1
|
Islam MR, Azmal M, Prima FS, Zaman B, Hossain MM, Mishu MA, Ghosh A. Retention of methicillin susceptibility in Staphylococcus aureus using natural adjuvant as an allosteric modifier of penicillin-binding protein 2a. Comput Biol Med 2024; 181:109070. [PMID: 39205340 DOI: 10.1016/j.compbiomed.2024.109070] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Revised: 08/21/2024] [Accepted: 08/22/2024] [Indexed: 09/04/2024]
Abstract
The emergence of methicillin-resistant Staphylococcus aureus (MRSA) poses a significant global public health challenge due to its resistance to conventional antibiotics, primarily mediated by the mutated penicillin-binding protein, PBP2a. This study aims to investigate the potential of phytochemicals derived from medicinal plants in the Indian subcontinent to serve as adjuvants, enhancing the efficacy of methicillin against MRSA through allosteric modification of PBP2a using molecular docking and molecular dynamics (MD) simulation. After comprehensive Absorption, Distribution, Metabolism, and Excretion (ADME) profiling, along with AMES and hepatotoxicity tests, 9 compounds were shortlisted as suitable adjuvant candidates. Among them, nimbolide, quercetin, emodin, daidzein, eriodictyol, luteolin, and apigenin exhibited strong binding affinity to the allosteric site of PBP2a, with docking scores ranging from -8.7 to -7.3 kcal/mol. These phytochemicals facilitated enhanced methicillin binding, as evidenced by improved docking scores ranging from -6.1 to -6.8 kcal/mol, compared to -5.6 kcal/mol for methicillin alone. Molecular dynamics simulations confirmed the stability and favorable conformations of phytochemical-PBP2a complexes. Quercetin and daidzein were identified as the most promising adjuvant candidates, forming stable and energetically favorable complexes with PBP2a. Experimental validation showed that quercetin, at 30 mg/mL, effectively retained methicillin's antibacterial efficacy against MRSA. This study underscores the potential of natural compounds in overcoming antibiotic resistance and suggests that phytochemical-antibiotic synergism could be a viable strategy to combat multidrug-resistant bacterial infections.
Collapse
Affiliation(s)
- Md Rubiath Islam
- Department of Biochemistry and Molecular Biology, Shahjalal University of Science and Technology, Sylhet, 3114, Bangladesh
| | - Mahir Azmal
- Department of Biochemistry and Molecular Biology, Shahjalal University of Science and Technology, Sylhet, 3114, Bangladesh
| | - Fatema Sultana Prima
- Department of Biochemistry and Molecular Biology, Shahjalal University of Science and Technology, Sylhet, 3114, Bangladesh
| | - Bushra Zaman
- Department of Biochemistry and Molecular Biology, Shahjalal University of Science and Technology, Sylhet, 3114, Bangladesh
| | - Md Muluk Hossain
- Department of Biochemistry and Molecular Biology, Shahjalal University of Science and Technology, Sylhet, 3114, Bangladesh
| | - Moshiul Alam Mishu
- Department of Biochemistry and Molecular Biology, Shahjalal University of Science and Technology, Sylhet, 3114, Bangladesh
| | - Ajit Ghosh
- Department of Biochemistry and Molecular Biology, Shahjalal University of Science and Technology, Sylhet, 3114, Bangladesh.
| |
Collapse
|
2
|
Nguyen VT, Birhanu BT, Miguel-Ruano V, Kim C, Batuecas M, Yang J, El-Araby AM, Jiménez-Faraco E, Schroeder VA, Alba A, Rana N, Sader S, Thomas CA, Feltzer R, Lee M, Fisher JF, Hermoso JA, Chang M, Mobashery S. Restoring susceptibility to β-lactam antibiotics in methicillin-resistant Staphylococcus aureus. Nat Chem Biol 2024:10.1038/s41589-024-01688-0. [PMID: 39060390 DOI: 10.1038/s41589-024-01688-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Accepted: 06/26/2024] [Indexed: 07/28/2024]
Abstract
Infections by Staphylococcus aureus have been treated historically with β-lactam antibiotics. However, these antibiotics have become obsolete in methicillin-resistant S. aureus by acquisition of the bla and mec operons. The presence of the β-lactam antibiotic is detected by the sensor domains of BlaR and/or MecR, and the information is transmitted to the cytoplasm, resulting in derepression of the antibiotic-resistance genes. We hypothesized that inhibition of the sensor domain would shut down this response system, and β-lactam susceptibility would be restored. An in silico search of 11 million compounds led to a benzimidazole-based hit and, ultimately, to the boronate 4. The X-ray structure of 4 is covalently engaged with the active-site serine of BlaR. Compound 4 potentiates by 16- to 4,096-fold the activities of oxacillin and of meropenem against methicillin-resistant S. aureus strains. The combination of 4 with oxacillin or meropenem shows efficacy in infected mice, validating the strategy.
Collapse
Affiliation(s)
- Van T Nguyen
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, IN, USA
| | - Biruk T Birhanu
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, IN, USA
| | - Vega Miguel-Ruano
- Department of Crystallography and Structural Biology, Instituto de Química-Física 'Blas Cabrera', Consejo Superior de Investigaciones Científicas, Madrid, Spain
| | - Choon Kim
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, IN, USA
| | - Mayte Batuecas
- Department of Crystallography and Structural Biology, Instituto de Química-Física 'Blas Cabrera', Consejo Superior de Investigaciones Científicas, Madrid, Spain
| | - Jingdong Yang
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, IN, USA
| | - Amr M El-Araby
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, IN, USA
| | - Eva Jiménez-Faraco
- Department of Crystallography and Structural Biology, Instituto de Química-Física 'Blas Cabrera', Consejo Superior de Investigaciones Científicas, Madrid, Spain
| | - Valerie A Schroeder
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, IN, USA
| | - Alejandra Alba
- Department of Crystallography and Structural Biology, Instituto de Química-Física 'Blas Cabrera', Consejo Superior de Investigaciones Científicas, Madrid, Spain
| | - Neha Rana
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, IN, USA
| | - Safaa Sader
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, IN, USA
| | - Caitlyn A Thomas
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, IN, USA
| | - Rhona Feltzer
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, IN, USA
| | - Mijoon Lee
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, IN, USA
| | - Jed F Fisher
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, IN, USA
| | - Juan A Hermoso
- Department of Crystallography and Structural Biology, Instituto de Química-Física 'Blas Cabrera', Consejo Superior de Investigaciones Científicas, Madrid, Spain
| | - Mayland Chang
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, IN, USA
| | - Shahriar Mobashery
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, IN, USA.
| |
Collapse
|
3
|
Watanabe S, Nsofor CA, Thitiananpakorn K, Tan XE, Aiba Y, Takenouchi R, Kiga K, Sasahara T, Miyanaga K, Veeranarayanan S, Shimamori Y, Lian AYS, Nguyen TM, Nguyen HM, Alessa O, Kumwenda GP, Jayathilake S, Revilleza JEC, Baranwal P, Nishikawa Y, Li FY, Kawaguchi T, Sankaranarayanan S, Arbaah M, Zhang Y, Maniruzzaman, Liu Y, Sarah H, Li J, Sugano T, Ho TMD, Batbold A, Nayanjin T, Cui L. Metabolic remodeling by RNA polymerase gene mutations is associated with reduced β-lactam susceptibility in oxacillin-susceptible MRSA. mBio 2024; 15:e0033924. [PMID: 38988221 PMCID: PMC11237739 DOI: 10.1128/mbio.00339-24] [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: 02/01/2024] [Accepted: 03/27/2024] [Indexed: 07/12/2024] Open
Abstract
The emergence of oxacillin-susceptible methicillin-resistant Staphylococcus aureus (OS-MRSA) has imposed further challenges to the clinical management of MRSA infections. When exposed to β-lactam antibiotics, these strains can easily acquire reduced β-lactam susceptibility through chromosomal mutations, including those in RNA polymerase (RNAP) genes such as rpoBC, which may then lead to treatment failure. Despite the increasing prevalence of such strains and the apparent challenges they pose for diagnosis and treatment, there is limited information available on the actual mechanisms underlying such chromosomal mutation-related transitions to reduced β-lactam susceptibility, as it does not directly associate with the expression of mecA. This study investigated the cellular physiology and metabolism of six missense mutants with reduced oxacillin susceptibility, each carrying respective mutations on RpoBH929P, RpoBQ645H, RpoCG950R, RpoCG498D, RpiAA64E, and FruBA211E, using capillary electrophoresis-mass spectrometry-based metabolomics analysis. Our results showed that rpoBC mutations caused RNAP transcription dysfunction, leading to an intracellular accumulation of ribonucleotides. These mutations also led to the accumulation of UDP-Glc/Gal and UDP-GlcNAc, which are precursors of UTP-associated peptidoglycan and wall teichoic acid. Excessive amounts of building blocks then contributed to the cell wall thickening of mutant strains, as observed in transmission electron microscopy, and ultimately resulted in decreased susceptibility to β-lactam in OS-MRSA. IMPORTANCE The emergence of oxacillin-susceptible methicillin-resistant Staphylococcus aureus (OS-MRSA) strains has created new challenges for treating MRSA infections. These strains can become resistant to β-lactam antibiotics through chromosomal mutations, including those in the RNA polymerase (RNAP) genes such as rpoBC, leading to treatment failure. This study investigated the mechanisms underlying reduced β-lactam susceptibility in four rpoBC mutants of OS-MRSA. The results showed that rpoBC mutations caused RNAP transcription dysfunction, leading to an intracellular accumulation of ribonucleotides and precursors of peptidoglycan as well as wall teichoic acid. This, in turn, caused thickening of the cell wall and ultimately resulted in decreased susceptibility to β-lactam in OS-MRSA. These findings provide insights into the mechanisms of antibiotic resistance in OS-MRSA and highlight the importance of continued research in developing effective treatments to combat antibiotic resistance.
Collapse
Affiliation(s)
- Shinya Watanabe
- Division of Bacteriology, Department of Infection and Immunity, Jichi Medical University, Tochigi, Japan
| | - Chijioke A Nsofor
- Division of Bacteriology, Department of Infection and Immunity, Jichi Medical University, Tochigi, Japan
- Department of Biotechnology, School of Biological Sciences, Federal University of Technology Owerri Nigeria, Owerri, Nigeria
| | - Kanate Thitiananpakorn
- Division of Bacteriology, Department of Infection and Immunity, Jichi Medical University, Tochigi, Japan
| | - Xin-Ee Tan
- Division of Bacteriology, Department of Infection and Immunity, Jichi Medical University, Tochigi, Japan
| | - Yoshifumi Aiba
- Division of Bacteriology, Department of Infection and Immunity, Jichi Medical University, Tochigi, Japan
| | - Remi Takenouchi
- School of Medicine, Jichi Medical University, Tochigi, Japan
| | - Kotaro Kiga
- Division of Bacteriology, Department of Infection and Immunity, Jichi Medical University, Tochigi, Japan
- Research Center for Drug and Vaccine Development, National Institute of Infectious Diseases, Tokyo, Japan
| | - Teppei Sasahara
- Division of Bacteriology, Department of Infection and Immunity, Jichi Medical University, Tochigi, Japan
| | - Kazuhiko Miyanaga
- Division of Bacteriology, Department of Infection and Immunity, Jichi Medical University, Tochigi, Japan
| | - Srivani Veeranarayanan
- Division of Bacteriology, Department of Infection and Immunity, Jichi Medical University, Tochigi, Japan
| | - Yuzuki Shimamori
- Division of Bacteriology, Department of Infection and Immunity, Jichi Medical University, Tochigi, Japan
| | - Adeline Yeo Syin Lian
- Division of Bacteriology, Department of Infection and Immunity, Jichi Medical University, Tochigi, Japan
| | - Thuy Minh Nguyen
- Division of Bacteriology, Department of Infection and Immunity, Jichi Medical University, Tochigi, Japan
| | - Huong Minh Nguyen
- Division of Bacteriology, Department of Infection and Immunity, Jichi Medical University, Tochigi, Japan
| | - Ola Alessa
- Division of Bacteriology, Department of Infection and Immunity, Jichi Medical University, Tochigi, Japan
| | | | - Sarangi Jayathilake
- Division of Bacteriology, Department of Infection and Immunity, Jichi Medical University, Tochigi, Japan
| | | | - Priyanka Baranwal
- Division of Bacteriology, Department of Infection and Immunity, Jichi Medical University, Tochigi, Japan
| | - Yutaro Nishikawa
- Division of Bacteriology, Department of Infection and Immunity, Jichi Medical University, Tochigi, Japan
| | - Feng-Yu Li
- Division of Bacteriology, Department of Infection and Immunity, Jichi Medical University, Tochigi, Japan
| | - Tomofumi Kawaguchi
- Division of Bacteriology, Department of Infection and Immunity, Jichi Medical University, Tochigi, Japan
| | - Sowmiya Sankaranarayanan
- Division of Bacteriology, Department of Infection and Immunity, Jichi Medical University, Tochigi, Japan
| | - Mahmoud Arbaah
- Division of Bacteriology, Department of Infection and Immunity, Jichi Medical University, Tochigi, Japan
| | - Yuancheng Zhang
- Division of Bacteriology, Department of Infection and Immunity, Jichi Medical University, Tochigi, Japan
| | - Maniruzzaman
- Division of Bacteriology, Department of Infection and Immunity, Jichi Medical University, Tochigi, Japan
| | - Yi Liu
- Division of Bacteriology, Department of Infection and Immunity, Jichi Medical University, Tochigi, Japan
| | - Hossain Sarah
- Division of Bacteriology, Department of Infection and Immunity, Jichi Medical University, Tochigi, Japan
| | - Junjie Li
- Division of Bacteriology, Department of Infection and Immunity, Jichi Medical University, Tochigi, Japan
| | - Takashi Sugano
- Division of Bacteriology, Department of Infection and Immunity, Jichi Medical University, Tochigi, Japan
| | - Thi My Duyen Ho
- Division of Bacteriology, Department of Infection and Immunity, Jichi Medical University, Tochigi, Japan
| | - Anujin Batbold
- Division of Bacteriology, Department of Infection and Immunity, Jichi Medical University, Tochigi, Japan
| | - Tergel Nayanjin
- Division of Bacteriology, Department of Infection and Immunity, Jichi Medical University, Tochigi, Japan
| | - Longzhu Cui
- Division of Bacteriology, Department of Infection and Immunity, Jichi Medical University, Tochigi, Japan
| |
Collapse
|
4
|
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: 7] [Impact Index Per Article: 7.0] [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
|
5
|
Nolan AC, Zeden MS, Kviatkovski I, Campbell C, Urwin L, Corrigan RM, Gründling A, O’Gara JP. Purine Nucleosides Interfere with c-di-AMP Levels and Act as Adjuvants To Re-Sensitize MRSA To β-Lactam Antibiotics. mBio 2023; 14:e0247822. [PMID: 36507833 PMCID: PMC9973305 DOI: 10.1128/mbio.02478-22] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Accepted: 11/17/2022] [Indexed: 12/14/2022] Open
Abstract
The purine-derived signaling molecules c-di-AMP and (p)ppGpp control mecA/PBP2a-mediated β-lactam resistance in methicillin-resistant Staphylococcus aureus (MRSA) raise the possibility that purine availability can control antibiotic susceptibility. Consistent with this, exogenous guanosine and xanthosine, which are fluxed through the GTP branch of purine biosynthesis, were shown to significantly reduce MRSA β-lactam resistance. In contrast, adenosine (fluxed to ATP) significantly increased oxacillin resistance, whereas inosine (which can be fluxed to ATP and GTP via hypoxanthine) only marginally increased oxacillin susceptibility. Furthermore, mutations that interfere with de novo purine synthesis (pur operon), transport (NupG, PbuG, PbuX) and the salvage pathway (DeoD2, Hpt) increased β-lactam resistance in MRSA strain JE2. Increased resistance of a nupG mutant was not significantly reversed by guanosine, indicating that NupG is required for guanosine transport, which is required to reduce β-lactam resistance. Suppressor mutants resistant to oxacillin/guanosine combinations contained several purine salvage pathway mutations, including nupG and hpt. Guanosine significantly increased cell size and reduced levels of c-di-AMP, while inactivation of GdpP, the c-di-AMP phosphodiesterase negated the impact of guanosine on β-lactam susceptibility. PBP2a expression was unaffected in nupG or deoD2 mutants, suggesting that guanosine-induced β-lactam susceptibility may result from dysfunctional c-di-AMP-dependent osmoregulation. These data reveal the therapeutic potential of purine nucleosides, as β-lactam adjuvants that interfere with the normal activation of c-di-AMP are required for high-level β-lactam resistance in MRSA. IMPORTANCE The clinical burden of infections caused by antimicrobial resistant (AMR) pathogens is a leading threat to public health. Maintaining the effectiveness of existing antimicrobial drugs or finding ways to reintroduce drugs to which resistance is widespread is an important part of efforts to address the AMR crisis. Predominantly, the safest and most effective class of antibiotics are the β-lactams, which are no longer effective against methicillin-resistant Staphylococcus aureus (MRSA). Here, we report that the purine nucleosides guanosine and xanthosine have potent activity as adjuvants that can resensitize MRSA to oxacillin and other β-lactam antibiotics. Mechanistically, exposure of MRSA to these nucleosides significantly reduced the levels of the cyclic dinucleotide c-di-AMP, which is required for β-lactam resistance. Drugs derived from nucleotides are widely used in the treatment of cancer and viral infections highlighting the clinical potential of using purine nucleosides to restore or enhance the therapeutic effectiveness of β-lactams against MRSA and potentially other AMR pathogens.
Collapse
Affiliation(s)
- Aaron C. Nolan
- Microbiology, School of Biological and Chemical Sciences, University of Galway, Ireland
| | - Merve S. Zeden
- Microbiology, School of Biological and Chemical Sciences, University of Galway, Ireland
| | - Igor Kviatkovski
- Section of Molecular Microbiology and Medical Research Council Centre for Molecular Bacteriology and Infection, Imperial College London, London, United Kingdom
| | - Christopher Campbell
- Microbiology, School of Biological and Chemical Sciences, University of Galway, Ireland
| | - Lucy Urwin
- The Florey Institute, School of Bioscience, University of Sheffield, Sheffield, United Kingdom
| | - Rebecca M. Corrigan
- The Florey Institute, School of Bioscience, University of Sheffield, Sheffield, United Kingdom
| | - Angelika Gründling
- Section of Molecular Microbiology and Medical Research Council Centre for Molecular Bacteriology and Infection, Imperial College London, London, United Kingdom
| | - James P. O’Gara
- Microbiology, School of Biological and Chemical Sciences, University of Galway, Ireland
| |
Collapse
|
6
|
G S, K D, P S, N B. DFT calculations, molecular docking, in vitro antimicrobial and antidiabetic studies of green synthesized Schiff bases: as Covid-19 inhibitor. J Biomol Struct Dyn 2023; 41:12997-13014. [PMID: 36752337 DOI: 10.1080/07391102.2023.2175039] [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: 11/29/2022] [Accepted: 01/11/2023] [Indexed: 02/09/2023]
Abstract
In this investigation, we synthesized Schiff bases 2-(2-methoxyphenoxy)-N-(4-methylbenzylidene)ethanamine, N-(4-methoxybenzylidene)-2-(2-methoxyphenoxy)ethanamine and 2-(2-methoxyphenoxy)-N-(4-nitrobenzylidene)ethanamine from 2-(2-methoxyphenoxy)ethanamine and various aromatic aldehydes by the environmentally friendly sonication method. The B3LYP method with a 6-311++G (d, p) basis set was used in the DFT calculation to obtain the optimized structure of the Schiff base MPEA-NIT. The compounds were tested in vitro for inhibition of bacterial growth (disc well method) and inhibition of α-amylase (starch-iodine method). The compounds tested showed inhibitory activities. In addition, they were subjected to PASS analysis, drug likeness, and bioactivity score predictions using online software. To confirm the experimental findings, molecular docking analyses of synthesized compounds on α-amylase (PDB ID: 1SMD), tRNA threonylcarbamoyladenosine (PDB ID: 5MVR), glycosyl transferase (PDB ID: 6D9T), and peptididoglycan D,D-transpeptidase (PDB ID: 6HZQ) were performed. The emergence of a new coronavirus epidemic necessitates the development of antiviral medications (SARS-CoV-2). Docking active site interactions were investigated to predict compounds' activity against COVID-19 by binding with the SARS-CoV-2 (PDB ID: 6Y84).Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Saranya G
- Department of Chemistry, Chikkaiah Naicker College, Erode, India
| | | | - Shanmugapriya P
- Department of Chemistry, KSR College of Engineering, Thiruchengode, India
| | - Bhuvaneshwari N
- Department of Chemistry, Chikkaiah Naicker College, Erode, India
| |
Collapse
|
7
|
Dina NE, Tahir MA, Bajwa SZ, Amin I, Valev VK, Zhang L. SERS-based antibiotic susceptibility testing: Towards point-of-care clinical diagnosis. Biosens Bioelectron 2023; 219:114843. [PMID: 36327563 DOI: 10.1016/j.bios.2022.114843] [Citation(s) in RCA: 17] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Revised: 08/09/2022] [Accepted: 10/19/2022] [Indexed: 11/06/2022]
Abstract
Emerging antibiotic resistant bacteria constitute one of the biggest threats to public health. Surface-enhanced Raman scattering (SERS) is highly promising for detecting such bacteria and for antibiotic susceptibility testing (AST). SERS is fast, non-destructive (can probe living cells) and it is technologically flexible (readily integrated with robotics and machine learning algorithms). However, in order to integrate into efficient point-of-care (PoC) devices and to effectively replace the current culture-based methods, it needs to overcome the challenges of reliability, cost and complexity. Recently, significant progress has been made with the emergence of both new questions and new promising directions of research and technological development. This article brings together insights from several representative SERS-based AST studies and approaches oriented towards clinical PoC biosensing. It aims to serve as a reference source that can guide progress towards PoC routines for identifying antibiotic resistant pathogens. In turn, such identification would help to trace the origin of sporadic infections, in order to prevent outbreaks and to design effective medical treatment and preventive procedures.
Collapse
Affiliation(s)
- Nicoleta Elena Dina
- Department of Molecular and Biomolecular Department, National Institute for Research and Development of Isotopic and Molecular Technologies, 400293, Cluj-Napoca, Romania.
| | - Muhammad Ali Tahir
- Shanghai Key Laboratory of Atmospheric Particle Pollution and Prevention, Department of Environmental Science & Engineering, Fudan University, Shanghai, 200433, People's Republic of China
| | - Sadia Z Bajwa
- National Institute for Biotechnology and Genetic Engineering (NIBGE), P.O. Box No. 577, Jhang Road, 38000, Faisalabad, Pakistan
| | - Imran Amin
- National Institute for Biotechnology and Genetic Engineering (NIBGE), P.O. Box No. 577, Jhang Road, 38000, Faisalabad, Pakistan
| | - Ventsislav K Valev
- Centre for Photonics and Photonic Materials, Department of Physics, University of Bath, Bath, BA2 7AY, United Kingdom; Centre for Therapeutic Innovation, University of Bath, Bath, United Kingdom; Centre for Nanoscience and Nanotechnology, University of Bath, Bath, United Kingdom.
| | - Liwu Zhang
- Shanghai Key Laboratory of Atmospheric Particle Pollution and Prevention, Department of Environmental Science & Engineering, Fudan University, Shanghai, 200433, People's Republic of China; Shanghai Institute of Pollution Control and Ecological Security, Shanghai, 200092, People's Republic of China.
| |
Collapse
|
8
|
Verma AK, Ahmed SF, Hossain MS, Bhojiya AA, Mathur A, Upadhyay SK, Srivastava AK, Vishvakarma NK, Barik M, Rahaman MM, Bahadur NM. Molecular docking and simulation studies of flavonoid compounds against PBP-2a of methicillin-resistant Staphylococcus aureus. J Biomol Struct Dyn 2022; 40:10561-10577. [PMID: 34243699 DOI: 10.1080/07391102.2021.1944911] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Methicillin-Resistant Staphylococcus aureus (MRSA), a pathogenic bacterium that causes life-threatening outbreaks such as community-onset and nosocomial infections as emerging 'superbug'. Time and motion study of its virulent property developed resistance against most of the antibiotics such as Vancomycin. Thereby, to curb this problem entails the development of new therapeutic agents. Plant-derived antimicrobial agents have recently piqued people's interest, so in this research, 186 flavonoids compound selected to unmask the best candidates that can act as potent inhibitors against the Penicillin Binding Protein-2a (PBP-2a) of MRSA. Molecular docking performed using PyRx and GOLD suite to determine the binding affinities and interactions between the phytochemicals and the PBP-2a. The selected candidates strongly interact with the different amino acid residues. The 30 ns molecular dynamics (MD) simulations with five top-ranked compounds such as Naringin, Hesperidin, Neohesperidin, Didymin and Icariin validated the docking interactions. These findings are also strongly supported by root-mean-square deviation, root-mean-square fluctuation and the radius of gyration. ADME/T analysis demonstrates that these candidates appear to be safer inhibitors. Our findings point to natural flavonoids as a promising and readily available source of adjuvant antimicrobial therapy against resistant strains in the future.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Abhishek Kumar Verma
- Department of Life Sciences, Faculty of Science and Technology, Mewar University, Gangrar, Rajasthan, India
| | - Sk Faisal Ahmed
- Department of Biotechnology and Genetic Engineering, Noakhali Science and Technology University, Noakhali, Bangladesh
| | - Md Shahadat Hossain
- Department of Biotechnology and Genetic Engineering, Noakhali Science and Technology University, Noakhali, Bangladesh
| | - Ali Asger Bhojiya
- Faculty of Agriculture and Veterinary Sciences, Mewar University, Gangrar, Rajasthan, India
| | - Ankita Mathur
- Department of Life Sciences, Faculty of Science and Technology, Mewar University, Gangrar, Rajasthan, India
| | - Sudhir K Upadhyay
- Department of Environmental Sciences, V.B.S. Purvanchal University, Jaunpur, Uttar Pradesh, India
| | | | | | - Mayadhar Barik
- Department of Life Sciences, Faculty of Science and Technology, Mewar University, Gangrar, Rajasthan, India.,Research Section, Mewar University, Gangrar, Rajasthan, India
| | | | - Newaz Mohammed Bahadur
- Department of Applied Chemistry and Chemical Engineering, Noakhali Science and Technology University, Noakhali, Bangladesh
| |
Collapse
|
9
|
Identification and Characterization of a Potential Antimicrobial Peptide Isolated from Soil Brevibacillus sp. WUL10 and Its Activity against MRSA Pathogens. Trop Med Infect Dis 2022; 7:tropicalmed7060093. [PMID: 35736972 PMCID: PMC9228811 DOI: 10.3390/tropicalmed7060093] [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: 05/16/2022] [Revised: 06/01/2022] [Accepted: 06/05/2022] [Indexed: 11/17/2022] Open
Abstract
Methicillin-resistant Staphylococcus aureus (MRSA) is a severe threat to public health globally. The development of novel agents has encountered the repeated mechanism of drug resistance. This study aimed to investigate an anti-MRSA substance isolated from a promising soil bacterium. The result showed that an isolate (WUL10) was in the Brevibacillus genus. The minimum inhibitory concentration (MIC) of the purified substance was 1 µg/mL against S. aureus TISTR 517 and MRSA strains. This substance showed the bactericidal effect at the concentration of 1–2 µg/mL against these bacterial indicators. The activity of the substance retained more than 95% when encountering high temperatures and a wide range of pH, but it was sensitive to proteolytic enzymes and SDS. It was identified as a novel antimicrobial peptide (KVLVKYLGGLLKLAALMV-COOH) with the predicted structure of α-helix. The substance could rupture the cell wall of the tested pathogen. MIC and MBC of the synthesized peptide were 16 and 64 µg/mL, respectively. The difference in the activity between the isolated and synthetic peptides might be from the synergistic effects of other AMPs in the purified substance. This novel AMP would provide an advantage for further development of anti-MRSA substances to manage the situation of antibiotic resistance.
Collapse
|
10
|
The Influence of Antibiotic Resistance on Innate Immune Responses to Staphylococcus aureus Infection. Antibiotics (Basel) 2022; 11:antibiotics11050542. [PMID: 35625186 PMCID: PMC9138074 DOI: 10.3390/antibiotics11050542] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Revised: 04/13/2022] [Accepted: 04/18/2022] [Indexed: 11/16/2022] Open
Abstract
Staphylococcus aureus (S. aureus) causes a broad range of infections and is associated with significant morbidity and mortality. S. aureus produces a diverse range of cellular and extracellular factors responsible for its invasiveness and ability to resist immune attack. In recent years, increasing resistance to last-line anti-staphylococcal antibiotics daptomycin and vancomycin has been observed. Resistant strains of S. aureus are highly efficient in invading a variety of professional and nonprofessional phagocytes and are able to survive inside host cells. Eliciting immune protection against antibiotic-resistant S. aureus infection is a global challenge, requiring both innate and adaptive immune effector mechanisms. Dendritic cells (DC), which sit at the interface between innate and adaptive immune responses, are central to the induction of immune protection against S. aureus. However, it has been observed that S. aureus has the capacity to develop further antibiotic resistance and acquire increased resistance to immunological recognition by the innate immune system. In this article, we review the strategies utilised by S. aureus to circumvent antibiotic and innate immune responses, especially the interaction between S. aureus and DC, focusing on how this relationship is perturbed with the development of antibiotic resistance.
Collapse
|
11
|
Qekwana DN, Odoi A, Oguttu JW. Efficacy Profiles of Antimicrobials Evaluated against Staphylococcus Species Isolated from Canine Clinical Specimens. Animals (Basel) 2021; 11:ani11113232. [PMID: 34827963 PMCID: PMC8614345 DOI: 10.3390/ani11113232] [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: 10/10/2021] [Revised: 11/04/2021] [Accepted: 11/07/2021] [Indexed: 11/16/2022] Open
Abstract
Simple Summary Clinical cases associated with staphylococci infections are common among dogs and cats. There is evidence to suggest that staphylococci infections are increasingly becoming unresponsive to commonly used antimicrobials. This negatively impacts the ability of these infections to be treated successfully. Although resistance among these organisms has been linked to several factors, including sharing the same mechanism of action or belonging to the same group, there is evidence to suggest that cross resistance can occur between unrelated antimicrobials. The findings of this study not only confirm that antimicrobials that belong to the same group share the same mechanism of resistance and similar antimicrobial efficacy against staphylococcal infections, but also show that cross resistance occurs between unrelated antimicrobials. This should be taken into consideration when selecting antimicrobials for inclusion in the susceptibility testing panel as well as for the treatment of staphylococci infections. Abstract Cross-resistance occurs between antimicrobials with either similar mechanisms of action and/or similar chemical structures, or even between unrelated antimicrobials. This study employed a multivariate approach to investigate the associations between the efficacy profile of antimicrobials and the clustering of eleven different antimicrobial agents based on their efficacy profile. Records of the susceptibility of 382 confirmed Staphylococcus species isolates against 15 antimicrobials based on the disc diffusion method were included in this study. Tetrachoric correlation coefficients were computed to assess the correlations of antimicrobial efficacy profiles against Staphylococcus aureus. Principal components analysis and factor analysis were used to assess the clustering of antimicrobial susceptibility profiles. Strong correlations were observed among aminoglycosides, penicillins, fluroquinolones, and lincosamides. Three main factors were extracted, with Factor 1 dominated by the susceptibility profile of enrofloxacin (factor loading (FL) = 0.859), gentamicin (FL = 0.898), tylosin (FL = 0.801), and ampicillin (FL = −0.813). Factor 2, on the other hand, was dominated by the susceptibility profile of clindamycin (FL = 0.927) and lincomycin-spectinomycin (FL = 0.848) and co-trimazole (FL = −0.693). Lastly, Factor 3 was dominated by the susceptibility profile of amoxicillin-clavulanic acid (FL = 0.848) and cephalothin (FL = 0.824). Antimicrobials belonging to the same category or class of antimicrobial, tended to exhibit similar efficacy profiles, therefore, laboratories must choose only one of the antimicrobials in each group to help reduce the cost of antimicrobial susceptibility tests.
Collapse
Affiliation(s)
- Daniel Nenene Qekwana
- Section of Veterinary Public Health, Department of Paraclinical Sciences, Faculty of Veterinary Science, University of Pretoria, Pretoria 0110, South Africa
- Correspondence:
| | - Agricola Odoi
- Biomedical and Diagnostic Sciences, College of Veterinary Medicine, University of Tennessee, Knoxville, TN 37996, USA;
| | - James Wabwire Oguttu
- Department of Agriculture and Animal Health, College of Agriculture and Environmental Sciences, University of South Africa, Johannesburg 709, South Africa;
| |
Collapse
|
12
|
Bahr G, González LJ, Vila AJ. Metallo-β-lactamases in the Age of Multidrug Resistance: From Structure and Mechanism to Evolution, Dissemination, and Inhibitor Design. Chem Rev 2021; 121:7957-8094. [PMID: 34129337 PMCID: PMC9062786 DOI: 10.1021/acs.chemrev.1c00138] [Citation(s) in RCA: 106] [Impact Index Per Article: 35.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Antimicrobial resistance is one of the major problems in current practical medicine. The spread of genes coding for resistance determinants among bacteria challenges the use of approved antibiotics, narrowing the options for treatment. Resistance to carbapenems, last resort antibiotics, is a major concern. Metallo-β-lactamases (MBLs) hydrolyze carbapenems, penicillins, and cephalosporins, becoming central to this problem. These enzymes diverge with respect to serine-β-lactamases by exhibiting a different fold, active site, and catalytic features. Elucidating their catalytic mechanism has been a big challenge in the field that has limited the development of useful inhibitors. This review covers exhaustively the details of the active-site chemistries, the diversity of MBL alleles, the catalytic mechanism against different substrates, and how this information has helped developing inhibitors. We also discuss here different aspects critical to understand the success of MBLs in conferring resistance: the molecular determinants of their dissemination, their cell physiology, from the biogenesis to the processing involved in the transit to the periplasm, and the uptake of the Zn(II) ions upon metal starvation conditions, such as those encountered during an infection. In this regard, the chemical, biochemical and microbiological aspects provide an integrative view of the current knowledge of MBLs.
Collapse
Affiliation(s)
- Guillermo Bahr
- Instituto de Biología Molecular y Celular de Rosario (IBR), CONICET, Universidad Nacional de Rosario, Ocampo y Esmeralda S/N, 2000 Rosario, Argentina
- Area Biofísica, Facultad de Ciencias Bioquímicas y Farmacéuticas, Universidad Nacional de Rosario, Suipacha 531, 2000 Rosario, Argentina
| | - Lisandro J. González
- Instituto de Biología Molecular y Celular de Rosario (IBR), CONICET, Universidad Nacional de Rosario, Ocampo y Esmeralda S/N, 2000 Rosario, Argentina
- Area Biofísica, Facultad de Ciencias Bioquímicas y Farmacéuticas, Universidad Nacional de Rosario, Suipacha 531, 2000 Rosario, Argentina
| | - Alejandro J. Vila
- Instituto de Biología Molecular y Celular de Rosario (IBR), CONICET, Universidad Nacional de Rosario, Ocampo y Esmeralda S/N, 2000 Rosario, Argentina
- Area Biofísica, Facultad de Ciencias Bioquímicas y Farmacéuticas, Universidad Nacional de Rosario, Suipacha 531, 2000 Rosario, Argentina
| |
Collapse
|
13
|
K R, Kakkassery JT, Raphael VP, Johnson R, K VT. In vitro antibacterial and in silico docking studies of two Schiff bases on Staphylococcus aureus and its target proteins. FUTURE JOURNAL OF PHARMACEUTICAL SCIENCES 2021. [DOI: 10.1186/s43094-021-00225-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
Abstract
Background
Schiff base compounds have extensive applications in various fields such as analytical, inorganic, organic, and biological fields. They have excellent pharmacology application prospects in the modern era and are widely used in the pharmaceutical industry. In the present work in vitro antibacterial and in silico docking studies of two Schiff base compounds 2,2’-(5,5-dimethylcyclohexane-1,3-diylidene)bis(azan-1-yl-1-ylidene)diphenol (DmChDp) and N,N’-(5,5-dimethylcyclohexane-1,3-diylidene)dianiline (DmChDa) were carried out against the bacterial strain Staphylococcus aureus and its target proteins.
Results
The tests proved that the ligands have potential antibacterial activity. In the computational analysis, the drug-like properties of the compounds were first pre-filtered using the Lipinski rule of five. Then, molecular docking study was conducted using the AutoDock 4.2 program, to establish the mechanism by which the molecules inhibit the growth of S. aureus. For this purpose, 6 different target proteins (PDB ID: 1T2P, 3U2D, 2W9S, 1N67, 2ZCO, and 4H8E) of S. aureus were selected. Both the Schiff bases showed a good binding affinity with the target protein dihydrofolate reductase enzyme (PDB ID: 2W9S) but in different sites. Maximum binding energies of about − 10.3 and − 10.2 kcal/mol were observed when DmChDp and DmChDa were docked with 2W9S.
Conclusion
Schiff base compounds DmChDp and DmChDa have appreciable growth-inhibitory power against S. aureus, which can be attributed to the deactivation of the enzyme, dihydrofolate reductase.
Collapse
|
14
|
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: 17.3] [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
|
15
|
Ross C, Syed B, Pak J, Jhanji V, Yamaki J, Sharma A. Stability Evaluation of Extemporaneously Compounded Vancomycin Ophthalmic Drops: Effect of Solvents and Storage Conditions. Pharmaceutics 2021; 13:pharmaceutics13020289. [PMID: 33672310 PMCID: PMC7926595 DOI: 10.3390/pharmaceutics13020289] [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: 02/04/2021] [Revised: 02/11/2021] [Accepted: 02/18/2021] [Indexed: 11/16/2022] Open
Abstract
Vancomycin is the drug of choice for methicillin-resistant Staphylococcus aureus keratitis and other ocular infections. Vancomycin ophthalmic drops are not commercially available and require compounding. The present study was designed to investigate the stability of vancomycin ophthalmic drops in normal saline, phosphate-buffered saline (PBS), and balanced salt solution (BSS) while stored at room temperature or under refrigeration. Vancomycin ophthalmic drops (50 mg/mL) were aseptically prepared from commercially available intravenous powder using PBS, BSS, and saline. Solutions were stored at room temperature and in a refrigerator for 28 days. The vancomycin stability was tested by a microbiology assay and high-performance liquid chromatography HPLC analysis immediately after formulation and at days 7, 14, and 28 after storage at room temperature or under refrigeration. The pH, turbidity was also tested. Vancomycin formulations in PBS, BSS and normal saline had initial pH of 5; 5.5; 3 respectively. The formulation in PBS developed turbidity and a slight decrease in pH upon storage. Microbiological assay did not show any change in zone of inhibition with any of the formulation upon storage either at room temperature or under refrigeration. HPLC analysis did not detect any decrease in vancomycin concentration or the accumulation of degraded products in any of the formulations upon storage either at room temperature or under refrigeration. Vancomycin ophthalmic drops prepared using PBS, BSS, and normal saline were stable up to the tested time point of 28 days, irrespective of their storage temperature.
Collapse
Affiliation(s)
- Christopher Ross
- Chapman University School of Pharmacy, Chapman University Irvine, Room 267, 9401 Jeronimo Road, Irvine, CA 92618, USA; (C.R.); (B.S.); (J.P.); (J.Y.)
| | - Basir Syed
- Chapman University School of Pharmacy, Chapman University Irvine, Room 267, 9401 Jeronimo Road, Irvine, CA 92618, USA; (C.R.); (B.S.); (J.P.); (J.Y.)
| | - Joanna Pak
- Chapman University School of Pharmacy, Chapman University Irvine, Room 267, 9401 Jeronimo Road, Irvine, CA 92618, USA; (C.R.); (B.S.); (J.P.); (J.Y.)
| | - Vishal Jhanji
- Department of Ophthalmology School of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA;
| | - Jason Yamaki
- Chapman University School of Pharmacy, Chapman University Irvine, Room 267, 9401 Jeronimo Road, Irvine, CA 92618, USA; (C.R.); (B.S.); (J.P.); (J.Y.)
| | - Ajay Sharma
- Chapman University School of Pharmacy, Chapman University Irvine, Room 267, 9401 Jeronimo Road, Irvine, CA 92618, USA; (C.R.); (B.S.); (J.P.); (J.Y.)
- Correspondence: ; Tel.: +1-714-516-5498
| |
Collapse
|
16
|
Badua AT, Boonyayatra S, Awaiwanont N, Gaban PBV, Mingala CN. Methicillin-resistant Staphylococcus aureus (MRSA) associated with mastitis among water buffaloes in the Philippines. Heliyon 2020; 6:e05663. [PMID: 33319108 PMCID: PMC7723804 DOI: 10.1016/j.heliyon.2020.e05663] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Revised: 10/12/2020] [Accepted: 12/01/2020] [Indexed: 11/04/2022] Open
Abstract
Methicillin-resistant Staphylococcus aureus (MRSA) from dairy animals could pose a public health concern in the population. The study was designed to determine the prevalence of S. aureus and MRSA associated with mastitis among water buffaloes in the central part of Luzon island, the Philippines, and to investigate its associated factors. Three hundred and eighty-four water buffaloes were examined for mastitis using California mastitis test (CMT). Composite milk samples (n = 93) were collected from buffaloes showing positive reaction with CMT. S. aureus was identified from milk samples using biochemical tests. Cefoxitin disk diffusion assay and PCR detecting mecA gene were performed to identify MRSA isolates. Disk diffusion assay was used to investigate the antimicrobial resistance against 9 antibiotics. The prevalence of S. aureus was 41.94% (39/93). MRSA isolates resistant to cefoxitin were at 25.81% (24/93) but only 37.5% (9/24) harbored the mecA gene. All 24 MRSA isolates were resistant to penicillin while the majority were susceptible to clindamycin, trimethoprim-sulfamethoxazole, gentamycin, tetracycline, rifampicin, ciprofloxacin and chloramphenicol with intermediate susceptibility to erythromycin. Furthermore, 37.5% of the isolates were found resistant to two or more antibiotics. Animal-level factor associated with MRSA infection was the history of mastitis (OR = 3.18, CI = 1.03-9.79, p = 0.040). Herd-level factors associated with the detection of MRSA in milk included herd size (OR = 4.24, CI = 1.05-17.07, p = 0.042) and the presence of other animals (OR = 0.15, CI = 0.04-0.58, p = 0.006). High prevalence of intramammary infection with S. aureus and MRSA in dairy buffaloes was observed in the region. This finding raises the concern of preventing zoonotic spread of MRSA.
Collapse
Affiliation(s)
- Alona T. Badua
- Department of Food Animal Clinic, Faculty of Veterinary Medicine, Chiang Mai University, Mae Hia, Muang, 50100 Chiang Mai, Thailand
- Department of Animal Science, College of Agriculture, Central Luzon State University, Science City of Muñoz 3120, Nueva Ecija, Philippines
| | - Sukolrat Boonyayatra
- Department of Food Animal Clinic, Faculty of Veterinary Medicine, Chiang Mai University, Mae Hia, Muang, 50100 Chiang Mai, Thailand
| | - Nattakarn Awaiwanont
- Department of Food Animal Clinic, Faculty of Veterinary Medicine, Chiang Mai University, Mae Hia, Muang, 50100 Chiang Mai, Thailand
| | - Paula Blanca V. Gaban
- Biosafety and Environment Section, Philippine Carabao Center National Headquarters and Gene Pool, Maharlika Highway, Science City of Muñoz 3120, Nueva Ecija, Philippines
| | - Claro N. Mingala
- Biosafety and Environment Section, Philippine Carabao Center National Headquarters and Gene Pool, Maharlika Highway, Science City of Muñoz 3120, Nueva Ecija, Philippines
- Department of Animal Science, College of Agriculture, Central Luzon State University, Science City of Muñoz 3120, Nueva Ecija, Philippines
| |
Collapse
|
17
|
Identification and characterization of mutations responsible for the β-lactam resistance in oxacillin-susceptible mecA-positive Staphylococcus aureus. Sci Rep 2020; 10:16907. [PMID: 33037239 PMCID: PMC7547103 DOI: 10.1038/s41598-020-73796-5] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2020] [Accepted: 09/20/2020] [Indexed: 12/14/2022] Open
Abstract
Staphylococcus aureus strains that are susceptible to the β-lactam antibiotic oxacillin despite carrying mecA (OS-MRSA) cause serious clinical problems globally because of their ability to easily acquire β-lactam resistance. Understanding the genetic mechanism(s) of acquisition of the resistance is therefore crucial for infection control management. For this purpose, a whole-genome sequencing-based analysis was performed using 43 clinical OS-MRSA strains and 100 mutants with reduced susceptibility to oxacillin (MICs 1.0–256 µg/mL) generated from 26 representative OS-MRSA strains. Genome comparison between the mutants and their respective parent strains identified a total of 141 mutations in 46 genes and 8 intergenic regions. Among them, the mutations are frequently found in genes related to RNA polymerase (rpoBC), purine biosynthesis (guaA, prs, hprT), (p)ppGpp synthesis (relSau), glycolysis (pykA, fbaA, fruB), protein quality control (clpXP, ftsH), and tRNA synthase (lysS, gltX), whereas no mutations existed in mec and bla operons. Whole-genome transcriptional profile of the resistant mutants demonstrated that expression of genes associated with purine biosynthesis, protein quality control, and tRNA synthesis were significantly inhibited similar to the massive transcription downregulation seen in S. aureus during the stringent response, while the levels of mecA expression and PBP2a production were varied. We conclude that a combination effect of mecA upregulation and stringent-like response may play an important role in acquisition of β-lactam resistance in OS-MRSA.
Collapse
|
18
|
Catechin isolated from cashew nut shell exhibits antibacterial activity against clinical isolates of MRSA through ROS-mediated oxidative stress. Appl Microbiol Biotechnol 2020; 104:8279-8297. [PMID: 32857200 DOI: 10.1007/s00253-020-10853-z] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Revised: 07/09/2020] [Accepted: 08/23/2020] [Indexed: 01/16/2023]
Abstract
Staphylococcus aureus causes severe infections and among all methicillin-resistant S. aureus (MRSA) remains a great challenge in spite of decade research of antibacterial compounds. Even though some synthetic antibiotics have been developed, they are not effective against MRSA, and hence, there is a search for natural, alternative and plant-based antibacterial compound. In this connection, catechin isolated from cashew nut shell was investigated for its antibacterial potential against MRSA. Catechin exhibited zone of inhibition (ZOI) and minimum inhibitory concentration (MIC) in a range of 15.1-19.5 mm and 78.1-156.2 μg/ml, respectively, against ATCC and clinical isolates of MRSA. Among all clinical isolates, clinical isolate-3 exhibited highest sensitivity to catechin. Catechin has arrested the growth of MRSA strains and also caused toxicity by membrane disruption which was illustrated by AO/EB fluorescence staining. Increased nucleic acid leakage (1.58-28.6-fold) and protein leakage (1.40-23.50-fold) was noticed in MRSA due to catechin treatment when compared to methicillin. Bacteria treated with catechin at its MIC showed 1.52-, 1.87- and 1.74-fold increase of ROS production in methicillin susceptible S. aureus (MSSA), MRSA and clinical isolate-3 strains, respectively, as compared to control. Superoxide dismutase (5.31-9.63 U/mg protein) and catalase (1573-3930 U/mg protein) were significantly decreased as compared to control in catechin-treated S. aureus. Thus, catechin exhibited antibacterial activity through oxidative stress by increased production of ROS and decreased antioxidant enzymes. Altogether results suggest that catechin is a promising lead compound with antibacterial potential against MRSA. KEY POINTS: • Catechin was isolated and identified as active compound in cashew nut shell. • Catechin exhibited antimicrobial activity against clinical isolates of MRSA. • Bacterial cell wall damage was caused by catechin in MRSA strains. • Catechin increased the oxidative stress in MRSA by intracellular ROS production.
Collapse
|
19
|
Sheam MM, Syed SB, Nain Z, Tang SS, Paul DK, Ahmed KR, Biswas SK. Community-acquired pneumonia: aetiology, antibiotic resistance and prospects of phage therapy. J Chemother 2020; 32:395-410. [PMID: 32820711 DOI: 10.1080/1120009x.2020.1807231] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Bacteria are the most common aetiological agents of community-acquired pneumonia (CAP) and use a variety of mechanisms to evade the host immune system. With the emerging antibiotic resistance, CAP-causing bacteria have now become resistant to most antibiotics. Consequently, significant morbimortality is attributed to CAP despite their varying rates depending on the clinical setting in which the patients being treated. Therefore, there is a pressing need for a safe and effective alternative or supplement to conventional antibiotics. Bacteriophages could be a ray of hope as they are specific in killing their host bacteria. Several bacteriophages had been identified that can efficiently parasitize bacteria related to CAP infection and have shown a promising protective effect. Thus, bacteriophages have shown immense possibilities against CAP inflicted by multidrug-resistant bacteria. This review provides an overview of common antibiotic-resistant CAP bacteria with a comprehensive summarization of the promising bacteriophage candidates for prospective phage therapy.
Collapse
Affiliation(s)
- Md Moinuddin Sheam
- Department of Biotechnology and Genetic Engineering, Faculty of Biological Sciences, Islamic University, Kushtia, Bangladesh
| | - Shifath Bin Syed
- Department of Biotechnology and Genetic Engineering, Faculty of Biological Sciences, Islamic University, Kushtia, Bangladesh
| | - Zulkar Nain
- Department of Biotechnology and Genetic Engineering, Faculty of Biological Sciences, Islamic University, Kushtia, Bangladesh.,Department of Genetic Engineering and Biotechnology, Faculty of Sciences and Engineering, East West University, Dhaka, Bangladesh
| | - Swee-Seong Tang
- Division of Microbiology, Institute of Biological Sciences, Faculty of Science, University of Malaya, Kuala Lumpur, Malaysia
| | - Dipak Kumar Paul
- Department of Applied Nutrition and Food Technology, Faculty of Biological Sciences, Islamic University, Kushtia, Bangladesh.,Central Laboratory, Islamic University, Kushtia, Bangladesh
| | - Kazi Rejvee Ahmed
- Department of Biotechnology and Genetic Engineering, Faculty of Biological Sciences, Islamic University, Kushtia, Bangladesh
| | - Sudhangshu Kumar Biswas
- Department of Biotechnology and Genetic Engineering, Faculty of Biological Sciences, Islamic University, Kushtia, Bangladesh.,Central Laboratory, Islamic University, Kushtia, Bangladesh
| |
Collapse
|
20
|
Thapa RK, Winther-Larsen HC, Diep DB, Tønnesen HH. Preformulation studies on novel garvicin KS peptides for topical applications. Eur J Pharm Sci 2020; 151:105333. [PMID: 32268197 DOI: 10.1016/j.ejps.2020.105333] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Revised: 03/30/2020] [Accepted: 03/30/2020] [Indexed: 11/18/2022]
Abstract
Antimicrobial peptides (AMPs) are emerging as a viable alternative to antibiotics attributable to their potent antimicrobial effects and low propensity for resistance development, especially in chronic infected wounds. The development of an optimized topical formulation of AMPs is thus warranted. Preformulation studies for determination of the suitability and optimization requirements of AMPs in topical formulation development are important. Therefore, we sought to investigate the preformulation studies for a novel bacteriocin garvicin KS (GarKS), which is composed of three peptides (GakA, GakB, and GakC). The effects of physiological fluids and varying temperatures on GarKS peptide stability were determined. The antimicrobial effects of the peptides and their combinations were evaluated in Staphylococcus aureus (methicillin sensitive and resistant strains). Furthermore, their effects on fibroblast viability and proliferation were determined. The GarKS peptides were stable in water and PBS at room and physiological temperatures, however, the peptides were significantly degraded in simulated wound fluid. The antimicrobial and fibroblast cell viability/proliferation effects of either individual GarKS peptides or their combinations varied. A careful consideration of the peptide stability, antimicrobial efficacy, and fibroblast viability/proliferation effects suggests GakA+GakB as a potent combination for the development of an optimized topical formulation of GarKS peptides.
Collapse
Affiliation(s)
- Raj Kumar Thapa
- Section for Pharmaceutics and Social Pharmacy, Department of Pharmacy, University of Oslo, P. O. Box 1068 Blindern, NO, 0316 Oslo, Norway.
| | - Hanne Cecilie Winther-Larsen
- Centre for Integrative Microbial Evolution (CIME) and Department of Pharmacology and Pharmaceutical Biosciences, University of Oslo, P. O. Box 1068 Blindern, NO, 0371 Oslo, Norway
| | - Dzung B Diep
- Faculty of Chemistry, Biotechnology, and Food Science, Norwegian University of Life Sciences, P.O. Box 5003, NO, 1432 Ås, Norway
| | - Hanne Hjorth Tønnesen
- Section for Pharmaceutics and Social Pharmacy, Department of Pharmacy, University of Oslo, P. O. Box 1068 Blindern, NO, 0316 Oslo, Norway
| |
Collapse
|
21
|
Diagnostic Values of Multiplex Loop-Mediated Isothermal Amplification and Multiplex Polymerase Chain Reaction for Detection of Methicillin-Resistant Staphylococcus aureus. Jundishapur J Microbiol 2020. [DOI: 10.5812/jjm.96682] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
Background: Methicillin-resistant Staphylococcus aureus (MRSA) has emerged as a significant pathogen in community and hospital environments and is associated with high mortality and morbidity. Both polymerase chain reaction (PCR) and loop-mediated isothermal amplification (LAMP) methods are sensitive and acceptable molecular methods for the diagnosis of infectious diseases. Objectives: This study aimed to develop detection assays for Staphylococcal mecA and spa using multiplex PCR and LAMP. Methods: Both methods were standardized, and detection limits were determined using serial dilutions of S. aureus DNA samples. Fifty-three clinical isolates of S. aureus were confirmed to the species level using biochemical tests and multiplex PCR and multiplex LAMP for the spa gene, while disk diffusion, minimum inhibitory concentration, and detection of mecA genes were used for the assessment of methicillin resistance. Results: The PCR could detect the mecA and spa genes at 1 fg/mL and 10 fg/mL of bacterial DNA, which were equal to 35 and 350 gene copy numbers, respectively. Similarly, multiplex LAMP detected the spa and mecA genes at 0.1 fg/mL and 1 fg/mL of bacterial DNA, which were equal to 3.5 and 35 genome copy numbers, respectively. According to MIC and disk diffusion methods, four (7.54%) cases were oxacillin-sensitive methicillin-resistant S. aureus, 16 isolates were methicillin-sensitive, and 37 isolates were methicillin-resistant. According to multiplex PCR, 47.75% of the isolates were mecA-positive while in multiplex LAMP, 41 (35.77%) isolates were mecA-positive. Conclusions: The sensitivity and specificity of the multiplex LAMP were higher than those of multiplex PCR and biochemical methods. Thus, we can apply the LAMP for the routine detection of MRSA.
Collapse
|
22
|
Smith JT, Amador S, McGonagle CJ, Needle D, Gibson R, Andam CP. Population genomics of Staphylococcus pseudintermedius in companion animals in the United States. Commun Biol 2020; 3:282. [PMID: 32503984 PMCID: PMC7275049 DOI: 10.1038/s42003-020-1009-y] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2019] [Accepted: 05/15/2020] [Indexed: 01/08/2023] Open
Abstract
Staphylococcus pseudintermedius is a commensal bacterium and a major opportunistic pathogen of dogs. The emergence of methicillin-resistant S. pseudintermedius (MRSP) is also becoming a serious concern. We carried out a population genomics study of 130 clinical S. pseudintermedius isolates from dogs and cats in the New England region of the United States. Results revealed the co-circulation of phylogenetically diverse lineages that have access to a large pool of accessory genes. Many MRSP and multidrug-resistant clones have emerged through multiple independent, horizontal acquisition of resistance determinants and frequent genetic exchange that disseminate DNA to the broader population. When compared to a Texas population, we found evidence of clonal expansion of MRSP lineages that have disseminated over large distances. These findings provide unprecedented insight into the diversification of a common cutaneous colonizer of man's oldest companion animal and the widespread circulation of multiple high-risk resistant clones.
Collapse
Affiliation(s)
- Joshua T Smith
- University of New Hampshire, Department of Molecular, Cellular and Biomedical Sciences, Durham, NH, 03824, USA
| | - Sharlene Amador
- University of New Hampshire, Department of Molecular, Cellular and Biomedical Sciences, Durham, NH, 03824, USA
| | - Colin J McGonagle
- University of New Hampshire, Department of Molecular, Cellular and Biomedical Sciences, Durham, NH, 03824, USA
| | - David Needle
- New Hampshire Veterinary Diagnostic Laboratory, Durham, NH, 03824, USA
| | - Robert Gibson
- New Hampshire Veterinary Diagnostic Laboratory, Durham, NH, 03824, USA
| | - Cheryl P Andam
- University of New Hampshire, Department of Molecular, Cellular and Biomedical Sciences, Durham, NH, 03824, USA.
| |
Collapse
|
23
|
Fergestad ME, Stamsås GA, Morales Angeles D, Salehian Z, Wasteson Y, Kjos M. Penicillin-binding protein PBP2a provides variable levels of protection toward different β-lactams in Staphylococcus aureus RN4220. Microbiologyopen 2020; 9:e1057. [PMID: 32419377 PMCID: PMC7424258 DOI: 10.1002/mbo3.1057] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2020] [Revised: 04/21/2020] [Accepted: 04/25/2020] [Indexed: 12/31/2022] Open
Abstract
Methicillin-resistant Staphylococcus aureus (MRSA) is resistant to most β-lactams due to the expression of an extra penicillin-binding protein, PBP2a, with low β-lactam affinity. It has long been known that heterologous expression of the PBP2a-encoding mecA gene in methicillin-sensitive S. aureus (MSSA) provides protection towards β-lactams, however, some reports suggest that the degree of protection can vary between different β-lactams. To test this more systematically, we introduced an IPTG-inducible mecA into the MSSA laboratory strain RN4220. We confirm, by growth assays as well as single-cell microfluidics time-lapse microscopy experiments, that PBP2a expression protects against β-lactams in S. aureus RN4220. By testing a panel of ten different β-lactams, we conclude that there is also a great variation in the level of protection conferred by PBP2a. Expression of PBP2a resulted in an only fourfold increase in minimum inhibitory concentration (MIC) for imipenem, while a 32-fold increase in MIC was observed for cefaclor and cephalexin. Interestingly, in our experimental setup, PBP2a confers the highest protection against cefaclor and cephalexin-two β-lactams that are known to have a high specific affinity toward the transpeptidase PBP3 of S. aureus. Notably, using a single-cell microfluidics setup we demonstrate a considerable phenotypic variation between cells upon β-lactam exposure and show that mecA-expressing S. aureus can survive β-lactam concentrations much higher than the minimal inhibitory concentrations. We discuss possible explanations and implications of these results including important aspects regarding treatment of infection.
Collapse
Affiliation(s)
- Marte Ekeland Fergestad
- Faculty of Chemistry, Biotechnology and Food Science, Norwegian University of Life Sciences, Ås, Norway.,Department of Paraclinical Sciences, Faculty of Veterinary Medicine, Norwegian University of Life Sciences, Ås, Norway
| | - Gro Anita Stamsås
- Faculty of Chemistry, Biotechnology and Food Science, Norwegian University of Life Sciences, Ås, Norway
| | - Danae Morales Angeles
- Faculty of Chemistry, Biotechnology and Food Science, Norwegian University of Life Sciences, Ås, Norway
| | - Zhian Salehian
- Faculty of Chemistry, Biotechnology and Food Science, Norwegian University of Life Sciences, Ås, Norway
| | - Yngvild Wasteson
- Department of Paraclinical Sciences, Faculty of Veterinary Medicine, Norwegian University of Life Sciences, Ås, Norway
| | - Morten Kjos
- Faculty of Chemistry, Biotechnology and Food Science, Norwegian University of Life Sciences, Ås, Norway
| |
Collapse
|
24
|
Qian Y, Allegretta G, Janardhanan J, Peng Z, Mahasenan KV, Lastochkin E, Gozun MMN, Tejera S, Schroeder VA, Wolter WR, Feltzer R, Mobashery S, Chang M. Exploration of the Structural Space in 4(3 H)-Quinazolinone Antibacterials. J Med Chem 2020; 63:5287-5296. [PMID: 32343145 DOI: 10.1021/acs.jmedchem.0c00153] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
We report herein the syntheses of 79 derivatives of the 4(3H)-quinazolinones and their structure-activity relationship (SAR) against methicillin-resistant Staphylococcus aureus (MRSA). Twenty one analogs were further evaluated in in vitro assays. Subsequent investigation of the pharmacokinetic properties singled out compound 73 ((E)-3-(5-carboxy-2-fluorophenyl)-2-(4-cyanostyryl)quinazolin-4(3H)-one) for further study. The compound synergized with piperacillin-tazobactam (TZP) both in vitro and in vivo in a clinically relevant mouse model of MRSA infection. The TZP combination lacks activity against MRSA, yet it synergized with compound 73 to kill MRSA in a bactericidal manner. The synergy is rationalized by the ability of the quinazolinones to bind to the allosteric site of penicillin-binding protein (PBP)2a, resulting in opening of the active site, whereby the β-lactam antibiotic now is enabled to bind to the active site in its mechanism of action. The combination effectively treats MRSA infection, for which many antibiotics (including TZP) have faced clinical obsolescence.
Collapse
Affiliation(s)
- Yuanyuan Qian
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, Indiana 46556, United States
| | - Giuseppe Allegretta
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, Indiana 46556, United States
| | - Jeshina Janardhanan
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, Indiana 46556, United States
| | - Zhihong Peng
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, Indiana 46556, United States
| | - Kiran V Mahasenan
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, Indiana 46556, United States
| | - Elena Lastochkin
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, Indiana 46556, United States
| | - Melissa Malia N Gozun
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, Indiana 46556, United States
| | - Sara Tejera
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, Indiana 46556, United States
| | - Valerie A Schroeder
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, Indiana 46556, United States
| | - William R Wolter
- Freimann Life Sciences Center, University of Notre Dame, Notre Dame, Indiana 46556, United States
| | - Rhona Feltzer
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, Indiana 46556, United States
| | - Shahriar Mobashery
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, Indiana 46556, United States
| | - Mayland Chang
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, Indiana 46556, United States
| |
Collapse
|
25
|
Gondokesumo ME, Kurniawan IM. Molecular docking study of sappan wood extract to inhibit PBP2A enzyme on methicillin-resistant Staphylococcus aureus (MRSA). J Basic Clin Physiol Pharmacol 2020; 30:/j/jbcpp.ahead-of-print/jbcpp-2019-0282/jbcpp-2019-0282.xml. [PMID: 31953996 DOI: 10.1515/jbcpp-2019-0282] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2019] [Accepted: 12/13/2019] [Indexed: 11/15/2022]
Abstract
Background PBP2a is a type of penicillin-binding proteins (PBPs) that cause resistivity in methicillin-resistant Staphylococcus aureus (MRSA) from β-lactam antibiotics. MRSA susceptible with cefttobiprole (fifth generation of cephalosporin as an anti-MRSA agent) which inhibits PBP2a and stops its growth. Contrary to its efficacy, ceftobiprole causes taste disturbance more than any other cephalosporins; furthermore, its mechanism is unknown. This study aims to explore an in silico study of a natural compound, which serves as a potential alternative to overcome MRSA with minimum adverse side effects. Methods A molecular docking study was performed using Molegro Virtual Docker version 5.5. Brazilin and proto-sappanins A-E are phytochemical compounds contained in sappan wood extract and are docked into the binding site of PBP2a (Protein Data Bank: ID 4DKI). Results Brazilin and proto-sappanins A-E have some interaction with Ser 403 amino acid residue which is an important interaction to inhibit PBP2a protein. The result of the molecular docking study showed that the MolDock score of proto-sappanins D and E is lower than that of methicillin but higher than that of its native ligand (ceftobiprole). Conclusions The results of this study suggest that proto-sappanins D and E have an excellent potential activity as an alternative to ceftobiprole in limiting MRSA growth through PBP2A enzyme inhibition.
Collapse
Affiliation(s)
- Marisca Evalina Gondokesumo
- Department of Biology Pharmacy, Faculty of Pharmacy, University of Surabaya, Jl. Raya Kalirungkut, Surabaya, East Java 60293, Indonesia, Phone: +62 31 2981110
| | - Ihsan Mulyadi Kurniawan
- Indonesia University of Education, Department of Biology Education, Bandung, West Java, Indonesia
| |
Collapse
|
26
|
Chisanga M, Linton D, Muhamadali H, Ellis DI, Kimber RL, Mironov A, Goodacre R. Rapid differentiation of Campylobacter jejuni cell wall mutants using Raman spectroscopy, SERS and mass spectrometry combined with chemometrics. Analyst 2020; 145:1236-1249. [DOI: 10.1039/c9an02026h] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
SERS was developed for intercellular and intracellular analyses. Using a series of cell wall mutants in C. jejuni we show cell wall versus cytoplasm differences.
Collapse
Affiliation(s)
- Malama Chisanga
- School of Chemistry
- Manchester Institute of Biotechnology
- University of Manchester
- Manchester
- UK
| | - Dennis Linton
- School of Biological Sciences
- Faculty of Biology
- Medicine and Health
- University of Manchester
- Manchester
| | - Howbeer Muhamadali
- Department of Biochemistry
- Institute of Integrative Biology
- University of Liverpool
- Liverpool
- UK
| | - David I. Ellis
- School of Chemistry
- Manchester Institute of Biotechnology
- University of Manchester
- Manchester
- UK
| | - Richard L. Kimber
- Department of Earth and Environmental Sciences
- University of Manchester
- Manchester
- UK
| | - Aleksandr Mironov
- EM Core Facility
- Faculty of Biology
- Medicine and Health
- University of Manchester
- Manchester
| | - Royston Goodacre
- Department of Biochemistry
- Institute of Integrative Biology
- University of Liverpool
- Liverpool
- UK
| |
Collapse
|
27
|
Lian ZJ, Lin TY, Yao CX, Su YL, Liao SH, Wu SM. Staphylococcus aureus strains exposed to copper indium sulfide quantum dots exhibit increased tolerance to penicillin G, tetracycline and ciprofloxacin. NEW J CHEM 2020. [DOI: 10.1039/c9nj05748j] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Copper indium sulfide, CuInS2 (CIS), semiconductor nanocrystals have the qualities of low toxicity, high absorption coefficient and near-infrared luminescence, and thus have attracted increasing attention due to their wide prospective applications in various fields.
Collapse
Affiliation(s)
- Zong-Juan Lian
- Department of Analytical Chemistry
- China Pharmaceutical University
- Nanjing 210009
- China
- Key Laboratory of Drug Quality Control and Pharmacovigilance
| | - Tian-Yang Lin
- Department of Analytical Chemistry
- China Pharmaceutical University
- Nanjing 210009
- China
- Key Laboratory of Drug Quality Control and Pharmacovigilance
| | - Cai-Xia Yao
- Department of Analytical Chemistry
- China Pharmaceutical University
- Nanjing 210009
- China
- Key Laboratory of Drug Quality Control and Pharmacovigilance
| | - Yi-Long Su
- Department of Analytical Chemistry
- China Pharmaceutical University
- Nanjing 210009
- China
- Key Laboratory of Drug Quality Control and Pharmacovigilance
| | - Sheng-Hua Liao
- Department of Analytical Chemistry
- China Pharmaceutical University
- Nanjing 210009
- China
- Key Laboratory of Drug Quality Control and Pharmacovigilance
| | - Sheng-Mei Wu
- Department of Analytical Chemistry
- China Pharmaceutical University
- Nanjing 210009
- China
- Key Laboratory of Drug Quality Control and Pharmacovigilance
| |
Collapse
|
28
|
Belluzo BS, Abriata LA, Giannini E, Mihovilcevic D, Dal Peraro M, Llarrull LI. An experiment-informed signal transduction model for the role of the Staphylococcus aureus MecR1 protein in β-lactam resistance. Sci Rep 2019; 9:19558. [PMID: 31862951 PMCID: PMC6925264 DOI: 10.1038/s41598-019-55923-z] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2019] [Accepted: 12/04/2019] [Indexed: 11/13/2022] Open
Abstract
The treatment of hospital- and community-associated infections by methicillin-resistant Staphylococcus aureus (MRSA) is a perpetual challenge. This Gram-positive bacterium is resistant specifically to β-lactam antibiotics, and generally to many other antibacterial agents. Its resistance mechanisms to β-lactam antibiotics are activated only when the bacterium encounters a β-lactam. This activation is regulated by the transmembrane sensor/signal transducer proteins BlaR1 and MecR1. Neither the transmembrane/metalloprotease domain, nor the complete MecR1 and BlaR1 proteins, are isolatable for mechanistic study. Here we propose a model for full-length MecR1 based on homology modeling, residue coevolution data, a new extensive experimental mapping of transmembrane topology, partial structures, molecular simulations, and available NMR data. Our model defines the metalloprotease domain as a hydrophilic transmembrane chamber effectively sealed by the apo-sensor domain. It proposes that the amphipathic helices inserted into the gluzincin domain constitute the route for transmission of the β-lactam-binding event in the extracellular sensor domain, to the intracellular and membrane-embedded zinc-containing active site. From here, we discuss possible routes for subsequent activation of proteolytic action. This study provides the first coherent model of the structure of MecR1, opening routes for future functional investigations on how β-lactam binding culminates in the proteolytic degradation of MecI.
Collapse
Affiliation(s)
- Bruno S Belluzo
- Instituto de Biología Molecular y Celular de Rosario (IBR, CONICET-UNR), Predio CONICET Rosario, 27 de Febrero 210 bis, 2000, Rosario, Argentina
| | - Luciano A Abriata
- Laboratory for Biomolecular Modeling - École Polytechnique Fédérale de Lausanne and Swiss Institute of Bioinformatics, CH-1015, Lausanne, Switzerland
| | - Estefanía Giannini
- Instituto de Biología Molecular y Celular de Rosario (IBR, CONICET-UNR), Predio CONICET Rosario, 27 de Febrero 210 bis, 2000, Rosario, Argentina
| | - Damila Mihovilcevic
- Instituto de Biología Molecular y Celular de Rosario (IBR, CONICET-UNR), Predio CONICET Rosario, 27 de Febrero 210 bis, 2000, Rosario, Argentina
| | - Matteo Dal Peraro
- Laboratory for Biomolecular Modeling - École Polytechnique Fédérale de Lausanne and Swiss Institute of Bioinformatics, CH-1015, Lausanne, Switzerland
| | - Leticia I Llarrull
- Instituto de Biología Molecular y Celular de Rosario (IBR, CONICET-UNR), Predio CONICET Rosario, 27 de Febrero 210 bis, 2000, Rosario, Argentina. .,Área Biofísica, Facultad de Ciencias Bioquímicas y Farmacéuticas, Universidad Nacional de Rosario, Suipacha 570, 2000, Rosario, Argentina.
| |
Collapse
|
29
|
Susceptibility of Methicillin-Resistant Staphylococcus aureus to Five Quinazolinone Antibacterials. Antimicrob Agents Chemother 2019; 64:AAC.01344-19. [PMID: 31611358 PMCID: PMC7187613 DOI: 10.1128/aac.01344-19] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2019] [Accepted: 10/08/2019] [Indexed: 11/22/2022] Open
Abstract
The in vitro activities of five quinazolinone antibacterials, compounds Q1 to Q5, were tested against 210 strains of methicillin-resistant Staphylococcus aureus (MRSA). The MIC50/MIC90 values (in μg/ml) were as follows: Q1, 0.5/2; Q2, 1/4; Q3, 2/4; Q4, 0.06/0.25; and Q5, 0.125/0.5. The in vitro activities of five quinazolinone antibacterials, compounds Q1 to Q5, were tested against 210 strains of methicillin-resistant Staphylococcus aureus (MRSA). The MIC50/MIC90 values (in μg/ml) were as follows: Q1, 0.5/2; Q2, 1/4; Q3, 2/4; Q4, 0.06/0.25; and Q5, 0.125/0.5. Several strains with high MIC values (from 8 to >32 μg/ml) for some of these compounds exhibited amino acid changes in the penicillin-binding proteins, which are targeted by these antibacterials.
Collapse
|
30
|
Speri E, Kim C, De Benedetti S, Qian Y, Lastochkin E, Fishovitz J, Fisher JF, Mobashery S. Cinnamonitrile Adjuvants Restore Susceptibility to β-Lactams against Methicillin-Resistant Staphylococcus aureus. ACS Med Chem Lett 2019; 10:1148-1153. [PMID: 31413798 PMCID: PMC6691485 DOI: 10.1021/acsmedchemlett.9b00169] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2019] [Accepted: 07/01/2019] [Indexed: 11/28/2022] Open
Abstract
β-Lactams are used routinely to treat Staphylococcus aureus infections. However, the emergence of methicillin-resistant S. aureus (MRSA) renders them clinically precarious. We describe a class of cinnamonitrile adjuvants that restore the activity of oxacillin (a penicillin member of the β-lactams) against MRSA. The lead adjuvants were tested against six important strains of MRSA, one vancomycin-intermediate S. aureus (VISA) strain, and one linezolid-resistant S. aureus strain. Five compounds out of 84 total compounds showed broad potentiation. At 8 μM (E)-3-(5-(3,4-dichlorobenzyl)-2-(trifluoromethoxy)phenyl)-2-(methylsulfonyl)acrylonitrile (26) potentiated oxacillin with a >4000-fold reduction of its MIC (from 256 to 0.06 mg·L-1). This class of adjuvants holds promise for reversal of the resistance phenotype of MRSA.
Collapse
Affiliation(s)
- Enrico Speri
- Department Chemistry and
Biochemistry, University of Notre Dame, Notre Dame, Indiana 46556, United States
| | - Choon Kim
- Department Chemistry and
Biochemistry, University of Notre Dame, Notre Dame, Indiana 46556, United States
| | - Stefania De Benedetti
- Department Chemistry and
Biochemistry, University of Notre Dame, Notre Dame, Indiana 46556, United States
| | - Yuanyuan Qian
- Department Chemistry and
Biochemistry, University of Notre Dame, Notre Dame, Indiana 46556, United States
| | - Elena Lastochkin
- Department Chemistry and
Biochemistry, University of Notre Dame, Notre Dame, Indiana 46556, United States
| | - Jennifer Fishovitz
- Department Chemistry and
Biochemistry, University of Notre Dame, Notre Dame, Indiana 46556, United States
| | - Jed F. Fisher
- Department Chemistry and
Biochemistry, University of Notre Dame, Notre Dame, Indiana 46556, United States
| | - Shahriar Mobashery
- Department Chemistry and
Biochemistry, University of Notre Dame, Notre Dame, Indiana 46556, United States
| |
Collapse
|
31
|
Amoako DG, Somboro AM, Abia ALK, Allam M, Ismail A, Bester L, Essack SY. Genomic analysis of methicillin-resistant Staphylococcus aureus isolated from poultry and occupational farm workers in Umgungundlovu District, South Africa. THE SCIENCE OF THE TOTAL ENVIRONMENT 2019; 670:704-716. [PMID: 30909047 DOI: 10.1016/j.scitotenv.2019.03.110] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/30/2018] [Revised: 03/06/2019] [Accepted: 03/07/2019] [Indexed: 06/09/2023]
Abstract
This study detected methicillin-resistant Staphylococcus aureus (MRSA) isolates circulating in poultry and farm workers at an intensive poultry production system in uMgungundlovu, South Africa and established the genetic relatedness and characteristics of the isolates using whole genome sequencing (WGS). A total of 145 S. aureus were isolated from poultry (120) and occupational workers (25) in the "farm to fork" continuum (farm, transport, slaughterhouse, and retail points). Twelve MRSA (12/145; 8.3%) isolates were found in the poultry food-chain. MRSA isolates were subjected to antibiotic susceptibility testing against a panel of 20 antibiotics using the broth dilution method and their whole genome was sequenced via the Illumina MiSeq. All the MRSA isolates were multi-drug resistant (MDR) and carried the mecA gene on the SCCmec mobile genetic element (MGE). The majority (11/12) of the MRSA isolates circulating between humans and animals in the continuum belonged to a human-associated clone, ST612-CC8-t1257-SCCmec_IVd (2B), previously reported in South Africa. Other MGEs present in the isolates included: plasmid replicons based on Rep 7 and 20, insertion sequences (IS1182), and prophages (phi2958PVL). Genomic analysis identified a distinct acquired antibiotic resistome in the clone, which accurately predicted the phenotypic antibiograms. Phylogenetic analysis clustered the isolates within the major cluster (I), suggesting the spread of the local dominant multidrug resistance MRSA clone ST612-CC8-t1257-SCCmec_IVd (2B) between humans and animals along the 'farm to fork' continuum. The findings of this study suggest the need to establish appropriate control measures to curb the spread of MDR-MRSA in the food chain.
Collapse
Affiliation(s)
- Daniel Gyamfi Amoako
- Antimicrobial Research Unit, College of Health Sciences, University of KwaZulu-Natal, Durban, South Africa; Biomedical Resource Unit, School of Laboratory Medicine and Medical Sciences, College of Health Sciences, University of KwaZulu-Natal, Durban, South Africa.
| | - Anou Moise Somboro
- Antimicrobial Research Unit, College of Health Sciences, University of KwaZulu-Natal, Durban, South Africa; Biomedical Resource Unit, School of Laboratory Medicine and Medical Sciences, College of Health Sciences, University of KwaZulu-Natal, Durban, South Africa.
| | - Akebe Luther King Abia
- Antimicrobial Research Unit, College of Health Sciences, University of KwaZulu-Natal, Durban, South Africa.
| | - Mushal Allam
- Sequencing Core Facility, National Institute for Communicable Diseases, National Health Laboratory Service, Pretoria, South Africa.
| | - Arshad Ismail
- Sequencing Core Facility, National Institute for Communicable Diseases, National Health Laboratory Service, Pretoria, South Africa.
| | - Linda Bester
- Biomedical Resource Unit, School of Laboratory Medicine and Medical Sciences, College of Health Sciences, University of KwaZulu-Natal, Durban, South Africa.
| | - Sabiha Yusuf Essack
- Antimicrobial Research Unit, College of Health Sciences, University of KwaZulu-Natal, Durban, South Africa.
| |
Collapse
|
32
|
Dweba CC, Zishiri OT, El Zowalaty ME. Isolation and Molecular Identification of Virulence, Antimicrobial and Heavy Metal Resistance Genes in Livestock-Associated Methicillin-Resistant Staphylococcus aureus. Pathogens 2019; 8:pathogens8020079. [PMID: 31207959 PMCID: PMC6630769 DOI: 10.3390/pathogens8020079] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2019] [Revised: 05/16/2019] [Accepted: 05/16/2019] [Indexed: 02/07/2023] Open
Abstract
Staphylococcus aureus is one of the most important pathogens of humans and animals. Livestock production contributes a significant proportion to the South African Gross Domestic Product. Consequently, the aim of this study was to determine for the first time the prevalence, virulence, antibiotic and heavy metal resistance in livestock-associated S. aureus isolated from South African livestock production systems. Microbial phenotypic methods were used to detect the presence of antibiotic and heavy metal resistance. Furthermore, molecular DNA based methods were used to genetically determine virulence as well as antibiotic and heavy metal resistance determinants. Polymerase chain reaction (PCR) confirmed 217 out of 403 (53.8%) isolates to be S. aureus. Kirby-Bauer disc diffusion method was conducted to evaluate antibiotic resistance and 90.8% of S. aureus isolates were found to be resistant to at least three antibiotics, and therefore, classified as multidrug resistant. Of the antibiotics tested, 98% of the isolates demonstrated resistance towards penicillin G. High resistance was shown against different heavy metals, with 90% (196/217), 88% (192/217), 86% (188/217) and 84% (183/217) of the isolates resistant to 1500 µg/mL concentration of Cadmium (Cd), Zinc (Zn), Lead (Pb) and Copper (Cu) respectively. A total of 10 antimicrobial resistance and virulence genetic determinants were screened for all livestock associated S. aureus isolates. Methicillin-resistant S. aureus (MRSA) isolates were identified, by the presence of mecC, in 27% of the isolates with a significant relationship (p < 0.001)) with the host animal. This is the first report of mecC positive LA-MRSA in South Africa and the African continent. The gene for tetracycline resistance (tetK) was the most frequently detected of the screened genes with an overall prevalence of 35% and the highest prevalence percentage was observed for goats (56.76%) followed by avian species (chicken, duck and wild birds) (42.5%). Virulence-associated genes were observed across all animal host species. The study reports the presence of luks/pv, a gene encoding the PVL toxin previously described to be a marker for community acquired-MRSA, suggesting the crossing of species between human and livestock. The high prevalence of S. aureus from the livestock indicates a major food security and healthcare threat. This threat is further compounded by the virulence of the pathogen, which causes numerous clinical manifestations. The phenomenon of co-selection is observed in this study as isolates exhibited resistance to both antibiotics and heavy metals. Further, all the screened antibiotic and heavy metal resistance genes did not correspond with the phenotypic resistance.
Collapse
Affiliation(s)
- Chumisa C Dweba
- Discipline of Genetics, School of Life Sciences, College of Agriculture Engineering and Sciences, University of KwaZulu-Natal, Durban 4000, South Africa.
| | - Oliver T Zishiri
- Discipline of Genetics, School of Life Sciences, College of Agriculture Engineering and Sciences, University of KwaZulu-Natal, Durban 4000, South Africa.
| | - Mohamed E El Zowalaty
- Infectious Diseases and Anti-Infective Therapy Research Group, Sharjah Medical Research Institute and College of Pharmacy, University of Sharjah, Sharjah 27272, UAE.
- Department of Infectious Diseases, St. Jude Children's Research Hospital, 262 Danny Thomas Pl, Memphis, TN 38105, USA.
| |
Collapse
|
33
|
Zaghouani M, Bögeholz LAK, Mercier E, Wintermeyer W, Roche SP. Total synthesis of (±)-fumimycin and analogues for biological evaluation as peptide deformylase inhibitors. Tetrahedron 2019; 75:3216-3230. [PMID: 31555018 PMCID: PMC6759494 DOI: 10.1016/j.tet.2019.03.037] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
A concise 7-step total synthesis of (±)-fumimycin in 11.6 % overall yield is reported. An acid-catalyzed intramolecular aza-Friedel-Crafts cyclization was developed to construct the benzofuranone skeleton of the natural product bearing an α,α-disubstituted amino acid moiety in a single step. Regioselective chlorination followed by a Suzuki-Miyaura cross-coupling rapidly enabled the preparation of a library of analogues which were evaluated against peptide deformylase for antibacterial activity.
Collapse
Affiliation(s)
- Mehdi Zaghouani
- Department of Chemistry and Biochemistry, Florida Atlantic University, Boca Raton, Florida 33431, USA
| | - Lena A. K. Bögeholz
- Department of Physical Biochemistry, Max Planck Institute for Biophysical Chemistry, Göttingen 37077, Germany
| | - Evan Mercier
- Department of Physical Biochemistry, Max Planck Institute for Biophysical Chemistry, Göttingen 37077, Germany
| | - Wolfgang Wintermeyer
- Department of Physical Biochemistry, Max Planck Institute for Biophysical Chemistry, Göttingen 37077, Germany
| | - Stéphane P. Roche
- Department of Chemistry and Biochemistry, Florida Atlantic University, Boca Raton, Florida 33431, USA
| |
Collapse
|
34
|
Galia L, Ligozzi M, Bertoncelli A, Mazzariol A. Real-time PCR assay for detection of Staphylococcus aureus, Panton-Valentine Leucocidin and Methicillin Resistance directly from clinical samples. AIMS Microbiol 2019; 5:138-146. [PMID: 31384708 PMCID: PMC6642910 DOI: 10.3934/microbiol.2019.2.138] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2018] [Accepted: 03/26/2019] [Indexed: 02/04/2023] Open
Abstract
Rapid detection of Methicillin Resistant Staphylococcus aureus (MRSA) is an important concern for both treatment and implementation of infection control policies. The present study provides an ‘in house’ real-time PCR assay to detect directly nuc, pvl, and mecA genes. The assay is able to perform identification of MRSA, Methicillin-Sensitive S. aureus, Methicillin-Resistant Coagulase Negative Staphylococci and the Panton-Valentine leukocidin virulence gene from rectal and pharyngeal swab samples in a screening context. We found an analytical sensitivity of this current Triplex PCR assay of 514 CFU/mL. Analytical specificity was tested with different Gram-positive and Gram-negative species and yielded no false-positive PCR signal. The sensitivity and specificity of the Triplex Real Time PCR were both 100% for these targets when compared with the culture and conventional methods. This assay is readily adaptable for routine use in a microbiology laboratory, as it will enable the implementation of timely and properly guided therapy and infection control strategies.
Collapse
Affiliation(s)
- Liliana Galia
- Department of Diagnostics and Public Health, University of Verona, Verona, Italy
| | - Marco Ligozzi
- Department of Diagnostics and Public Health, University of Verona, Verona, Italy
| | - Anna Bertoncelli
- Department of Diagnostics and Public Health, University of Verona, Verona, Italy
| | - Annarita Mazzariol
- Department of Diagnostics and Public Health, University of Verona, Verona, Italy
| |
Collapse
|
35
|
Lounsbury N, Reeber MG, Mina G, Chbib C. A Mini-Review on Ceftaroline in Bacteremia Patients with Methicillin-Resistant Staphylococcus aureus (MRSA) Infections. Antibiotics (Basel) 2019; 8:antibiotics8010030. [PMID: 30897759 PMCID: PMC6466573 DOI: 10.3390/antibiotics8010030] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2019] [Revised: 03/09/2019] [Accepted: 03/16/2019] [Indexed: 11/16/2022] Open
Abstract
OBJECTIVE The objective of this review is to describe the outcomes of patients treated with ceftaroline in the non-Food and Drug Administration (FDA) approved indication of methicillin-resistant Staphylococcus aureus (MRSA) infections in both pediatric and adult populations. DATA SOURCES A systematic overview was conducted by searching PubMed, Medline, and The Cochrane Library up to January 2019. STUDY SELECTION AND DATA EXTRACTION All English-language clinical trials and case reports related to the efficacy of ceftaroline in new, not-yet-approved FDA indications in MRSA infections in pediatric or adult populations. DATA SYNTHESIS In the case of MRSA bacteremia (MRSAB) infections, three different randomized studies in pediatric patients showed effectiveness of ceftaroline. When used in the case of adult populations with MRSA bacteremia, a small trial of 16 patients showed 50% clinical success in patients with acute bacterial skin and skin structure infections versus 63% clinical success in patients with community-acquired bacterial pneumonia. Another case series of six refractory case reports showed 50% clinical success of ceftaroline in patients with MRSA. CONCLUSIONS Although there are few case reports and limited data to date, ceftaroline fosamil should continue to be studied as an alternative therapy in MRSA infections in both pediatric and adult populations. Clinical success rates of ceftaroline were, in most cases, considered high when treating patients with MRSA infection. More clinical trials need to be studied. In the specific case of MRSA bacteremia, the treatment options remain few and ceftaroline should be extensively studied for the salvage treatment of MRSAB.
Collapse
Affiliation(s)
- Nicole Lounsbury
- Department of Pharmaceutical Sciences, Larkin University College of Pharmacy, 18301 North Miami Ave, Miami, FL 33169, USA.
| | - Mary G Reeber
- Department of Pharmaceutical Sciences, Larkin University College of Pharmacy, 18301 North Miami Ave, Miami, FL 33169, USA.
| | - Georges Mina
- Pharmacy department, Jackson Memorial Hospital, 1611 NW 12th Ave, Miami, FL 33136, USA.
| | - Christiane Chbib
- Department of Pharmaceutical Sciences, Larkin University College of Pharmacy, 18301 North Miami Ave, Miami, FL 33169, USA.
| |
Collapse
|
36
|
Bhawini A, Pandey P, Dubey AP, Zehra A, Nath G, Mishra MN. RelQ Mediates the Expression of β-Lactam Resistance in Methicillin-Resistant Staphylococcus aureus. Front Microbiol 2019; 10:339. [PMID: 30915038 PMCID: PMC6421274 DOI: 10.3389/fmicb.2019.00339] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2018] [Accepted: 02/08/2019] [Indexed: 11/18/2022] Open
Abstract
An induced stringent response, which is established by an increased level of (p)ppGpp, is required for the expression of β-lactam resistance in methicillin-resistant Staphylococcus aureus (MRSA). However, it is not clear whether RSH (enzyme mediating stringent response to amino acid starvation) or small alarmone synthetases (SASs) are involved in the maintenance of (p)ppGpp level in response to β-lactams. Since the S. aureus genome encodes two active SASs (RelP and RelQ), their contribution to the expression of β-lactam resistance in MRSA was investigated. It was determined that relQ deletion renders community-associated MRSA (CA-MRSA) sensitive to β-lactams by negatively affecting the expression of mecA, and induction of (p)ppGpp synthesis by mupirocin bypasses the requirement of relQ for the expression of high-level β-lactam resistance. Surprisingly, relP deletion increased the level of β-lactam resistance. Such contradictory observations could be attributed to the fact that relQ promoter is ~5-fold stronger than the relP and is induced by oxacillin as well as deletion of either of the SASs, while relP promoter responds only to oxacillin. The stronger promoter activity of relQ, coupled with the inducibility of the relQ promoter in response to the lack of relP, results in efficient expression of relQ in the relP-deleted background. This positively affects mecA expression and renders the ΔrelP strain highly resistant. These findings indicate an important role for RelQ in the expression of high-level β-lactam resistance in MRSA.
Collapse
Affiliation(s)
- Ajita Bhawini
- Department of Microbiology, Institute of Medical Sciences, Banaras Hindu University, Varanasi, India
| | - Parul Pandey
- School of Biotechnology, Faculty of Science, Banaras Hindu University, Varanasi, India
| | | | - Aafreen Zehra
- Biotechnology Division, CSIR-Central Institute of Medicinal and Aromatic Plants, Lucknow, India
| | - Gopal Nath
- Department of Microbiology, Institute of Medical Sciences, Banaras Hindu University, Varanasi, India
| | - Mukti Nath Mishra
- Department of Microbiology, Institute of Medical Sciences, Banaras Hindu University, Varanasi, India.,Biotechnology Division, CSIR-Central Institute of Medicinal and Aromatic Plants, Lucknow, India
| |
Collapse
|
37
|
Speri E, Fishovitz J, Mobashery S. Structure-activity relationship of the cinnamamide family of antibiotic potentiators for methicillin-resistant Staphylococcus aureus (MRSA). MEDCHEMCOMM 2018; 9:2008-2016. [PMID: 30746061 DOI: 10.1039/c8md00479j] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/24/2018] [Accepted: 10/19/2018] [Indexed: 11/21/2022]
Abstract
Methicillin-resistant Staphylococcus aureus (MRSA) is a global public health threat. MRSA has evolved a complex set of biochemical processes that mobilize the organism for inducible resistance on challenge by β-lactam antibiotics. Interfering pharmacologically with this machinery has the potential to reverse the β-lactam-resistance phenotype, whereby susceptibility to obsolete antibiotics would be restored. We describe herein our discovery of one class of such agents, the cinnamamide family of antibiotic potentiators. A hit compound of the class (compound 1) showed modest potentiation of the activity of oxacillin, a penicillin antibiotic, against an MRSA strain. A total of 50 analogues of compound 1 were prepared and screened. Seven of these compounds showed more dramatic potentiation of the antibacterial activity, which lowered the minimal-inhibitory concentrations (MICs) for the antibiotic by as much as 64- to 128-fold.
Collapse
Affiliation(s)
- Enrico Speri
- Department of Chemistry and Biochemistry , University of Notre Dame , Notre Dame , IN 46556 , USA .
| | - Jennifer Fishovitz
- 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 .
| |
Collapse
|
38
|
Frederick TE, Peng JW. A gratuitous β-Lactamase inducer uncovers hidden active site dynamics of the Staphylococcus aureus BlaR1 sensor domain. PLoS One 2018; 13:e0197241. [PMID: 29771929 PMCID: PMC5957439 DOI: 10.1371/journal.pone.0197241] [Citation(s) in RCA: 5] [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: 12/31/2017] [Accepted: 04/28/2018] [Indexed: 11/18/2022] Open
Abstract
Increasing evidence shows that active sites of proteins have non-trivial conformational dynamics. These dynamics include active site residues sampling different local conformations that allow for multiple, and possibly novel, inhibitor binding poses. Yet, active site dynamics garner only marginal attention in most inhibitor design efforts and exert little influence on synthesis strategies. This is partly because synthesis requires a level of atomic structural detail that is frequently missing in current characterizations of conformational dynamics. In particular, while the identity of the mobile protein residues may be clear, the specific conformations they sample remain obscure. Here, we show how an appropriate choice of ligand can significantly sharpen our abilities to describe the interconverting binding poses (conformations) of protein active sites. Specifically, we show how 2-(2'-carboxyphenyl)-benzoyl-6-aminopenicillanic acid (CBAP) exposes otherwise hidden dynamics of a protein active site that binds β-lactam antibiotics. When CBAP acylates (binds) the active site serine of the β-lactam sensor domain of BlaR1 (BlaRS), it shifts the time scale of the active site dynamics to the slow exchange regime. Slow exchange enables direct characterization of inter-converting protein and bound ligand conformations using NMR methods. These methods include chemical shift analysis, 2-d exchange spectroscopy, off-resonance ROESY of the bound ligand, and reduced spectral density mapping. The active site architecture of BlaRS is shared by many β-lactamases of therapeutic interest, suggesting CBAP could expose functional motions in other β-lactam binding proteins. More broadly, CBAP highlights the utility of identifying chemical probes common to structurally homologous proteins to better expose functional motions of active sites.
Collapse
Affiliation(s)
- Thomas E. Frederick
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, IN, United States of America
| | - Jeffrey W. Peng
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, IN, United States of America
- Department of Physics, University of Notre Dame, Notre Dame, IN, United States of America
| |
Collapse
|
39
|
Zhan XY, Zhu QY. Evolution of methicillin-resistant Staphylococcus aureus: Evidence of positive selection in a penicillin-binding protein (PBP) 2a coding gene mecA. INFECTION GENETICS AND EVOLUTION 2018; 59:16-22. [PMID: 29413881 DOI: 10.1016/j.meegid.2018.01.018] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/24/2017] [Revised: 01/19/2018] [Accepted: 01/22/2018] [Indexed: 11/15/2022]
Abstract
Methicillin-resistant Staphylococcus aureus (S. aureus) (MRSA) represents more and more S. aureus infections. MecA, the novel coding gene of penicillin-binding protein (PBP) 2a of MRSA, is the key resistance factor of β-lactam, but little is known about the evolution of this gene. Given the crucial role of mecA in S. aureus physiology and β-lactam resistance, the selective forces may contribute to adaptation of the bacteria to the special environments such as its host or antibiotics. To understand the evolution of this gene, we screened GenBank database and analyzed mecA of 249 S. aureus strains. Twenty-nine unique alleles with 26 unique amino acid sequences were identified. Phylogenetic analysis showed three main groups of mecA in the global S. aureus strains. Analysis of these alleles using codon-substitution models (M8, M3, and M2a) and likelihood ratio tests (LRTs) of the codeML package and a random-effects likelihood (REL) method of HyPhy package for the site-specific ratio of nonsynonymous to synonymous substitution rates suggested that fourteen sites in the allosteric domain of PBP2a have been subjected to strong positive selection pressure. Mutations of two positive selection sites (N146K and E239K) were reported to be essential for ceftaroline- or L-695, 256-resistant. Further study indicated that the positive selection pressure might be more likely related to the host's inflammatory or immune response during S. aureus infection. Our studies provide the first evidence of positive Darwinian selection in the mecA of S. aureus, contributing to a better understanding of the adaptive mechanism of this bacterium.
Collapse
Affiliation(s)
- Xiao-Yong Zhan
- Guangzhou KingMed Center for Clinical Laboratory, Guangzhou 510300, China; KingMed School of Laboratory Medicine, Guangzhou Medical University, Guangzhou 510300, China; The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou 510080, China.
| | - Qing-Yi Zhu
- Guangzhou KingMed Center for Clinical Laboratory, Guangzhou 510300, China; KingMed School of Laboratory Medicine, Guangzhou Medical University, Guangzhou 510300, China
| |
Collapse
|
40
|
Dahal M, Schwan WR. Management of methicillin-resistant Staphylococcus aureus mediated ventilator-associated pneumonia. CURRENT TRENDS IN MICROBIOLOGY 2018; 12:95-107. [PMID: 31341354 PMCID: PMC6656405] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Prevention strategies and clinical management of methicillin- resistant Staphylococcus aureus (MRSA) infections in ventilated patients who develop ventilator-associated pneumonia (VAP) are important. Since MRSA are the most frequently isolated bacteria in patients with VAP, and a significant cause of morbidity and mortality in intubated patients, rapid diagnosis and early treatment could reduce mortality. This review will examine preventive steps (i.e. screening ventilated patients for MRSA, decolonization, and hand washing), assessing clinical presentations before the results of culture are obtained to direct empiric treatment, and the appropriate antibiotic therapy upon culture confirmation of MRSA that could help in the management of VAP.
Collapse
|
41
|
Vermote A, Van Calenbergh S. Small-Molecule Potentiators for Conventional Antibiotics against Staphylococcus aureus. ACS Infect Dis 2017; 3:780-796. [PMID: 28889735 DOI: 10.1021/acsinfecdis.7b00084] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Antimicrobial resistance constitutes a global health problem, while the discovery and development of novel antibiotics is stagnating. Methicillin-resistant Staphylococcus aureus, responsible for the establishment of recalcitrant, biofilm-related infections, is a well-known and notorious example of a highly resistant micro-organism. Since resistance development is unavoidable with conventional antibiotics that target bacterial viability, it is vital to develop alternative treatment options on top. Strategies aimed at more subtle manipulation of bacterial behavior have recently attracted attention. Here, we provide a literature overview of several small-molecule potentiators for antibiotics, identified for the treatment of Staphylococcus aureus infection. Typically, these potentiators are not bactericidal by themselves and function by reversing resistance mechanisms, by attenuating Staphylococcus aureus virulence, and/or by interfering with quorum sensing.
Collapse
Affiliation(s)
- Arno Vermote
- Laboratory for Medicinal Chemistry, Ghent University, Ottergemsesteenweg 460, B-9000 Ghent, Belgium
| | - Serge Van Calenbergh
- Laboratory for Medicinal Chemistry, Ghent University, Ottergemsesteenweg 460, B-9000 Ghent, Belgium
| |
Collapse
|
42
|
Xu N, Cheng H, Xu J, Li F, Gao B, Li Z, Gao C, Huo K, Fu J, Xiong W. Silver-loaded nanotubular structures enhanced bactericidal efficiency of antibiotics with synergistic effect in vitro and in vivo. Int J Nanomedicine 2017; 12:731-743. [PMID: 28184157 PMCID: PMC5291465 DOI: 10.2147/ijn.s123648] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Antibiotic-resistant bacteria have become a major issue due to the long-term use and abuse of antibiotics in treatments in clinics. The combination therapy of antibiotics and silver (Ag) nanoparticles is an effective way of both enhancing the antibacterial effect and decreasing the usage of antibiotics. Although the method has been proved to be effective in vitro, no in vivo tests have been carried out at present. Herein, we described a combination therapy of local delivery of Ag and systemic antibiotics treatment in vitro in an infection model of rat. Ag nanoparticle-loaded TiO2 nanotube (NT) arrays (Ag-NTs) were fabricated on titanium implants for a customized release of Ag ion. The antibacterial properties of silver combined with antibiotics vancomycin, rifampin, gentamicin, and levofloxacin, respectively, were tested in vitro by minimum inhibitory concentration (MIC) assay, disk diffusion assay, and antibiofilm formation test. Enhanced antibacterial activity of combination therapy was observed for all the chosen bacterial strains, including gram-negative Escherichia coli (ATCC 25922), gram-positive Staphylococcus aureus (ATCC 25923), and methicillin-resistant Staphylococcus aureus (MRSA; ATCC 33591 and ATCC 43300). Moreover, after a relative short (3 weeks) combinational treatment, animal experiments in vivo further proved the synergistic antibacterial effect by X-ray and histological and immunohistochemical analyses. These results demonstrated that the combination of Ag nanoparticles and antibiotics significantly enhanced the antibacterial effect both in vitro and in vivo through the synergistic effect. The strategy is promising for clinical application to reduce the usage of antibiotics and shorten the administration time of implant-associated infection.
Collapse
Affiliation(s)
- Na Xu
- The State Key Laboratory of Refractories and Metallurgy, School of Materials and Metallurgy, Wuhan University of Science and Technology; Institute of Biology and Medicine, Wuhan University of Science and Technology
| | - Hao Cheng
- Orthopaedic Department, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China; Biomaterials Innovation Research Center, Division of Biomedical Engineering, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Jiangwen Xu
- The State Key Laboratory of Refractories and Metallurgy, School of Materials and Metallurgy, Wuhan University of Science and Technology
| | - Feng Li
- Orthopaedic Department, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Biao Gao
- The State Key Laboratory of Refractories and Metallurgy, School of Materials and Metallurgy, Wuhan University of Science and Technology
| | - Zi Li
- Orthopaedic Department, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Chenghao Gao
- Orthopaedic Department, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Kaifu Huo
- Wuhan National Laboratory for Optoelectronics, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Jijiang Fu
- The State Key Laboratory of Refractories and Metallurgy, School of Materials and Metallurgy, Wuhan University of Science and Technology; Institute of Biology and Medicine, Wuhan University of Science and Technology
| | - Wei Xiong
- Orthopaedic Department, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| |
Collapse
|
43
|
Stefano GB, Samuel J, Kream RM. Antibiotics May Trigger Mitochondrial Dysfunction Inducing Psychiatric Disorders. Med Sci Monit 2017; 23:101-106. [PMID: 28063266 PMCID: PMC5240889 DOI: 10.12659/msm.899478] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Clinical usage of several classes of antibiotics is associated with moderate to severe side effects due to the promotion of mitochondrial dysfunction. We contend that this may be due to perturbation of unique evolutionary relationships that link selective biochemical and molecular aspects of mitochondrial biology to conserved enzymatic processes derived from bacterial progenitors. Operationally, stereo-selective conformational matching between mitochondrial respiratory complexes, cytosolic and nuclear signaling complexes appears to support the conservation of a critically important set of chemical messengers required for existential regulation of homeostatic cellular processes. Accordingly, perturbation of normative mitochondrial function by select classes of antibiotics is certainly reflective of the high degree of evolutionary pressure designed to maintain ongoing bidirectional signaling processes between cellular compartments. These issues are of critical importance in evaluating potentially severe side effects of antibiotics on complex behavioral functions mediated by CNS neuronal groups. The CNS is extremely dependent on delivery of molecular oxygen for maintaining a required level of metabolic activity, as reflected by the high concentration of neuronal mitochondria. Thus, it is not surprising to find several distinct behavioral abnormalities conforming to established psychiatric criteria that are associated with antibiotic usage in humans. The manifestation of acute and/or chronic psychiatric conditions following antibiotic usage may provide unique insights into key etiological factors of major psychiatric syndromes that involve rundown of cellular bioenergetics via mitochondrial dysfunction. Thus, a potential window of opportunity exists for development of novel therapeutic agents targeting diminished mitochondrial function as a factor in severe behavioral disorders.
Collapse
Affiliation(s)
- George B Stefano
- Department of Research, MitoGenetics Research Institute, Farmingdale, NY, USA
| | - Joshua Samuel
- Department of Research, MitoGenetics Research Institute, Farmingdale, NY, USA
| | - Richard M Kream
- Department of Research, MitoGenetics Research Institute, Farmingdale, NY, USA
| |
Collapse
|
44
|
Lee SH, Wang H, Labroli M, Koseoglu S, Zuck P, Mayhood T, Gill C, Mann P, Sher X, Ha S, Yang SW, Mandal M, Yang C, Liang L, Tan Z, Tawa P, Hou Y, Kuvelkar R, DeVito K, Wen X, Xiao J, Batchlett M, Balibar CJ, Liu J, Xiao J, Murgolo N, Garlisi CG, Sheth PR, Flattery A, Su J, Tan C, Roemer T. TarO-specific inhibitors of wall teichoic acid biosynthesis restore β-lactam efficacy against methicillin-resistant staphylococci. Sci Transl Med 2016; 8:329ra32. [PMID: 26962156 DOI: 10.1126/scitranslmed.aad7364] [Citation(s) in RCA: 75] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
The widespread emergence of methicillin-resistant Staphylococcus aureus (MRSA) has dramatically eroded the efficacy of current β-lactam antibiotics and created an urgent need for new treatment options. We report an S. aureus phenotypic screening strategy involving chemical suppression of the growth inhibitory consequences of depleting late-stage wall teichoic acid biosynthesis. This enabled us to identify early-stage pathway-specific inhibitors of wall teichoic acid biosynthesis predicted to be chemically synergistic with β-lactams. We demonstrated by genetic and biochemical means that each of the new chemical series discovered, herein named tarocin A and tarocin B, inhibited the first step in wall teichoic acid biosynthesis (TarO). Tarocins do not have intrinsic bioactivity but rather demonstrated potent bactericidal synergy in combination with broad-spectrum β-lactam antibiotics against diverse clinical isolates of methicillin-resistant staphylococci as well as robust efficacy in a murine infection model of MRSA. Tarocins and other inhibitors of wall teichoic acid biosynthesis may provide a rational strategy to develop Gram-positive bactericidal β-lactam combination agents active against methicillin-resistant staphylococci.
Collapse
Affiliation(s)
- Sang Ho Lee
- Merck Research Laboratories, Kenilworth, NJ 07033, USA
| | - Hao Wang
- Merck Research Laboratories, Kenilworth, NJ 07033, USA
| | - Marc Labroli
- Merck Research Laboratories, West Point, PA 19486, USA
| | | | - Paul Zuck
- Merck Research Laboratories, West Point, PA 19486, USA
| | - Todd Mayhood
- Merck Research Laboratories, Kenilworth, NJ 07033, USA
| | - Charles Gill
- Merck Research Laboratories, Kenilworth, NJ 07033, USA
| | - Paul Mann
- Merck Research Laboratories, Kenilworth, NJ 07033, USA
| | - Xinwei Sher
- Merck Research Laboratories, Boston, MA 02115, USA
| | - Sookhee Ha
- Merck Research Laboratories, Kenilworth, NJ 07033, USA
| | - Shu-Wei Yang
- Merck Research Laboratories, Kenilworth, NJ 07033, USA
| | - Mihir Mandal
- Merck Research Laboratories, Kenilworth, NJ 07033, USA
| | | | - Lianzhu Liang
- Merck Research Laboratories, Kenilworth, NJ 07033, USA
| | - Zheng Tan
- Merck Research Laboratories, Kenilworth, NJ 07033, USA
| | - Paul Tawa
- Merck Research Laboratories, Kenilworth, NJ 07033, USA
| | - Yan Hou
- Merck Research Laboratories, Kenilworth, NJ 07033, USA
| | | | | | - Xiujuan Wen
- Merck Research Laboratories, Kenilworth, NJ 07033, USA
| | - Jing Xiao
- Merck Research Laboratories, Kenilworth, NJ 07033, USA
| | | | | | - Jenny Liu
- Merck Research Laboratories, Kenilworth, NJ 07033, USA
| | - Jianying Xiao
- Merck Research Laboratories, Kenilworth, NJ 07033, USA
| | | | | | - Payal R Sheth
- Merck Research Laboratories, Kenilworth, NJ 07033, USA
| | - Amy Flattery
- Merck Research Laboratories, Kenilworth, NJ 07033, USA
| | - Jing Su
- Merck Research Laboratories, Kenilworth, NJ 07033, USA.
| | | | - Terry Roemer
- Merck Research Laboratories, Kenilworth, NJ 07033, USA.
| |
Collapse
|
45
|
Waters EM, Rudkin JK, Coughlan S, Clair GC, Adkins JN, Gore S, Xia G, Black NS, Downing T, O'Neill E, Kadioglu A, O'Gara JP. Redeploying β-Lactam Antibiotics as a Novel Antivirulence Strategy for the Treatment of Methicillin-Resistant Staphylococcus aureus Infections. J Infect Dis 2016; 215:80-87. [PMID: 28077586 DOI: 10.1093/infdis/jiw461] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2016] [Accepted: 09/08/2016] [Indexed: 12/13/2022] Open
Abstract
Innovative approaches to the use of existing antibiotics is an important strategy in efforts to address the escalating antimicrobial resistance crisis. We report a new approach to the treatment of methicillin-resistant Staphylococcus aureus (MRSA) infections by demonstrating that oxacillin can be used to significantly attenuate the virulence of MRSA despite the pathogen being resistant to this drug. Using mechanistic in vitro assays and in vivo models of invasive pneumonia and sepsis, we show that oxacillin-treated MRSA strains are significantly attenuated in virulence. This effect is based primarily on the oxacillin-dependent repression of the accessory gene regulator quorum-sensing system and altered cell wall architecture, which in turn lead to increased susceptibility to host killing of MRSA. Our data indicate that β-lactam antibiotics should be included in the treatment regimen as an adjunct antivirulence therapy for patients with MRSA infections. This would represent an important change to current clinical practice for treatment of MRSA infection, with the potential to significantly improve patient outcomes in a safe, cost-effective manner.
Collapse
Affiliation(s)
- Elaine M Waters
- Department of Clinical Infection, Microbiology and Immunology, Institute of Infection and Global Health, University of Liverpool
| | | | - Simone Coughlan
- School of Mathematics, Statistics, and Applied Mathematics, National University of Ireland, Galway
| | - Geremy C Clair
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, Washington
| | - Joshua N Adkins
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, Washington
| | - Suzanna Gore
- Department of Clinical Infection, Microbiology and Immunology, Institute of Infection and Global Health, University of Liverpool
| | - Guoqing Xia
- Division of Infection, Immunity and Respiratory Medicine, Faculty of Biology, Medicine and Health, University of Manchester, United Kingdom
| | - Nikki S Black
- Department of Microbiology, School of Natural Sciences
| | - Tim Downing
- School of Mathematics, Statistics, and Applied Mathematics, National University of Ireland, Galway.,School of Biotechnology, Dublin City University
| | - Eoghan O'Neill
- Department of Clinical Microbiology, Royal College of Surgeons in Ireland, Connolly Hospital, Dublin
| | - Aras Kadioglu
- Department of Clinical Infection, Microbiology and Immunology, Institute of Infection and Global Health, University of Liverpool
| | | |
Collapse
|
46
|
Lavanya P, Ramaiah S, Anbarasu A. A Molecular Docking and Dynamics Study to Screen Potent Anti-Staphylococcal Compounds Against Ceftaroline Resistant MRSA. J Cell Biochem 2016; 117:542-8. [PMID: 26252252 DOI: 10.1002/jcb.25307] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2015] [Accepted: 07/28/2015] [Indexed: 01/16/2023]
Abstract
World Health Organization reports that methicillin-resistant Staphylococcus aureus (MRSA) is the origin of higher proportion of hospital acquired infections. In order to combat the effect of MRSA infection, an ideal drug should stimulate the allosteric exposure of active site, prompting penicillin binding proteins (PBP2a) to bind with that particular compound. Ceftaroline shows high binding affinity towards PBP2a and also confers resistance against degrading enzymes. Recently, two amino acid alterations in the allosteric site of PBP2a, asparagine (N) to lysine (K) at position 146 and glutamic acid (E) to lysine at position 150 are reported to confer resistance against ceftaroline resulting in the rise of ceftaroline-resistant MRSA strains. The present study focuses on the identification of potential ligands that can effectively bind with allosteric site of PBP2a, that leads to the access of active site and entry of a β-lactam antibiotic for effective inhibition. The results obtained from our study will be useful for designing effective compounds with potential therapeutic effects against ceftaroline resistant MRSA strains.
Collapse
Affiliation(s)
- P Lavanya
- Medical & Biological Computing Laboratory, School of Biosciences and Technology, VIT University, Vellore, 632014, Tamil Nadu, India
| | - Sudha Ramaiah
- Medical & Biological Computing Laboratory, School of Biosciences and Technology, VIT University, Vellore, 632014, Tamil Nadu, India
| | - Anand Anbarasu
- Medical & Biological Computing Laboratory, School of Biosciences and Technology, VIT University, Vellore, 632014, Tamil Nadu, India
| |
Collapse
|
47
|
Janardhanan J, Chang M, Mobashery S. The oxadiazole antibacterials. Curr Opin Microbiol 2016; 33:13-17. [PMID: 27239942 DOI: 10.1016/j.mib.2016.05.009] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2016] [Revised: 05/09/2016] [Accepted: 05/11/2016] [Indexed: 01/22/2023]
Abstract
The oxadiazoles are a class of antibacterials discovered by in silico docking and scoring of compounds against the X-ray structure of a penicillin-binding protein. These antibacterials exhibit activity against Gram-positive bacteria, including against methicillin-resistant Staphylococcus aureus (MRSA) and vancomycin-resistant Enterococcus (VRE). They show in vivo efficacy in murine models of peritonitis/sepsis and neutropenic thigh MRSA infection. They are bactericidal and orally bioavailable. The oxadiazoles show promise in treatment of MRSA infection.
Collapse
Affiliation(s)
- Jeshina Janardhanan
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, IN 46556, United States
| | - Mayland Chang
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, IN 46556, United States.
| | - Shahriar Mobashery
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, IN 46556, United States.
| |
Collapse
|
48
|
Mann PA, Müller A, Wolff KA, Fischmann T, Wang H, Reed P, Hou Y, Li W, Müller CE, Xiao J, Murgolo N, Sher X, Mayhood T, Sheth PR, Mirza A, Labroli M, Xiao L, McCoy M, Gill CJ, Pinho MG, Schneider T, Roemer T. Chemical Genetic Analysis and Functional Characterization of Staphylococcal Wall Teichoic Acid 2-Epimerases Reveals Unconventional Antibiotic Drug Targets. PLoS Pathog 2016; 12:e1005585. [PMID: 27144276 PMCID: PMC4856313 DOI: 10.1371/journal.ppat.1005585] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2015] [Accepted: 03/29/2016] [Indexed: 11/18/2022] Open
Abstract
Here we describe a chemical biology strategy performed in Staphylococcus aureus and Staphylococcus epidermidis to identify MnaA, a 2-epimerase that we demonstrate interconverts UDP-GlcNAc and UDP-ManNAc to modulate substrate levels of TarO and TarA wall teichoic acid (WTA) biosynthesis enzymes. Genetic inactivation of mnaA results in complete loss of WTA and dramatic in vitro β-lactam hypersensitivity in methicillin-resistant S. aureus (MRSA) and S. epidermidis (MRSE). Likewise, the β-lactam antibiotic imipenem exhibits restored bactericidal activity against mnaA mutants in vitro and concomitant efficacy against 2-epimerase defective strains in a mouse thigh model of MRSA and MRSE infection. Interestingly, whereas MnaA serves as the sole 2-epimerase required for WTA biosynthesis in S. epidermidis, MnaA and Cap5P provide compensatory WTA functional roles in S. aureus. We also demonstrate that MnaA and other enzymes of WTA biosynthesis are required for biofilm formation in MRSA and MRSE. We further determine the 1.9Å crystal structure of S. aureus MnaA and identify critical residues for enzymatic dimerization, stability, and substrate binding. Finally, the natural product antibiotic tunicamycin is shown to physically bind MnaA and Cap5P and inhibit 2-epimerase activity, demonstrating that it inhibits a previously unanticipated step in WTA biosynthesis. In summary, MnaA serves as a new Staphylococcal antibiotic target with cognate inhibitors predicted to possess dual therapeutic benefit: as combination agents to restore β-lactam efficacy against MRSA and MRSE and as non-bioactive prophylactic agents to prevent Staphylococcal biofilm formation. Staphylococcus aureus and Staphylococcus epidermidis cause life-threatening infections that are commonly acquired in hospitals as well as the community and remain difficult to treat with current antibiotics. In part, this is due to the emergence of methicillin-resistant S. aureus and S. epidermidis (MRSA and MRSE), which exhibit broad resistance to β-lactams such as penicillin and other members of this important founding class of antibiotics. Compounding this problem, Staphylococci commonly colonize the surface of catheters and other medical devices, forming bacterial communities that are intrinsically resistant to antibiotics. Here we functionally characterize a family of 2-epimerases, named MnaA and Cap5P, that we demonstrate by genetic, biochemical, and X-ray crystallography means are essential for wall teichoic acid biosynthesis and that upon their genetic inactivation render methicillin-resistant Staphylococci unable to form biofilms as well as broadly hypersusceptible to β-lactam antibiotics both in vitro and in a host infection setting. WTA 2-epimerases therefore constitute a novel class of methicillin-resistant Staphylococcal drug targets.
Collapse
Affiliation(s)
- Paul A. Mann
- Merck Research Laboratories, Kenilworth New Jersey, United States of America
| | - Anna Müller
- Institute for Pharmaceutical Microbiology, University of Bonn, Bonn, Germany
- German Centre for Infection Research (DZIF), partner site Bonn-Cologne, Bonn, Germany
| | - Kerstin A. Wolff
- Merck Research Laboratories, Kenilworth New Jersey, United States of America
| | - Thierry Fischmann
- Merck Research Laboratories, Kenilworth New Jersey, United States of America
| | - Hao Wang
- Merck Research Laboratories, Kenilworth New Jersey, United States of America
| | - Patricia Reed
- Laboratory of Bacterial Cell Biology, Instituto de Tecnologia Química e Biológica, Universidade Nova de Lisboa, Oeiras, Portugal
| | - Yan Hou
- Merck Research Laboratories, Kenilworth New Jersey, United States of America
| | - Wenjin Li
- PharmaCenter Bonn, Pharmaceutical Institute, Pharmaceutical Chemistry, University of Bonn, Bonn, Germany
| | - Christa E. Müller
- PharmaCenter Bonn, Pharmaceutical Institute, Pharmaceutical Chemistry, University of Bonn, Bonn, Germany
| | - Jianying Xiao
- Merck Research Laboratories, Kenilworth New Jersey, United States of America
| | - Nicholas Murgolo
- Merck Research Laboratories, Kenilworth New Jersey, United States of America
| | - Xinwei Sher
- Merck Research Laboratories, Kenilworth New Jersey, United States of America
| | - Todd Mayhood
- Merck Research Laboratories, Kenilworth New Jersey, United States of America
| | - Payal R. Sheth
- Merck Research Laboratories, Kenilworth New Jersey, United States of America
| | - Asra Mirza
- Merck Research Laboratories, Kenilworth New Jersey, United States of America
| | - Marc Labroli
- Merck Research Laboratories, Kenilworth New Jersey, United States of America
| | - Li Xiao
- Merck Research Laboratories, Kenilworth New Jersey, United States of America
| | - Mark McCoy
- Merck Research Laboratories, Kenilworth New Jersey, United States of America
| | - Charles J. Gill
- Merck Research Laboratories, Kenilworth New Jersey, United States of America
| | - Mariana G. Pinho
- Laboratory of Bacterial Cell Biology, Instituto de Tecnologia Química e Biológica, Universidade Nova de Lisboa, Oeiras, Portugal
| | - Tanja Schneider
- Institute for Pharmaceutical Microbiology, University of Bonn, Bonn, Germany
- German Centre for Infection Research (DZIF), partner site Bonn-Cologne, Bonn, Germany
| | - Terry Roemer
- Merck Research Laboratories, Kenilworth New Jersey, United States of America
- * E-mail:
| |
Collapse
|
49
|
Barbour PM, Wang W, Chang L, Pickard KL, Rais R, Slusher BS, Wang X. Property-Guided Synthesis of Aza-Tricyclic Indolines: Development of Gold Catalysis En Route. Adv Synth Catal 2016; 358:1482-1490. [PMID: 28360827 PMCID: PMC5367856 DOI: 10.1002/adsc.201501101] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Antibiotic resistance is a worldwide public health threat that needs to be addressed by improved antibiotic stewardship and continuing development of new chemical entities to treat resistant bacterial infections. Compounds that work alongside known antibiotics as combination therapies offer an efficient and sustainable approach to counteract antibiotic resistance in bacteria. Guided by property-based analysis, a series of aza-tricyclic indolines (ATIs) were synthesized to optimize their physiochemical properties as novel combination therapies with β-lactams to treat methicillin-resistant S. aureus (MRSA) infections. A novel and highly efficient gold-catalyzed tandem cyclization was developed to facilitate the synthesis of these ATIs. One guanidine-containing ATI was discovered to possess both improved anti-MRSA activity and lower mammalian toxicity both in vitro and in vivo. In addition, it also showed significantly enhanced aqueous solubility and metabolic stability. These results indicated that the ATIs are a novel class of anti-MRSA agents suitable for further evaluations as adjuvant therapy in animal model studies.
Collapse
Affiliation(s)
- Patrick M. Barbour
- Department of Chemistry and Biochemistry, University of Colorado, Boulder, CO 80309, USA
| | - Wei Wang
- Department of Chemistry and Biochemistry, University of Colorado, Boulder, CO 80309, USA
| | - Le Chang
- Department of Chemistry and Biochemistry, University of Colorado, Boulder, CO 80309, USA
| | - Kasey L. Pickard
- Department of Chemistry and Biochemistry, University of Colorado, Boulder, CO 80309, USA
| | - Rana Rais
- Department of Neurology and Johns Hopkins Drug Discovery, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Barbara S. Slusher
- Department of Neurology and Johns Hopkins Drug Discovery, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Xiang Wang
- Department of Chemistry and Biochemistry, University of Colorado, Boulder, CO 80309, USA
| |
Collapse
|
50
|
Co-encapsulation of multi-lipids and polymers enhances the performance of vancomycin in lipid–polymer hybrid nanoparticles: In vitro and in silico studies. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2016; 61:616-30. [DOI: 10.1016/j.msec.2015.12.053] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/21/2015] [Revised: 11/16/2015] [Accepted: 12/23/2015] [Indexed: 12/19/2022]
|