1
|
Dimas DA, Kumar V, Mandal PS, Masterson JM, Tonelli M, Singh S. Chemoenzymatic Modification of Daptomycin: Aromatic Group Installation on Trp1. Chembiochem 2024; 25:e202400503. [PMID: 39019798 PMCID: PMC11576237 DOI: 10.1002/cbic.202400503] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2024] [Revised: 07/13/2024] [Accepted: 07/17/2024] [Indexed: 07/19/2024]
Abstract
Daptomycin is a cyclic lipodepsipeptide antibiotic used to treat infections caused by Gram-positive pathogens, including multi-drug resistant strains such as methicillin-resistant Staphylococcus aureus (MRSA) and vancomycin-resistant enterococci (VRE). The emergence of daptomycin-resistant bacterial strains has renewed interest in generating daptomycin analogs. Previous studies have shown that replacing the tryptophan of daptomycin with aromatic groups can generate analogs with enhanced potency. Additionally, we have demonstrated that aromatic prenyltransferases can attach diverse groups to the tryptophan of daptomycin. Here, we report the use of the prenyltransferase CdpNPT to derivatize the tryptophan of daptomycin with a library of benzylic and heterocyclic pyrophosphates. An analytical-scale study revealed that CdpNPT can transfer various aromatic groups onto daptomycin. Subsequent scaled-up and purified reactions indicated that the enzyme can attach aromatic groups to N1, C2, C5 and C6 positions of Trp1 of daptomycin. In vitro antibacterial activity assays using six of these purified compounds identified aromatic substituted daptomycin analogs show potency against both daptomycin-susceptible and resistant strains of Gram-positive bacteria. These findings suggest that installing aromatic groups on the Trp1 of daptomycin can lead to the generation of potent daptomycin analogs.
Collapse
Affiliation(s)
- Dustin A Dimas
- Department of Chemistry and Biochemistry, University of Oklahoma, Stephenson Life Sciences Research Center, 101 Stephenson Parkway, Norman, Oklahoma, 73019, United States
| | - Vikas Kumar
- Department of Chemistry and Biochemistry, University of Oklahoma, Stephenson Life Sciences Research Center, 101 Stephenson Parkway, Norman, Oklahoma, 73019, United States
| | - Prashant S Mandal
- Department of Chemistry and Biochemistry, University of Oklahoma, Stephenson Life Sciences Research Center, 101 Stephenson Parkway, Norman, Oklahoma, 73019, United States
| | - Johanna M Masterson
- Department of Chemistry and Biochemistry, University of Oklahoma, Stephenson Life Sciences Research Center, 101 Stephenson Parkway, Norman, Oklahoma, 73019, United States
| | - Marco Tonelli
- National Magnetic Resonance Facility at Madison, University of Wisconsin-Madison, 411 Babcock Drive, Madison, Wisconsin, 45005, United States
| | - Shanteri Singh
- Department of Chemistry and Biochemistry, University of Oklahoma, Stephenson Life Sciences Research Center, 101 Stephenson Parkway, Norman, Oklahoma, 73019, United States
| |
Collapse
|
2
|
Brdová D, Ruml T, Viktorová J. Mechanism of staphylococcal resistance to clinically relevant antibiotics. Drug Resist Updat 2024; 77:101147. [PMID: 39236354 DOI: 10.1016/j.drup.2024.101147] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Revised: 08/17/2024] [Accepted: 08/26/2024] [Indexed: 09/07/2024]
Abstract
Staphylococcus aureus, a notorious pathogen with versatile virulence, poses a significant challenge to current antibiotic treatments due to its ability to develop resistance mechanisms against a variety of clinically relevant antibiotics. In this comprehensive review, we carefully dissect the resistance mechanisms employed by S. aureus against various antibiotics commonly used in clinical settings. The article navigates through intricate molecular pathways, elucidating the mechanisms by which S. aureus evades the therapeutic efficacy of antibiotics, such as β-lactams, vancomycin, daptomycin, linezolid, etc. Each antibiotic is scrutinised for its mechanism of action, impact on bacterial physiology, and the corresponding resistance strategies adopted by S. aureus. By synthesising the knowledge surrounding these resistance mechanisms, this review aims to serve as a comprehensive resource that provides a foundation for the development of innovative therapeutic strategies and alternative treatments for S. aureus infections. Understanding the evolving landscape of antibiotic resistance is imperative for devising effective countermeasures in the battle against this formidable pathogen.
Collapse
Affiliation(s)
- Daniela Brdová
- Department of Biochemistry and Microbiology, University of Chemistry and Technology, Technicka 3, Prague 16628, Czech Republic.
| | - Tomáš Ruml
- Department of Biochemistry and Microbiology, University of Chemistry and Technology, Technicka 3, Prague 16628, Czech Republic.
| | - Jitka Viktorová
- Department of Biochemistry and Microbiology, University of Chemistry and Technology, Technicka 3, Prague 16628, Czech Republic.
| |
Collapse
|
3
|
Madison CL, Steinert ASJ, Luedeke CE, Hajjafar N, Srivastava P, Berti AD, Bayer AS, Kebriaei R. It takes two to tango: Preserving daptomycin efficacy against daptomycin-resistant MRSA using daptomycin-phage co-therapy. Microbiol Spectr 2024; 12:e0067924. [PMID: 39470283 PMCID: PMC11619598 DOI: 10.1128/spectrum.00679-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Accepted: 09/23/2024] [Indexed: 10/30/2024] Open
Abstract
Daptomycin (DAP) is a cyclic lipopeptide antibiotic that targets the cell membrane and is commonly used for the treatment of multidrug-resistant Staphylococcus aureus infections, especially methicillin-resistant strains (MRSA). Emergence of DAP resistance during DAP therapy is increasingly reported; however, the mechanisms by which resistance occurs are diverse. Using phages in combination with antibiotics is of high interest to circumvent antibiotic resistance, due to their minimal side effects, accessibility, and synergistic effects with antibiotics. Here, we have investigated the relationship between DAP resistance and sensitivity to phage Sb-1 in a cohort of 14 clinical MRSA strains. This cohort consists of four isogenic pairs (a clinical DAP-susceptible parent and its isogenic DAP-resistant variant isolated during DAP treatment), and three DAP-susceptible/DAP-resistant strain pairs are created by traditional serial passaging in the presence of increasing DAP concentrations. Efficiency of plating (EOP) and bacteriophage plaque sizes were recorded, and synergy between the antibacterial agents was tested using the phage-antibiotic combination (PAC) checkerboard method. Plaque sizes and EOP significantly increased (P = 0.019 and P = 0.008, respectively) as strains developed resistance to DAP. The PAC checkerboards conveyed less antagonistic patterns in DAP-resistant mutant strains compared with their DAP-susceptible counterparts. Antagonism was displayed in 71.4% (5/7) of the DAP-susceptible strains, while it was only present in 14.3% (1/7) of the DAP-resistant mutants. Transmission electron microscopy images showed significantly thicker cell walls in DAP-susceptible strains exposed to DAP monotherapy compared with either growth control or DAP-phage (P = 0.0002 and 0.0007, respectively). These data indicate increased sensitivity to phage Sb-1 concurrent with the emergence of DAP resistance. IMPORTANCE Multidrug-resistant Staphylococcus aureus is a threat to the health care system, especially cross-resistance between daptomycin (DAP) and glycopeptides through various mutations such as mprF (which is involved in the modification of membrane phospholipids in some bacteria) and yycG (part of a two-component regulatory system in bacteria that is important for regulating cell wall biosynthesis and other cellular processes) has been reported previously. Our current study shows adjunctive treatment with phage in DAP-resistant strains will lead to synergistic activity and larger phage plaque sizes, translating to elevated lytic performance. The addition of bacteriophage to standard-of-care antibiotic therapies for multidrug-resistant S. aureus infections has the potential to hinder, and possibly revert, resistance to antibiotics. Applying this strategy can potentially lead to the preservation of the current antibiotics. Verification of this salutary outcome in relevant ex vivo and in vivo models of endovascular infections is required to validate translatability.
Collapse
Affiliation(s)
- Casey L. Madison
- P3 Research Laboratory, Division of Pharmaceutics and Pharmacology, College of Pharmacy, The Ohio State University, Columbus, Ohio, USA
| | - Anja S. J. Steinert
- P3 Research Laboratory, Division of Pharmaceutics and Pharmacology, College of Pharmacy, The Ohio State University, Columbus, Ohio, USA
| | - Corrin E. Luedeke
- P3 Research Laboratory, Division of Pharmaceutics and Pharmacology, College of Pharmacy, The Ohio State University, Columbus, Ohio, USA
| | - Neeka Hajjafar
- P3 Research Laboratory, Division of Pharmaceutics and Pharmacology, College of Pharmacy, The Ohio State University, Columbus, Ohio, USA
| | - Prakhar Srivastava
- P3 Research Laboratory, Division of Pharmaceutics and Pharmacology, College of Pharmacy, The Ohio State University, Columbus, Ohio, USA
| | - Andrew D. Berti
- Department of Pharmacy Practice, College of Pharmacy, Wayne State University, Detroit, Michigan, USA
| | - Arnold S. Bayer
- David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, USA
- The Lundquist Institute at Harbor-UCLA Medical Center, Torrance, California, USA
| | - Razieh Kebriaei
- P3 Research Laboratory, Division of Pharmaceutics and Pharmacology, College of Pharmacy, The Ohio State University, Columbus, Ohio, USA
| |
Collapse
|
4
|
Xu Y, Xiao Y, Zhao H, Wang B, Yu J, Shang Y, Zhou Y, Wu X, Guo Y, Yu F. Phenotypic and genetic characterization of daptomycin non-susceptible Staphylococcus aureus strains selected by adaptive laboratory evolution. Front Cell Infect Microbiol 2024; 14:1453233. [PMID: 39512591 PMCID: PMC11540788 DOI: 10.3389/fcimb.2024.1453233] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2024] [Accepted: 09/30/2024] [Indexed: 11/15/2024] Open
Abstract
Background Daptomycin non-susceptible Staphylococcus aureus (DNS) strains pose a serious clinical threat, yet their characteristics remain poorly understood. Methods DNS derivatives were generated by exposing S. aureus strains to subinhibitory concentrations of daptomycin. Competition experiment and growth kinetics experiment were used to observe the growth of bacteria. Galleria mellonella larvae and mouse skin abscess models were used to observe the virulence of bacteria. Transmission electron microscopy (TEM), cytochrome C experiment and biofilm formation experiment were used to observe the drug resistance phenotype. And homologous recombination was used to study the role of mutations. Results Phenotypic profiling of DNS strains revealed impaired growth, increased cell wall thickness, enhanced biofilm formation, reduced negative surface charge, and attenuated virulence compared to their wild-type strains. Whole genome sequencing identified mutations in mprF, cls2, and saeR in DNS strains. Allelic replacement experiments validated the roles of MprF L341F and Cls2 F60S substitutions in augmenting daptomycin non-susceptibility in Newman. Deletion of saeR in the NewmanMprFL341F strain and complementation of saeR in the Newman-DNS strain did not directly alter daptomycin susceptibility. However, the deletion of saeR was found to enhance competitive fitness under daptomycin pressure. Conclusion This work validates adaptive laboratory evolution (ALE) for modeling clinical DNS strains and uncovers contributions of mprF, cls2, and saeR mutations to the adaptation and resistance mechanisms of S. aureus against daptomycin. These findings enrich our understanding of how S. aureus acquired resistance to daptomycin, thus paving the way for the development of more effective treatment strategies and offering potential molecular markers for resistance surveillance.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Yinjuan Guo
- Department of Clinical Laboratory Medicine, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Fangyou Yu
- Department of Clinical Laboratory Medicine, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| |
Collapse
|
5
|
McNeil JC, Sommer LM, Joseph M, Hulten KG, Kaplan SL. Penicillin susceptibility among Staphylococcus aureus skin and soft tissue infections at a children's hospital. Microbiol Spectr 2024; 12:e0086924. [PMID: 39248483 PMCID: PMC11448063 DOI: 10.1128/spectrum.00869-24] [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: 04/05/2024] [Accepted: 08/13/2024] [Indexed: 09/10/2024] Open
Abstract
Shortly after its introduction into clinical practice, Staphylococcus aureus isolates gained resistance to penicillin via the acquisition of β-lactamases. A number of centers have recently described an increase in the proportion of invasive methicillin-susceptible S. aureus (MSSA), which are also susceptible to penicillin (PSSA). Little data are available regarding the prevalence or impact of PSSA in skin and soft tissue infections (SSTI). Community-acquired MSSA SSTI isolates were obtained through a surveillance study at Texas Children's Hospital from January 2017 to December 2021. A total of 200 random isolates underwent PCR for blaZ β-lactamase; blaZ-negative isolates then underwent penicillin susceptibility testing using macrobroth dilution. Isolates which were blaZ negative and had a penicillin MIC ≤0.125 µg/mL were regarded as PSSA with the remainder regarded as penicillin-resistant MSSA (PR-MSSA). All PSSA underwent multilocus sequence typing. Medical records were reviewed. The median age of subjects was 4.2 years (IQR: 1.6-10.5). PSSA accounted for 9% of isolates during the study period. PSSA and PR-MSSA cases were similar with respect to age, demographics, and rates of prior antibiotic exposure. PSSA isolates less often had vancomycin MIC ≥1.5 µg/mL. Furthermore, 39% of PSSA were variants of sequence type 1. In multivariable analyses, penicillin susceptibility was independently associated with both hospital admission and surgical intervention. PSSA account for a small but significant proportion of MSSA SSTI in children. Clinically distinguishing patients with PSSA and PR-MSSA SSTI is challenging. However, PSSA SSTI were independently associated with higher rates of hospital admission as well as the need for surgical intervention suggesting a significant clinical impact.IMPORTANCEThe vast majority of Staphylococcus aureus in the US are penicillin resistant with most clinical labs no longer reporting penicillin susceptibility for this organism. A number of centers, however, have reported increasing penicillin susceptibility among invasive S. aureus infections. Skin and soft tissue infections (SSTI) are far more common than invasive infections, yet the frequency and impact of penicillin-susceptible S. aureus (PSSA) in this population are uncertain. Through active surveillance at a children's hospital, we found that 9% of methicillin-susceptible S. aureus SSTI isolates were PSSA. PSSA were independently associated with hospital admission for the management of SSTI as well as the need for debridement in the operating room. Given that most SSTI are managed in the outpatient setting, these findings suggest a clinical impact of this phenotype and the need for a reassessment of the value in susceptibility testing and potentially even treatment with penicillin.
Collapse
Affiliation(s)
- J. Chase McNeil
- Department of Pediatrics, Division of Infectious Diseases, Baylor College of Medicine and Texas Children’s Hospital, Houston, Texas, USA
| | - Lauren M. Sommer
- Department of Pediatrics, Division of Infectious Diseases, Baylor College of Medicine and Texas Children’s Hospital, Houston, Texas, USA
| | - Marritta Joseph
- Department of Pediatrics, Division of Infectious Diseases, Baylor College of Medicine and Texas Children’s Hospital, Houston, Texas, USA
| | - Kristina G. Hulten
- Department of Pediatrics, Division of Infectious Diseases, Baylor College of Medicine and Texas Children’s Hospital, Houston, Texas, USA
| | - Sheldon L. Kaplan
- Department of Pediatrics, Division of Infectious Diseases, Baylor College of Medicine and Texas Children’s Hospital, Houston, Texas, USA
| |
Collapse
|
6
|
Freeman CD, Hansen T, Urbauer R, Wilkinson BJ, Singh VK, Hines KM. Defective pgsA contributes to increased membrane fluidity and cell wall thickening in Staphylococcus aureus with high-level daptomycin resistance. mSphere 2024; 9:e0011524. [PMID: 38752757 PMCID: PMC11332330 DOI: 10.1128/msphere.00115-24] [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: 02/14/2024] [Accepted: 04/17/2024] [Indexed: 05/28/2024] Open
Abstract
Daptomycin is a membrane-targeting last-resort antimicrobial therapeutic for the treatment of infections caused by methicillin- and/or vancomycin-resistant Staphylococcus aureus. In the rare event of failed daptomycin therapy, the source of resistance is often attributable to mutations directly within the membrane phospholipid biosynthetic pathway of S. aureus or in the regulatory systems that control cell envelope response and membrane homeostasis. Here we describe the structural changes to the cell envelope in a daptomycin-resistant isolate of S. aureus strain N315 that has acquired mutations in the genes most commonly reported associated with daptomycin resistance: mprF, yycG, and pgsA. In addition to the decreased phosphatidylglycerol (PG) levels that are the hallmark of daptomycin resistance, the mutant with high-level daptomycin resistance had increased branched-chain fatty acids (BCFAs) in its membrane lipids, increased membrane fluidity, and increased cell wall thickness. However, the successful utilization of isotope-labeled straight-chain fatty acids (SCFAs) in lipid synthesis suggested that the aberrant BCFA:SCFA ratio arose from upstream alteration in fatty acid synthesis rather than a structural preference in PgsA. Transcriptomics studies revealed that expression of pyruvate dehydrogenase (pdhB) was suppressed in the daptomycin-resistant isolate, which is known to increase BCFA levels. While complementation with an additional copy of pdhB had no effect, complementation of the pgsA mutation resulted in increased PG formation, reduction in cell wall thickness, restoration of normal BCFA levels, and increased daptomycin susceptibility. Collectively, these results demonstrate that pgsA contributes to daptomycin resistance through its influence on membrane fluidity and cell wall thickness, in addition to phosphatidylglycerol levels. IMPORTANCE The cationic lipopeptide antimicrobial daptomycin has become an essential tool for combating infections with Staphylococcus aureus that display reduced susceptibility to β-lactams or vancomycin. Since daptomycin's activity is based on interaction with the negatively charged membrane of S. aureus, routes to daptomycin-resistance occur through mutations in the lipid biosynthetic pathway surrounding phosphatidylglycerols and the regulatory systems that control cell envelope homeostasis. Therefore, there are many avenues to achieve daptomycin resistance and several different, and sometimes contradictory, phenotypes of daptomycin-resistant S. aureus, including both increased and decreased cell wall thickness and membrane fluidity. This study is significant because it demonstrates the unexpected influence of a lipid biosynthesis gene, pgsA, on membrane fluidity and cell wall thickness in S. aureus with high-level daptomycin resistance.
Collapse
Affiliation(s)
| | - Tayte Hansen
- Department of Microbiology and Immunology, A.T. Still University of Health Sciences, Kirksville, Missouri, USA
| | - Ramona Urbauer
- Department of Chemistry, University of Georgia, Athens, Georgia, USA
| | - Brian J. Wilkinson
- School of Biological Sciences, Illinois State University, Normal, Illinois, USA
| | - Vineet K. Singh
- Department of Microbiology and Immunology, A.T. Still University of Health Sciences, Kirksville, Missouri, USA
| | - Kelly M. Hines
- Department of Chemistry, University of Georgia, Athens, Georgia, USA
| |
Collapse
|
7
|
Cao R, Su H, Wei Z, He Z, Pan T, Li Y, Sun B. An induced mutation of ABC-transporter component VraF(K84E) contributes to vancomycin resistance and virulence in Staphylococcus aureus strain MW2. Int J Med Microbiol 2024; 315:151624. [PMID: 38838390 DOI: 10.1016/j.ijmm.2024.151624] [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: 02/04/2024] [Revised: 05/22/2024] [Accepted: 05/23/2024] [Indexed: 06/07/2024] Open
Abstract
Staphylococcus aureus is a notorious pathogen responsible for various severe diseases. Due to the emergence of drug-resistant strains, the prevention and treatment of S. aureus infections have become increasingly challenging. Vancomycin is considered to be one of the last-resort drugs for treating most methicillin-resistant S. aureus (MRSA), so it is of great significance to further reveal the mechanism of vancomycin resistance. VraFG is one of the few important ABC (ATP-binding cassette) transporters in S. aureus that can form TCS (two-component systems)/ABC transporter modules. ABC transporters can couple the energy released from ATP hydrolysis to translocate solutes across the cell membrane. In this study, we obtained a strain with decreased vancomycin susceptibility after serial passaging and selection. Subsequently, whole-genome sequencing was performed on this laboratory-derived strain MWA2 and a novel single point mutation was discovered in vraF gene, leading to decreased sensitivity to vancomycin and daptomycin. Furthermore, the mutation reduces autolysis of S. aureus and downregulates the expression of lytM, isaA, and atlA. Additionally, we observed that the mutant has a less net negative surface charge than wild-type strain. We also noted an increase in the expression of the dlt operon and mprF gene, which are associated with cell surface charge and serve to hinder the binding of cationic peptides by promoting electrostatic repulsion. Moreover, this mutation has been shown to enhance hemolytic activity, expand subcutaneous abscesses, reflecting an increased virulence. This study confirms the impact of a point mutation of VraF on S. aureus antibiotic resistance and virulence, contributing to a broader understanding of ABC transporter function and providing new targets for treating S. aureus infections.
Collapse
Affiliation(s)
- Ruobing Cao
- Institute of Advanced Technology, University of Science and Technology of China, Hefei, China
| | - Huimin Su
- Department of Oncology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui 230027, China
| | - Zichun Wei
- Institute of Advanced Technology, University of Science and Technology of China, Hefei, China
| | - Zhien He
- Department of Oncology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui 230027, China
| | - Ting Pan
- Department of Oncology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui 230027, China
| | - Yujie Li
- Department of Oncology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui 230027, China.
| | - Baolin Sun
- Institute of Advanced Technology, University of Science and Technology of China, Hefei, China; Department of Oncology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui 230027, China.
| |
Collapse
|
8
|
Donnelly J, McDermott H, Gash S, O'Connor C, O'Connell K, O'Donnell S, Dinesh B, Burns K, Fitzpatrick F. Getting to the heart of the matter-are two agents really better than one for the treatment of staphylococcal infective endocarditis? Int J Infect Dis 2024; 142:106975. [PMID: 38395218 DOI: 10.1016/j.ijid.2024.02.018] [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: 11/09/2023] [Revised: 02/18/2024] [Accepted: 02/19/2024] [Indexed: 02/25/2024] Open
Abstract
The recently published European Society of Cardiology guidelines for infective endocarditis management recommends daptomycin combination therapy for the treatment of staphylococcal endocarditis in severe penicillin allergy, rather than daptomycin monotherapy. We discuss the evidence base behind this recommendation, highlighting concerns regarding the lack of robust clinical studies, increased cost and logistical considerations, and adverse effects of combination therapy. Although further studies are required to elucidate the role of combination vs monotherapy in these patients, we propose a pragmatic management approach to reduce the risk of adverse antimicrobial side effects and limit costs, while aiming to maintain treatment efficacy.
Collapse
Affiliation(s)
- James Donnelly
- Department of Clinical Microbiology, Beaumont Hospital, Dublin, Ireland.
| | - Helene McDermott
- Department of Clinical Microbiology, Beaumont Hospital, Dublin, Ireland
| | - Sadhbh Gash
- Department of Pharmacy, Beaumont Hospital, Dublin, Ireland
| | - Ciara O'Connor
- Department of Clinical Microbiology, Beaumont Hospital, Dublin, Ireland
| | - Karina O'Connell
- Department of Clinical Microbiology, Beaumont Hospital, Dublin, Ireland; Department of Clinical Microbiology, The Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Sinead O'Donnell
- Department of Clinical Microbiology, Beaumont Hospital, Dublin, Ireland; Department of Clinical Microbiology, The Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Binu Dinesh
- Department of Clinical Microbiology, Beaumont Hospital, Dublin, Ireland; Department of Clinical Microbiology, The Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Karen Burns
- Department of Clinical Microbiology, Beaumont Hospital, Dublin, Ireland; Department of Clinical Microbiology, The Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Fidelma Fitzpatrick
- Department of Clinical Microbiology, Beaumont Hospital, Dublin, Ireland; Department of Clinical Microbiology, The Royal College of Surgeons in Ireland, Dublin, Ireland
| |
Collapse
|
9
|
Wagner TM, Pöntinen AK, Al Rubaye M, Sundsfjord A, Hegstad K. Adaptive cell wall thickening in Enterococcus faecalis is associated with decreased vancomycin susceptibility. Clin Microbiol Infect 2024; 30:396.e1-396.e5. [PMID: 38065364 DOI: 10.1016/j.cmi.2023.12.002] [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: 05/05/2023] [Revised: 11/28/2023] [Accepted: 12/02/2023] [Indexed: 12/25/2023]
Abstract
OBJECTIVES Enterococcus faecalis can adopt both a commensal and a nosocomial lifestyle, resisting numerous antibiotics. In this study, we aim to investigate the relationship between the cell wall (CW) thickness and decreased susceptibility to vancomycin (VD) in van-gene negative clinical isolates of E. faecalis (nMIC 8 = 2, nMIC 4 = 3, ST30, ST40, and ST59). METHODS The CW thickness was assessed in VD strains and compared with vancomycin susceptible isolates of the same sequence type (ST) (Vancomycin susceptible [VS]; nMIC 2 = 5). The VD and VS strains were subjected to serial passage (evolved [ev]) with and without vancomycin selection. Subsequent measurements of CW thickness and vancomycin MICs were performed. RESULTS The VD strains exhibited increased CW thickness when compared with ST-related VS strains (ΔCW thickness VD vs. VS ST30 25 nm, ST59 15 nm, and ST40 1 nm). Serial passages without vancomycin selection led to a decrease in CW thickness and vancomycin MIC in VD strains (ΔCW thickness VD vs. evVD ST30 22 nm, ST59 3 nm, and ST40 2 nm). Serial passages with vancomycin selection caused an increase in CW thickness and vancomycin MIC in ST-related VS strains (ΔCW thickness VS vs. evVS ST30 22 nm, ST59 16 nm, and ST40 1 nm). DISCUSSION Adaptive changes in CW thickness were observed in response to vancomycin exposure. Increased CW thickness correlated with decreased vancomycin susceptibility, whereas decreased CW thickness correlated with increased vancomycin susceptibility. Core single nucleotide polymorphisms in the evolved mutants were mostly found in genes encoding proteins associated with the cytoplasm or the cytoplasmic membrane. The potential relevance of these adaptive changes is underlined by the observed phenotypes in clinical isolates. Our findings emphasize the importance of monitoring adaptive changes, as vancomycin-resistant enterococci infections are a growing concern.
Collapse
Affiliation(s)
- Theresa Maria Wagner
- Research Group for Host-Microbe Interactions, Department of Medical Biology, Faculty of Health Sciences, UiT The Arctic University of Norway, Tromsø, Norway
| | - Anna K Pöntinen
- Norwegian National Advisory Unit on Detection of Antimicrobial Resistance, Department of Microbiology and Infection Control, University Hospital of North Norway, Tromsø, Norway; Department of Biostatistics, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Mushtaq Al Rubaye
- Research Group for Host-Microbe Interactions, Department of Medical Biology, Faculty of Health Sciences, UiT The Arctic University of Norway, Tromsø, Norway
| | - Arnfinn Sundsfjord
- Research Group for Host-Microbe Interactions, Department of Medical Biology, Faculty of Health Sciences, UiT The Arctic University of Norway, Tromsø, Norway; Norwegian National Advisory Unit on Detection of Antimicrobial Resistance, Department of Microbiology and Infection Control, University Hospital of North Norway, Tromsø, Norway
| | - Kristin Hegstad
- Research Group for Host-Microbe Interactions, Department of Medical Biology, Faculty of Health Sciences, UiT The Arctic University of Norway, Tromsø, Norway; Norwegian National Advisory Unit on Detection of Antimicrobial Resistance, Department of Microbiology and Infection Control, University Hospital of North Norway, Tromsø, Norway.
| |
Collapse
|
10
|
Faure A, Manuse S, Gonin M, Grangeasse C, Jault JM, Orelle C. Daptomycin avoids drug resistance mediated by the BceAB transporter in Streptococcus pneumoniae. Microbiol Spectr 2024; 12:e0363823. [PMID: 38214521 PMCID: PMC10846014 DOI: 10.1128/spectrum.03638-23] [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: 10/12/2023] [Accepted: 12/19/2023] [Indexed: 01/13/2024] Open
Abstract
Drug-resistant bacteria are a serious threat to human health as antibiotics are gradually losing their clinical efficacy. Comprehending the mechanism of action of antimicrobials and their resistance mechanisms plays a key role in developing new agents to fight antimicrobial resistance. The lipopeptide daptomycin is an antibiotic that selectively disrupts Gram-positive bacterial membranes, thereby showing slower resistance development than many classical drugs. Consequently, it is often used as a last resort antibiotic to preserve its use as one of the least potent antibiotics at our disposal. The mode of action of daptomycin has been debated but was recently found to involve the formation of a tripartite complex between undecaprenyl precursors of cell wall biosynthesis and the anionic phospholipid phosphatidylglycerol. BceAB-type ABC transporters are known to confer resistance to antimicrobial peptides that sequester some precursors of the peptidoglycan, such as the undecaprenyl pyrophosphate or lipid II. The expression of these transporters is upregulated by dedicated two-component regulatory systems in the presence of antimicrobial peptides that are recognized by the system. Here, we investigated whether daptomycin evades resistance mediated by the BceAB transporter from the bacterial pathogen Streptococcus pneumoniae. Although daptomycin can bind to the transporter, our data showed that the BceAB transporter does not mediate resistance to the drug and its expression is not induced in its presence. These findings show that the pioneering membrane-active daptomycin has the potential to escape the resistance mechanism mediated by BceAB-type transporters and confirm that the development of this class of compounds has promising clinical applications.IMPORTANCEAntibiotic resistance is rising in all parts of the world. New resistance mechanisms are emerging and dangerously spreading, threatening our ability to treat common infectious diseases. Daptomycin is an antimicrobial peptide that is one of the last antibiotics approved for clinical use. Understanding the resistance mechanisms toward last-resort antibiotics such as daptomycin is critical for the success of future antimicrobial therapies. BceAB-type ABC transporters confer resistance to antimicrobial peptides that target precursors of cell-wall synthesis. In this study, we showed that the BceAB transporter from the human pathogen Streptococcus pneumoniae does not confer resistance to daptomycin, suggesting that this drug and other calcium-dependent lipopeptide antibiotics have the potential to evade the action of this type of ABC transporters in other bacterial pathogens.
Collapse
Affiliation(s)
- Agathe Faure
- Molecular Microbiology and Structural Biochemistry (MMSB), UMR 5086 CNRS/University of Lyon, Lyon, France
| | - Sylvie Manuse
- Molecular Microbiology and Structural Biochemistry (MMSB), UMR 5086 CNRS/University of Lyon, Lyon, France
| | - Mathilde Gonin
- Molecular Microbiology and Structural Biochemistry (MMSB), UMR 5086 CNRS/University of Lyon, Lyon, France
| | - Christophe Grangeasse
- Molecular Microbiology and Structural Biochemistry (MMSB), UMR 5086 CNRS/University of Lyon, Lyon, France
| | - Jean-Michel Jault
- Molecular Microbiology and Structural Biochemistry (MMSB), UMR 5086 CNRS/University of Lyon, Lyon, France
| | - Cédric Orelle
- Molecular Microbiology and Structural Biochemistry (MMSB), UMR 5086 CNRS/University of Lyon, Lyon, France
| |
Collapse
|
11
|
Tebano G, Zaghi I, Baldasso F, Calgarini C, Capozzi R, Salvadori C, Cricca M, Cristini F. Antibiotic Resistance to Molecules Commonly Prescribed for the Treatment of Antibiotic-Resistant Gram-Positive Pathogens: What Is Relevant for the Clinician? Pathogens 2024; 13:88. [PMID: 38276161 PMCID: PMC10819222 DOI: 10.3390/pathogens13010088] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Revised: 01/13/2024] [Accepted: 01/15/2024] [Indexed: 01/27/2024] Open
Abstract
Antibiotic resistance in Gram-positive pathogens is a relevant concern, particularly in the hospital setting. Several antibiotics are now available to treat these drug-resistant pathogens, such as daptomycin, dalbavancin, linezolid, tedizolid, ceftaroline, ceftobiprole, and fosfomycin. However, antibiotic resistance can also affect these newer molecules. Overall, this is not a frequent phenomenon, but it is a growing concern in some settings and can compromise the effectiveness of these molecules, leaving few therapeutic options. We reviewed the available evidence about the epidemiology of antibiotic resistance to these antibiotics and the main molecular mechanisms of resistance, particularly methicillin-resistant Sthaphylococcus aureus, methicillin-resistant coagulase-negative staphylococci, vancomycin-resistant Enterococcus faecium, and penicillin-resistant Streptococcus pneumoniae. We discussed the interpretation of susceptibility tests when minimum inhibitory concentrations are not available. We focused on the risk of the emergence of resistance during treatment, particularly for daptomycin and fosfomycin, and we discussed the strategies that can be implemented to reduce this phenomenon, which can lead to clinical failure despite appropriate antibiotic treatment. The judicious use of antibiotics, epidemiological surveillance, and infection control measures is essential to preserving the efficacy of these drugs.
Collapse
Affiliation(s)
- Gianpiero Tebano
- Infectious Diseases Unit, AUSL Romagna, Ravenna Hospital, 48121 Ravenna, Italy; (I.Z.); (C.C.); (C.S.)
| | - Irene Zaghi
- Infectious Diseases Unit, AUSL Romagna, Ravenna Hospital, 48121 Ravenna, Italy; (I.Z.); (C.C.); (C.S.)
- Unit of Microbiology, The Greater Romagna Area Hub Laboratory, 47522 Cesena, Italy;
| | - Francesco Baldasso
- Infectious Diseases Unit, AUSL Romagna, Forlì and Cesena Hospitals, 47121 Forlì and Cesena, Italy; (F.B.); (R.C.); (F.C.)
- Department of Medical and Surgical Sciences (DIMEC), Alma Mater Studiorum, University of Bologna, 40126 Bologna, Italy
| | - Chiara Calgarini
- Infectious Diseases Unit, AUSL Romagna, Ravenna Hospital, 48121 Ravenna, Italy; (I.Z.); (C.C.); (C.S.)
- Department of Medical and Surgical Sciences (DIMEC), Alma Mater Studiorum, University of Bologna, 40126 Bologna, Italy
| | - Roberta Capozzi
- Infectious Diseases Unit, AUSL Romagna, Forlì and Cesena Hospitals, 47121 Forlì and Cesena, Italy; (F.B.); (R.C.); (F.C.)
- Department of Medical and Surgical Sciences (DIMEC), Alma Mater Studiorum, University of Bologna, 40126 Bologna, Italy
| | - Caterina Salvadori
- Infectious Diseases Unit, AUSL Romagna, Ravenna Hospital, 48121 Ravenna, Italy; (I.Z.); (C.C.); (C.S.)
| | - Monica Cricca
- Unit of Microbiology, The Greater Romagna Area Hub Laboratory, 47522 Cesena, Italy;
- Department of Medical and Surgical Sciences (DIMEC), Alma Mater Studiorum, University of Bologna, 40126 Bologna, Italy
| | - Francesco Cristini
- Infectious Diseases Unit, AUSL Romagna, Forlì and Cesena Hospitals, 47121 Forlì and Cesena, Italy; (F.B.); (R.C.); (F.C.)
| |
Collapse
|
12
|
Boutet-Dubois A, Magnan C, Lienard A, Pouget C, Bouchet F, Marchandin H, Larcher R, Lavigne JP, Pantel A. In Vivo-Acquired Resistance to Daptomycin during Methicillin-Resistant Staphylococcus aureus Bacteremia. Antibiotics (Basel) 2023; 12:1647. [PMID: 38136681 PMCID: PMC10740961 DOI: 10.3390/antibiotics12121647] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Revised: 11/11/2023] [Accepted: 11/16/2023] [Indexed: 12/24/2023] Open
Abstract
Daptomycin (DAP) represents an interesting alternative to treat methicillin-resistant Staphylococcus aureus (MRSA) infections. Different mechanisms of DAP resistance have been described; however, in vivo-acquired resistance is uncharacterized. This study described the phenotypic and genotypic evolution of MRSA strains that became resistant to DAP in two unrelated patients with bacteremia under DAP treatment, in two hospitals in the South of France. DAP MICs were determined using broth microdilution method on the pairs of isogenic (DAP-S/DAP-R) S. aureus isolated from bloodstream cultures. Whole genome sequencing was carried out using Illumina MiSeq Sequencing system. The two cases revealed DAP-R acquisition by MRSA strains within three weeks in patients treated by DAP. The isolates belonged to the widespread ST5 (patient A) and ST8 (patient B) lineages and were of spa-type t777 and t622, respectively. SNP analysis comparing each DAP-S/DAP-R pair confirmed that the isolates were isogenic. The causative mutations were identified in MprF (Multiple peptide resistance Factor) protein: L826F (Patient A) and S295L (Patient B), and in Cls protein: R228H (Patient B). These proteins encoded both proteins of the lipid biosynthetic enzymes. The resistance to DAP is particularly poorly described whereas DAP is highly prescribed to treat MRSA. Our study highlights the non-systematic cross-resistance between DAP and glycopeptides and the importance of monitoring DAP MIC in persistent MRSA bacteremia.
Collapse
Affiliation(s)
- Adeline Boutet-Dubois
- VBIC, INSERM U1047, Department of Microbiology and Hospital Hygiene, University of Montpellier, CHU Nîmes, 30029 Nîmes, France; (A.B.-D.); (C.M.); (C.P.); (A.P.)
| | - Chloé Magnan
- VBIC, INSERM U1047, Department of Microbiology and Hospital Hygiene, University of Montpellier, CHU Nîmes, 30029 Nîmes, France; (A.B.-D.); (C.M.); (C.P.); (A.P.)
| | - Alexi Lienard
- Laboratory of Medical Biology, CH Bassin de Thau, 34207 Sète, France;
| | - Cassandra Pouget
- VBIC, INSERM U1047, Department of Microbiology and Hospital Hygiene, University of Montpellier, CHU Nîmes, 30029 Nîmes, France; (A.B.-D.); (C.M.); (C.P.); (A.P.)
| | - Flavien Bouchet
- Department of Internal Medicine-Infectiology, CH Bassin de Thau, 34207 Sète, France;
| | - Hélène Marchandin
- HydroSciences Montpellier, Department of Microbiology and Hospital Hygiene, University of Montpellier, CNRS, IRD, CHU Nîmes, 30029 Nîmes, France;
| | - Romaric Larcher
- Department of Infectious Diseases, CHU Nîmes, 30029 Nîmes, France;
| | - Jean-Philippe Lavigne
- VBIC, INSERM U1047, Department of Microbiology and Hospital Hygiene, University of Montpellier, CHU Nîmes, 30029 Nîmes, France; (A.B.-D.); (C.M.); (C.P.); (A.P.)
| | - Alix Pantel
- VBIC, INSERM U1047, Department of Microbiology and Hospital Hygiene, University of Montpellier, CHU Nîmes, 30029 Nîmes, France; (A.B.-D.); (C.M.); (C.P.); (A.P.)
| |
Collapse
|
13
|
Kirchner L, Marciniak T, Ziebuhr W, Scherf-Clavel O, Holzgrabe U. The solvent- and surface-dependent adsorption of the lipopeptide antibiotic daptomycin: The general necessity of adsorption tests. J Pharm Biomed Anal 2023; 235:115657. [PMID: 37619296 DOI: 10.1016/j.jpba.2023.115657] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Revised: 08/15/2023] [Accepted: 08/16/2023] [Indexed: 08/26/2023]
Abstract
The impact of poor or non-reproducible analyte recoveries due to non-specific drug adsorption on various analytical assays is often underestimated. Even internationally approved guidelines for pharmaceutical analysis such as the EMA guideline on bioanalytical method validation, the ICH guideline M10 on bioanalytical method validation and study sample analysis or the FDA bioanalytical method validation guidance do not adequately encourage more detailed investigations. Furthermore, other areas of research in which the concentration of active pharmaceutical compounds plays a crucial role, for example screening for minimal inhibitory concentrations of bacterial isolates, are potentially affected as well. The aim of this study was to demonstrate the general necessity of drug adsorption tests, using the lipopeptide antibiotic daptomycin as an example. A wide range of typical materials used in processing samples in pharmaceutical and biological analysis, as well as various solvents and biological matrices were included in the experiments. A fully validated LC-MS/MS method was applied for the determination of daptomycin concentrations, which were subsequently used to calculate the recovery. Recovery results (n = 3) ranged from 0.00% to 102.12% with a maximum relative standard deviation of 12.78%. These findings demonstrate that recovery can vary greatly depending on the solvent and the contact material, indicating the need to be optimized and, if applicable, validated. Hence, high reproducibility can only be achieved if all materials (and their manufacturers) used in a method are specified, not just those used in steps considered critical.
Collapse
Affiliation(s)
- Lukas Kirchner
- University of Würzburg, Institute for Pharmacy and Food Chemistry, 97074 Würzburg, Germany.
| | - Tessa Marciniak
- University of Würzburg, Institute for Molecular Infection Biology, 97080 Würzburg, Germany.
| | - Wilma Ziebuhr
- University of Würzburg, Institute for Molecular Infection Biology, 97080 Würzburg, Germany.
| | - Oliver Scherf-Clavel
- University of Würzburg, Institute for Pharmacy and Food Chemistry, 97074 Würzburg, Germany; Aalen University, Faculty of Chemistry, Beethovenstraße 1, 73430 Aalen, Germany.
| | - Ulrike Holzgrabe
- University of Würzburg, Institute for Pharmacy and Food Chemistry, 97074 Würzburg, Germany.
| |
Collapse
|
14
|
Zhang R, Ashford NK, Li A, Ross DH, Werth BJ, Xu L. High-throughput analysis of lipidomic phenotypes of methicillin-resistant Staphylococcus aureus by coupling in situ 96-well cultivation and HILIC-ion mobility-mass spectrometry. Anal Bioanal Chem 2023; 415:6191-6199. [PMID: 37535099 PMCID: PMC11059195 DOI: 10.1007/s00216-023-04890-6] [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: 07/21/2022] [Revised: 07/23/2023] [Accepted: 07/25/2023] [Indexed: 08/04/2023]
Abstract
Antimicrobial resistance is a major threat to human health as resistant pathogens spread globally, and the development of new antimicrobials is slow. Since many antimicrobials function by targeting cell wall and membrane components, high-throughput lipidomics for bacterial phenotyping is of high interest for researchers to unveil lipid-mediated pathways when dealing with a large number of lab-selected or clinical strains. However, current practice for lipidomic analysis requires the cultivation of bacteria on a large scale, which does not replicate the growth conditions for high-throughput bioassays that are normally carried out in 96-well plates, such as susceptibility tests, growth curve measurements, and biofilm quantitation. Analysis of bacteria grown under the same condition as other bioassays would better inform the differences in susceptibility and other biological metrics. In this work, a high-throughput method for cultivation and lipidomic analysis of antimicrobial-resistant bacteria was developed for standard 96-well plates exemplified by methicillin-resistant Staphylococcus aureus (MRSA). By combining a 30-mm liquid chromatography (LC) column with ion mobility (IM) separation, elution time could be dramatically shortened to 3.6 min for a single LC run without losing major lipid features. Peak capacity was largely rescued by the addition of the IM dimension. Through multi-linear calibration, the deviation of retention time can be limited to within 5%, making database-based automatic lipid identification feasible. This high-throughput method was further validated by characterizing the lipidomic phenotypes of antimicrobial-resistant mutants derived from the MRSA strain, W308, grown in a 96-well plate.
Collapse
Affiliation(s)
- Rutan Zhang
- Department of Medicinal Chemistry, University of Washington, Seattle, WA, 98195, USA
| | - Nate K Ashford
- Department of Pharmacy, University of Washington, Seattle, WA, 98195, USA
| | - Amy Li
- Department of Medicinal Chemistry, University of Washington, Seattle, WA, 98195, USA
| | - Dylan H Ross
- Department of Medicinal Chemistry, University of Washington, Seattle, WA, 98195, USA
- Biological Sciences Division, Pacific Northwest National Laboratory, WA, 99352, Richland, USA
| | - Brian J Werth
- Department of Pharmacy, University of Washington, Seattle, WA, 98195, USA
| | - Libin Xu
- Department of Medicinal Chemistry, University of Washington, Seattle, WA, 98195, USA.
| |
Collapse
|
15
|
Giulieri SG, Guérillot R, Holmes NE, Baines SL, Hachani A, Hayes AS, Daniel DS, Seemann T, Davis JS, Van Hal S, Tong SYC, Stinear TP, Howden BP. A statistical genomics framework to trace bacterial genomic predictors of clinical outcomes in Staphylococcus aureus bacteremia. Cell Rep 2023; 42:113069. [PMID: 37703880 DOI: 10.1016/j.celrep.2023.113069] [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: 12/01/2022] [Revised: 06/29/2023] [Accepted: 08/18/2023] [Indexed: 09/15/2023] Open
Abstract
Outcomes of severe bacterial infections are determined by the interplay between host, pathogen, and treatments. While human genomics has provided insights into host factors impacting Staphylococcus aureus infections, comparatively little is known about S. aureus genotypes and disease severity. Building on the hypothesis that bacterial pathoadaptation is a key outcome driver, we developed a genome-wide association study (GWAS) framework to identify adaptive mutations associated with treatment failure and mortality in S. aureus bacteremia (1,358 episodes). Our research highlights the potential of vancomycin-selected mutations and vancomycin minimum inhibitory concentration (MIC) as key explanatory variables to predict infection severity. The contribution of bacterial variation was much lower for clinical outcomes (heritability <5%); however, GWASs allowed us to identify additional, MIC-independent candidate pathogenesis loci. Using supervised machine learning, we were able to quantify the predictive potential of these adaptive signatures. Our statistical genomics framework provides a powerful means to capture adaptive mutations impacting severe bacterial infections.
Collapse
Affiliation(s)
- Stefano G Giulieri
- Department of Microbiology and Immunology, The University of Melbourne at the Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia; Victorian Infectious Disease Service, The Royal Melbourne Hospital at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia; Department of Infectious Diseases, The University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia; Department of Infectious Diseases, Austin Health, Heidelberg, VIC 3084, Australia.
| | - Romain Guérillot
- Department of Microbiology and Immunology, The University of Melbourne at the Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia
| | - Natasha E Holmes
- Department of Infectious Diseases, The University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia; Department of Infectious Diseases, Austin Health, Heidelberg, VIC 3084, Australia
| | - Sarah L Baines
- Department of Microbiology and Immunology, The University of Melbourne at the Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia; Centre for Pathogen Genomics, The University of Melbourne, Melbourne, VIC 3000, Australia
| | - Abderrahman Hachani
- Department of Microbiology and Immunology, The University of Melbourne at the Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia
| | - Ashleigh S Hayes
- Department of Microbiology and Immunology, The University of Melbourne at the Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia
| | - Diane S Daniel
- Department of Microbiology and Immunology, The University of Melbourne at the Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia
| | - Torsten Seemann
- Centre for Pathogen Genomics, The University of Melbourne, Melbourne, VIC 3000, Australia
| | - Joshua S Davis
- Department of Infectious Diseases, John Hunter Hospital, New Lambton Heights, NSW 2305, Australia; Menzies School of Health Research, Charles Darwin University, Casuarina, NT 0810, Australia
| | - Sebastiaan Van Hal
- Department of Infectious Diseases and Microbiology, Royal Prince Alfred Hospital, Camperdown, NSW 2050, Australia; Central Clinical School, University of Sydney, Camperdown, NSW 2050, Australia
| | - Steven Y C Tong
- Victorian Infectious Disease Service, The Royal Melbourne Hospital at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia; Department of Infectious Diseases, The University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia
| | - Timothy P Stinear
- Department of Microbiology and Immunology, The University of Melbourne at the Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia
| | - Benjamin P Howden
- Department of Microbiology and Immunology, The University of Melbourne at the Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia; Department of Infectious Diseases, Austin Health, Heidelberg, VIC 3084, Australia; Centre for Pathogen Genomics, The University of Melbourne, Melbourne, VIC 3000, Australia
| |
Collapse
|
16
|
Fait A, Andersson DI, Ingmer H. Evolutionary history of Staphylococcus aureus influences antibiotic resistance evolution. Curr Biol 2023; 33:3389-3397.e5. [PMID: 37494936 DOI: 10.1016/j.cub.2023.06.082] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Revised: 05/05/2023] [Accepted: 06/29/2023] [Indexed: 07/28/2023]
Abstract
Antibiotic resistance often confers a fitness cost to the resistant cell and thus raises key questions of how resistance is maintained in the absence of antibiotics and, if lost, whether cells are genetically primed for re-evolving resistance. To address these questions, we have examined vancomycin-intermediate Staphylococcus aureus (VISA) strains that arise during vancomycin therapy. VISA strains harbor a broad spectrum of mutations, and they are known to be unstable both in patients and in the laboratory. Here, we show that loss of resistance in VISA strains is correlated with a fitness increase and is attributed to adaptive mutations, leaving the initial VISA-adaptive mutations intact. Importantly, upon a second exposure to vancomycin, such revertants evolve significantly faster to become VISA, and they reach higher resistance levels than vancomycin-naive cells. Further, we find that sub-lethal concentrations of vancomycin stabilize the VISA phenotype, as do the human β-defensin 3 (hBD-3) and the bacteriocin nisin that both, like vancomycin, bind to the peptidoglycan building block, lipid II. Thus, factors binding lipid II may stabilize VISA both in vivo and in vitro, and in case resistance is lost, mutations remain that predispose to resistance development. These findings may explain why VISA infections often are re-occurring and suggest that previous vancomycin adaptation should be considered a risk factor when deciding on antimicrobial chemotherapy.
Collapse
Affiliation(s)
- Anaëlle Fait
- Department of Veterinary and Animal Sciences, University of Copenhagen, 1870 Frederiksberg, Denmark; Department of Environmental Systems Science, ETH Zürich, 8092 Zürich, Switzerland
| | - Dan I Andersson
- Department of Medical Biochemistry and Microbiology, Uppsala University, 751 23 Uppsala, Sweden
| | - Hanne Ingmer
- Department of Veterinary and Animal Sciences, University of Copenhagen, 1870 Frederiksberg, Denmark.
| |
Collapse
|
17
|
Woldeyohannis NN, Desta AF. Fate of antimicrobial resistance genes (ARG) and ARG carriers in struvite production process from human urine. JOURNAL OF ENVIRONMENTAL SCIENCE AND HEALTH. PART A, TOXIC/HAZARDOUS SUBSTANCES & ENVIRONMENTAL ENGINEERING 2023; 58:783-792. [PMID: 37469114 DOI: 10.1080/10934529.2023.2235246] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Revised: 06/17/2023] [Accepted: 06/26/2023] [Indexed: 07/21/2023]
Abstract
Struvite, a human urine-derived fertilizer types, is characterized by its low water solubility that renders it a slow-releasing eco-friendly fertilizer. Knowing the fate of antibiotic resistance genes in struvite is important since human urine carries microorganisms, viruses and mobilomes. In this study, urine samples were collected and struvite production was done using MgCl2. From the fresh, stored urine and struvite, DNA was extracted and metagenomic sequencing was done using Illumina HiSeqX. Metagenome-derived genome sequence analysis revealed the dominance of phages of Streptococcus, Bacillus and Escherichia, with nearly 50% abundance of streptococcus phage in fresh urine. Increased antibiotic resistance genes were found in the stored urine than in fresh and struvite samples. The top five resistance genes in all the three samples were to aminoglycosides, carbapenem, chloramphenicol, erythromycin and efflux pump, with key carrying pathogens including Acinetobacter, Aeromonas and Enterococcus. The identified families for carbapenem, aminoglycoside resistance and efflux pump were shown persistent in struvite with a shift in gene families. The detection of resistance-gene-laden mobilomes, including the last-resort antibiotics in the struvite sample, requires due attention before the implementation of struvite as fertilizer. Further optimization of the struvite production process with regard to the minimization of mobilomes is recommended.
Collapse
Affiliation(s)
- Nebiyat N Woldeyohannis
- Microbial, Cellular and Molecular Biology Department, Addis Ababa University, Addis Ababa, Ethiopia
| | - Adey F Desta
- Microbial, Cellular and Molecular Biology Department, Addis Ababa University, Addis Ababa, Ethiopia
| |
Collapse
|
18
|
Douglas EJA, Wulandari SW, Lovell SD, Laabei M. Novel antimicrobial strategies to treat multi-drug resistant Staphylococcus aureus infections. Microb Biotechnol 2023; 16:1456-1474. [PMID: 37178319 PMCID: PMC10281381 DOI: 10.1111/1751-7915.14268] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Revised: 04/12/2023] [Accepted: 04/18/2023] [Indexed: 05/15/2023] Open
Abstract
Antimicrobial resistance is a major obstacle for the treatment of infectious diseases and currently represents one of the most significant threats to global health. Staphylococcus aureus remains a formidable human pathogen with high mortality rates associated with severe systemic infections. S. aureus has become notorious as a multidrug resistant bacterium, which when combined with its extensive arsenal of virulence factors that exacerbate disease, culminates in an incredibly challenging pathogen to treat clinically. Compounding this major health issue is the lack of antibiotic discovery and development, with only two new classes of antibiotics approved for clinical use in the last 20 years. Combined efforts from the scientific community have reacted to the threat of dwindling treatment options to combat S. aureus disease in several innovative and exciting developments. This review describes current and future antimicrobial strategies aimed at treating staphylococcal colonization and/or disease, examining therapies that show significant promise at the preclinical development stage to approaches that are currently being investigated in clinical trials.
Collapse
|
19
|
Stone MC, Mychack A, Coe KA, Walker S. Combining Signal Peptidase and Lipoprotein Processing Inhibitors Overcomes Ayr Resistance in Staphylococcus aureus. Antimicrob Agents Chemother 2023; 67:e0011523. [PMID: 37097175 PMCID: PMC10190671 DOI: 10.1128/aac.00115-23] [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: 02/02/2023] [Accepted: 03/30/2023] [Indexed: 04/26/2023] Open
Abstract
Antibiotic resistance in bacterial pathogens is an ongoing public health concern. The arylomycins are a class of natural product antibiotics that target the type I signal peptidase, which carries out the terminal step in protein secretion. Here, we used transposon sequencing (Tn-Seq) to profile the effects of the optimized arylomycin derivative G0775 in Staphylococcus aureus. Our transposon libraries include both upregulation and inactivation mutants, allowing us to identify resistance mechanisms and targets for synergism. We identified several cell envelope pathways that, when inactivated, sensitize S. aureus to the arylomycin G0775. These pathways include the lipoprotein processing pathway, and we have shown that inhibitors of this pathway synergize with G0775 even though lipoprotein processing is nonessential in S. aureus. Moreover, we found that blocking this pathway completely reverses Ayr resistance, which is a major resistance mechanism to arylomycins, including G0775. Our Tn-Seq data also showed that upregulation of mprF and several other genes is protective against G0775. Because a subset of these genes was previously found in a Tn-Seq profile of the clinically important antibiotic daptomycin, we tested a set of daptomycin-nonsusceptible clinical isolates with gain-of-function mutations in mprF for susceptibility to arylomycin G0775. Despite structural and mechanistic differences between these antibiotics, we observed similar decreases in susceptibility. Taken together, our results highlight how Tn-Seq profiles that include both gene inactivation and upregulation can identify targets, antibiotic resistance mechanisms, and strategies to overcome resistance.
Collapse
Affiliation(s)
- Madeleine C. Stone
- Department of Microbiology, Blavatnik Institute, Harvard Medical School, Boston, Massachusetts, USA
| | - Aaron Mychack
- Department of Microbiology, Blavatnik Institute, Harvard Medical School, Boston, Massachusetts, USA
| | - Kathryn A. Coe
- Department of Microbiology, Blavatnik Institute, Harvard Medical School, Boston, Massachusetts, USA
| | - Suzanne Walker
- Department of Microbiology, Blavatnik Institute, Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
20
|
Ali Alghamdi B, Al-Johani I, Al-Shamrani JM, Musamed Alshamrani H, Al-Otaibi BG, Almazmomi K, Yusnoraini Yusof N. Antimicrobial resistance in methicillin-resistant staphylococcus aureus. Saudi J Biol Sci 2023; 30:103604. [PMID: 36936699 PMCID: PMC10018568 DOI: 10.1016/j.sjbs.2023.103604] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Revised: 02/02/2023] [Accepted: 02/19/2023] [Indexed: 03/02/2023] Open
Abstract
In the medical community, antibiotics are revered as a miracle because they stop diseases brought on by pathogenic bacteria. Antibiotics have become the cornerstone of contemporary medical advancements ever since penicillin was discovered. Antibiotic resistance developed among germs quickly, placing a strain in the medical field. Methicillin-resistant Staphylococcus aureus (MRSA), Since 1961, has emerged as the major general antimicrobial resistant bacteria (AMR) worldwide. MRSA can easily transmit across the hospital system and has mostly gained resistance to medications called beta-lactamases. This enzyme destroys the cell wall of beta-lactam antibiotics resulting in resistance against that respective antibiotic. Daptomycin, linezolid and vancomycin were previously used to treat MRSA infections. However, due to mutations and Single nucleotide polymorphisms (SNPs) in Open reading frames (ORFs) and SCCmec machinery of respective antibody, MRSA developed resistance against those antibiotics. The MRSA strains (USA300, CC398, CC130 etc.), when their pan-genomes were analyzed were found the genes involved in invoking resistance against the antibiotics as well as the epidemiology of that respective strain. PENC (penicillin plus potassium clavulanate) is the new antibiotic showing potential in treatment of MRSA though it is itself resistant against penicillin alone. In this review, our main focus is on mechanism of development of AMR in MRSA, how different ORFs are involved in evoking resistance in MRSA and what is the core-genome of different antimicrobial resistant MRSA.
Collapse
Affiliation(s)
- Bandar Ali Alghamdi
- Department of Cardiac Surgery, King Fahad Armed Forces Hospital, Jeddah, Saudi Arabia
| | - Intisar Al-Johani
- Department of Biotechnology, Taif University, Taif City, Saudi Arabia
| | | | - Hussein Musamed Alshamrani
- Directorate of Health Affairs in Qunfudah Center (Namerah Primary Health care) Pharmacy Department, Saudi Arabia
| | | | - Kholod Almazmomi
- Department of Biotechnology, Taif University, Taif City, Saudi Arabia
| | - Nik Yusnoraini Yusof
- Institute for Research in Molecular Medicine (INFORMM), Health Campus, Universiti Sains Malaysia, Kubang Kerian 16150, Kelantan, Malaysia
- Corresponding author at.: Institute for Research in Molecular Medicine (INFORMM) Universiti Sains Malaysia Kubang Kerian, Kelantan 16150, Malaysia.
| |
Collapse
|
21
|
Kirchner L, Marciniak T, Ziebuhr W, Scherf-Clavel O, Holzgrabe U. Development, validation and application of a selective and sensitive LC-MS/MS method for the quantification of daptomycin in a suspension of Mammaliicoccus sciuri in Mueller-Hinton broth. J Pharm Biomed Anal 2023; 227:115293. [PMID: 36822068 DOI: 10.1016/j.jpba.2023.115293] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Revised: 02/06/2023] [Accepted: 02/12/2023] [Indexed: 02/17/2023]
Abstract
Reports of therapy failures related to the use of daptomycin (DAP) are steadily increasing. This is mainly due to emerging DAP resistance for which, however, the underlying mechanism is often unknown. To elucidate the mode of action of novel DAP resistance traits it is indispensable to reliably detect and quantify DAP in complex matrices such as bacterial culture media. In this study, we established a selective and sensitive quantification method for DAP upon growth of a DAP resistant Mammaliicoccus sciuri strain in Mueller-Hinton medium. The method combined methanol-induced protein precipitation followed by high performance liquid chromatography with gradient elution coupled to mass spectrometry (LC-MS/MS) using daptomycin-d5 as internal standard. All validation parameters met the acceptance criteria of the European Medicines Agency (EMA) guideline on bioanalytical method validation. We successfully applied this highly selective and sensitive LC-MS/MS method for the quantification of DAP in in vitro studies addressing DAP resistance mechanisms in Gram-positive bacteria.
Collapse
Affiliation(s)
- Lukas Kirchner
- University of Würzburg, Institute for Pharmacy and Food Chemistry, 97074 Würzburg, Germany.
| | - Tessa Marciniak
- University of Würzburg, Institute for Molecular Infection Biology, 97080 Würzburg, Germany.
| | - Wilma Ziebuhr
- University of Würzburg, Institute for Molecular Infection Biology, 97080 Würzburg, Germany.
| | - Oliver Scherf-Clavel
- University of Würzburg, Institute for Pharmacy and Food Chemistry, 97074 Würzburg, Germany.
| | - Ulrike Holzgrabe
- University of Würzburg, Institute for Pharmacy and Food Chemistry, 97074 Würzburg, Germany.
| |
Collapse
|
22
|
Staphylococcus aureus Cell Wall Phenotypic Changes Associated with Biofilm Maturation and Water Availability: A Key Contributing Factor for Chlorine Resistance. Int J Mol Sci 2023; 24:ijms24054983. [PMID: 36902413 PMCID: PMC10003762 DOI: 10.3390/ijms24054983] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Revised: 02/25/2023] [Accepted: 03/02/2023] [Indexed: 03/08/2023] Open
Abstract
Staphylococcus aureus biofilms are resistant to both antibiotics and disinfectants. As Staphylococci cell walls are an important defence mechanism, we sought to examine changes to the bacterial cell wall under different growth conditions. Cell walls of S. aureus grown as 3-day hydrated biofilm, 12-day hydrated biofilm, and 12-day dry surface biofilm (DSB) were compared to cell walls of planktonic organisms. Additionally, proteomic analysis using high-throughput tandem mass tag-based mass spectrometry was performed. Proteins involved in cell wall synthesis in biofilms were upregulated in comparison to planktonic growth. Bacterial cell wall width (measured by transmission electron microscopy) and peptidoglycan production (detected using a silkworm larva plasma system) increased with biofilm culture duration (p < 0.001) and dehydration (p = 0.002). Similarly, disinfectant tolerance was greatest in DSB, followed by 12-day hydrated biofilm and then 3-day biofilm, and it was least in the planktonic bacteria--suggesting that changes to the cell wall may be a key factor for S. aureus biofilm biocide resistance. Our findings shed light on possible new targets to combat biofilm-related infections and hospital dry surface biofilms.
Collapse
|
23
|
García de la Mària C, Cañas MA, Fernández-Pittol M, Dahl A, García-González J, Hernández-Meneses M, Cuervo G, Moreno A, Miró JM, Marco F. Emerging issues on Staphylococcus aureus endocarditis and the role in therapy of daptomycin plus fosfomycin. Expert Rev Anti Infect Ther 2023; 21:281-293. [PMID: 36744387 DOI: 10.1080/14787210.2023.2174969] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
INTRODUCTION Methicillin-resistant and -susceptible Staphylococcus aureus (MRSA/MSSA) infections are a major global health-care problem. Bacteremia with S. aureus exhibits high rates of morbidity and mortality and can cause complicated infections such as infective endocarditis (IE). The emerging resistance profile of S. aureus is worrisome, and several international agencies have appealed for new treatment approaches to be developed. AREAS COVERED Daptomycin presents a rapid bactericidal effect against MRSA and has been considered at least as effective as vancomycin in treating MRSA bacteremia. However, therapy failure is often related to deep-seated infections, e.g. endocarditis, with high bacterial inocula and daptomycin regimens <10 mg/kg/day. Current antibiotic options for treating invasive S. aureus infections have limitations in monotherapy. Daptomycin in combination with other antibiotics, e.g. fosfomycin, may be effective in improving clinical outcomes in patients with MRSA IE. EXPERT OPINION Exploring therapeutic combinations has shown fosfomycin to have a unique mechanism of action and to be the most effective option in preventing the onset of resistance to and optimizing the efficacy of daptomycin, suggesting the synergistic combination of fosfomycin with daptomycin is a useful alternative treatment option for MSSA or MRSA IE.
Collapse
Affiliation(s)
- Cristina García de la Mària
- Infectious Diseases Service, Hospital Clinic - Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS). University of Barcelona, Barcelona, Spain
| | - Maria-Alexandra Cañas
- Infectious Diseases Service, Hospital Clinic - Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS). University of Barcelona, Barcelona, Spain
| | | | - Anders Dahl
- Infectious Diseases Service, Hospital Clinic - Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS). University of Barcelona, Barcelona, Spain.,Department of Cardiology, Bispebjerg University Hospital, Copenhagen, Denmark
| | - Javier García-González
- Infectious Diseases Service, Hospital Clinic - Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS). University of Barcelona, Barcelona, Spain
| | - Marta Hernández-Meneses
- Infectious Diseases Service, Hospital Clinic - Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS). University of Barcelona, Barcelona, Spain
| | - Guillermo Cuervo
- Infectious Diseases Service, Hospital Clinic - Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS). University of Barcelona, Barcelona, Spain
| | - Asunción Moreno
- Infectious Diseases Service, Hospital Clinic - Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS). University of Barcelona, Barcelona, Spain
| | - Jose M Miró
- Infectious Diseases Service, Hospital Clinic - Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS). University of Barcelona, Barcelona, Spain.,CIBERINFEC, Instituto de Salud Carlos III, Madrid, Spain
| | - Francesc Marco
- Microbiology Department, Centre Diagnòstic Biomèdic (CDB) Hospital Clínic, Barcelona, Spain.,ISGlobal, Hospital Clínic - Universitat de Barcelona, Barcelona, Spain
| |
Collapse
|
24
|
Kumar G, Kiran Tudu A. Tackling multidrug-resistant Staphylococcus aureus by natural products and their analogues acting as NorA efflux pump inhibitors. Bioorg Med Chem 2023; 80:117187. [PMID: 36731248 DOI: 10.1016/j.bmc.2023.117187] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Revised: 01/23/2023] [Accepted: 01/23/2023] [Indexed: 01/29/2023]
Abstract
Staphylococcus aureus (S. aureus) is a pathogen responsible for various community and hospital-acquired infections with life-threatening complications like bacteraemia, endocarditis, meningitis, liver abscess, and spinal cord epidural abscess. Antibiotics have been used to treat microbial infections since the introduction of penicillin in 1940. In recent decades, the abuse and misuse of antibiotics in humans, animals, plants, and fungi, including the treatment of non-microbial diseases, have led to the rapid emergence of multidrug-resistant pathogens with increased virulence. Bacteria have developed several complementary mechanisms to avoid the effects of antibiotics. These mechanisms include chemical transformations and enzymatic inactivation of antibiotics, modification of antibiotics' target site, and reduction of intracellular antibiotics concentration by changes in membrane permeability or by the overexpression of efflux pumps (EPs). The strategy to check antibiotic resistance includes synthesis of the antibiotic analogues, or antibiotics are given in combination with the adjuvant. The inhibitors of multidrug EPs are considered promising alternative therapeutic options with the potential to revive the effects of antibiotics and reduce bacterial virulence. Natural products played a vital role in drug discovery and significantly contributed to the area of infectious diseases. Also, natural products provide lead compounds that sometimes need modification based on structural and biological properties to meet the drug criteria. This review discusses natural products and their derived compounds as NorA efflux pump inhibitors (EPIs).
Collapse
Affiliation(s)
- Gautam Kumar
- Department of Natural Products, Chemical Sciences, National Institute of Pharmaceutical Education and Research-Hyderabad, Hyderabad, Balanagar, Telangana 500037, India.
| | - Asha Kiran Tudu
- Department of Natural Products, Chemical Sciences, National Institute of Pharmaceutical Education and Research-Hyderabad, Hyderabad, Balanagar, Telangana 500037, India
| |
Collapse
|
25
|
Munoz-Torres JR, Martínez-González SB, Lozano-Luján AD, Martínez-Vázquez MC, Velasco-Elizondo P, Garza-Veloz I, Martinez-Fierro ML. Biological properties and surgical applications of the human amniotic membrane. Front Bioeng Biotechnol 2023; 10:1067480. [PMID: 36698632 PMCID: PMC9868191 DOI: 10.3389/fbioe.2022.1067480] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Accepted: 12/22/2022] [Indexed: 01/11/2023] Open
Abstract
The amniotic membrane (AM) is the inner part of the placenta. It has been used therapeutically for the last century. The biological proprieties of AM include immunomodulatory, anti-scarring, anti-microbial, pro or anti-angiogenic (surface dependent), and tissue growth promotion. Because of these, AM is a functional tissue for the treatment of different pathologies. The AM is today part of the treatment for various conditions such as wounds, ulcers, burns, adhesions, and skin injury, among others, with surgical resolution. This review focuses on the current surgical areas, including gynecology, plastic surgery, gastrointestinal, traumatology, neurosurgery, and ophthalmology, among others, that use AM as a therapeutic option to increase the success rate of surgical procedures. Currently there are articles describing the mechanisms of action of AM, some therapeutic implications and the use in surgeries of specific surgical areas, this prevents knowing the therapeutic response of AM when used in surgeries of different organs or tissues. Therefore, we described the use of AM in various surgical specialties along with the mechanisms of action, helping to improve the understanding of the therapeutic targets and achieving an adequate perspective of the surgical utility of AM with a particular emphasis on regenerative medicine.
Collapse
|
26
|
Soundararajan M, Marincola G, Liong O, Marciniak T, Wencker FDR, Hofmann F, Schollenbruch H, Kobusch I, Linnemann S, Wolf SA, Helal M, Semmler T, Walther B, Schoen C, Nyasinga J, Revathi G, Boelhauve M, Ziebuhr W. Farming Practice Influences Antimicrobial Resistance Burden of Non-Aureus Staphylococci in Pig Husbandries. Microorganisms 2022; 11:microorganisms11010031. [PMID: 36677324 PMCID: PMC9865537 DOI: 10.3390/microorganisms11010031] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 12/15/2022] [Accepted: 12/19/2022] [Indexed: 12/24/2022] Open
Abstract
Non-aureus staphylococci (NAS) are ubiquitous bacteria in livestock-associated environments where they may act as reservoirs of antimicrobial resistance (AMR) genes for pathogens such as Staphylococcus aureus. Here, we tested whether housing conditions in pig farms could influence the overall AMR-NAS burden. Two hundred and forty porcine commensal and environmental NAS isolates from three different farm types (conventional, alternative, and organic) were tested for phenotypic antimicrobial susceptibility and subjected to whole genome sequencing. Genomic data were analysed regarding species identity and AMR gene carriage. Seventeen different NAS species were identified across all farm types. In contrast to conventional farms, no AMR genes were detectable towards methicillin, aminoglycosides, and phenicols in organic farms. Additionally, AMR genes to macrolides and tetracycline were rare among NAS in organic farms, while such genes were common in conventional husbandries. No differences in AMR detection existed between farm types regarding fosfomycin, lincosamides, fusidic acid, and heavy metal resistance gene presence. The combined data show that husbandry conditions influence the occurrence of resistant and multidrug-resistant bacteria in livestock, suggesting that changing husbandry practices may be an appropriate means of limiting the spread of AMR bacteria on farms.
Collapse
Affiliation(s)
| | - Gabriella Marincola
- Institute of Molecular Infection Biology, University of Würzburg, 97080 Würzburg, Germany
| | - Olivia Liong
- Institute of Molecular Infection Biology, University of Würzburg, 97080 Würzburg, Germany
| | - Tessa Marciniak
- Institute of Molecular Infection Biology, University of Würzburg, 97080 Würzburg, Germany
| | - Freya D. R. Wencker
- Institute of Molecular Infection Biology, University of Würzburg, 97080 Würzburg, Germany
| | - Franka Hofmann
- Department of Agriculture; South Westphalia University of Applied Sciences, 59494 Soest, Germany
| | - Hannah Schollenbruch
- Department of Agriculture; South Westphalia University of Applied Sciences, 59494 Soest, Germany
| | - Iris Kobusch
- Department of Agriculture; South Westphalia University of Applied Sciences, 59494 Soest, Germany
| | - Sabrina Linnemann
- Department of Agriculture; South Westphalia University of Applied Sciences, 59494 Soest, Germany
| | - Silver A. Wolf
- Genome Sequencing and Genomic Epidemiology, Robert Koch Institute, 13353 Berlin, Germany
| | - Mustafa Helal
- Genome Sequencing and Genomic Epidemiology, Robert Koch Institute, 13353 Berlin, Germany
| | - Torsten Semmler
- Genome Sequencing and Genomic Epidemiology, Robert Koch Institute, 13353 Berlin, Germany
| | - Birgit Walther
- Advanced Light and Electron Microscopy (ZBS4), Robert Koch Institute, 13353 Berlin, Germany
| | - Christoph Schoen
- Institute of Hygiene and Microbiology, University of Würzburg, 97080 Würzburg, Germany
| | - Justin Nyasinga
- Department of Pathology, Aga-Khan-University Hospital Nairobi, Nairobi, Kenya
- Department of Biomedical Sciences and Technology, The Technical University of Kenya, Nairobi, Kenya
| | - Gunturu Revathi
- Department of Pathology, Aga-Khan-University Hospital Nairobi, Nairobi, Kenya
| | - Marc Boelhauve
- Department of Agriculture; South Westphalia University of Applied Sciences, 59494 Soest, Germany
| | - Wilma Ziebuhr
- Institute of Molecular Infection Biology, University of Würzburg, 97080 Würzburg, Germany
- Correspondence: ; Tel.: +49-(0)931-31-2578
| |
Collapse
|
27
|
Greenfield ML, Martin LM, Joodaki F. Computing Individual Area per Head Group Reveals Lipid Bilayer Dynamics. J Phys Chem B 2022; 126:10697-10711. [PMID: 36475708 DOI: 10.1021/acs.jpcb.2c04633] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Lipid bilayers express a range of phases from solid-like to gel-like to liquid-like as a function of temperature and lipid surface concentration. The area occupied per lipid head group serves as one useful indicator of the bilayer phase, in conjunction with the two-dimensional radial distribution function (i.e., structure factor) within the bilayer. Typically, the area per head group is determined by dividing the bilayer area equally among all head groups. Such an approach is less satisfactory for a multicomponent set of diverse lipids. In this work, area determination is performed on a lipid-by-lipid basis by attributing to a lipid the volume that surrounds each atom. Voronoi tessellation provides this division of the interfacial region on a per-atom basis. The method is applied to a multicomponent system of water, NaCl, and 19 phospholipid types that was devised recently [Langmuir2022, 38, 9481-9499] as a computational representation of the Gram-positive Staphylococcus aureus phospholipid bilayer. Results demonstrate that lipids and water molecules occupy similar extents of area within the interfacial region; ascribing area only to head groups implicitly incorporates assumptions about head group hydration. Results further show that lipid tails provide non-negligible contributions to area on the membrane side of the bilayer-water interface. Results for minimum and maximum area of individual lipids reveal that spontaneous fluctuations displace head groups more than 10 Å from the interfacial region during an NPT simulation at 310 K, leading to a zero contribution to total area at some times. Total area fluctuations and fluctuations per individual lipid relax with a correlation time of ∼10 ns. The method complements density profile as an approach to quantify the structure and dynamics of computational lipid bilayers.
Collapse
Affiliation(s)
- Michael L Greenfield
- Department of Chemical Engineering, 360 Fascitelli Center for Advanced Engineering, University of Rhode Island, Kingston, Rhode Island02881, United States
| | - Lenore M Martin
- Department of Cell and Molecular Biology, University of Rhode Island, 120 Flagg Road, Kingston, Rhode Island02881, United States
| | - Faramarz Joodaki
- Department of Chemical Engineering, 360 Fascitelli Center for Advanced Engineering, University of Rhode Island, Kingston, Rhode Island02881, United States
| |
Collapse
|
28
|
Moreira R, Taylor SD. The impact of lysyl-phosphatidylglycerol on the interaction of daptomycin with model membranes. Org Biomol Chem 2022; 20:9319-9329. [PMID: 36129316 DOI: 10.1039/d2ob01384c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Daptomycin is an important clinical antibiotic for which resistance is rising. Daptomycin resistant strains of S. aureus often have increased 1,2-diacyl-sn-glycero-3-[phospho-1-(3-lysyl(1-glycerol))] (lysyl-PG) and mutations to the proteins directly involved in the synthesis and translocation of lysyl-PG are implicated in resistance mechanisms. To study the interaction of daptomycin with lysyl-DMPG-containing model membranes a new stereospecific and regioselective synthesis of lysyl-DMPG was developed. Studies on model membranes containing lysyl-DMPG demonstrate that: (1) daptomycin is not significantly repelled by the cationic charge of lysyl-DMPG; (2) daptomycin binds less avidly to lysyl-DMPG compared to DMPG; (3) the presence of lysyl-DMPG does not impact the membrane bound backbone conformation of daptomycin in a significant way; (4) lysyl-DMPG increases oligomer formation; (5) lysyl-DMPG does not impact model membrane fluidity at lysyl-PG : PG ratios that are relevant to daptomycin resistance. The results of these studies suggest that increased lysyl-PG content does not confer resistance to daptomycin by altering membrane fluidity or reducing membrane affinity but may confer resistance by altering the structure of daptomycin oligomers.
Collapse
Affiliation(s)
- Ryan Moreira
- Department of Chemistry, University of Waterloo, 200 University Avenue West, Waterloo, Ontario, N2L 3G1, Canada.
| | - Scott D Taylor
- Department of Chemistry, University of Waterloo, 200 University Avenue West, Waterloo, Ontario, N2L 3G1, Canada.
| |
Collapse
|
29
|
Nguyen AH, Hood KS, Mileykovskaya E, Miller WR, Tran TT. Bacterial cell membranes and their role in daptomycin resistance: A review. Front Mol Biosci 2022; 9:1035574. [PMID: 36452455 PMCID: PMC9702088 DOI: 10.3389/fmolb.2022.1035574] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Accepted: 10/26/2022] [Indexed: 11/16/2022] Open
Abstract
Lipids play a major role in bacterial cells. Foremost, lipids are the primary constituents of the cell membrane bilayer, providing structure and separating the cell from the surrounding environment. This makes the lipid bilayer a prime target for antimicrobial peptides and membrane-acting antibiotics such as daptomycin. In response, bacteria have evolved mechanisms by which the membrane can be adapted to resist attack by these antimicrobial compounds. In this review, we focus on the membrane phospholipid changes associated with daptomycin resistance in enterococci, Staphylococcus aureus, and the Viridans group streptococci.
Collapse
Affiliation(s)
- April H. Nguyen
- Center for Infectious Diseases Research, Houston Methodist Research Institute, Houston, TX, United States,Division of Infectious Diseases, Department of Medicine, Houston Methodist Hospital, Houston, TX, United States
| | - Kara S. Hood
- Center for Infectious Diseases Research, Houston Methodist Research Institute, Houston, TX, United States,Division of Infectious Diseases, Department of Medicine, Houston Methodist Hospital, Houston, TX, United States
| | - Eugenia Mileykovskaya
- Department of Biochemistry and Molecular Biology, McGovern Medical School, University of Texas Health Science Center, Houston, TX, United States
| | - William R. Miller
- Center for Infectious Diseases Research, Houston Methodist Research Institute, Houston, TX, United States,Division of Infectious Diseases, Department of Medicine, Houston Methodist Hospital, Houston, TX, United States
| | - Truc T. Tran
- Center for Infectious Diseases Research, Houston Methodist Research Institute, Houston, TX, United States,Division of Infectious Diseases, Department of Medicine, Houston Methodist Hospital, Houston, TX, United States,*Correspondence: Truc T. Tran,
| |
Collapse
|
30
|
Schultz JR, Costa SK, Jachak GR, Hegde P, Zimmerman M, Pan Y, Josten M, Ejeh C, Hammerstad T, Sahl HG, Pereira PM, Pinho MG, Dartois V, Cheung A, Aldrich CC. Identification of 5-(Aryl/Heteroaryl)amino-4-quinolones as Potent Membrane-Disrupting Agents to Combat Antibiotic-Resistant Gram-Positive Bacteria. J Med Chem 2022; 65:13910-13934. [PMID: 36219779 PMCID: PMC9826610 DOI: 10.1021/acs.jmedchem.2c01151] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Nosocomial infections caused by resistant Gram-positive organisms are on the rise, presumably due to a combination of factors including prolonged hospital exposure, increased use of invasive procedures, and pervasive antibiotic therapy. Although antibiotic stewardship and infection control measures are helpful, newer agents against multidrug-resistant (MDR) Gram-positive bacteria are urgently needed. Here, we describe our efforts that led to the identification of 5-amino-4-quinolone 111 with exceptionally potent Gram-positive activity with minimum inhibitory concentrations (MICs) ≤0.06 μg/mL against numerous clinical isolates. Preliminary mechanism of action and resistance studies demonstrate that the 5-amino-4-quinolones are bacteriostatic, do not select for resistance, and selectively disrupt bacterial membranes. While the precise molecular mechanism has not been elucidated, the lead compound is nontoxic displaying a therapeutic index greater than 500, is devoid of hemolytic activity, and has attractive physicochemical properties (clog P = 3.8, molecular weight (MW) = 441) that warrant further investigation of this promising antibacterial scaffold for the treatment of Gram-positive infections.
Collapse
Affiliation(s)
- John R Schultz
- Department of Medicinal Chemistry, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Stephen K Costa
- Department of Microbiology & Immunology, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire 03755, United States
| | - Gorakhnath R Jachak
- Department of Medicinal Chemistry, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Pooja Hegde
- Department of Medicinal Chemistry, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Matthew Zimmerman
- Center for Discovery and Innovation, Hackensack Meridian Health, Nutley, New Jersey 07110, United States
| | - Yan Pan
- Center for Discovery and Innovation, Hackensack Meridian Health, Nutley, New Jersey 07110, United States
| | - Michaele Josten
- Institute for Pharmaceutical Microbiology and Institute for Medical Microbiology, Immunology, and Parasitology, University of Bonn, D-53115 Bonn, Germany
| | - Chinedu Ejeh
- Department of Microbiology & Immunology, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire 03755, United States
| | - Travis Hammerstad
- Department of Medicinal Chemistry, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Hans Georg Sahl
- Institute for Pharmaceutical Microbiology and Institute for Medical Microbiology, Immunology, and Parasitology, University of Bonn, D-53115 Bonn, Germany
| | - Pedro M Pereira
- Bacterial Cell Biology Laboratory, Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Avenida da República (EAN), 2781-901 Oeiras, Portugal
| | - Mariana G Pinho
- Bacterial Cell Biology Laboratory, Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Avenida da República (EAN), 2781-901 Oeiras, Portugal
| | - Véronique Dartois
- Center for Discovery and Innovation, Hackensack Meridian Health, Nutley, New Jersey 07110, United States
| | - Ambrose Cheung
- Department of Microbiology & Immunology, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire 03755, United States
| | - Courtney C Aldrich
- Department of Medicinal Chemistry, University of Minnesota, Minneapolis, Minnesota 55455, United States
| |
Collapse
|
31
|
Lade H, Joo HS, Kim JS. Molecular Basis of Non-β-Lactam Antibiotics Resistance in Staphylococcus aureus. Antibiotics (Basel) 2022; 11:1378. [PMID: 36290036 PMCID: PMC9598170 DOI: 10.3390/antibiotics11101378] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Revised: 10/04/2022] [Accepted: 10/06/2022] [Indexed: 11/22/2022] Open
Abstract
Methicillin-resistant Staphylococcus aureus (MRSA) is one of the most successful human pathogens with the potential to cause significant morbidity and mortality. MRSA has acquired resistance to almost all β-lactam antibiotics, including the new-generation cephalosporins, and is often also resistant to multiple other antibiotic classes. The expression of penicillin-binding protein 2a (PBP2a) is the primary basis for β-lactams resistance by MRSA, but it is coupled with other resistance mechanisms, conferring resistance to non-β-lactam antibiotics. The multiplicity of resistance mechanisms includes target modification, enzymatic drug inactivation, and decreased antibiotic uptake or efflux. This review highlights the molecular basis of resistance to non-β-lactam antibiotics recommended to treat MRSA infections such as macrolides, lincosamides, aminoglycosides, glycopeptides, oxazolidinones, lipopeptides, and others. A thorough understanding of the molecular and biochemical basis of antibiotic resistance in clinical isolates could help in developing promising therapies and molecular detection methods of antibiotic resistance.
Collapse
Affiliation(s)
- Harshad Lade
- Department of Laboratory Medicine, Hallym University College of Medicine, Kangdong Sacred Heart Hospital, Seoul 05355, Korea
| | - Hwang-Soo Joo
- Department of Biotechnology, College of Engineering, Duksung Women’s University, Seoul 01369, Korea
| | - Jae-Seok Kim
- Department of Laboratory Medicine, Hallym University College of Medicine, Kangdong Sacred Heart Hospital, Seoul 05355, Korea
| |
Collapse
|
32
|
Poshvina DV, Dilbaryan DS, Kasyanov SP, Sadykova VS, Lapchinskaya OA, Rogozhin EA, Vasilchenko AS. Staphylococcus aureus is able to generate resistance to novel lipoglycopeptide antibiotic gausemycin A. Front Microbiol 2022; 13:963979. [PMID: 36246291 PMCID: PMC9558223 DOI: 10.3389/fmicb.2022.963979] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Accepted: 09/14/2022] [Indexed: 11/13/2022] Open
Abstract
Gausemycin A is the first member of the novel lipoglycopeptides family produced by Streptomyces roseoflavus INA-Ac-5812. Gausemycin A has a pronounced bactericidal activity against methicillin-resistant Staphylococcus aureus. However, the ability of S. aureus to be resistant to gausemycin A has not been investigated yet. Using serial passaging, we have obtained the resistant variant S. aureus 5812R, which is 80 times more resistant compared to the parent strain. Susceptibility testing of S. aureus 5812R revealed the acquisition of cross-resistance to daptomycin, cefazolin, tetracycline, and gentamicin, while the resistance to vancomycin, nisin, and ramoplanin was absent. Whole genome sequencing revealed single nucleotide polymorphism (SNP) and deletions in S. aureus 5812R, among which are genes encoding efflux pump (sepA), the two-component Kdp system (kdpE), and the component of isoprenoid biosynthesis pathway (hepT). Phenotypically, S. aureus 5812R resembles a small-colony variant, as it is slow-growing, forms small colonies, and is deficient in pigments. Profiling of fatty acids (FA) composition constituting the cytoplasmic membrane of S. aureus 5812R revealed the prevalence of anteiso-branched FA, while straight FA was slightly less present. The evidence also showed that the gausemycin A-resistant strain has increased expression of the cls2 gene of the cardiolipin synthase. The performed checkerboard assay pointed out that the combination of gausemycin A and ciprofloxacin showed a synergistic effect against S. aureus 5812R.
Collapse
Affiliation(s)
- Darya V. Poshvina
- Laboratory of Antimicrobial Resistance, Institute of Environmental and Agricultural Biology (X-BIO), Tyumen State University, Tyumen, Russia
| | - Diana S. Dilbaryan
- Laboratory of Antimicrobial Resistance, Institute of Environmental and Agricultural Biology (X-BIO), Tyumen State University, Tyumen, Russia
| | - Sergey P. Kasyanov
- A.V. Zhirmunsky National Scientific Center of Marine Biology, Vladivostok, Russia
| | | | | | - Eugene A. Rogozhin
- Gause Institute of New Antibiotics, Moscow, Russia
- Shemyakin and Ovchinnikov Institute of Bioorganic Chemistry Russian Academy of Sciences, Moscow, Russia
| | - Alexey S. Vasilchenko
- Laboratory of Antimicrobial Resistance, Institute of Environmental and Agricultural Biology (X-BIO), Tyumen State University, Tyumen, Russia
- *Correspondence: Alexey S. Vasilchenko
| |
Collapse
|
33
|
Huang S, Fu Y, Mo A. Electrophoretic-deposited MXene titanium coatings in regulating bacteria and cell response for peri-implantitis. Front Chem 2022; 10:991481. [PMID: 36247682 PMCID: PMC9558740 DOI: 10.3389/fchem.2022.991481] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Accepted: 08/16/2022] [Indexed: 11/16/2022] Open
Abstract
Background: Two-dimensional(2D)MXenes have continued to receive increasing interest from researchers due to their graphene-like properties, in addition to their versatile properties for applications in electronic devices, power generation, sensors, drug delivery, and biomedicine. However, their construction and biological properties as titanium coatings to prevent peri-implantitis are still unclear. Materials and methods: In this work, few-layer Ti3C2Tx MXene coatings with different thicknesses at varied depositing voltages (30, 40, and 50 V) were constructed by anodic electrophoretic deposition without adding any electrolytic ions. In vitro cytocompatibility assay was performed on preosteoblasts (MC3T3-E1) cell lines after the characterization of the coating. Meanwhile, the antibacterial activity against bacteria which are closely related to peri-implantitis including Staphylococcus aureus (S. aureus) and its drug-resistant strain MRSA was further investigated. Results: MXene-coated titanium models with different thicknesses were successfully assembled by analyzing the results of characterization. The compounding of Ti3C2Tx could significantly improve the initial adhesion and proliferation of MC3T3-E1 cells. Moreover, the coating can effectively inhibit the adhesion and cell activity of S. aureus and MRSA, and MRSA expressed greater restricting behavior than S. aureus. The ability to promote antibacterial activity is proportional to the content of Ti3C2Tx. Its antioxidant capacity to reduce ROS in the culture environment and bacterial cells was first revealed. Conclusion: In summary, this work shows a new avenue for MXene-based nano-biomaterials under the clinical problem of multiple antibiotic resistance.
Collapse
Affiliation(s)
- Si Huang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Sichuan University, Chengdu, China
- Department of Implantology, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Yu Fu
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Sichuan University, Chengdu, China
- Department of Implantology, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Anchun Mo
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Sichuan University, Chengdu, China
- Department of Implantology, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- *Correspondence: Anchun Mo,
| |
Collapse
|
34
|
Tsai CE, Yang CJ, Chuang YC, Wang JT, Sheng WH, Chen YC, Chang SC. Evaluation of the synergistic effect of ceftaroline against methicillin-resistant Staphylococcus aureus. Int J Infect Dis 2022; 122:230-236. [PMID: 35640827 DOI: 10.1016/j.ijid.2022.05.057] [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: 02/24/2022] [Revised: 04/22/2022] [Accepted: 05/25/2022] [Indexed: 10/18/2022] Open
Abstract
OBJECTIVES We aimed to determine the synergistic effects of ceftaroline (CPT) in combination with daptomycin (DAP), vancomycin (VAN), or linezolid (LNZ) against various methicillin-resistant Staphylococcus aureus (MRSA) strains. METHODS MRSA strains randomly selected from 2014 to 2018 were studied. Checkerboard titration and in vitro time-kill analyses were used to determine the synergistic activities of the antibiotic combinations. RESULTS A total of 10 genetically distinct MRSA strains were included in this study. The checkerboard titration analysis revealed that the CPT-DAP, CPT-VAN, and CPT-LNZ combinations had a synergistic effect against 30%, 10%, and 10% of the selected MRSA strains, respectively. Using time-kill analysis, we showed that CPT-DAP exhibited a significant synergistic and sustained bactericidal effect against both DAP-susceptible (Δ colony-forming units/ml, -5.79; P = 0.0495) and DAP-resistant (Δ colony-forming units/ml, -6.40; P = 0.0463) MRSA strains at a concentration of 0.5 × the minimum inhibitory concentration of CPT plus 0.5 × the minimum inhibitory concentration of DAP. No synergistic bactericidal effects were observed for the CPT-VAN and CPT-LNZ combinations against the selected strains. CONCLUSION The CPT-DAP combination showed better synergistic activity than the CPT-VAN and CPT-LNZ combinations against the enrolled MRSA strains. DAP, rather than VAN or LNZ, might be a better choice for CPT combination in the treatment of MRSA infections.
Collapse
Affiliation(s)
- Cheng-En Tsai
- School of Medicine, National Taiwan University College of Medicine, Taipei City, Taiwan
| | - Chia-Jui Yang
- Department of Internal Medicine, Far Eastern Memorial Hospital, New Taipei City, Taiwan; School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Yu-Chung Chuang
- Department of Internal Medicine, National Taiwan University Hospital, Taipei City, Taiwan.
| | - Jann-Tay Wang
- Department of Internal Medicine, National Taiwan University Hospital, Taipei City, Taiwan
| | - Wang-Huei Sheng
- Department of Internal Medicine, National Taiwan University Hospital, Taipei City, Taiwan
| | - Yee-Chun Chen
- Department of Internal Medicine, National Taiwan University Hospital, Taipei City, Taiwan
| | - Shan-Chwen Chang
- Department of Internal Medicine, National Taiwan University Hospital, Taipei City, Taiwan
| |
Collapse
|
35
|
Douglas EJA, Alkhzem AH, Wonfor T, Li S, Woodman TJ, Blagbrough IS, Laabei M. Antibacterial activity of novel linear polyamines against Staphylococcus aureus. Front Microbiol 2022; 13:948343. [PMID: 36071957 PMCID: PMC9441809 DOI: 10.3389/fmicb.2022.948343] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Accepted: 08/04/2022] [Indexed: 01/11/2023] Open
Abstract
New therapeutic options are urgently required for the treatment of Staphylococcus aureus infections. Accordingly, we sought to exploit the vulnerability of S. aureus to naturally occurring polyamines. We have developed and tested the anti-staphylococcal activity of three novel linear polyamines based on spermine and norspermine. Using a panel of genetically distinct and clinically relevant multidrug resistant S. aureus isolates, including the polyamine resistant USA300 strain LAC, compound AHA-1394 showed a greater than 128-fold increase in inhibition against specific S. aureus strains compared to the most active natural polyamine. Furthermore, we show that AHA-1394 has superior biofilm prevention and biofilm dispersal properties compared to natural polyamines while maintaining minimal toxicity toward human HepG2 cells. We examined the potential of S. aureus to gain resistance to AHA-1394 following in vitro serial passage. Whole genome sequencing of two stable resistant mutants identified a gain of function mutation (S337L) in the phosphatidylglycerol lysyltransferase mprF gene. Inactivation of mutant mprF confirmed the importance of this allele to AHA-1394 resistance. Importantly, AHA-1394 resistant mutants showed a marked decrease in relative fitness and increased generation time. Intriguingly, mprF::S337L contributed to altered surface charge only in the USA300 background whereas increased cell wall thickness was observed in both USA300 and SH1000. Lastly, we show that AHA-1394 displays a particular proclivity for antibiotic potentiation, restoring sensitivity of MRSA and VRSA isolates to daptomycin, oxacillin and vancomycin. Together this study shows that polyamine derivatives are impressive drug candidates that warrant further investigation.
Collapse
Affiliation(s)
- Edward J. A. Douglas
- Department of Biology and Biochemistry, University of Bath, Bath, United Kingdom
| | - Abdulaziz H. Alkhzem
- Department of Pharmacy and Pharmacology, University of Bath, Bath, United Kingdom
| | - Toska Wonfor
- Department of Biology and Biochemistry, University of Bath, Bath, United Kingdom
| | - Shuxian Li
- Department of Biology and Biochemistry, University of Bath, Bath, United Kingdom
| | - Timothy J. Woodman
- Department of Pharmacy and Pharmacology, University of Bath, Bath, United Kingdom
| | - Ian S. Blagbrough
- Department of Pharmacy and Pharmacology, University of Bath, Bath, United Kingdom
| | - Maisem Laabei
- Department of Biology and Biochemistry, University of Bath, Bath, United Kingdom
- *Correspondence: Maisem Laabei,
| |
Collapse
|
36
|
Joodaki F, Martin LM, Greenfield ML. Generation and Computational Characterization of a Complex Staphylococcus aureus Lipid Bilayer. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2022; 38:9481-9499. [PMID: 35901279 DOI: 10.1021/acs.langmuir.2c00483] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Studies indicate a crucial cell membrane role in the antibiotic resistance of Staphylococcus aureus. To simulate its membrane structure and dynamics, a complex molecular-scale computational representation of the S. aureus lipid bilayer was developed. Phospholipid types and their amounts were optimized by reverse Monte Carlo to represent characterization data from the literature, leading to 19 different phospholipid types that combine three headgroups [phosphatidylglycerol, lysyl-phosphatidylglycerol (LPG), and cardiolipin] and 10 tails, including iso- and anteiso-branched saturated chains. The averaged lipid bilayer thickness was 36.7 Å, and area per headgroup was 67.8 Å2. Phosphorus and nitrogen density profiles showed that LPG headgroups tended to be bent and oriented more parallel to the bilayer plane. The water density profile showed that small amounts reached the membrane center. Carbon density profiles indicated hydrophobic interactions for all lipids in the middle of the bilayer. Bond vector order parameters along each tail demonstrated different C-H ordering even within distinct lipids of the same type; however, all tails followed similar trends in average order parameter. These complex simulations further revealed bilayer insights beyond those attainable with monodisperse, unbranched lipids. Longer tails often extended into the opposite leaflet. Carbon at and beyond a branch showed significantly decreased ordering compared to carbon in unbranched tails; this feature arose in every branched lipid. Diverse tail lengths distributed these disordered methyl groups throughout the middle third of the bilayer. Distributions in mobility and ordering reveal diverse properties that cannot be obtained with monodisperse lipids.
Collapse
Affiliation(s)
- Faramarz Joodaki
- Department of Chemical Engineering, University of Rhode Island, 360 Fascitelli Center for Advanced Engineering, Kingston, Rhode Island 02881, United States
| | - Lenore M Martin
- Department of Cell and Molecular Biology, University of Rhode Island, 120 Flagg Road, Kingston, Rhode Island 02881, United States
| | - Michael L Greenfield
- Department of Chemical Engineering, University of Rhode Island, 360 Fascitelli Center for Advanced Engineering, Kingston, Rhode Island 02881, United States
| |
Collapse
|
37
|
Dysregulation of Cell Envelope Homeostasis in Staphylococcus aureus Exposed to Solvated Lignin. Appl Environ Microbiol 2022; 88:e0054822. [PMID: 35852361 PMCID: PMC9361832 DOI: 10.1128/aem.00548-22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
Lignin is an aromatic plant cell wall polymer that facilitates water transport through the vasculature of plants and is generated in large quantities as an inexpensive by-product of pulp and paper manufacturing and biorefineries. Although lignin's ability to reduce bacterial growth has been reported previously, its hydrophobicity complicates the ability to examine its biological effects on living cells in aqueous growth media. We recently described the ability to solvate lignin in Good's buffers with neutral pH, a breakthrough that allowed examination of lignin's antimicrobial effects against the human pathogen Staphylococcus aureus. These analyses showed that lignin damages the S. aureus cell membrane, causes increased cell clustering, and inhibits growth synergistically with tunicamycin, a teichoic acid synthesis inhibitor. In the present study, we examined the physiological and transcriptomic responses of S. aureus to lignin. Intriguingly, lignin restored the susceptibility of genetically resistant S. aureus isolates to penicillin and oxacillin, decreased intracellular pH, impaired normal cell division, and rendered cells more resistant to detergent-induced lysis. Additionally, transcriptome sequencing (RNA-Seq) differential expression (DE) analysis of lignin-treated cultures revealed significant gene expression changes (P < 0.05 with 5% false discovery rate [FDR]) related to the cell envelope, cell wall physiology, fatty acid metabolism, and stress resistance. Moreover, a pattern of concurrent up- and downregulation of genes within biochemical pathways involved in transmembrane transport and cell wall physiology was observed, which likely reflects an attempt to tolerate or compensate for lignin-induced damage. Together, these results represent the first comprehensive analysis of lignin's antibacterial activity against S. aureus. IMPORTANCE S. aureus is a leading cause of skin and soft tissue infections. The ability of S. aureus to acquire genetic resistance to antibiotics further compounds its ability to cause life-threatening infections. While the historical response to antibiotic resistance has been to develop new antibiotics, bacterial pathogens are notorious for rapidly acquiring genetic resistance mechanisms. As such, the development of adjuvants represents a viable way of extending the life span of current antibiotics to which pathogens may already be resistant. Here, we describe the phenotypic and transcriptomic response of S. aureus to treatment with lignin. Our results demonstrate that lignin extracted from sugarcane and sorghum bagasse restores S. aureus susceptibility to β-lactams, providing a premise for repurposing these antibiotics in treatment of resistant S. aureus strains, possibly in the form of topical lignin/β-lactam formulations.
Collapse
|
38
|
Akunuri R, Unnissa T, Vadakattu M, Bujji S, Mahammad Ghouse S, Madhavi Yaddanapudi V, Chopra S, Nanduri S. Bacterial Pyruvate Kinase: A New Potential Target to Combat Drug‐Resistant
Staphylococcus aureus
Infections. ChemistrySelect 2022. [DOI: 10.1002/slct.202201403] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Affiliation(s)
- Ravikumar Akunuri
- Department of Chemical Sciences National Institute of Pharmaceutical Education and Research (NIPER) Hyderabad 500 037, Telangana State India
| | - Tanveer Unnissa
- Department of Chemical Sciences National Institute of Pharmaceutical Education and Research (NIPER) Hyderabad 500 037, Telangana State India
| | - Manasa Vadakattu
- Department of Chemical Sciences National Institute of Pharmaceutical Education and Research (NIPER) Hyderabad 500 037, Telangana State India
| | - Sushmitha Bujji
- Department of Chemical Sciences National Institute of Pharmaceutical Education and Research (NIPER) Hyderabad 500 037, Telangana State India
| | - Shaik Mahammad Ghouse
- Department of Chemical Sciences National Institute of Pharmaceutical Education and Research (NIPER) Hyderabad 500 037, Telangana State India
| | - Venkata Madhavi Yaddanapudi
- Department of Chemical Sciences National Institute of Pharmaceutical Education and Research (NIPER) Hyderabad 500 037, Telangana State India
| | - Sidharth Chopra
- Division of Molecular Microbiology and Immunology CSIR-Central Drug Research Institute (CDRI) Sitapur Road, Sector 10, Janakipuram Extension Lucknow 226 031, Uttar Pradesh India
| | - Srinivas Nanduri
- Department of Chemical Sciences National Institute of Pharmaceutical Education and Research (NIPER) Hyderabad 500 037, Telangana State India
| |
Collapse
|
39
|
Sulaiman JE, Long L, Qian PY, Lam H. Proteome profiling of evolved methicillin-resistant Staphylococcus aureus strains with distinct daptomycin tolerance and resistance phenotypes. Front Microbiol 2022; 13:970146. [PMID: 35992709 PMCID: PMC9386379 DOI: 10.3389/fmicb.2022.970146] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Accepted: 07/13/2022] [Indexed: 12/04/2022] Open
Abstract
Methicillin-resistant Staphylococcus aureus (MRSA) is a highly dangerous pathogen, and daptomycin has been increasingly used to treat its infections in clinics. Recently, several groups have shown that tolerance and resistance of microbes can evolve rapidly under cyclic antibiotic exposure. We have previously shown that the same tolerance and resistance development occurs in MRSA treated with daptomycin in an adaptive laboratory evolution (ALE) experiment. In the present study, we performed proteomic analysis to compare six daptomycin-tolerant and resistant MRSA strains that were evolved from the same ancestral strain. The strain with a higher tolerance level than the others had the most different proteome and response to antibiotic treatment, resembling those observed in persister cells, which are small subpopulations of bacteria that survive lethal antibiotics treatment. By comparing the proteome changes across strains with similar phenotypes, we identified the key proteins that play important roles in daptomycin tolerance and resistance in MRSA. We selected two candidates to be confirmed by gene overexpression analysis. Overexpression of EcsA1 and FabG, which were up-regulated in all of the tolerant evolved strains, led to increased daptomycin tolerance in wild-type MRSA. The proteomics data also suggested that cell wall modulations were implicated in both resistance and tolerance, but in different ways. While the resistant strains had peptidoglycan changes and a more positive surface charge to directly repel daptomycin, the tolerant strains possessed different cell wall changes that do not involve the peptidoglycan nor alterations of the surface charge. Overall, our study showed the differential proteome profiles among multiple tolerant and resistant strains, pinpointed the key proteins for the two phenotypes and revealed the differences in cell wall modulations between the daptomycin-tolerant/resistant strains.
Collapse
Affiliation(s)
- Jordy Evan Sulaiman
- Department of Chemical and Biological Engineering, The Hong Kong University of Science and Technology, Kowloon, Hong Kong SAR, China
| | - Lexin Long
- Department of Ocean Science and Hong Kong Branch of Southern Marine Science and Engineering Guangdong Laboratory (Guangzhou), The Hong Kong University of Science and Technology, Kowloon, Hong Kong SAR, China
| | - Pei-Yuan Qian
- Department of Ocean Science and Hong Kong Branch of Southern Marine Science and Engineering Guangdong Laboratory (Guangzhou), The Hong Kong University of Science and Technology, Kowloon, Hong Kong SAR, China
- Southern Marine Science and Engineering Guangdong Laboratory, Guangzhou, China
| | - Henry Lam
- Department of Chemical and Biological Engineering, The Hong Kong University of Science and Technology, Kowloon, Hong Kong SAR, China
- *Correspondence: Henry Lam,
| |
Collapse
|
40
|
Hofer W, Oueis E, Fayad AA, Deschner F, Andreas A, de Carvalho LP, Hüttel S, Bernecker S, Pätzold L, Morgenstern B, Zaburannyi N, Bischoff M, Stadler M, Held J, Herrmann J, Müller R. Regio‐ and Stereoselective Epoxidation and Acidic Epoxide Opening of Antibacterial and Antiplasmodial Chlorotonils Yield Highly Potent Derivatives. Angew Chem Int Ed Engl 2022; 61:e202202816. [PMID: 35485800 PMCID: PMC9400904 DOI: 10.1002/anie.202202816] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Indexed: 11/12/2022]
Abstract
The rise of antimicrobial resistance poses a severe threat to public health. The natural product chlorotonil was identified as a new antibiotic targeting multidrug resistant Gram‐positive pathogens and Plasmodium falciparum. Although chlorotonil shows promising activities, the scaffold is highly lipophilic and displays potential biological instabilities. Therefore, we strived towards improving its pharmaceutical properties by semisynthesis. We demonstrated stereoselective epoxidation of chlorotonils and epoxide ring opening in moderate to good yields providing derivatives with significantly enhanced solubility. Furthermore, in vivo stability of the derivatives was improved while retaining their nanomolar activity against critical human pathogens (e.g. methicillin‐resistant Staphylococcus aureus and P. falciparum). Intriguingly, we showed further superb activity for the frontrunner molecule in a mouse model of S. aureus infection.
Collapse
Affiliation(s)
- Walter Hofer
- Microbial Natural Products Helmholtz Institute for Pharmaceutical Research Saarland (HIPS) Helmholtz Centre for Infection Research (HZI) and Department of Pharmacy at Saarland University Campus Building E8.1 66123 Saarbrücken Germany
- German Centre for Infection Research (DZIF) Braunschweig Germany
| | - Emilia Oueis
- Microbial Natural Products Helmholtz Institute for Pharmaceutical Research Saarland (HIPS) Helmholtz Centre for Infection Research (HZI) and Department of Pharmacy at Saarland University Campus Building E8.1 66123 Saarbrücken Germany
- Department of Chemistry Khalifa University of Science and Technology 127788 Abu Dhabi United Arab Emirates
- American University of Beirut Faculty of Medicine DTS Bldg, Second Floor, Room 215-B Beirut Lebanon
| | - Antoine Abou Fayad
- Microbial Natural Products Helmholtz Institute for Pharmaceutical Research Saarland (HIPS) Helmholtz Centre for Infection Research (HZI) and Department of Pharmacy at Saarland University Campus Building E8.1 66123 Saarbrücken Germany
- Department of Experimental Pathology Immunology and Microbiology Center for Infectious Disease Research (CIDR) WHO Collaborating Center for Reference and Research on Bacterial Pathogens American University of Beirut Faculty of Medicine DTS Bldg, Second Floor, Room 215-B Beirut Lebanon
| | - Felix Deschner
- Microbial Natural Products Helmholtz Institute for Pharmaceutical Research Saarland (HIPS) Helmholtz Centre for Infection Research (HZI) and Department of Pharmacy at Saarland University Campus Building E8.1 66123 Saarbrücken Germany
- German Centre for Infection Research (DZIF) Braunschweig Germany
| | - Anastasia Andreas
- Microbial Natural Products Helmholtz Institute for Pharmaceutical Research Saarland (HIPS) Helmholtz Centre for Infection Research (HZI) and Department of Pharmacy at Saarland University Campus Building E8.1 66123 Saarbrücken Germany
- German Centre for Infection Research (DZIF) Braunschweig Germany
| | - Laìs Pessanha de Carvalho
- German Centre for Infection Research (DZIF) Braunschweig Germany
- Institute of Tropical Medicine Eberhard Karls University Tübingen Wilhelmstraße 27 72074 Tübingen Germany
| | - Stephan Hüttel
- German Centre for Infection Research (DZIF) Braunschweig Germany
- Microbial Drugs Helmholtz Centre for Infection Research (HZI) Inhoffenstraße 7 38124 Braunschweig Germany
| | - Steffen Bernecker
- Microbial Drugs Helmholtz Centre for Infection Research (HZI) Inhoffenstraße 7 38124 Braunschweig Germany
| | - Linda Pätzold
- Institute for Medical Microbiology and Hygiene Saarland University 66421 Homburg Germany
| | - Bernd Morgenstern
- Inorganic Solid State Chemistry Saarland University Campus 66123 Saarbrücken Germany
| | - Nestor Zaburannyi
- Microbial Natural Products Helmholtz Institute for Pharmaceutical Research Saarland (HIPS) Helmholtz Centre for Infection Research (HZI) and Department of Pharmacy at Saarland University Campus Building E8.1 66123 Saarbrücken Germany
| | - Markus Bischoff
- Institute for Medical Microbiology and Hygiene Saarland University 66421 Homburg Germany
| | - Marc Stadler
- German Centre for Infection Research (DZIF) Braunschweig Germany
- Microbial Drugs Helmholtz Centre for Infection Research (HZI) Inhoffenstraße 7 38124 Braunschweig Germany
| | - Jana Held
- German Centre for Infection Research (DZIF) Braunschweig Germany
- Institute of Tropical Medicine Eberhard Karls University Tübingen Wilhelmstraße 27 72074 Tübingen Germany
- Centre de Recherches Médicales de Lambaréné Lambaréné Gabon
| | - Jennifer Herrmann
- Microbial Natural Products Helmholtz Institute for Pharmaceutical Research Saarland (HIPS) Helmholtz Centre for Infection Research (HZI) and Department of Pharmacy at Saarland University Campus Building E8.1 66123 Saarbrücken Germany
- German Centre for Infection Research (DZIF) Braunschweig Germany
| | - Rolf Müller
- Microbial Natural Products Helmholtz Institute for Pharmaceutical Research Saarland (HIPS) Helmholtz Centre for Infection Research (HZI) and Department of Pharmacy at Saarland University Campus Building E8.1 66123 Saarbrücken Germany
- German Centre for Infection Research (DZIF) Braunschweig Germany
| |
Collapse
|
41
|
Chen H, Yang N, Yu L, Li J, Zhang H, Zheng Y, Xu M, Liu Y, Yang Y, Li J. Synergistic Microbicidal Effect of AUR and PEITC Against Staphylococcus aureus Skin Infection. Front Cell Infect Microbiol 2022; 12:927289. [PMID: 35774400 PMCID: PMC9237442 DOI: 10.3389/fcimb.2022.927289] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2022] [Accepted: 05/11/2022] [Indexed: 11/13/2022] Open
Abstract
Given the increasing prevalence of Staphylococcus aureus antibiotic resistance, there is an urgent need to repurpose approved drugs with known pharmacology and toxicology as an alternative therapeutic strategy. We have reported that the sustained monotherapy of auranofin (AUR) inevitably resulted in reduced susceptibility or even the emergence of resistance to AUR in S. aureus. However, whether drug combination could increase antibacterial activity while preventing AUR resistance is still unknown. Here, we focused on the important role of AUR combined with phenethyl isothiocyanate (PEITC) in skin infection and determined the synergistic antimicrobial effect on S. aureus by using checkerboard assays and time-kill kinetics analysis. This synergistic antimicrobial activity correlated with increased reactive oxygen species (ROS) generation, disruption of bacterial cell structure, and inhibition of biofilm formation. We also showed that AUR synergized with PEITC effectively restored the susceptibility to AUR via regulating thioredoxin reductase (TrxR) and rescued mice from subcutaneous abscesses through eliminating S. aureus pathogens, including methicillin-resistant S. aureus (MRSA). Collectively, our study indicated that the AUR and PEITC combination had a synergistic antimicrobial impact on S. aureus in vitro and in vivo. These results suggest that AUR and PEITC treatment may be a promising option for S. aureus infection.
Collapse
Affiliation(s)
- Haoran Chen
- Department of Infectious Disease, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Ning Yang
- Department of Infectious Disease, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Liang Yu
- Department of Infectious Disease, The First Affiliated Hospital of Anhui Medical University, Hefei, China
- Anhui Center for Surveillance of Bacterial Resistance, Hefei, China
- Institute of Bacterial Resistance, Anhui Medical University, Hefei, China
| | - Jiajia Li
- The Center for Scientific Research, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Hui Zhang
- Department of Infectious Disease, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Yahong Zheng
- Department of Infectious Disease, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Mengran Xu
- Department of Infectious Disease, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Yanyan Liu
- Department of Infectious Disease, The First Affiliated Hospital of Anhui Medical University, Hefei, China
- Anhui Center for Surveillance of Bacterial Resistance, Hefei, China
- Institute of Bacterial Resistance, Anhui Medical University, Hefei, China
| | - Yi Yang
- Department of Infectious Disease, The First Affiliated Hospital of Anhui Medical University, Hefei, China
- *Correspondence: Yi Yang, ; Jiabin Li,
| | - Jiabin Li
- Department of Infectious Disease, The First Affiliated Hospital of Anhui Medical University, Hefei, China
- Anhui Center for Surveillance of Bacterial Resistance, Hefei, China
- Institute of Bacterial Resistance, Anhui Medical University, Hefei, China
- *Correspondence: Yi Yang, ; Jiabin Li,
| |
Collapse
|
42
|
Santos AL, Liu D, Reed AK, Wyderka AM, van Venrooy A, Li JT, Li VD, Misiura M, Samoylova O, Beckham JL, Ayala-Orozco C, Kolomeisky AB, Alemany LB, Oliver A, Tegos GP, Tour JM. Light-activated molecular machines are fast-acting broad-spectrum antibacterials that target the membrane. SCIENCE ADVANCES 2022; 8:eabm2055. [PMID: 35648847 PMCID: PMC9159576 DOI: 10.1126/sciadv.abm2055] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Accepted: 04/14/2022] [Indexed: 06/01/2023]
Abstract
The increasing occurrence of antibiotic-resistant bacteria and the dwindling antibiotic research and development pipeline have created a pressing global health crisis. Here, we report the discovery of a distinctive antibacterial therapy that uses visible (405 nanometers) light-activated synthetic molecular machines (MMs) to kill Gram-negative and Gram-positive bacteria, including methicillin-resistant Staphylococcus aureus, in minutes, vastly outpacing conventional antibiotics. MMs also rapidly eliminate persister cells and established bacterial biofilms. The antibacterial mode of action of MMs involves physical disruption of the membrane. In addition, by permeabilizing the membrane, MMs at sublethal doses potentiate the action of conventional antibiotics. Repeated exposure to antibacterial MMs is not accompanied by resistance development. Finally, therapeutic doses of MMs mitigate mortality associated with bacterial infection in an in vivo model of burn wound infection. Visible light-activated MMs represent an unconventional antibacterial mode of action by mechanical disruption at the molecular scale, not existent in nature and to which resistance development is unlikely.
Collapse
Affiliation(s)
- Ana L. Santos
- Department of Chemistry, Rice University, Houston, TX 77005, USA
- IdISBA–Fundación de Investigación Sanitaria de las Islas Baleares, Palma, Spain
| | - Dongdong Liu
- Department of Chemistry, Rice University, Houston, TX 77005, USA
| | - Anna K. Reed
- Department of Chemistry, Rice University, Houston, TX 77005, USA
| | - Aaron M. Wyderka
- Department of Chemistry, Rice University, Houston, TX 77005, USA
| | | | - John T. Li
- Department of Chemistry, Rice University, Houston, TX 77005, USA
| | - Victor D. Li
- Department of Chemistry, Rice University, Houston, TX 77005, USA
| | - Mikita Misiura
- Department of Chemistry, Rice University, Houston, TX 77005, USA
| | - Olga Samoylova
- Department of Chemistry, Rice University, Houston, TX 77005, USA
| | - Jacob L. Beckham
- Department of Chemistry, Rice University, Houston, TX 77005, USA
| | | | | | - Lawrence B. Alemany
- Department of Chemistry, Rice University, Houston, TX 77005, USA
- Shared Equipment Authority, Rice University, Houston, TX 77005, USA
| | - Antonio Oliver
- IdISBA–Fundación de Investigación Sanitaria de las Islas Baleares, Palma, Spain
- Servicio de Microbiologia, Hospital Universitari Son Espases, Palma, Spain
| | - George P. Tegos
- Office of Research, Reading Hospital, Tower Health, 420 S. Fifth Avenue, West Reading, PA 19611, USA
| | - James M. Tour
- Department of Chemistry, Rice University, Houston, TX 77005, USA
- Smalley-Curl Institute, Rice University, Houston, TX 77005, USA
- Department of Materials Science and NanoEngineering, Rice University, Houston, TX 77005, USA
- NanoCarbon Center and the Welch Institute for Advanced Materials, Rice University, Houston, TX 77005, USA
| |
Collapse
|
43
|
Müller R, Hofer W, Oueis E, Abou Fayad A, Deschner F, Andreas A, de Carvalho LP, Hüttel S, Bernecker S, Pätzold L, Morgenstern B, Zaburannyi N, Bischoff M, Stadler M, Held J, Herrmann J. Regio‐ and Stereoselective Epoxidation and Acidic Epoxide Opening of Antibacterial and Antiplasmodial Chlorotonils Yield Highly Potent Derivatives. Angew Chem Int Ed Engl 2022. [DOI: 10.1002/ange.202202816] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Affiliation(s)
- Rolf Müller
- Helmholtz-Institute for Pharmaceutical Research Saarland Microbial Natural Products Campus Building E8.1 66123 Saarbrücken GERMANY
| | - Walter Hofer
- Helmholtz-Institut fur Pharmazeutische Forschung Saarland Microbial Natural Products Campus Building E8.1 66123 Saarbrücken GERMANY
| | - Emilia Oueis
- Khalifa University of Science and Technology Department of Chemistry 127788 Abu Dhabi UNITED ARAB EMIRATES
| | - Antoine Abou Fayad
- American University of Beirut Department of Experimental Pathology, Immunology and Microbiology Beirut LEBANON
| | - Felix Deschner
- Helmholtz-Institut fur Pharmazeutische Forschung Saarland Microbial Natural Products Campus Building E8.1 66123 Saarbrücken GERMANY
| | - Anastasia Andreas
- Helmholtz-Institut fur Pharmazeutische Forschung Saarland Microbial Natural Products Campus Building E8.1 66123 Saarbrücken GERMANY
| | - Laìs Pessanha de Carvalho
- University of Tübingen: Eberhard Karls Universitat Tubingen Institute of Tropical Medicine Wilhelmstraße 27 72074 Tübingen GERMANY
| | - Stephan Hüttel
- Helmholtz Centre for Infection Research: Helmholtz-Zentrum fur Infektionsforschung GmbH Microbial Drugs Inhoffenstraße 7 38124 Braunschweig GERMANY
| | - Steffen Bernecker
- HZI: Helmholtz-Zentrum fur Infektionsforschung GmbH Microbial Drugs Inhoffenstraße 7 38124 Braunschweig GERMANY
| | - Linda Pätzold
- Universität des Saarlandes: Universitat des Saarlandes Institute for Medical Microbiology and Hygiene 66421 Homburg GERMANY
| | - Bernd Morgenstern
- Universität des Saarlandes: Universitat des Saarlandes Inorganic Solid State Chemistry 66123 Saarbrücken GERMANY
| | - Nestor Zaburannyi
- Helmholtz-Institut fur Pharmazeutische Forschung Saarland Microbial Natural Products 66123 Saarbrücken GERMANY
| | - Markus Bischoff
- Universität des Saarlandes: Universitat des Saarlandes Institute for Medical Microbiology and Hygiene 66421 Homburg GERMANY
| | - Marc Stadler
- HZI: Helmholtz-Zentrum fur Infektionsforschung GmbH Microbial Drugs Inhoffenstraße 7 38124 Braunschweig GERMANY
| | - Jana Held
- Eberhard Karls Universität Tübingen: Eberhard Karls Universitat Tubingen Institute of Tropical Medicine Wilhelmstraße 27 72074 Tübingen GERMANY
| | - Jennifer Herrmann
- Helmholtz-Institut fur Pharmazeutische Forschung Saarland Microbial Natural Products Campus Building E8.1 66123 Saarbrücken GERMANY
| |
Collapse
|
44
|
Streifel AC, Varley CD, Ham Y, Sikka MK, Lewis JS. The challenge of antibiotic selection in prosthetic joint infections due to Corynebacterium striatum: a case report. BMC Infect Dis 2022; 22:290. [PMID: 35346085 PMCID: PMC8962155 DOI: 10.1186/s12879-022-07270-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2021] [Accepted: 03/13/2022] [Indexed: 12/18/2022] Open
Abstract
Background Corynebacterium striatum is a gram-positive facultative anaerobe found in the environment and human flora that has historically been considered a contaminant. More recently, Corynebacterium striatum has been implicated in human infections, including respiratory infections, endocarditis, and bone and joint infections, particularly those involving hardware or implanted devices. Case presentation A 65-year-old man presented for washout of his left total knee arthroplasty following a revision 20 days prior. The patient underwent debridement of his left total knee and revision of the left total femur arthroplasty. Daptomycin was initiated empirically due to a previous rash from vancomycin. Operative tissue cultures grew Staphylococcus haemolyticus, Staphylococcus epidermidis and Corynebacterium striatum. Given concern for daptomycin resistance and the reliability of vancomycin susceptibility, daptomycin was discontinued and vancomycin initiated following a graded challenge. Within a few days, the patient developed a diffuse, blanching, erythematous, maculopapular rash and daptomycin was restarted. Over the next 72 h, his rash progressed and he met criteria for drug reaction with eosinophilia and systemic symptoms (DRESS) syndrome. Daptomycin was stopped and oral linezolid initiated; rash improved. C. striatum returned with susceptibility to gentamicin, linezolid, vancomycin and daptomycin. Due to concern for adverse effects on long-term linezolid, daptomycin was restarted and was tolerated for 20 days, at which point purulent drainage from incision increased. The patient underwent another arthroplasty revision and washout. Operative cultures from this surgery were again positive for C. striatum. Repeat C. striatum susceptibilities revealed resistance to daptomycin but retained susceptibility to linezolid. Daptomycin was again changed to linezolid. He completed six weeks of linezolid followed by linezolid 600 mg daily for suppression and ultimately opted for disarticulation. Conclusions C. striatum has historically been regarded as a contaminant, particularly when grown in tissue culture in the setting of prosthetic joint infection. Based on the available literature and susceptibility patterns, the most appropriate first-line therapy is vancomycin or linezolid. Treatment with daptomycin should be avoided, even when isolates appear susceptible, due to the risk of development of high-level resistance (MIC > 256 µg/mL) and clinical failure.
Collapse
Affiliation(s)
- Amber C Streifel
- Department of Pharmacy, Oregon Health and Science University, 3181 SW Sam Jackson Park Road, Portland, OR, USA.
| | - Cara D Varley
- Department of Medicine, Division of Infectious Diseases, Oregon Health and Science University, 3181 SW Sam Jackson Park Road, Portland, OR, USA.,School of Public Health, Oregon Health and Science University-Portland State University, 3181 SW Sam Jackson Park Road, Portland, OR, USA
| | - YoungYoon Ham
- Department of Pharmacy, Oregon Health and Science University, 3181 SW Sam Jackson Park Road, Portland, OR, USA
| | - Monica K Sikka
- Department of Pharmacy, Oregon Health and Science University, 3181 SW Sam Jackson Park Road, Portland, OR, USA
| | - James S Lewis
- Department of Pharmacy, Oregon Health and Science University, 3181 SW Sam Jackson Park Road, Portland, OR, USA.,Department of Medicine, Division of Infectious Diseases, Oregon Health and Science University, 3181 SW Sam Jackson Park Road, Portland, OR, USA
| |
Collapse
|
45
|
Hong XB, Yu ZL, Fu HB, Cai ZH, Chen J. Daptomycin and linezolid for severe methicillin-resistant Staphylococcus aureus psoas abscess and bacteremia: A case report and review of the literature. World J Clin Cases 2022; 10:2550-2558. [PMID: 35434080 PMCID: PMC8968589 DOI: 10.12998/wjcc.v10.i8.2550] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 11/18/2021] [Accepted: 01/26/2022] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Vancomycin remains a first-line treatment drug as per the treatment guidelines for methicillin-resistant Staphylococcus aureus (MRSA) bacteremia. However, a number of gram-positive cocci have developed resistance to several drugs, including glycopeptides. Therefore, there is an urgent need for effective and innovative antibacterial drugs to treat patients with infections caused by drug-resistant bacteria.
CASE SUMMARY A 24-year-old male was admitted to hospital owing to lumbago, fever, and hematuria. Computed tomography (CT) results showed an abscess in the psoas major muscle of the patient. Repeated abscess drainage and blood culture suggested MRSA, and vancomycin was initiated. However, after day 10, CT scans showed abscesses in the lungs and legs of the patient. Therefore, treatment was switched to daptomycin. Linezolid was also added considering inflammation in the lungs. After 10 d of the dual-drug anti-MRSA treatment, culture of the abscess drainage turned negative for MRSA. On day 28, the patient was discharged without any complications.
CONCLUSION This case indicates that daptomycin combined with linezolid is an effective remedy for bacteremia caused by MRSA with pulmonary complications.
Collapse
Affiliation(s)
- Xiao-Bing Hong
- Department of Pharmacy, The Second Affiliated Hospital of Shantou University of Medical College, Shantou 515041, Guangdong Province, China
| | - Ze-Lin Yu
- Department of Pharmacy, The Second Affiliated Hospital of Shantou University of Medical College, Shantou 515041, Guangdong Province, China
| | - Hong-Bo Fu
- Department of Pharmacy, The Second Affiliated Hospital of Shantou University of Medical College, Shantou 515041, Guangdong Province, China
| | - Ze-Hong Cai
- Department of Pharmacology, Shantou University Medical College, Shantou 515041, Guangdong Province, China
| | - Jie Chen
- Department of Pharmacy, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou 510080, Guangdong Province, China
| |
Collapse
|
46
|
Proteomic Correlates of Enhanced Daptomycin Activity following β-Lactam Preconditioning in Daptomycin-Resistant, Methicillin-Resistant Staphylococcus aureus. Antimicrob Agents Chemother 2022; 66:e0201721. [PMID: 35041502 DOI: 10.1128/aac.02017-21] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Clinical treatment options for daptomycin (DAP)-resistant (DAP-R), methicillin-resistant Staphylococcus aureus (MRSA) infections are relatively limited. Current therapeutic strategies often take advantage of potential synergistic activity of DAP plus β-lactams; however, the mechanisms underlying their combinatorial efficacy are likely complex and remain incompletely understood. We recently showed that in vitro β-lactam passaging can resensitize DAP-R strains to a DAP-susceptible (DAP-S) phenotype. To further investigate the implications of selected β-lactam pretreatments on DAP plus β-lactam combination efficacy, we utilized DAP-R strain D712. We studied six such combinations, featuring β-lactams with a broad range of penicillin-binding protein-targeting profiles (PBP-1 to -4), using DAP-R strain D712. Of note, preconditioning with each β-lactam antibiotic (sequential exposures), followed by DAP exposure, yielded significantly enhanced in vitro activity compared to either DAP treatment alone or simultaneous exposures to both antibiotics. To explore the underpinnings of these outcomes, proteomic analyses were performed, with or without β-lactam preconditioning. Relative proteomic quantitation comparing β-lactam pretreatments (versus untreated controls) identified differential modulation of several well-known metabolic, cellular, and biosynthetic processes, i.e., the autolytic and riboflavin biosynthetic pathways. Moreover, these differential proteomic readouts with β-lactam preconditioning were not PBP target specific. Taken together, these studies suggest that the cellular response to β-lactam preconditioning in DAP-R MRSA leads to distinct and complex changes in the proteome that appear to resensitize such strains to DAP-mediated killing.
Collapse
|
47
|
Slavetinsky CJ, Hauser JN, Gekeler C, Slavetinsky J, Geyer A, Kraus A, Heilingbrunner D, Wagner S, Tesar M, Krismer B, Kuhn S, Ernst CM, Peschel A. Sensitizing Staphylococcus aureus to antibacterial agents by decoding and blocking the lipid flippase MprF. eLife 2022; 11:66376. [PMID: 35044295 PMCID: PMC8806190 DOI: 10.7554/elife.66376] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Accepted: 01/18/2022] [Indexed: 11/13/2022] Open
Abstract
The pandemic of antibiotic resistance represents a major human health threat demanding new antimicrobial strategies. MprF is the synthase and flippase of the phospholipid lysyl-phosphatidylglycerol that increases virulence and resistance of methicillin-resistant Staphylococcus aureus (MRSA) and other pathogens to cationic host defense peptides and antibiotics. With the aim to design MprF inhibitors that could sensitize MRSA to antimicrobial agents and support the clearance of staphylococcal infections with minimal selection pressure, we developed MprF-targeting monoclonal antibodies, which bound and blocked the MprF flippase subunit. Antibody M-C7.1 targeted a specific loop in the flippase domain that proved to be exposed at both sides of the bacterial membrane, thereby enhancing the mechanistic understanding of bacterial lipid translocation. M-C7.1 rendered MRSA susceptible to host antimicrobial peptides and antibiotics such as daptomycin, and it impaired MRSA survival in human phagocytes. Thus, MprF inhibitors are recommended for new anti-virulence approaches against MRSA and other bacterial pathogens.
Collapse
Affiliation(s)
| | | | - Cordula Gekeler
- Department of Infection Biology, Eberhard Karls University Tübingen
| | | | - André Geyer
- Department of Infection Biology, Eberhard Karls University Tübingen
| | | | | | - Samuel Wagner
- Cluster of Excellence 'Controlling Microbes to Fight Infections', University of Tübingen
| | | | - Bernhard Krismer
- Department of Infection Biology, Eberhard Karls University Tübingen
| | - Sebastian Kuhn
- Department of Infection Biology, Eberhard Karls University Tübingen
| | - Christoph M Ernst
- Department of Molecular Biology and Center for Computational and Integrative Biology, Broad Institute
| | - Andreas Peschel
- Department of Infection Biology, Eberhard Karls University Tübingen
| |
Collapse
|
48
|
Phenotypic and genetic changes associated with the seesaw effect in MRSA strain N315 in a bioreactor model. J Glob Antimicrob Resist 2022; 28:249-253. [PMID: 35085792 PMCID: PMC9576143 DOI: 10.1016/j.jgar.2022.01.013] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Revised: 01/17/2022] [Accepted: 01/19/2022] [Indexed: 11/20/2022] Open
Abstract
Objectives: Over the past decade, daptomycin treatment of methicillin-resistant Staphylococcus aureus (MRSA) infections has led to the emergence of daptomycin nonsusceptible (DAP-NS) MRSA strains and a subsequent interest in combinatorial antibiotic therapies. We investigated the phenotypic and genetic changes associated with the seesaw effect, which describes the correlation between daptomycin resistance and increased β-lactam susceptibility in DAP-NS MRSA and the reverse phenomenon of DAP-NS strains acquiring renewed susceptibility to daptomycin after β-lactam exposure. Methods: A continuous bioreactor model was used to study the effects of incremental doses of daptomycin followed by oxacillin on MRSA strain N315. Minimum inhibitory concentrations for daptomycin and oxacillin were determined for the bioreactor-derived samples. Transmission electron microscopy and cytochrome C binding assays were used to measure cell wall thickness and cell membrane charge, respectively, in the bioreactor-derived samples. Whole-genome sequencing was used to identify mutations associated with the seesaw effect. Results: Although daptomycin resistance conferred enhanced susceptibility to oxacillin, oxacillin treatment of DAP-NS strains was accompanied by a lowered minimum inhibitory concentration for daptomycin. Additionally, there was a reduction in relative positive cell surface charge and cell wall thickness. However, the mutations acquired in our DAP-NS populations were not accompanied by additional genomic changes after treatment with oxacillin, implicating alternative mechanisms for the seesaw effect. Conclusion: In this study, we successfully produced and characterized the seesaw effect in MRSA strain N315 in a unique bioreactor model.
Collapse
|
49
|
Wu S, Zhang J, Peng Q, Liu Y, Lei L, Zhang H. The Role of Staphylococcus aureus YycFG in Gene Regulation, Biofilm Organization and Drug Resistance. Antibiotics (Basel) 2021; 10:antibiotics10121555. [PMID: 34943766 PMCID: PMC8698359 DOI: 10.3390/antibiotics10121555] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Revised: 12/09/2021] [Accepted: 12/16/2021] [Indexed: 02/05/2023] Open
Abstract
Antibiotic resistance is a serious global health concern that may have significant social and financial consequences. Methicillin-resistant Staphylococcus aureus (MRSA) infection is responsible for substantial morbidity and leads to the death of 21.8% of infected patients annually. A lack of novel antibiotics has prompted the exploration of therapies targeting bacterial virulence mechanisms. The two-component signal transduction system (TCS) enables microbial cells to regulate gene expression and the subsequent metabolic processes that occur due to environmental changes. The YycFG TCS in S. aureus is essential for bacterial viability, the regulation of cell membrane metabolism, cell wall synthesis and biofilm formation. However, the role of YycFG-associated biofilm organization in S. aureus antimicrobial drug resistance and gene regulation has not been discussed in detail. We reviewed the main molecules involved in YycFG-associated cell wall biosynthesis, biofilm development and polysaccharide intercellular adhesin (PIA) accumulation. Two YycFG-associated regulatory mechanisms, accessory gene regulator (agr) and staphylococcal accessory regulator (SarA), were also discussed. We highlighted the importance of biofilm formation in the development of antimicrobial drug resistance in S. aureus infections. Data revealed that inhibition of the YycFG pathway reduced PIA production, biofilm formation and bacterial pathogenicity, which provides a potential target for the management of MRSA-induced infections.
Collapse
Affiliation(s)
- Shizhou Wu
- Department of Orthopedics, West China Hospital, Sichuan University, Chengdu 610041, China; (S.W.); (J.Z.); (Q.P.)
| | - Junqi Zhang
- Department of Orthopedics, West China Hospital, Sichuan University, Chengdu 610041, China; (S.W.); (J.Z.); (Q.P.)
| | - Qi Peng
- Department of Orthopedics, West China Hospital, Sichuan University, Chengdu 610041, China; (S.W.); (J.Z.); (Q.P.)
| | - Yunjie Liu
- West China School of Public Health, Sichuan University, Chengdu 610041, China;
| | - Lei Lei
- West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
- Correspondence: (L.L.); (H.Z.)
| | - Hui Zhang
- Department of Orthopedics, West China Hospital, Sichuan University, Chengdu 610041, China; (S.W.); (J.Z.); (Q.P.)
- Correspondence: (L.L.); (H.Z.)
| |
Collapse
|
50
|
Pleiotropic Effects of Statins: New Therapeutic Approaches to Chronic, Recurrent Infection by Staphylococcus aureus. Pharmaceutics 2021; 13:pharmaceutics13122047. [PMID: 34959329 PMCID: PMC8706520 DOI: 10.3390/pharmaceutics13122047] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2021] [Revised: 11/25/2021] [Accepted: 11/28/2021] [Indexed: 01/01/2023] Open
Abstract
An emergent approach to bacterial infection is the use of host rather than bacterial-directed strategies. This approach has the potential to improve efficacy in especially challenging infection settings, including chronic, recurrent infection due to intracellular pathogens. For nearly two decades, the pleiotropic effects of statin drugs have been examined for therapeutic usefulness beyond the treatment of hypercholesterolemia. Interest originated after retrospective studies reported decreases in the risk of death due to bacteremia or sepsis for those on a statin regimen. Although subsequent clinical trials have yielded mixed results and earlier findings have been questioned for biased study design, in vitro and in vivo studies have provided clear evidence of protective mechanisms that include immunomodulatory effects and the inhibition of host cell invasion. Ultimately, the benefits of statins in an infection setting appear to require attention to the underlying host response and to the timing of the dosage. From this examination of statin efficacy, additional novel host-directed strategies may produce adjunctive therapeutic approaches for the treatment of infection where traditional antimicrobial therapy continues to yield poor outcomes. This review focuses on the opportunistic pathogen, Staphylococcus aureus, as a proof of principle in examining the promise and limitations of statins in recalcitrant infection.
Collapse
|