBP1), providing mechanistic clues about its function and regulation during cell division. The structure reveals the pedestal domain, the transpeptidase domain, and most of the linker connecting to the "penicillin-binding protein and serine/threonine kinase associated" (PASTA) domains, but not its two PASTA domains, despite their presence in the construct. To address this absence, the structure of the PASTA domains was determined at 1.5 Å resolution. Extensive molecular-dynamics simulations interpret the PASTA domains of saPBP1 as conformationally mobile and separated from the transpeptidase domain. This conclusion was confirmed by SAXS experiments on the full-length protein in solution. A series of crystallographic complexes with β-lactam antibiotics (as inhibitors) and penta-Gly (as a substrate mimetic) allowed the molecular characterization of both inhibition by antibiotics and binding for the donor and acceptor peptidoglycan strands. Mass-spectrometry experiments with synthetic peptidoglycan fragments revealed binding by PASTA domains in coordination with the remaining domains. The observed mobility of the PASTA domain in saPBP1 could play a crucial role for in vivo interaction with its glycosyltransferase partner in the membrane or with other components of the divisome machinery, as well as for coordination of transpeptidation and polymerization processes in the bacterial divisome.
Collapse
Affiliation(s)
- Siseth Martínez-Caballero
- Department of Crystallography and Structural Biology, Institute of Physical Chemistry "Rocasolano", CSIC, 28006 Madrid, Spain
| | - Kiran V Mahasenan
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, IN 46556, USA
| | - Choon Kim
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, IN 46556, USA
| | - Rafael Molina
- Department of Crystallography and Structural Biology, Institute of Physical Chemistry "Rocasolano", CSIC, 28006 Madrid, Spain
| | - Rhona Feltzer
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, IN 46556, USA
| | - Mijoon Lee
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, IN 46556, USA
| | - Renee Bouley
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, IN 46556, USA
| | - Dusan Hesek
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, IN 46556, USA
| | - Jed F Fisher
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, IN 46556, USA
| | - Inés G Muñoz
- Structural Biology Programme, Spanish National Cancer Research Center (CNIO), 28029 Madrid, Spain
| | - Mayland Chang
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, IN 46556, USA
| | - Shahriar Mobashery
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, IN 46556, USA
| | - Juan A Hermoso
- Department of Crystallography and Structural Biology, Institute of Physical Chemistry "Rocasolano", CSIC, 28006 Madrid, Spain
| |
Collapse
|
16
|
Masters EA, Muthukrishnan G, Ho L, Gill AL, de Mesy Bentley KL, Galloway CA, McGrath JL, Awad HA, Gill SR, Schwarz EM. Staphylococcus aureus Cell Wall Biosynthesis Modulates Bone Invasion and Osteomyelitis Pathogenesis. Front Microbiol 2021; 12:723498. [PMID: 34484165 PMCID: PMC8415456 DOI: 10.3389/fmicb.2021.723498] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2021] [Accepted: 07/15/2021] [Indexed: 11/13/2022] Open
Abstract
Staphylococcus aureus invasion of the osteocyte lacuno-canalicular network (OLCN) is a novel mechanism of bacterial persistence and immune evasion in chronic osteomyelitis. Previous work highlighted S. aureus cell wall transpeptidase, penicillin binding protein 4 (PBP4), and surface adhesin, S. aureus surface protein C (SasC), as critical factors for bacterial deformation and propagation through nanopores in vitro, representative of the confined canaliculi in vivo. Given these findings, we hypothesized that cell wall synthesis machinery and surface adhesins enable durotaxis- and haptotaxis-guided invasion of the OLCN, respectively. Here, we investigated select S. aureus cell wall synthesis mutants (Δpbp3, Δatl, and ΔmreC) and surface adhesin mutants (ΔclfA and ΔsasC) for nanopore propagation in vitro and osteomyelitis pathogenesis in vivo. In vitro evaluation in the microfluidic silicon membrane-canalicular array (μSiM-CA) showed pbp3, atl, clfA, and sasC deletion reduced nanopore propagation. Using a murine model for implant-associated osteomyelitis, S. aureus cell wall synthesis proteins were found to be key modulators of S. aureus osteomyelitis pathogenesis, while surface adhesins had minimal effects. Specifically, deletion of pbp3 and atl decreased septic implant loosening and S. aureus abscess formation in the medullary cavity, while deletion of surface adhesins showed no significant differences. Further, peri-implant osteolysis, osteoclast activity, and receptor activator of nuclear factor kappa-B ligand (RANKL) production were decreased following pbp3 deletion. Most notably, transmission electron microscopy (TEM) imaging of infected bone showed that pbp3 was the only gene herein associated with decreased submicron invasion of canaliculi in vivo. Together, these results demonstrate that S. aureus cell wall synthesis enzymes are critical for OLCN invasion and osteomyelitis pathogenesis in vivo.
Collapse
Affiliation(s)
- Elysia A Masters
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, United States.,Department of Biomedical Engineering, University of Rochester Medical Center, Rochester, NY, United States
| | - Gowrishankar Muthukrishnan
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, United States.,Department of Orthopaedics, University of Rochester Medical Center, Rochester, NY, United States
| | - Lananh Ho
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, United States.,Department of Biomedical Engineering, University of Rochester Medical Center, Rochester, NY, United States
| | - Ann Lindley Gill
- Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, NY, United States
| | - Karen L de Mesy Bentley
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, United States.,Department of Orthopaedics, University of Rochester Medical Center, Rochester, NY, United States.,Department of Pathology and Laboratory Medicine, University of Rochester Medical Center, Rochester, NY, United States
| | - Chad A Galloway
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, United States.,Department of Pathology and Laboratory Medicine, University of Rochester Medical Center, Rochester, NY, United States
| | - James L McGrath
- Department of Biomedical Engineering, University of Rochester Medical Center, Rochester, NY, United States
| | - Hani A Awad
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, United States.,Department of Biomedical Engineering, University of Rochester Medical Center, Rochester, NY, United States
| | - Steven R Gill
- Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, NY, United States
| | - Edward M Schwarz
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, United States.,Department of Biomedical Engineering, University of Rochester Medical Center, Rochester, NY, United States.,Department of Orthopaedics, University of Rochester Medical Center, Rochester, NY, United States.,Department of Pathology and Laboratory Medicine, University of Rochester Medical Center, Rochester, NY, United States
| |
Collapse
|
17
|
Deghady AM, Hussein RK, Alhamzani AG, Mera A. Density Functional Theory and Molecular Docking Investigations of the Chemical and Antibacterial Activities for 1-(4-Hydroxyphenyl)-3-phenylprop-2-en-1-one. Molecules 2021; 26:molecules26123631. [PMID: 34198585 PMCID: PMC8231836 DOI: 10.3390/molecules26123631] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2021] [Revised: 06/10/2021] [Accepted: 06/12/2021] [Indexed: 01/05/2023] Open
Abstract
The present investigation informs a descriptive study of 1-(4-Hydroxyphenyl) -3-phenylprop-2-en-1-one compound, by using density functional theory at B3LYP method with 6-311G** basis set. The oxygen atoms and π-system revealed a high chemical reactivity for the title compound as electron donor spots and active sites for an electrophilic attack. Quantum chemical parameters such as hardness (η), softness (S), electronegativity (χ), and electrophilicity (ω) were yielded as descriptors for the molecule’s chemical behavior. The optimized molecular structure was obtained, and the experimental data were matched with geometrical analysis values describing the molecule’s stable structure. The computed FT-IR and Raman vibrational frequencies were in good agreement with those observed experimentally. In a molecular docking study, the inhibitory potential of the studied molecule was evaluated against the penicillin-binding proteins of Staphylococcus aureus bacteria. The carbonyl group in the molecule was shown to play a significant role in antibacterial activity, four bonds were formed by the carbonyl group with the key protein of the bacteria (three favorable hydrogen bonds plus one van der Waals bond) out of six interactions. The strong antibacterial activity was also indicated by the calculated high binding energy (−7.40 kcal/mol).
Collapse
Affiliation(s)
- Ahmed M. Deghady
- Basic Science Department, Higher Technological Institute, 10th of Ramadan City 44629, Egypt;
| | - Rageh K. Hussein
- Physics Department, College of Science, Imam Mohammad Ibn Saud Islamic University (IMSIU), Riyadh 11623, Saudi Arabia
- Correspondence:
| | - Abdulrahman G. Alhamzani
- Chemistry Department, College of Science, Imam Mohammad Ibn Saud Islamic University (IMSIU), Riyadh 11623, Saudi Arabia;
| | - Abeer Mera
- Physics Department, College of Arts and Science, Prince Sattam Bin Abdulaziz University, Wadi Addawasir 11991, Saudi Arabia;
- Physics Department, Faculty of Science, Kafrelsheikh University, Kafrelsheikh 33516, Egypt
| |
Collapse
|
18
|
Kussmann M, Obermueller M, Karer M, Kriz R, Chen RY, Hohl L, Schneider L, Burgmann H, Traby L, Vossen MG. Synergistic Effect of Cefazolin Plus Fosfomycin Against Staphylococcus aureus in vitro and in vivo in an Experimental Galleria mellonella Model. Front Pharmacol 2021; 12:685807. [PMID: 34045971 PMCID: PMC8144499 DOI: 10.3389/fphar.2021.685807] [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: 03/25/2021] [Accepted: 04/26/2021] [Indexed: 11/13/2022] Open
Abstract
Objectives: This study investigated the synergistic in vitro and in vivo activity of cefazolin plus fosfomycin against methicillin-susceptible and methicillin-resistant S. aureus (MSSA and MRSA) to provide the basis for a potential treatment alternative. Methods: Antimicrobial susceptibility and in vitro synergy tests were performed with five MSSA and five MRSA isolates using the broth microdilution and chequerboard assays, respectively. The in vivo efficacy of cefazolin plus fosfomycin for the treatment of MRSA infections was assessed using the Galleria mellonella survival assay. Results: Using fractional inhibitory concentration index (FICI), the evaluated combination of cefazolin plus fosfomycin showed synergistic in vitro activity against all MSSA and MRSA isolates tested. In addition, cefazolin susceptibility was recovered in all MRSA isolates except one fosfomycin-resistant strain when combined with fosfomycin at readily achievable concentrations. The G. mellonella survival assay demonstrated highly synergistic in vivo activity of cefazolin plus fosfomycin, resulting in a 44–52% reduction in mortality when compared to cefazolin-alone and fosfomycin-alone, respectively. Conclusion: If susceptibility to fosfomycin is either confirmed or can be assumed based on local resistance patterns, combination therapy with cefazolin plus fosfomycin could be a valuable treatment option for empirical as well as targeted therapy of S. aureus and MRSA infections. Future studies proving the clinical significance of this combination therapy are therefore warranted.
Collapse
Affiliation(s)
- Manuel Kussmann
- Department of Medicine I, Division of Infectious Diseases and Tropical Medicine, Medical University of Vienna, Vienna, Austria
| | - Markus Obermueller
- Department of Medicine I, Division of Infectious Diseases and Tropical Medicine, Medical University of Vienna, Vienna, Austria
| | - Matthias Karer
- Department of Medicine I, Division of Infectious Diseases and Tropical Medicine, Medical University of Vienna, Vienna, Austria.,Department of Clinical Pharmacology, Medical University Vienna, Vienna, Austria
| | - Richard Kriz
- Department of Medicine I, Division of Infectious Diseases and Tropical Medicine, Medical University of Vienna, Vienna, Austria
| | - Rui-Yang Chen
- Department of Medicine I, Division of Infectious Diseases and Tropical Medicine, Medical University of Vienna, Vienna, Austria
| | - Lena Hohl
- Department of Medicine I, Division of Infectious Diseases and Tropical Medicine, Medical University of Vienna, Vienna, Austria
| | - Lisa Schneider
- Department of Medicine I, Division of Infectious Diseases and Tropical Medicine, Medical University of Vienna, Vienna, Austria
| | - Heinz Burgmann
- Department of Medicine I, Division of Infectious Diseases and Tropical Medicine, Medical University of Vienna, Vienna, Austria
| | - Ludwig Traby
- Department of Medicine I, Division of Infectious Diseases and Tropical Medicine, Medical University of Vienna, Vienna, Austria
| | - Matthias G Vossen
- Department of Medicine I, Division of Infectious Diseases and Tropical Medicine, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
19
|
Nain‐Perez A, Barbosa LCA, Araujo MH, Martins JPA, Takahashi JA, Oliveira G, Diniz R, Heller L, Hoenke S, Csuk R. Antibacterial and Cytotoxic Activity of Ruthenium‐
p
‐cymene Complexes with 2‐Methylquinolin‐8‐ol Derivatives. ChemistrySelect 2021. [DOI: 10.1002/slct.202100733] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Affiliation(s)
- Amalyn Nain‐Perez
- Department of Chemistry Universidade Federal de Minas Gerais Av. Pres. Antônio Carlos 6627. Campus Pampulha. CEP 31270-901. Belo Horizonte. MG. Brazil
- Department of Chemistry and Molecular Biology University of Gothenburg Kemivägen 10 41296, Göteborg Sweden
| | - Luiz C. A. Barbosa
- Department of Chemistry Universidade Federal de Minas Gerais Av. Pres. Antônio Carlos 6627. Campus Pampulha. CEP 31270-901. Belo Horizonte. MG. Brazil
- Department of Chemistry Universidade Federal de Viçosa Av. P. H. Rolfs, s/n, CEP 36570-900 Viçosa, MG Brazil
| | - Maria H. Araujo
- Department of Chemistry Universidade Federal de Minas Gerais Av. Pres. Antônio Carlos 6627. Campus Pampulha. CEP 31270-901. Belo Horizonte. MG. Brazil
| | - João P. A. Martins
- Department of Chemistry Universidade Federal de Minas Gerais Av. Pres. Antônio Carlos 6627. Campus Pampulha. CEP 31270-901. Belo Horizonte. MG. Brazil
| | - Jacqueline A. Takahashi
- Department of Chemistry Universidade Federal de Minas Gerais Av. Pres. Antônio Carlos 6627. Campus Pampulha. CEP 31270-901. Belo Horizonte. MG. Brazil
| | - Geane Oliveira
- Department of Chemistry Universidade Federal de Minas Gerais Av. Pres. Antônio Carlos 6627. Campus Pampulha. CEP 31270-901. Belo Horizonte. MG. Brazil
| | - Renata Diniz
- Department of Chemistry Universidade Federal de Minas Gerais Av. Pres. Antônio Carlos 6627. Campus Pampulha. CEP 31270-901. Belo Horizonte. MG. Brazil
| | - Lucie Heller
- Organic Chemistry Martin-Luther-University Halle-Wittenberg Kurt-Mothes-Str.2. D-06120 Halle (Saale Germany
| | - Sophie Hoenke
- Organic Chemistry Martin-Luther-University Halle-Wittenberg Kurt-Mothes-Str.2. D-06120 Halle (Saale Germany
| | - René Csuk
- Organic Chemistry Martin-Luther-University Halle-Wittenberg Kurt-Mothes-Str.2. D-06120 Halle (Saale Germany
| |
Collapse
|
20
|
Fisher JF, Mobashery S. β-Lactams against the Fortress of the Gram-Positive Staphylococcus aureus Bacterium. Chem Rev 2021; 121:3412-3463. [PMID: 33373523 PMCID: PMC8653850 DOI: 10.1021/acs.chemrev.0c01010] [Citation(s) in RCA: 52] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The biological diversity of the unicellular bacteria-whether assessed by shape, food, metabolism, or ecological niche-surely rivals (if not exceeds) that of the multicellular eukaryotes. The relationship between bacteria whose ecological niche is the eukaryote, and the eukaryote, is often symbiosis or stasis. Some bacteria, however, seek advantage in this relationship. One of the most successful-to the disadvantage of the eukaryote-is the small (less than 1 μm diameter) and nearly spherical Staphylococcus aureus bacterium. For decades, successful clinical control of its infection has been accomplished using β-lactam antibiotics such as the penicillins and the cephalosporins. Over these same decades S. aureus has perfected resistance mechanisms against these antibiotics, which are then countered by new generations of β-lactam structure. This review addresses the current breadth of biochemical and microbiological efforts to preserve the future of the β-lactam antibiotics through a better understanding of how S. aureus protects the enzyme targets of the β-lactams, the penicillin-binding proteins. The penicillin-binding proteins are essential enzyme catalysts for the biosynthesis of the cell wall, and understanding how this cell wall is integrated into the protective cell envelope of the bacterium may identify new antibacterials and new adjuvants that preserve the efficacy of the β-lactams.
Collapse
Affiliation(s)
- Jed F Fisher
- Department of Chemistry and Biochemistry, McCourtney Hall, University of Notre Dame, Notre Dame Indiana 46556, United States
| | - Shahriar Mobashery
- Department of Chemistry and Biochemistry, McCourtney Hall, University of Notre Dame, Notre Dame Indiana 46556, United States
| |
Collapse
|
21
|
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
|
22
|
Rashid M, Ikram M, Haider A, Naz S, Haider J, Ul-Hamid A, Shahzadi A, Aqeel M. Photocatalytic, dye degradation, and bactericidal behavior of Cu-doped ZnO nanorods and their molecular docking analysis. Dalton Trans 2021; 49:8314-8330. [PMID: 32515772 DOI: 10.1039/d0dt01397h] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Nanostructures of Cu-doped ZnO (Cu:ZnO) were prepared with the chemical precipitation technique with an aim to enhance the photocatalytic and antibacterial properties of ZnO. Phase constitution, the presence of functional groups, optical properties, elemental composition, surface morphology and microstructure were evaluated using an X-ray diffractometer (XRD), Fourier transform infrared spectroscopy (FTIR), UV-Vis spectrophotometer, energy dispersive X-ray spectroscopy (EDS), field emission scanning electron microscope (FESEM) and high resolution transmission electron microscope (HR-TEM), respectively. Emission spectra were obtained with a photoluminescence (PL) spectroscope whereas interlayer d-spacing was estimated through HR-TEM. ZnO consisted of a hexagonal wurtzite structure. The crystallinity of the sample was observed to increase with increasing doping concentration. The addition of Cu to ZnO served to transform nanoclusters into nanorods as revealed during SEM analysis. Catalytic activity enhanced due to the formation of nanorods, and UV-Vis absorption spectra showed that methylene blue (MB) degraded more efficiently with ZnO nanoclusters compared to the NaBH4 reagent. In addition, the doped NPs showed enhanced bacterial efficiency for G +ve. Finally, a molecular docking study was undertaken to highlight the importance of the binding interactions of the Cu-doped ZnO nanorods with β-lactamase and beta-ketoacyl-acyl carrier protein synthase III (FabH) as possible enzyme targets. This research indicates that Cu-doped Zn nanorods are a highly efficient photocatalyst and can be aptly employed for wastewater treatment and antibacterial applications.
Collapse
Affiliation(s)
- Mehak Rashid
- Solar Cell Applications Research Lab, Department of Physics, Government College University Lahore, 54000, Punjab, Pakistan.
| | | | | | | | | | | | | | | |
Collapse
|
23
|
Desai NC, Vaja DV, Monapara JD, Manga V, Vani T. Synthesis, biological evaluation, and molecular docking studies of novel pyrazole, pyrazoline‐clubbed pyridine as potential antimicrobial agents. J Heterocycl Chem 2021. [DOI: 10.1002/jhet.4208] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Affiliation(s)
- Nisheeth C. Desai
- Division of Medicinal Chemistry, Department of Chemistry (DST‐FIST Sponsored) Mahatma Gandhi Campus, Maharaja Krishnakumarsinhji Bhavnagar University Bhavnagar India
| | - Darshita V. Vaja
- Division of Medicinal Chemistry, Department of Chemistry (DST‐FIST Sponsored) Mahatma Gandhi Campus, Maharaja Krishnakumarsinhji Bhavnagar University Bhavnagar India
| | - Jahnvi D. Monapara
- Division of Medicinal Chemistry, Department of Chemistry (DST‐FIST Sponsored) Mahatma Gandhi Campus, Maharaja Krishnakumarsinhji Bhavnagar University Bhavnagar India
| | - Vijjulatha Manga
- Molecular Modeling and Medicinal Chemistry Group, Department of Chemistry University College of Science, Osmania University Hyderabad India
| | - Tamalapakula Vani
- Molecular Modeling and Medicinal Chemistry Group, Department of Chemistry University College of Science, Osmania University Hyderabad India
| |
Collapse
|
24
|
Bitla S, Sagurthi SR, Dhanavath R, Puchakayala MR, Birudaraju S, Gayatri AA, Bhukya VK, Atcha KR. Design and synthesis of triazole conjugated novel 2,5-diaryl substituted 1,3,4-oxadiazoles as potential antimicrobial and anti-fungal agents. J Mol Struct 2020. [DOI: 10.1016/j.molstruc.2020.128705] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
25
|
Masters EA, de Mesy Bentley KL, Gill AL, Hao SP, Galloway CA, Salminen AT, Guy DR, McGrath JL, Awad HA, Gill SR, Schwarz EM. Identification of Penicillin Binding Protein 4 (PBP4) as a critical factor for Staphylococcus aureus bone invasion during osteomyelitis in mice. PLoS Pathog 2020; 16:e1008988. [PMID: 33091079 PMCID: PMC7608983 DOI: 10.1371/journal.ppat.1008988] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2020] [Revised: 11/03/2020] [Accepted: 09/17/2020] [Indexed: 12/03/2022] Open
Abstract
Staphylococcus aureus infection of bone is challenging to treat because it colonizes the osteocyte lacuno-canalicular network (OLCN) of cortical bone. To elucidate factors involved in OLCN invasion and identify novel drug targets, we completed a hypothesis-driven screen of 24 S. aureus transposon insertion mutant strains for their ability to propagate through 0.5 μm-sized pores in the Microfluidic Silicon Membrane Canalicular Arrays (μSiM-CA), developed to model S. aureus invasion of the OLCN. This screen identified the uncanonical S. aureus transpeptidase, penicillin binding protein 4 (PBP4), as a necessary gene for S. aureus deformation and propagation through nanopores. In vivo studies revealed that Δpbp4 infected tibiae treated with vancomycin showed a significant 12-fold reduction in bacterial load compared to WT infected tibiae treated with vancomycin (p<0.05). Additionally, Δpbp4 infected tibiae displayed a remarkable decrease in pathogenic bone-loss at the implant site with and without vancomycin therapy. Most importantly, Δpbp4 S. aureus failed to invade and colonize the OLCN despite high bacterial loads on the implant and in adjacent tissues. Together, these results demonstrate that PBP4 is required for S. aureus colonization of the OLCN and suggest that inhibitors may be synergistic with standard of care antibiotics ineffective against bacteria within the OLCN.
Collapse
Affiliation(s)
- Elysia A. Masters
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, United States of America
- Department of Biomedical Engineering, University of Rochester Medical Center, Rochester, NY, United States of America
| | - Karen L. de Mesy Bentley
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, United States of America
- Department of Pathology and Laboratory Medicine, University of Rochester Medical Center, Rochester, NY, United States of America
- Department of Orthopaedics, University of Rochester Medical Center, Rochester, NY, United States of America
| | - Ann Lindley Gill
- Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, NY, United States of America
| | - Stephanie P. Hao
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, United States of America
| | - Chad A. Galloway
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, United States of America
- Department of Pathology and Laboratory Medicine, University of Rochester Medical Center, Rochester, NY, United States of America
| | - Alec T. Salminen
- Department of Biomedical Engineering, University of Rochester Medical Center, Rochester, NY, United States of America
| | - Diamond R. Guy
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, United States of America
| | - James L. McGrath
- Department of Biomedical Engineering, University of Rochester Medical Center, Rochester, NY, United States of America
| | - Hani A. Awad
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, United States of America
- Department of Biomedical Engineering, University of Rochester Medical Center, Rochester, NY, United States of America
| | - Steven R. Gill
- Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, NY, United States of America
| | - Edward M. Schwarz
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, United States of America
- Department of Biomedical Engineering, University of Rochester Medical Center, Rochester, NY, United States of America
- Department of Orthopaedics, University of Rochester Medical Center, Rochester, NY, United States of America
| |
Collapse
|
26
|
Fusani L, Palmer DS, Somers DO, Wall ID. Exploring Ligand Stability in Protein Crystal Structures Using Binding Pose Metadynamics. J Chem Inf Model 2020; 60:1528-1539. [PMID: 31910338 PMCID: PMC7145342 DOI: 10.1021/acs.jcim.9b00843] [Citation(s) in RCA: 78] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
![]()
Identification of
correct protein–ligand binding poses is
important in structure-based drug design and crucial for the evaluation
of protein–ligand binding affinity. Protein–ligand coordinates are commonly obtained from
crystallography experiments that provide a static model of an ensemble
of conformations. Binding pose metadynamics (BPMD) is an enhanced
sampling method that allows for an efficient assessment of ligand
stability in solution. Ligand poses that are unstable under the bias
of the metadynamics simulation are expected to be infrequently occupied
in the energy landscape, thus making minimal contributions to the
binding affinity. Here, the robustness of the method is studied using
crystal structures with ligands known to be incorrectly modeled, as
well as 63 structurally diverse crystal structures with ligand fit
to electron density from the Twilight database. Results show that
BPMD can successfully differentiate compounds whose binding pose is
not supported by the electron density from those with well-defined
electron density.
Collapse
Affiliation(s)
- Lucia Fusani
- Molecular Design UK, GSK Medicines Research Centre, Gunnels Wood Road, Stevenage, Hertfordshire SG1 2NY, U.K.,Department of Pure and Applied Chemistry, University of Strathclyde, 295 Cathedral Street, Glasgow G11XL, U.K
| | - David S Palmer
- Department of Pure and Applied Chemistry, University of Strathclyde, 295 Cathedral Street, Glasgow G11XL, U.K
| | - Don O Somers
- Protein, Cellular and Structural Sciences, GSK Medicines Research Centre, Gunnels Wood Road, Stevenage, Hertfordshire SG1 2NY, U.K
| | - Ian D Wall
- Molecular Design UK, GSK Medicines Research Centre, Gunnels Wood Road, Stevenage, Hertfordshire SG1 2NY, U.K
| |
Collapse
|
27
|
New compounds for a good old class: Synthesis of two Β-lactam bearing cephalosporins and their evaluation with a multidisciplinary approach. Bioorg Med Chem 2020; 28:115302. [PMID: 31932194 DOI: 10.1016/j.bmc.2019.115302] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2019] [Revised: 12/13/2019] [Accepted: 12/31/2019] [Indexed: 11/21/2022]
Abstract
Antimicrobial resistance is spreading massively in the world and is becoming one of the main health threats of the 21st century. One of the possible strategies to overcome this problem is to modify the known classes of antibiotics in a rational way, with the aim of tuning their efficacy. In this paper, we present the synthesis and the evaluation of the biological activity of a series of two β-lactam bearing cephalosporin derivatives, in which an additional isolated azetidinone ring, bearing different substituents, is joined to the classical cephalosporanic nucleus by a chain of variable length. A computational approach has been also applied in order to predict the molecular interactions between some representative derivatives and selected penicillin-binding proteins, the natural targets of β-lactam antibiotics. All these derivatives are active against Gram-positive bacteria, with MIC100 comparable or even better than that of the reference antibiotic ceftriaxone, and show no or very low cytotoxic activity on different cell lines. Overall, these molecules appear to be able to exert their activity in particular against microorganisms belonging to some of the species more involved in the development of multidrug resistance.
Collapse
|
28
|
Design and synthesis of imidazolo-1, 2,3-triazoles hybrid compounds by microwave-assisted method: Evaluation as an antioxidant and antimicrobial agents and molecular docking studies. J Mol Struct 2019. [DOI: 10.1016/j.molstruc.2018.11.029] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
|
29
|
Hsu YP, Hall E, Booher G, Murphy B, Radkov AD, Yablonowski J, Mulcahey C, Alvarez L, Cava F, Brun YV, Kuru E, VanNieuwenhze MS. Fluorogenic D-amino acids enable real-time monitoring of peptidoglycan biosynthesis and high-throughput transpeptidation assays. Nat Chem 2019; 11:335-341. [PMID: 30804500 PMCID: PMC6444347 DOI: 10.1038/s41557-019-0217-x] [Citation(s) in RCA: 69] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2018] [Accepted: 01/10/2019] [Indexed: 01/09/2023]
Abstract
Peptidoglycan (PG) is an essential cell wall component that maintains the morphology and viability of nearly all bacteria. Its biosynthesis requires periplasmic transpeptidation reactions which construct peptide cross-linkages between polysaccharide chains to endow mechanical strength. However, tracking transpeptidation reaction in vivo and in vitro is challenging, mainly due to the lack of efficient, biocompatible probes. Here, we report the design, synthesis, and application of rotor-fluorogenic D-amino acids (RfDAAs) enabling real-time, continuous tracking of transpeptidation reactions. These probes enable monitoring PG biosynthesis in real time through visualizing transpeptidase reactions in live cells, as well as real-time activity assays of D,D-, L,D-transpeptidases, and sortases in vitro. The unique ability of RfDAAs to become fluorescent when incorporated into PG provides a powerful new tool to study PG biosynthesis with high temporal resolution and prospectively enable high-throughput screening for inhibitors of PG biosynthesis.
Collapse
Affiliation(s)
- Yen-Pang Hsu
- Department of Molecular and Cellular Biochemistry, Indiana University, Bloomington, IN, USA
| | - Edward Hall
- Department of Chemistry, Indiana University, Bloomington, IN, USA.,Department of Chemistry, Hanover College, Hanover, IN, USA
| | - Garrett Booher
- Department of Molecular and Cellular Biochemistry, Indiana University, Bloomington, IN, USA
| | - Brennan Murphy
- Department of Chemistry, Indiana University, Bloomington, IN, USA
| | - Atanas D Radkov
- Department of Chemistry, Indiana University, Bloomington, IN, USA.,Department of Biophysics and Biochemistry, University of California San Francisco, San Francisco, CA, USA
| | - Jacob Yablonowski
- Department of Molecular and Cellular Biochemistry, Indiana University, Bloomington, IN, USA
| | - Caitlyn Mulcahey
- Department of Molecular and Cellular Biochemistry, Indiana University, Bloomington, IN, USA
| | - Laura Alvarez
- Department of Molecular Biology, Umeå University, Umeå, Sweden
| | - Felipe Cava
- Department of Molecular Biology, Umeå University, Umeå, Sweden
| | - Yves V Brun
- Department of Biology, Indiana University, Bloomington, IN, USA. .,Département de Microbiologie, Infectiologie et Immunologie, Université de Montréal, Pavillon Roger-Gaudry, C.P. 6128, Succursale Centre-ville, Montréal, Canada.
| | - Erkin Kuru
- Department of Molecular and Cellular Biochemistry, Indiana University, Bloomington, IN, USA. .,Department of Genetics, Harvard Medical School, Boston, MA, USA.
| | - Michael S VanNieuwenhze
- Department of Molecular and Cellular Biochemistry, Indiana University, Bloomington, IN, USA. .,Department of Chemistry, Indiana University, Bloomington, IN, USA.
| |
Collapse
|
30
|
Alexander JAN, Chatterjee SS, Hamilton SM, Eltis LD, Chambers HF, Strynadka NCJ. Structural and kinetic analyses of penicillin-binding protein 4 (PBP4)-mediated antibiotic resistance in Staphylococcus aureus. J Biol Chem 2018; 293:19854-19865. [PMID: 30366985 DOI: 10.1074/jbc.ra118.004952] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2018] [Revised: 10/24/2018] [Indexed: 01/08/2023] Open
Abstract
Methicillin-resistant Staphylococcus aureus (MRSA) causes serious community-acquired and nosocomial infections worldwide. MRSA strains are resistant to a variety of antibiotics, including the classic penicillin and cephalosporin classes of β-lactams, making them intractable to treatment. Although β-lactam resistance in MRSA has been ascribed to the acquisition and activity of penicillin-binding protein 2a (PBP2a, encoded by mecA), it has recently been observed that resistance can also be mediated by penicillin-binding protein 4 (PBP4). Previously, we have shown that broad-spectrum β-lactam resistance can arise following serial passaging of a mecA-negative COL strain of S. aureus, creating the CRB strain. This strain has two missense mutations in pbp4 and a mutation in the pbp4 promoter, both of which play an instrumental role in β-lactam resistance. To better understand PBP4's role in resistance, here we have characterized its kinetics and structure with clinically relevant β-lactam antibiotics. We present the first crystallographic PBP4 structures of apo and acyl-enzyme intermediate forms complexed with three late-generation β-lactam antibiotics: ceftobiprole, ceftaroline, and nafcillin. In parallel, we characterized the structural and kinetic effects of the PBP4 mutations present in the CRB strain. Localized within the transpeptidase active-site cleft, the two substitutions appear to have different effects depending on the drug. With ceftobiprole, the missense mutations impaired the Km value 150-fold, decreasing the proportion of inhibited PBP4. However, ceftaroline resistance appeared to be mediated by other factors, possibly including mutation of the pbp4 promoter. Our findings provide evidence that S. aureus CRB has at least two PBP4-mediated resistance mechanisms.
Collapse
Affiliation(s)
- J Andrew N Alexander
- From the Department of Biochemistry and Molecular Biology.,the Centre for Blood Research, and
| | - Som S Chatterjee
- the Division of Infectious Disease, Department of Medicine, San Francisco General Hospital, San Francisco, California 94110
| | - Stephanie M Hamilton
- the Division of Infectious Disease, Department of Medicine, San Francisco General Hospital, San Francisco, California 94110
| | - Lindsay D Eltis
- From the Department of Biochemistry and Molecular Biology.,the Department of Microbiology and Immunology, The University of British Columbia, Vancouver, British Columbia V6T 1Z3, Canada and
| | - Henry F Chambers
- the Division of Infectious Disease, Department of Medicine, San Francisco General Hospital, San Francisco, California 94110
| | - Natalie C J Strynadka
- From the Department of Biochemistry and Molecular Biology, .,the Centre for Blood Research, and
| |
Collapse
|
31
|
Ealand CS, Machowski EE, Kana BD. β-lactam resistance: The role of low molecular weight penicillin binding proteins, β-lactamases and ld-transpeptidases in bacteria associated with respiratory tract infections. IUBMB Life 2018; 70:855-868. [PMID: 29717815 DOI: 10.1002/iub.1761] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2018] [Accepted: 04/04/2018] [Indexed: 02/02/2023]
Abstract
Disruption of peptidoglycan (PG) biosynthesis in the bacterial cell wall by β-lactam antibiotics has transformed therapeutic options for bacterial infections. These antibiotics target the transpeptidase domains in penicillin binding proteins (PBPs), which can be classified into high and low molecular weight (LMW) counterparts. While the essentiality of the former has been extensively demonstrated, the physiological roles of LMW PBPs remain poorly understood. Herein, we review the function of LMW PBPs, β-lactamases and ld-transpeptidases (Ldts) in pathogens associated with respiratory tract infections. More specifically, we explore their roles in mediating β-lactam resistance. Using a comparative genomics approach, we identified a high degree of genetic redundancy for LMW PBPs which retain the motifs, SxxN, SxN and KTG required for catalytic activity. Differences in domain architecture suggest distinct physiological roles, possibly related to bacterial cell cycle and/or adaptation to various environmental conditions. Many of the LMW PBPs play an important role in β-lactam resistance either through mutation or variation in abundance. In all of the bacterial genomes assessed, at least one β-lactamase homologue is present, suggesting that enzymatic degradation of β-lactams is a highly conserved resistance mechanism. Furthermore, the presence of Ldt homologues in the majority of species surveyed suggests that alternative PG crosslinking may further mediate β-lactam drug resistance. A deeper understanding of the interplay between these different mechanisms of β-lactam resistance will provide a framework for new therapeutics, which are urgently required given the rapid emergence of antimicrobial resistance. © 2018 IUBMB Life, 70(9):855-868, 2018.
Collapse
Affiliation(s)
- Christopher S Ealand
- DST/NRF Centre of Excellence for Biomedical TB Research, School of Pathology, Faculty of Health Sciences, University of the Witwatersrand and the National Health Laboratory Service, Johannesburg, South Africa
| | - Edith E Machowski
- DST/NRF Centre of Excellence for Biomedical TB Research, School of Pathology, Faculty of Health Sciences, University of the Witwatersrand and the National Health Laboratory Service, Johannesburg, South Africa
| | - Bavesh D Kana
- DST/NRF Centre of Excellence for Biomedical TB Research, School of Pathology, Faculty of Health Sciences, University of the Witwatersrand and the National Health Laboratory Service, Johannesburg, South Africa.,MRC-CAPRISA HIV-TB Pathogenesis and Treatment Research Unit, Centre for the AIDS Programme of Research in South Africa, CAPRISA, Durban, South Africa
| |
Collapse
|
32
|
Xue H, Wu Z, Qiao D, Tong C, Zhao X. Global acquisition of genetic material from different bacteria into the staphylococcal cassette chromosome elements of a Staphylococcus epidermidis isolate. Int J Antimicrob Agents 2017; 50:581-587. [DOI: 10.1016/j.ijantimicag.2017.06.015] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2016] [Accepted: 06/24/2017] [Indexed: 10/19/2022]
|
33
|
β-Lactam Antibiotics with a High Affinity for PBP2 Act Synergistically with the FtsZ-Targeting Agent TXA707 against Methicillin-Resistant Staphylococcus aureus. Antimicrob Agents Chemother 2017. [PMID: 28630190 DOI: 10.1128/aac.00863-17] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Methicillin-resistant Staphylococcus aureus (MRSA) is a multidrug-resistant pathogen that poses a significant risk to global health today. We have developed a promising new FtsZ-targeting agent (TXA707) with potent activity against MRSA isolates resistant to current standard-of-care antibiotics. We present here results that demonstrate differing extents of synergy between TXA707 and a broad range of β-lactam antibiotics (including six cephalosporins, two penicillins, and two carbapenems) against MRSA. To explore whether there is a correlation between the extent of synergy and the preferential antibacterial target of each β-lactam, we determined the binding affinities of the β-lactam antibiotics for each of the four native penicillin-binding proteins (PBPs) of S. aureus using a fluorescence anisotropy competition assay. A comparison of the resulting PBP binding affinities with our corresponding synergy results reveals that β-lactams with a high affinity for PBP2 afford the greatest degree of synergy with TXA707 against MRSA. In addition, we present fluorescence and electron microscopy studies that suggest a potential mechanism underlying the synergy between TXA707 and the β-lactam antibiotics. In this connection, our microscopy results show a disruption of septum formation in TXA707-treated MRSA cells, with a concomitant mislocalization of the PBPs from midcell to nonproductive peripheral sites. Viewed as a whole, our results indicate that PBP2-targeting β-lactam antibiotics are optimal synergistic partners with FtsZ-targeting agents for use in combination therapy of MRSA infections.
Collapse
|
34
|
Verdino A, Vigliotta G, Giordano D, Caputo I, Soriente A, De Rosa M, Marabotti A. Synthesis and biological evaluation of the progenitor of a new class of cephalosporin analogues, with a particular focus on structure-based computational analysis. PLoS One 2017; 12:e0181563. [PMID: 28749999 PMCID: PMC5531512 DOI: 10.1371/journal.pone.0181563] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2017] [Accepted: 07/03/2017] [Indexed: 02/04/2023] Open
Abstract
We present the synthesis and biological evaluation of the prototype of a new class of cephalosporins, containing an additional isolated beta lactam ring with two phenyl substituents. This new compound is effective against Gram positive microorganisms, with a potency similar to that of ceftriaxone, a cephalosporin widely used in clinics and taken as a reference, and with no cytotoxicity against two different human cell lines, even at a concentration much higher than the minimal inhibitory concentration tested. Additionally, a deep computational analysis has been conducted with the aim of understanding the contribution of its moieties to the binding energy towards several penicillin-binding proteins from both Gram positive and Gram negative bacteria. All these results will help us developing derivatives of this compound with improved chemical and biological properties, such as a broader spectrum of action and/or an increased affinity towards their molecular targets.
Collapse
Affiliation(s)
- Anna Verdino
- Department of Chemistry and Biology "A. Zambelli", University of Salerno, Fisciano (SA), Italy
| | - Giovanni Vigliotta
- Department of Chemistry and Biology "A. Zambelli", University of Salerno, Fisciano (SA), Italy
| | - Deborah Giordano
- Department of Chemistry and Biology "A. Zambelli", University of Salerno, Fisciano (SA), Italy
| | - Ivana Caputo
- Department of Chemistry and Biology "A. Zambelli", University of Salerno, Fisciano (SA), Italy
| | - Annunziata Soriente
- Department of Chemistry and Biology "A. Zambelli", University of Salerno, Fisciano (SA), Italy
| | - Margherita De Rosa
- Department of Chemistry and Biology "A. Zambelli", University of Salerno, Fisciano (SA), Italy
- * E-mail: (MDR); (AM)
| | - Anna Marabotti
- Department of Chemistry and Biology "A. Zambelli", University of Salerno, Fisciano (SA), Italy
- * E-mail: (MDR); (AM)
| |
Collapse
|
35
|
Park M, Rafii F. Exposure to β-lactams results in the alteration of penicillin-binding proteins in Clostridium perfringens. Anaerobe 2017; 45:78-85. [PMID: 28185856 DOI: 10.1016/j.anaerobe.2017.02.004] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2016] [Revised: 02/03/2017] [Accepted: 02/05/2017] [Indexed: 10/20/2022]
|
36
|
High-Level Resistance of Staphylococcus aureus to β-Lactam Antibiotics Mediated by Penicillin-Binding Protein 4 (PBP4). Antimicrob Agents Chemother 2017; 61:AAC.02727-16. [PMID: 28373193 DOI: 10.1128/aac.02727-16] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2016] [Accepted: 03/26/2017] [Indexed: 02/05/2023] Open
Abstract
Penicillin-binding protein 4 (PBP4), a nonessential, low-molecular-weight penicillin-binding protein of Staphylococcus aureus, has been implicated in low-level resistance to β-lactam antibiotics, although the mechanism is unknown. Mutations in PBP4 and its promoter were identified in a laboratory-generated mutant strain, CRB, which expresses high-level resistance to β-lactams, including resistance to the new-generation cephalosporins active against methicillin-resistant strains of S. aureus These mutations did not appreciably alter the β-lactam antibiotic binding affinity of purified recombinant mutant PBP4 compared to that of wild-type PBP4. Compared to the susceptible parent strain, COLnex, the CRB strain produces a highly cross-linked cell wall peptidoglycan, indicative of increased transpeptidase activity. The pbp4 promoter mutation of CRB was associated with greatly increased amounts of PBP4 in membranes compared to those in the COLnex parent. Replacement of the native promoter of COLnex with the mutant promoter of CRB resulted in increased amounts of PBP4 in membranes and a highly cross-linked cell wall. PBP4 can be repurposed to provide essential transpeptidase activity in vivo and confer high-level resistance to β-lactam antibiotics, such as ceftobiprole and ceftaroline.
Collapse
|
37
|
Kuok CF, Hoi SO, Hoi CF, Chan CH, Fong IH, Ngok CK, Meng LR, Fong P. Synergistic antibacterial effects of herbal extracts and antibiotics on methicillin-resistant Staphylococcus aureus: A computational and experimental study. Exp Biol Med (Maywood) 2017; 242:731-743. [PMID: 28118725 DOI: 10.1177/1535370216689828] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
Antibiotic resistance has become a serious global concern, and the discovery of antimicrobial herbal constituents may provide valuable solutions to overcome the problem. In this study, the effects of therapies combining antibiotics and four medicinal herbs on methicillin-resistant Staphylococcus aureus (MRSA) were investigated. Specifically, the synergistic effects of Magnolia officinalis, Verbena officinalis, Momordica charantia, and Daphne genkwa in combination with oxacillin or gentamicin against methicillin-resistant (ATCC43300) and methicillin-susceptible (ATCC25923) S. aureus were examined. In vitro susceptibility and synergistic testing were performed to measure the minimum inhibitory concentration and fractional inhibitory concentration (FIC) index of the antibiotics and medicinal herbs against MRSA and methicillin-susceptible S. aureus. To identify the active constituents in producing these synergistic effects, in silico molecular docking was used to investigate the binding affinities of 139 constituents of the four herbs to the two common MRSA inhibitory targets, penicillin binding proteins 2a (PBP2a) and 4 (PBP4). The physicochemical and absorption, distribution, metabolism, and excretion properties and drug safety profiles of these compounds were also analyzed. D. genkwa extract potentiated the antibacterial effects of oxacillin against MRSA, as indicated by an FIC index value of 0.375. M. officinalis and V. officinalis produced partial synergistic effects when combined with oxacillin, whereas M. charantia was found to have no beneficial effects in inhibiting MRSA. Overall, tiliroside, pinoresinol, magnatriol B, and momorcharaside B were predicted to be PBP2a or PBP4 inhibitors with good drug-like properties. This study identifies compounds that deserve further investigation with the aim of developing therapeutic agents to modulate the effect of antibiotics on MRSA. Impact statement Antibiotic resistant is a well-known threat to global health and methicillin-resistant Staphylococcus aureus is one of the most significant ones. These resistant bacteria kill thousands of people every year and therefore a new effective antimicrobial treatment is necessary. This study identified the herbs and their associated bioactive ingredients that can potential the effects of current antibiotics. These herbs have long history of human usage in China and have well-defined monograph in the Chinese Pharmacopeia. These indicate their relatively high clinical safety and may have a quicker drug development process than that of a new novel antibiotic. Based on the results of this study, the authors will perform further in vitro and animal studies, aiming to accumulate significant data for the application of clinical trial.
Collapse
Affiliation(s)
- Chiu-Fai Kuok
- School of Health Sciences, Macao Polytechnic Institute, Macao 999078, China
| | - Sai-On Hoi
- School of Health Sciences, Macao Polytechnic Institute, Macao 999078, China
| | - Chi-Fai Hoi
- School of Health Sciences, Macao Polytechnic Institute, Macao 999078, China
| | - Chi-Hong Chan
- School of Health Sciences, Macao Polytechnic Institute, Macao 999078, China
| | - Io-Hong Fong
- School of Health Sciences, Macao Polytechnic Institute, Macao 999078, China
| | - Cheong-Kei Ngok
- School of Health Sciences, Macao Polytechnic Institute, Macao 999078, China
| | - Li-Rong Meng
- School of Health Sciences, Macao Polytechnic Institute, Macao 999078, China
| | - Pedro Fong
- School of Health Sciences, Macao Polytechnic Institute, Macao 999078, China
| |
Collapse
|
38
|
Lahiri SD, Alm RA. Identification of non-PBP2a resistance mechanisms in Staphylococcus aureus after serial passage with ceftaroline: involvement of other PBPs. J Antimicrob Chemother 2016; 71:3050-3057. [PMID: 27494915 DOI: 10.1093/jac/dkw282] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2016] [Revised: 05/12/2016] [Accepted: 06/11/2016] [Indexed: 12/16/2023] Open
Abstract
OBJECTIVES Ceftaroline (the active metabolite of ceftaroline fosamil) is a cephalosporin that possesses activity against MRSA due to its differentiating high affinity for PBP2a. It is known that PBP2a sequence variations, including some outside of the transpeptidase-binding pocket, impact ceftaroline susceptibility and recent evidence suggests involvement of non-PBP2a mechanisms in ceftaroline resistance. This study evaluated the potential of ceftaroline to select for resistant Staphylococcus aureus clones during serial passage. METHODS Selection experiments were performed by up to 20 daily passages of three S. aureus isolates (two MRSA and one MSSA) in broth with increasing selective pressure. Mutants that emerged were tested for changes in ceftaroline susceptibility and genetically characterized. RESULTS The MSSA isolate developed mutations in PBP2 and PBP3 that increased the ceftaroline MIC by 16-fold and increased the MICs of other β-lactams. A Glu447Lys substitution in the PBP2a transpeptidase pocket in one MRSA isolate elevated the ceftaroline MIC to 8 mg/L. Selective pressure in a ceftaroline-resistant MRSA isolate generated mutations in LytD, as well as changes in the pbp4 promoter previously shown to result in PBP4 overexpression, the one PBP not inhibited by ceftaroline. Elevated ceftaroline MIC was reversed when tested in combination with extremely low levels of methicillin or meropenem that could inhibit the function of PBP4. CONCLUSIONS These studies demonstrate that resistance to ceftaroline can be manifested through numerous mechanisms. Further, they support a hypothesis where PBP4 can functionally provide the essential transpeptidase activity required for MRSA cell wall biogenesis when PBP2a is inhibited.
Collapse
Affiliation(s)
- Sushmita D Lahiri
- Infection Innovative Medicines Unit, AstraZeneca R&D Boston, Waltham, MA, USA
| | - Richard A Alm
- Infection Innovative Medicines Unit, AstraZeneca R&D Boston, Waltham, MA, USA
| |
Collapse
|
39
|
PBP 4 Mediates High-Level Resistance to New-Generation Cephalosporins in Staphylococcus aureus. Antimicrob Agents Chemother 2016; 60:3934-41. [PMID: 27067335 DOI: 10.1128/aac.00358-16] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2016] [Accepted: 04/08/2016] [Indexed: 12/21/2022] Open
Abstract
Staphylococcus aureus is an important cause of both hospital- and community-associated methicillin-resistant S. aureus (MRSA) infections worldwide. β-Lactam antibiotics are the drugs of choice to treat S. aureus infections, but resistance to these and other antibiotics make treatment problematic. High-level β-lactam resistance of S. aureus has always been attributed to the horizontally acquired penicillin binding protein 2a (PBP 2a) encoded by the mecA gene. Here, we show that S. aureus can also express high-level resistance to β-lactams, including new-generation broad-spectrum cephalosporins that are active against methicillin-resistant strains, through an uncanonical core genome-encoded penicillin binding protein, PBP 4, a nonessential enzyme previously considered not to be important for staphylococcal β-lactam resistance. Our results show that PBP 4 can mediate high-level resistance to β-lactams.
Collapse
|
40
|
In Silico Characterization of the Binding Affinity of Dendrimers to Penicillin-Binding Proteins (PBPs): Can PBPs be Potential Targets for Antibacterial Dendrimers? Appl Biochem Biotechnol 2016; 178:1546-66. [DOI: 10.1007/s12010-015-1967-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2015] [Accepted: 12/22/2015] [Indexed: 10/22/2022]
|
41
|
Greninger AL, Chatterjee SS, Chan LC, Hamilton SM, Chambers HF, Chiu CY. Whole-Genome Sequencing of Methicillin-Resistant Staphylococcus aureus Resistant to Fifth-Generation Cephalosporins Reveals Potential Non-mecA Mechanisms of Resistance. PLoS One 2016; 11:e0149541. [PMID: 26890675 PMCID: PMC4758708 DOI: 10.1371/journal.pone.0149541] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2015] [Accepted: 02/02/2016] [Indexed: 01/28/2023] Open
Abstract
Fifth-generation cephalosporins, ceftobiprole and ceftaroline, are promising drugs for treatment of bacterial infections from methicillin-resistant Staphylococcus aureus (MRSA). These antibiotics are able to bind native PBP2a, the penicillin-binding protein encoded by the mecA resistance determinant that mediates broad class resistance to nearly all other beta-lactam antibiotics, at clinically achievable concentrations. Mechanisms of resistance to ceftaroline based on mecA mutations have been previously described. Here we compare the genomes of 11 total parent-daughter strains of Staphylococcus aureus for which specific selection by serial passaging with ceftaroline or ceftobiprole was used to identify novel non-mecA mechanisms of resistance. All 5 ceftaroline-resistant strains, derived from 5 different parental strains, contained mutations directly upstream of the pbp4 gene (coding for the PBP4 protein), including four with the same thymidine insertion located 377 nucleotides upstream of the promoter site. In 4 of 5 independent ceftaroline-driven selections, we also isolated mutations to the same residue (Asn138) in PBP4. In addition, mutations in additional candidate genes such as ClpX endopeptidase, PP2C protein phosphatase and transcription terminator Rho, previously undescribed in the context of resistance to ceftaroline or ceftobiprole, were detected in multiple selections. These genomic findings suggest that non-mecA mechanisms, while yet to be encountered in the clinical setting, may also be important in mediating resistance to 5th-generation cephalosporins.
Collapse
Affiliation(s)
- Alexander L. Greninger
- Department of Laboratory Medicine, University of California San Francisco, San Francisco, California, United States of America
- UCSF-Abbott Viral Diagnostics and Discovery Center, San Francisco, California, United States of America
| | - Som S. Chatterjee
- Division of Infectious Diseases, Department of Medicine, San Francisco General Hospital, San Francisco, California, United States of America
| | - Liana C. Chan
- Division of Infectious Diseases, Department of Medicine, San Francisco General Hospital, San Francisco, California, United States of America
| | - Stephanie M. Hamilton
- Division of Infectious Diseases, Department of Medicine, San Francisco General Hospital, San Francisco, California, United States of America
| | - Henry F. Chambers
- Division of Infectious Diseases, Department of Medicine, San Francisco General Hospital, San Francisco, California, United States of America
| | - Charles Y. Chiu
- Department of Laboratory Medicine, University of California San Francisco, San Francisco, California, United States of America
- UCSF-Abbott Viral Diagnostics and Discovery Center, San Francisco, California, United States of America
| |
Collapse
|
42
|
The Staphylococcus aureus Chaperone PrsA Is a New Auxiliary Factor of Oxacillin Resistance Affecting Penicillin-Binding Protein 2A. Antimicrob Agents Chemother 2015; 60:1656-66. [PMID: 26711778 DOI: 10.1128/aac.02333-15] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2015] [Accepted: 12/15/2015] [Indexed: 12/17/2022] Open
Abstract
Expression of the methicillin-resistant S. aureus (MRSA) phenotype results from the expression of the extra penicillin-binding protein 2A (PBP2A), which is encoded by mecA and acquired horizontally on part of the SCCmec cassette. PBP2A can catalyze dd-transpeptidation of peptidoglycan (PG) because of its low affinity for β-lactam antibiotics and can functionally cooperate with the PBP2 transglycosylase in the biosynthesis of PG. Here, we focus upon the role of the membrane-bound PrsA foldase protein as a regulator of β-lactam resistance expression. Deletion of prsA altered oxacillin resistance in three different SCCmec backgrounds and, more importantly, caused a decrease in PBP2A membrane amounts without affecting mecA mRNA levels. The N- and C-terminal domains of PrsA were found to be critical features for PBP2A protein membrane levels and oxacillin resistance. We propose that PrsA has a role in posttranscriptional maturation of PBP2A, possibly in the export and/or folding of newly synthesized PBP2A. This additional level of control in the expression of the mecA-dependent MRSA phenotype constitutes an opportunity to expand the strategies to design anti-infective agents.
Collapse
|
43
|
Distinctive Binding of Avibactam to Penicillin-Binding Proteins of Gram-Negative and Gram-Positive Bacteria. Antimicrob Agents Chemother 2015; 60:752-6. [PMID: 26574008 PMCID: PMC4750707 DOI: 10.1128/aac.02102-15] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2015] [Accepted: 11/11/2015] [Indexed: 11/21/2022] Open
Abstract
Avibactam is a novel non-β-lactam β-lactamase inhibitor that covalently acylates a variety of β-lactamases, causing inhibition. Although avibactam presents limited antibacterial activity, its acylation ability toward bacterial penicillin-binding proteins (PBPs) was investigated. Staphylococcus aureus was of particular interest due to the reported β-lactamase activity of PBP4. The binding of avibactam to PBPs was measured by adding increasing concentrations to membrane preparations of a variety of Gram-positive and Gram-negative bacteria prior to addition of the fluorescent reagent Bocillin FL. Relative binding (measured here as the 50% inhibitory concentration [IC50]) to PBPs was estimated by quantification of fluorescence after gel electrophoresis. Avibactam was found to selectively bind to some PBPs. In Escherichia coli, Pseudomonas aeruginosa, Haemophilus influenzae, and S. aureus, avibactam primarily bound to PBP2, with IC50s of 0.92, 1.1, 3.0, and 51 μg/ml, respectively, whereas binding to PBP3 was observed in Streptococcus pneumoniae (IC50, 8.1 μg/ml). Interestingly, avibactam was able to significantly enhance labeling of S. aureus PBP4 by Bocillin FL. In PBP competition assays with S. aureus, where avibactam was used at a fixed concentration in combination with varied amounts of ceftazidime, the apparent IC50 of ceftazidime was found to be very similar to that determined for ceftazidime when used alone. In conclusion, avibactam is able to covalently bind to some bacterial PBPs. Identification of those PBP targets may allow the development of new diazabicyclooctane derivatives with improved affinity for PBPs or new combination therapies that act on multiple PBP targets.
Collapse
|
44
|
Ashraf Z, Bais A, Manir MM, Niazi U. Novel Penicillin Analogues as Potential Antimicrobial Agents; Design, Synthesis and Docking Studies. PLoS One 2015; 10:e0135293. [PMID: 26267242 PMCID: PMC4534092 DOI: 10.1371/journal.pone.0135293] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2015] [Accepted: 07/20/2015] [Indexed: 11/27/2022] Open
Abstract
A number of penicillin derivatives (4a-h) were synthesized by the condensation of 6-amino penicillinic acid (6-APA) with non-steroidal anti-inflammatory drugs as antimicrobial agents. In silico docking study of these analogues was performed against Penicillin Binding Protein (PDBID 1CEF) using AutoDock Tools 1.5.6 in order to investigate the antimicrobial data on structural basis. Penicillin binding proteins function as either transpeptidases or carboxypeptidases and in few cases demonstrate transglycosylase activity in bacteria. The excellent antibacterial potential was depicted by compounds 4c and 4e against Escherichia coli, Staphylococcus epidermidus and Staphylococcus aureus compared to the standard amoxicillin. The most potent penicillin derivative 4e exhibited same activity as standard amoxicillin against S. aureus. In the enzyme inhibitory assay the compound 4e inhibited E. coli MurC with an IC50 value of 12.5 μM. The docking scores of these compounds 4c and 4e also verified their greater antibacterial potential. The results verified the importance of side chain functionalities along with the presence of central penam nucleus. The binding affinities calculated from docking results expressed in the form of binding energies ranges from -7.8 to -9.2kcal/mol. The carboxylic group of penam nucleus in all these compounds is responsible for strong binding with receptor protein with the bond length ranges from 3.4 to 4.4 Ǻ. The results of present work ratify that derivatives 4c and 4e may serve as a structural template for the design and development of potent antimicrobial agents.
Collapse
Affiliation(s)
- Zaman Ashraf
- Department of Chemistry, Allama Iqbal Open University, H-8, Islamabad, Pakistan
- Department of Biology, College of Natural Sciences, Kongju National University, Kongju, South Korea
- * E-mail:
| | - Abdul Bais
- Department of Chemistry, Allama Iqbal Open University, H-8, Islamabad, Pakistan
| | | | - Umar Niazi
- Atta ur Rehman School of Modeling and Simulation, National University of Science and Technology, Islamabad, Pakistan
| |
Collapse
|
45
|
Gautam S, Kim T, Spiegel DA. Chemical probes reveal an extraseptal mode of cross-linking in Staphylococcus aureus. J Am Chem Soc 2015; 137:7441-7. [PMID: 26035224 DOI: 10.1021/jacs.5b02972] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Staphylococcus aureus is an important human pathogen and a model organism for studying cell wall synthesis in Gram-positive cocci. The prevailing model of cell wall biogenesis in cocci holds that peptidoglycan synthesis (i.e., transglycosylation and cross-linking) is restricted spatially to the septal cross-wall and temporally to cell division. Previously, we developed a method for visualizing cross-linking in S. aureus using fluorescently tagged mimics of the endogenous substrate of penicillin-binding proteins (PBPs). These probes are incorporated into the cell wall of S. aureus specifically by PBP4, allowing localization of the enzyme's cross-linking activity in vivo with precise spatial and temporal resolution. Here, using this methodology, we have discovered that PBP4 is active not only at the septum, but unexpectedly at the peripheral wall as well. These results challenge the long-held belief that peptidoglycan synthesis is restricted to the septum in spherical bacteria, and instead indicate the presence of two spatiotemporally distinct modes of cross-linking in S. aureus: one at the septum during cell division, and another at the peripheral wall between divisions.
Collapse
Affiliation(s)
- Samir Gautam
- §Department of Chemistry, Yale University, 225 Prospect Street, New Haven, Connecticut 06511, United States
| | - Taehan Kim
- §Department of Chemistry, Yale University, 225 Prospect Street, New Haven, Connecticut 06511, United States
| | - David A Spiegel
- §Department of Chemistry, Yale University, 225 Prospect Street, New Haven, Connecticut 06511, United States
| |
Collapse
|
46
|
Weichenberger CX, Afonine PV, Kantardjieff K, Rupp B. The solvent component of macromolecular crystals. ACTA CRYSTALLOGRAPHICA. SECTION D, BIOLOGICAL CRYSTALLOGRAPHY 2015; 71:1023-38. [PMID: 25945568 PMCID: PMC4427195 DOI: 10.1107/s1399004715006045] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/05/2014] [Accepted: 03/25/2015] [Indexed: 11/10/2022]
Abstract
The mother liquor from which a biomolecular crystal is grown will contain water, buffer molecules, native ligands and cofactors, crystallization precipitants and additives, various metal ions, and often small-molecule ligands or inhibitors. On average, about half the volume of a biomolecular crystal consists of this mother liquor, whose components form the disordered bulk solvent. Its scattering contributions can be exploited in initial phasing and must be included in crystal structure refinement as a bulk-solvent model. Concomitantly, distinct electron density originating from ordered solvent components must be correctly identified and represented as part of the atomic crystal structure model. Herein, are reviewed (i) probabilistic bulk-solvent content estimates, (ii) the use of bulk-solvent density modification in phase improvement, (iii) bulk-solvent models and refinement of bulk-solvent contributions and (iv) modelling and validation of ordered solvent constituents. A brief summary is provided of current tools for bulk-solvent analysis and refinement, as well as of modelling, refinement and analysis of ordered solvent components, including small-molecule ligands.
Collapse
Affiliation(s)
- Christian X. Weichenberger
- Center for Biomedicine, European Academy of Bozen/Bolzano (EURAC), Viale Druso 1, Bozen/Bolzano, I-39100 Südtirol/Alto Adige, Italy
| | - Pavel V. Afonine
- Physical Biosciences Division, Lawrence Berkeley National Laboratory (LBNL), 1 Cyclotron Road, Mail Stop 64R0121, Berkeley, CA 94720, USA
| | - Katherine Kantardjieff
- College of Science and Mathematics, California State University, San Marcos, CA 92078, USA
| | - Bernhard Rupp
- Department of Forensic Crystallography, k.-k. Hofkristallamt, 991 Audrey Place, Vista, CA 92084, USA
- Department of Genetic Epidemiology, Medical University of Innsbruck, Schöpfstrasse 41, A-6020 Innsbruck, Austria
| |
Collapse
|
47
|
Bansal A, Kar D, Murugan RA, Mallick S, Dutta M, Pandey SD, Chowdhury C, Ghosh AS. A putative low-molecular-mass penicillin-binding protein (PBP) of Mycobacterium smegmatis exhibits prominent physiological characteristics of DD-carboxypeptidase and beta-lactamase. MICROBIOLOGY-SGM 2015; 161:1081-1091. [PMID: 25750082 DOI: 10.1099/mic.0.000074] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/11/2014] [Accepted: 03/04/2015] [Indexed: 11/18/2022]
Abstract
DD-carboxypeptidases (DD-CPases) are low-molecular-mass (LMM) penicillin-binding proteins (PBPs) that are mainly involved in peptidoglycan remodelling, but little is known about the dd-CPases of mycobacteria. In this study, a putative DD-CPase of Mycobacterium smegmatis, MSMEG_2433 is characterized. The gene for the membrane-bound form of MSMEG_2433 was cloned and expressed in Escherichia coli in its active form, as revealed by its ability to bind to the Bocillin-FL (fluorescent penicillin). Interestingly, in vivo expression of MSMEG_2433 could restore the cell shape oddities of the septuple PBP mutant of E. coli, which was a prominent physiological characteristic of DD-CPases. Moreover, expression of MSMEG_2433 in trans elevated beta-lactam resistance in PBP deletion mutants (ΔdacAdacC) of E. coli, strengthening its physiology as a dd-CPase. To confirm the biochemical reason behind such physiological behaviours, a soluble form of MSMEG_2433 (sMSMEG_2433) was created, expressed and purified. In agreement with the observed physiological phenomena, sMSMEG_2433 exhibited DD-CPase activity against artificial and peptidoglycan-mimetic DD-CPase substrates. To our surprise, enzymic analyses of MSMEG_2433 revealed efficient deacylation for beta-lactam substrates at physiological pH, which is a unique characteristic of beta-lactamases. In addition to the MSMEG_2433 active site that favours dd-CPase activity, in silico analyses also predicted the presence of an omega-loop-like region in MSMEG_2433, which is an important determinant of its beta-lactamase activity. Based on the in vitro, in vivo and in silico studies, we conclude that MSMEG_2433 is a dual enzyme, possessing both DD-CPase and beta-lactamase activities.
Collapse
Affiliation(s)
- Ankita Bansal
- Department of Biotechnology, Indian Institute of Technology, Kharagpur, West Bengal PIN-721302, India
| | - Debasish Kar
- Department of Biotechnology, Indian Institute of Technology, Kharagpur, West Bengal PIN-721302, India
| | - Rajagopal A Murugan
- Department of Biotechnology, Indian Institute of Technology, Kharagpur, West Bengal PIN-721302, India
| | - Sathi Mallick
- Department of Biotechnology, Indian Institute of Technology, Kharagpur, West Bengal PIN-721302, India
| | - Mouparna Dutta
- Department of Biotechnology, Indian Institute of Technology, Kharagpur, West Bengal PIN-721302, India
| | - Satya Deo Pandey
- Department of Biotechnology, Indian Institute of Technology, Kharagpur, West Bengal PIN-721302, India
| | - Chiranjit Chowdhury
- Department of Biotechnology, Indian Institute of Technology, Kharagpur, West Bengal PIN-721302, India
| | - Anindya S Ghosh
- Department of Biotechnology, Indian Institute of Technology, Kharagpur, West Bengal PIN-721302, India
| |
Collapse
|
48
|
Qiao Y, Lebar MD, Schirner K, Schaefer K, Tsukamoto H, Kahne D, Walker S. Detection of lipid-linked peptidoglycan precursors by exploiting an unexpected transpeptidase reaction. J Am Chem Soc 2014; 136:14678-81. [PMID: 25291014 PMCID: PMC4210121 DOI: 10.1021/ja508147s] [Citation(s) in RCA: 85] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
![]()
Penicillin-binding
proteins (PBPs) are involved in the synthesis
and remodeling of bacterial peptidoglycan (PG). Staphylococcus
aureus expresses four PBPs. Genetic studies in S.
aureus have implicated PBP4 in the formation of highly cross-linked
PG, but biochemical studies have not reached a consensus on its primary
enzymatic activity. Using synthetic Lipid II, we show here that PBP4
preferentially acts as a transpeptidase (TP) in vitro. Moreover, it is the PBP primarily responsible for incorporating
exogenous d-amino acids into cellular PG, implying that it
also has TP activity in vivo. Notably, PBP4 efficiently
exchanges d-amino acids not only into PG polymers but also
into the PG monomers Lipid I and Lipid II. This is the first demonstration
that any TP domain of a PBP can activate the PG monomer building blocks.
Exploiting the promiscuous TP activity of PBP4, we developed a simple,
highly sensitive assay to detect cellular pools of lipid-linked PG
precursors, which are of notoriously low abundance. This method, which
addresses a longstanding problem, is useful for assessing how genetic
and pharmacological perturbations affect precursor levels, and may
facilitate studies to elucidate antibiotic mechanism of action.
Collapse
Affiliation(s)
- Yuan Qiao
- Chemical Biology Program, Harvard University , Cambridge, Massachusetts 02138, United States
| | | | | | | | | | | | | |
Collapse
|
49
|
Pournajaf A, Ardebili A, Goudarzi L, Khodabandeh M, Narimani T, Abbaszadeh H. PCR-based identification of methicillin-resistant Staphylococcus aureus strains and their antibiotic resistance profiles. Asian Pac J Trop Biomed 2014; 4:S293-7. [PMID: 25183100 DOI: 10.12980/apjtb.4.2014c423] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2014] [Accepted: 02/19/2014] [Indexed: 10/25/2022] Open
Abstract
OBJECTIVE To evaluated the PCR for mecA gene compared with the conventional oxacillin disk diffusion method for methicillin-resistant Staphylococcus aureus (S. aureus) identification. METHODS A total of 292 S. aureus strains were isolated from various clinical specimens obtained from hospitalized patients. Susceptibility test to several antimicrobial agents was performed by disk diffusion agar according to Clinical and Laboratory Standards Institute guidelines. The PCR amplification of the mecA gene was carried out in all the clinical isolates. RESULTS Among antibiotics used in our study, penicillin showed the least anti-staphylococcal activity and vancomycin was the most effective. The rate of methicillin-resistant S. aureus prevalence determined by oxacillin disk diffusion method was 47.6%; whereas, 45.1% of S. aureus isolates were mecA- positive in the PCR assay. CONCLUSIONS This study is suggestive that the PCR for detection of mecA gene is a fast, accurate and valuable diagnostic tool, particularly in hospitals in areas where methicillin-resistant S. aureus is endemic.
Collapse
Affiliation(s)
- Abazar Pournajaf
- Department of Microbiology, Faculty of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Abdollah Ardebili
- Department of Microbiology, Faculty of Medicine, Tehran University of Medical Sciences, Tehran, Iran ; Department of Microbiology, Faculty of Medicine, Golestan University of Medical Sciences, Gorgan, Iran
| | - Leyla Goudarzi
- Department of Microbiology, Faculty of Medicine, Shahid Beheshti University of Medical Science, Tehran, Iran
| | - Mahmoud Khodabandeh
- Department of Pediatric Infectious Disease, Faculty of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Tahmineh Narimani
- Department of Microbiology, Faculty of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Hassan Abbaszadeh
- Department of Pharmacology, Faculty of Medicine, Iran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
50
|
Spanu V, Scarano C, Cossu F, Pala C, Spanu C, De Santis EPL. Antibiotic Resistance Traits and Molecular Subtyping ofStaphylococcus aureusIsolated from Raw Sheep Milk Cheese. J Food Sci 2014; 79:M2066-71. [DOI: 10.1111/1750-3841.12590] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2014] [Accepted: 07/24/2014] [Indexed: 12/16/2022]
Affiliation(s)
- Vincenzo Spanu
- Dept. of Veterinary Medicine; Univ. of Sassari; Via Vienna 2 07100 Sassari Italy
| | - Christian Scarano
- Dept. of Veterinary Medicine; Univ. of Sassari; Via Vienna 2 07100 Sassari Italy
| | - Francesca Cossu
- Dept. of Veterinary Medicine; Univ. of Sassari; Via Vienna 2 07100 Sassari Italy
| | - Carlo Pala
- Dept. of Veterinary Medicine; Univ. of Sassari; Via Vienna 2 07100 Sassari Italy
| | - Carlo Spanu
- Dept. of Veterinary Medicine; Univ. of Sassari; Via Vienna 2 07100 Sassari Italy
| | | |
Collapse
|