1
|
Herzberg C, van Hasselt JGC. Pharmacodynamics of interspecies interactions in polymicrobial infections. NPJ Biofilms Microbiomes 2025; 11:20. [PMID: 39837846 PMCID: PMC11751299 DOI: 10.1038/s41522-024-00621-6] [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: 11/03/2023] [Accepted: 11/25/2024] [Indexed: 01/23/2025] Open
Abstract
The pharmacodynamic response of bacterial pathogens to antibiotics can be influenced by interactions with other bacterial species in polymicrobial infections (PMIs). Understanding the complex eco-evolutionary dynamics of PMIs and their impact on antimicrobial treatment response represents a step towards developing improved treatment strategies for PMIs. Here, we investigated how interspecies interactions in a multi-species bacterial community affect the pharmacodynamic response to antimicrobial treatment. To this end, we developed an in silico model which combined agent-based modeling with ordinary differential equations. Our analyses suggest that both interspecies interactions, modifying either drug sensitivity or bacterial growth rate, and drug-specific pharmacological properties drive the bacterial pharmacodynamic response. Furthermore, lifestyle of the bacterial population and the range of interactions can influence the impact of species interactions. In conclusion, this study provides a foundation for the design of antimicrobial treatment strategies for PMIs which leverage the effects of interspecies interactions.
Collapse
Affiliation(s)
- C Herzberg
- Leiden Academic Centre for Drug Research, Leiden University, Leiden, The Netherlands
| | - J G C van Hasselt
- Leiden Academic Centre for Drug Research, Leiden University, Leiden, The Netherlands.
| |
Collapse
|
2
|
Zhang Q, Zhen M, Wang X, Zhao F, Dong Y, Wang X, Gao S, Wang J, Shi W, Zhang Y. Antibiotic exposure enriches streptococci carrying resistance genes in periodontitis plaque biofilms. PeerJ 2025; 13:e18835. [PMID: 39850835 PMCID: PMC11756365 DOI: 10.7717/peerj.18835] [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: 10/09/2024] [Accepted: 12/18/2024] [Indexed: 01/25/2025] Open
Abstract
Background Periodontitis is not always satisfactorily treated with conventional scaling and root planing, and adjunctive use of antibiotics is required in clinical practice. Therefore, it is important for clinicians to understand the diversity and the antibiotic resistance of subgingival microbiota when exposed to different antibiotics. Materials and Methods In this study, subgingival plaques were collected from 10 periodontitis patients and 11 periodontally healthy volunteers, and their microbiota response to selective pressure of four antibiotics (amoxicillin, metronidazole, clindamycin, and tetracycline) were evaluated through 16S rRNA gene amplicon and metagenomic sequencing analysis. Additionally, sensitive and resistant strains were isolated and cultured in vitro for resistance evaluation. Results Cultivation of subgingival microbiota revealed the oral microbiota from periodontitis patients were more resistant to antibiotics than that of healthy. Significant differences were also observed for the microbial community between with and without antibiotics (especially amoxicillin and tetracycline) treated in periodontitis group. Conclusion Overall, after the two antibiotics (amoxicillin and tetracycline) exposed, the oral subgingival microbiota in periodontitis patients exhibited different diversity and composition. Streptococcus may account for oral biofilm-specific antibiotic resistance in periodontitis. This provides information for personalized treatment of periodontitis.
Collapse
Affiliation(s)
- Qian Zhang
- Central Laboratory, Peking University School and Hospital of Stomatology & National Center for Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, Beijing, China
| | - Min Zhen
- Department of Periodontology, Peking University School and Hospital of Stomatology & National Center for Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, Beijing, China
| | - Xiaochen Wang
- CAS Key Laboratory of Animal Ecology and Conservation Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - FengXiang Zhao
- College of Food Science & Nutritional Engineering, China Agricultural University, Beijing, China
| | - Yang Dong
- State Key Laboratory of Animal Biotech Breeding, College of Biological Sciences, China Agricultural University, Beijing, China
| | - Xiaoya Wang
- College of Food Science & Nutritional Engineering, China Agricultural University, Beijing, China
| | - Shengtao Gao
- College of Food Science & Nutritional Engineering, China Agricultural University, Beijing, China
| | - Jinfeng Wang
- College of Food Science & Nutritional Engineering, China Agricultural University, Beijing, China
| | - Wenyu Shi
- State Key Laboratory of Animal Biotech Breeding, College of Biological Sciences, China Agricultural University, Beijing, China
| | - Yifei Zhang
- Department of Dental Materials, Peking University School and Hospital of Stomatology & National Center for Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, Beijing, China
| |
Collapse
|
3
|
Wardell SJT, Yung DBY, Gupta A, Bostina M, Overhage J, Hancock REW, Pletzer D. DJK-5, an anti-biofilm peptide, increases Staphylococcus aureus sensitivity to colistin killing in co-biofilms with Pseudomonas aeruginosa. NPJ Biofilms Microbiomes 2025; 11:8. [PMID: 39779734 PMCID: PMC11711674 DOI: 10.1038/s41522-024-00637-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Accepted: 12/11/2024] [Indexed: 01/30/2025] Open
Abstract
Chronic infections represent a significant global health and economic challenge. Biofilms, which are bacterial communities encased in an extracellular polysaccharide matrix, contribute to approximately 80% of these infections. In particular, pathogens such as Pseudomonas aeruginosa and Staphylococcus aureus are frequently co-isolated from the sputum of patients with cystic fibrosis and are commonly found in chronic wound infections. Within biofilms, bacteria demonstrate a remarkable increase in resistance and tolerance to antimicrobial treatment. We investigated the efficacy of combining the last-line antibiotic colistin with a membrane- and stringent stress response-targeting anti-biofilm peptide DJK-5 against co-biofilms comprised of multidrug-resistant P. aeruginosa and methicillin-resistant S. aureus (MRSA). Colistin lacks canonical activity against S. aureus. However, our study revealed that under co-biofilm conditions, the antibiofilm peptide DJK-5 synergized with colistin against S. aureus. Similar enhancement was observed when daptomycin, a cyclic lipopeptide against Gram-positive bacteria, was combined with DJK-5, resulting in increased activity against P. aeruginosa. The combinatorial treatment induced morphological changes in both P. aeruginosa and S. aureus cell shape and size within co-biofilms. Importantly, our findings also demonstrate synergistic activity against both P. aeruginosa and S. aureus in a murine subcutaneous biofilm-like abscess model. In conclusion, combinatorial treatments with colistin or daptomycin and the anti-biofilm peptide DJK-5 show significant potential for targeting co-biofilm infections. These findings offer promising avenues for developing new therapeutic approaches to combat complex chronic infections.
Collapse
Affiliation(s)
- Samuel J T Wardell
- Department of Microbiology and Immunology, University of Otago, Dunedin, New Zealand
- Maurice Wilkins Centre for Molecular Biodiscovery, The University of Auckland, Auckland, New Zealand
| | - Deborah B Y Yung
- Department of Microbiology and Immunology, University of Otago, Dunedin, New Zealand
- Maurice Wilkins Centre for Molecular Biodiscovery, The University of Auckland, Auckland, New Zealand
| | - Anupriya Gupta
- Department of Microbiology and Immunology, University of Otago, Dunedin, New Zealand
| | - Mihnea Bostina
- Department of Microbiology and Immunology, University of Otago, Dunedin, New Zealand
- Maurice Wilkins Centre for Molecular Biodiscovery, The University of Auckland, Auckland, New Zealand
| | - Joerg Overhage
- Department of Health Sciences, Carleton University, Ottawa, ON, Canada
| | - Robert E W Hancock
- Centre for Microbial Diseases and Immunity Research, Department of Microbiology and Immunology, University of British Columbia, Vancouver, BC, Canada
| | - Daniel Pletzer
- Department of Microbiology and Immunology, University of Otago, Dunedin, New Zealand.
- Maurice Wilkins Centre for Molecular Biodiscovery, The University of Auckland, Auckland, New Zealand.
| |
Collapse
|
4
|
Nazeer RR, Askenasy I, Swain JEV, Welch M. Contribution of the infection ecosystem and biogeography to antibiotic failure in vivo. NPJ ANTIMICROBIALS AND RESISTANCE 2024; 2:45. [PMID: 39649078 PMCID: PMC11618093 DOI: 10.1038/s44259-024-00063-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Accepted: 11/11/2024] [Indexed: 12/10/2024]
Abstract
The acquisition of antibiotic resistance in bacteria, though a deeply concerning international issue, is reasonably well-understood at a mechanistic level. Less well-understood is why bacteria that are sensitive in vitro to well-established and widely-used antibiotics sometimes fail to respond to these agents in vivo. This is a particularly common problem in chronic, polymicrobial infection scenarios. Here, we discuss this in vitro-in vivo disconnect from the perspective of the bacterium, focusing in particular on how infection micro/macro-environment, biogeography, and the presence of co-habiting species affect the response to antibiotics. Using selected exemplars, we also consider interventions that might improve treatment outcomes, as well as ecologically 'eubiotic' approaches that have less of an impact on the patient's commensal microflora. In our view, the accrued data strongly suggest that we need a more comprehensive understanding of the in situ microbiology at infection sites.
Collapse
Affiliation(s)
| | - Isabel Askenasy
- Department of Biochemistry, University of Cambridge, Cambridge, UK
| | | | - Martin Welch
- Department of Biochemistry, University of Cambridge, Cambridge, UK
| |
Collapse
|
5
|
Herzberg C, van Meegen EN, van Hasselt JGC. Interplay of virulence factors shapes ecology and treatment outcomes in polymicrobial infections. Math Biosci 2024; 377:109293. [PMID: 39245301 DOI: 10.1016/j.mbs.2024.109293] [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: 05/07/2024] [Revised: 08/11/2024] [Accepted: 09/04/2024] [Indexed: 09/10/2024]
Abstract
Polymicrobial infections, caused by a community of multiple micro-organisms, are often associated with increased infection severity and poorer patient outcomes. The design of improved antimicrobial treatment strategies for PMIs can be supported by an understanding of their ecological and evolutionary dynamics. Bacterial species present in polymicrobial infections can produce virulence factors to inhibit host immune responses, such as neutrophil recruitment and phagocytosis. The presence of virulence factors can indirectly affect other bacterial species acting as a type of host-mediated interspecies interaction. The aim of this study was to assess how bacterial virulence factors targeting neutrophil function influence ecology and treatment outcomes of PMIs. An agent-based model was constructed which describes a dual-species bacterial population in the presence of neutrophils and a bacteriostatic drug. Our analysis has revealed unforeseen dynamics of the interplay of multiple virulence factors acting as interspecies interaction. We found that the distribution of two phagocytosis-inhibiting virulence factors amongst species can impact whether they have a mutually protective effect for both species. The addition of a virulence factor inhibiting neutrophil recruitment was found to reduce the protective effect of phagocytosis-inhibiting virulence factors. Furthermore we demonstrate the importance of virulence strength of a species relative to other virulent species to determine the fate of a species. We conclude that virulence factors are an important driver of population dynamics in polymicrobial infections, and may be a relevant therapeutic target for treatment of polymicrobial infections.
Collapse
Affiliation(s)
- C Herzberg
- Leiden Academic Centre for Drug Research, Leiden University, Einsteinweg 55, 2333CC Leiden, The Netherlands
| | - E N van Meegen
- Leiden Academic Centre for Drug Research, Leiden University, Einsteinweg 55, 2333CC Leiden, The Netherlands
| | - J G C van Hasselt
- Leiden Academic Centre for Drug Research, Leiden University, Einsteinweg 55, 2333CC Leiden, The Netherlands.
| |
Collapse
|
6
|
Shah R, Narh JK, Urlaub M, Jankiewicz O, Johnson C, Livingston B, Dahl JU. Pseudomonas aeruginosa kills Staphylococcus aureus in a polyphosphate-dependent manner. mSphere 2024; 9:e0068624. [PMID: 39365057 PMCID: PMC11520310 DOI: 10.1128/msphere.00686-24] [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: 08/22/2024] [Accepted: 09/16/2024] [Indexed: 10/05/2024] Open
Abstract
Due to their frequent coexistence in many polymicrobial infections, including in patients with cystic fibrosis or burn/chronic wounds, many studies have investigated the mechanistic details of the interaction between the opportunistic pathogens Pseudomonas aeruginosa and Staphylococcus aureus. P. aeruginosa rapidly outcompetes S. aureus under in vitro cocultivation conditions, which is mediated by several of P. aeruginosa's virulence factors. Here, we report that polyphosphate (polyP), an efficient stress defense system and virulence factor in P. aeruginosa, plays a role in the pathogen's ability to inhibit and kill S. aureus in a contact-independent manner. We show that P. aeruginosa cells characterized by low polyP levels are less detrimental to S. aureus growth and survival while the Gram-positive pathogen is significantly more compromised by the presence of P. aeruginosa cells that produce high levels of polyP. The polyP-dependent phenotype of P. aeruginosa-mediated killing of S. aureus could at least in part be direct, as polyP was detected in the spent media and causes significant damage to the S. aureus cell envelope. However, more likely is that polyP's effects are indirect through modulating the production of one of P. aeruginosa's virulence factors, pyocyanin. We show that pyocyanin production in P. aeruginosa occurs polyP-dependently and harms S. aureus through membrane damage and potentially the generation of reactive oxygen species, resulting in the increased expression of antioxidant enzymes. In summary, our study adds a new component to the list of biomolecules that the Gram-negative pathogen P. aeruginosa generates to compete with S. aureus for resources.IMPORTANCEHow do interactions between microorganisms shape the course of polymicrobial infections? Previous studies have provided evidence that the two opportunistic pathogens Pseudomonas aeruginosa and Staphylococcus aureus generate molecules that modulate their interaction with potentially significant impact on disease outcomes. Our study identified the biopolymer polyphosphate (polyP) as a new effector molecule that impacts P. aeruginosa's interaction with S. aureus. We show that P. aeruginosa kills S. aureus in a polyP-dependent manner, which occurs primarily through the polyP-dependent production of the P. aeruginosa virulence factor pyocyanin. Our findings add a new role for polyP to an already extensive list of functions. A more in-depth understanding of how polyP influences interspecies interactions is critical, as targeting polyP synthesis in bacteria such as P. aeruginosa may have a significant impact on other microorganisms and potentially result in dynamic changes in the microbial composition.
Collapse
Affiliation(s)
- Ritika Shah
- School of Biological Sciences, Illinois State University, Microbiology, Normal, Illinois, USA
| | - Julius Kwesi Narh
- School of Biological Sciences, Illinois State University, Microbiology, Normal, Illinois, USA
| | - Magdalena Urlaub
- School of Biological Sciences, Illinois State University, Microbiology, Normal, Illinois, USA
| | - Olivia Jankiewicz
- School of Biological Sciences, Illinois State University, Microbiology, Normal, Illinois, USA
| | - Colton Johnson
- School of Biological Sciences, Illinois State University, Microbiology, Normal, Illinois, USA
| | - Barry Livingston
- School of Biological Sciences, Illinois State University, Microbiology, Normal, Illinois, USA
| | - Jan-Ulrik Dahl
- School of Biological Sciences, Illinois State University, Microbiology, Normal, Illinois, USA
| |
Collapse
|
7
|
Guo J, Shu X, Yu S, Guo C, Shen G, Chen L, Zhou J, Xiao J, Guo H, Chen Y, Zeng Z, Wang P. Injectable hydrogel microsphere-bomb for MRSA-infected chronic osteomyelitis. J Control Release 2024; 376:337-353. [PMID: 39413850 DOI: 10.1016/j.jconrel.2024.10.021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Revised: 10/08/2024] [Accepted: 10/13/2024] [Indexed: 10/18/2024]
Abstract
Biofilm and bone tissue defect induced by the bacterial infection severely impede chronic osteomyelitis treatment. It is critical to break though the densely and obstinate biofilm so that the target drugs can deliver to the infected bone more effectively. Herein, an acoustically responsive multifunctional hydrogel microsphere-bomb (EMgel) was designed and prepared by microfluidic technology, which could be injected to the focus of bone infection, and blasted into the nidus deeply to destroy the bacterial biofilm matrix barrier under penetrating ultrasound, so the encapsulated natural polyphenolic EGCG and bioactive MoS2 released to repair the damaged bone. The results proved the hydrogel microsphere-bomb exhibited controlling drug release, favorable antibacterial (as high as 99 %), high biofilm resistance, fascinating antioxidation, good cytocompatibility, and osteogenic differentiation. The acoustically responsive microsphere-bomb further proved their fantastic ability to eradicate biofilm and promote bone regeneration in the Methicillin-resistant Staphylococcus aureus (MRSA) infected chronic osteomyelitis model due to the synergy effects of EGCG and bioactive MoS2. Especially, immunohistochemical staining showed lower inflammatory reaction and higher expression of OCN in EMgel group treated with ultrasound wave. This study presents a new design of hydrogel microsphere-based intelligence drug delivery for osteomyelitis treatment, which exhibit great promising potential for dealing with chronic orthopedic infections, drug delivery system and tissue engineering.
Collapse
Affiliation(s)
- Jiayi Guo
- Department of Ultrasound, The Third Affiliated Hospital, Southern Medical University, Guangzhou 510630, China; Institute of Biological and Medical Engineering, Guangdong Academy of Sciences, Guangzhou 510316, China
| | - Xian Shu
- Department of Ultrasound, The Third Affiliated Hospital, Southern Medical University, Guangzhou 510630, China
| | - Shan Yu
- Institute of Biological and Medical Engineering, Guangdong Academy of Sciences, Guangzhou 510316, China
| | - Cuiping Guo
- Institute of Biological and Medical Engineering, Guangdong Academy of Sciences, Guangzhou 510316, China
| | - Guangxin Shen
- Foshan Clinical Medical School of Guangzhou University of Chinese Medicine, Guangdong Province, Foshan 528031, China
| | - Longsheng Chen
- Institute of Biological and Medical Engineering, Guangdong Academy of Sciences, Guangzhou 510316, China
| | - Jiayi Zhou
- Department of Ultrasound, The Third Affiliated Hospital, Southern Medical University, Guangzhou 510630, China
| | - Jiangwei Xiao
- Institute of Biological and Medical Engineering, Guangdong Academy of Sciences, Guangzhou 510316, China
| | - Huilong Guo
- Institute of Biological and Medical Engineering, Guangdong Academy of Sciences, Guangzhou 510316, China
| | - Yi Chen
- Department of Ultrasound, The Third Affiliated Hospital, Southern Medical University, Guangzhou 510630, China
| | - Zhiwen Zeng
- Institute of Biological and Medical Engineering, Guangdong Academy of Sciences, Guangzhou 510316, China; Guangdong Key Lab of Medical Electronic Instruments and Polymer Material Products, Guangzhou 510500, China; National Engineering Research Center for Healthcare Devices, Guangzhou 510500, China.
| | - Ping Wang
- Department of Ultrasound, The Third Affiliated Hospital, Southern Medical University, Guangzhou 510630, China.
| |
Collapse
|
8
|
Preijers T, Muller AE, Abdulla A, de Winter BCM, Koch BCP, Sassen SDT. Dose Individualisation of Antimicrobials from a Pharmacometric Standpoint: The Current Landscape. Drugs 2024; 84:1167-1178. [PMID: 39240531 PMCID: PMC11512831 DOI: 10.1007/s40265-024-02084-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/11/2024] [Indexed: 09/07/2024]
Abstract
Successful antimicrobial therapy depends on achieving optimal drug concentrations within individual patients. Inter-patient variability in pharmacokinetics (PK) and differences in pathogen susceptibility (reflected in the minimum inhibitory concentration, [MIC]) necessitate personalised approaches. Dose individualisation strategies aim to address this challenge, improving treatment outcomes and minimising the risk of toxicity and antimicrobial resistance. Therapeutic drug monitoring (TDM), with the application of population pharmacokinetic (popPK) models, enables model-informed precision dosing (MIPD). PopPK models mathematically describe drug behaviour across populations and can be combined with patient-specific TDM data to optimise dosing regimens. The integration of machine learning (ML) techniques promises to further enhance dose individualisation by identifying complex patterns within extensive datasets. Implementing these approaches involves challenges, including rigorous model selection and validation to ensure suitability for target populations. Understanding the relationship between drug exposure and clinical outcomes is crucial, as is striking a balance between model complexity and clinical usability. Additionally, regulatory compliance, outcome measurement, and practical considerations for software implementation will be addressed. Emerging technologies, such as real-time biosensors, hold the potential for revolutionising TDM by enabling continuous monitoring, immediate and frequent dose adjustments, and near patient testing. The ongoing integration of TDM, advanced modelling techniques, and ML within the evolving digital health care landscape offers a potential for enhancing antimicrobial therapy. Careful attention to model development, validation, and ethical considerations of the applied techniques is paramount for successfully optimising antimicrobial treatment for the individual patient.
Collapse
Affiliation(s)
- Tim Preijers
- Department of Hospital Pharmacy, Erasmus MC, University Medical Centre Rotterdam, Rotterdam, The Netherlands
- Rotterdam Clinical Pharmacometrics Group, Erasmus MC, Rotterdam, The Netherlands
| | - Anouk E Muller
- Department of Medical Microbiology and Infectious Diseases, Erasmus University Medical Centre, Rotterdam, The Netherlands
- Department of Medical Microbiology, Haaglanden Medisch Centrum, The Hague, The Netherlands
- Centre for Antimicrobial Treatment Optimization Rotterdam (CATOR), Rotterdam, The Netherlands
| | - Alan Abdulla
- Department of Hospital Pharmacy, Erasmus MC, University Medical Centre Rotterdam, Rotterdam, The Netherlands
- Rotterdam Clinical Pharmacometrics Group, Erasmus MC, Rotterdam, The Netherlands
- Centre for Antimicrobial Treatment Optimization Rotterdam (CATOR), Rotterdam, The Netherlands
| | - Brenda C M de Winter
- Department of Hospital Pharmacy, Erasmus MC, University Medical Centre Rotterdam, Rotterdam, The Netherlands
- Rotterdam Clinical Pharmacometrics Group, Erasmus MC, Rotterdam, The Netherlands
- Centre for Antimicrobial Treatment Optimization Rotterdam (CATOR), Rotterdam, The Netherlands
| | - Birgit C P Koch
- Department of Hospital Pharmacy, Erasmus MC, University Medical Centre Rotterdam, Rotterdam, The Netherlands.
- Rotterdam Clinical Pharmacometrics Group, Erasmus MC, Rotterdam, The Netherlands.
- Centre for Antimicrobial Treatment Optimization Rotterdam (CATOR), Rotterdam, The Netherlands.
| | - Sebastiaan D T Sassen
- Department of Hospital Pharmacy, Erasmus MC, University Medical Centre Rotterdam, Rotterdam, The Netherlands
- Rotterdam Clinical Pharmacometrics Group, Erasmus MC, Rotterdam, The Netherlands
- Centre for Antimicrobial Treatment Optimization Rotterdam (CATOR), Rotterdam, The Netherlands
| |
Collapse
|
9
|
Ma HR, Xu HZ, Kim K, Anderson DJ, You L. Private benefit of β-lactamase dictates selection dynamics of combination antibiotic treatment. Nat Commun 2024; 15:8337. [PMID: 39333122 PMCID: PMC11436977 DOI: 10.1038/s41467-024-52711-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Accepted: 09/19/2024] [Indexed: 09/29/2024] Open
Abstract
β-lactam antibiotics have been prescribed for most bacterial infections since their discovery. However, resistance to β-lactams, mediated by β-lactamase (Bla) enzymes such as extended spectrum β-lactamases (ESBLs), has become widespread. Bla inhibitors can restore the efficacy of β-lactams against resistant bacteria, an approach which preserves existing antibiotics despite declining industry investment. However, the effects of combination treatment on selection for β-lactam resistance are not well understood. Bla production confers both private benefits for resistant cells and public benefits which faster-growing sensitive cells can also exploit. These benefits may be differentially impacted by Bla inhibitors, leading to non-intuitive selection dynamics. In this study, we demonstrate strain-to-strain variation in effective combination doses, with complex growth dynamics in mixed populations. Using modeling, we derive a criterion for the selection outcome of combination treatment, dependent on the burden and effective private benefit of Bla production. We then use engineered strains and natural isolates to show that strong private benefits of Bla are associated with increased selection for resistance. Finally, we demonstrate that this parameter can be coarsely estimated using high-throughput phenotyping of clonal populations. Our analysis shows that quantifying the phenotypic responses of bacteria to combination treatment can facilitate resistance-minimizing optimization of treatment.
Collapse
Affiliation(s)
- Helena R Ma
- Department of Biomedical Engineering, Duke University, Durham, NC, USA
- Center for Quantitative Biodesign, Duke University, Durham, NC, USA
| | - Helen Z Xu
- Department of Biology, Duke University, Durham, NC, USA
- Department of Computer Science, Duke University, Durham, NC, USA
| | - Kyeri Kim
- Department of Biomedical Engineering, Duke University, Durham, NC, USA
- Center for Quantitative Biodesign, Duke University, Durham, NC, USA
| | - Deverick J Anderson
- Division of Infectious Diseases, Department of Medicine, Duke University School of Medicine, Durham, NC, USA
- Duke Center for Antimicrobial Stewardship and Infection Prevention, Duke University School of Medicine, Durham, NC, USA
| | - Lingchong You
- Department of Biomedical Engineering, Duke University, Durham, NC, USA.
- Center for Quantitative Biodesign, Duke University, Durham, NC, USA.
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, NC, USA.
| |
Collapse
|
10
|
Mandell JB, Gish C, Cappellini AJ, Parker DM, Brothers KM, Ma D, Urish KL. Methicillin resistant Staphylococcus aureus mazEF expression promotes infections by influencing cellular growth, antibiotic sensitivity, and formation of biofilms. Sci Rep 2024; 14:21269. [PMID: 39261496 PMCID: PMC11390869 DOI: 10.1038/s41598-024-70829-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2024] [Accepted: 08/21/2024] [Indexed: 09/13/2024] Open
Abstract
Staphylococcus aureus infections are hard to treat due to the emergence of antibiotic resistant strains, as well as their ability to form biofilms. The MazEF toxin-antitoxin system is thought play a role in bacterial biofilm phenotype as well as antibiotic resistance. In S. aureus, the physiologic function of the mazEF gene in the disease transition from acute to chronic infection is not well understood. In methicillin resistant S. aureus (MRSA), loss of mazF expression results in loss of resistance to first generation cephalosporins. mazF::tn displayed sensitivity while the isogenic wild type (WT) remained resistant. mazF::tn displayed significantly increased growth of biofilms on metal implants over 48 h compared to WT and the complemented transposon mutant. mazF::tn biofilms displayed significantly decreased antibiotic tolerance to vancomycin and cefazolin in comparison to WT and complement biofilms. Mice given mazF::tn in a sepsis model displayed less abscess burden and increased survival (100%) when treated with cefazolin compared to WT bacteremia treated with cefazolin (20%). mazF::tn periprosthetic joint infections displayed increased biofilm burden at acute time points and decreased biofilm burden at chronic time points. Our data suggests MazEF in MRSA is responsible for controlling growth of biofilms, antibiotic tolerance, and influence chronic infections in vivo.
Collapse
Affiliation(s)
- Jonathan B Mandell
- Arthritis and Arthroplasty Design Laboratory, Department of Orthopaedic Surgery, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Ophthalmology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Charles Gish
- Arthritis and Arthroplasty Design Laboratory, Department of Orthopaedic Surgery, University of Pittsburgh, Pittsburgh, PA, USA
| | - Alex J Cappellini
- Arthritis and Arthroplasty Design Laboratory, Department of Orthopaedic Surgery, University of Pittsburgh, Pittsburgh, PA, USA
| | - Dana M Parker
- Arthritis and Arthroplasty Design Laboratory, Department of Orthopaedic Surgery, University of Pittsburgh, Pittsburgh, PA, USA
| | - Kimberly M Brothers
- Arthritis and Arthroplasty Design Laboratory, Department of Orthopaedic Surgery, University of Pittsburgh, Pittsburgh, PA, USA
| | - Dongzhu Ma
- Arthritis and Arthroplasty Design Laboratory, Department of Orthopaedic Surgery, University of Pittsburgh, Pittsburgh, PA, USA
| | - Kenneth L Urish
- Arthritis and Arthroplasty Design Laboratory, Department of Orthopaedic Surgery, University of Pittsburgh, Pittsburgh, PA, USA.
- The Bone and Joint Center, Magee Womens Hospital, University of Pittsburgh Medical Center, Pittsburgh, PA, USA.
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, USA.
- Clinical and Translational Science Institute, University of Pittsburgh, Pittsburgh, PA, USA.
- Department of Biomedical Engineering, Carnegie Mellon University, Pittsburgh, PA, USA.
| |
Collapse
|
11
|
Haeberle AL, Greenwood-Quaintance KE, Zar S, Johnson S, Patel R, Willett JLE. Genotypic and phenotypic characterization of Enterococcus faecalis isolates from periprosthetic joint infections. Microbiol Spectr 2024; 12:e0056524. [PMID: 38912797 PMCID: PMC11302728 DOI: 10.1128/spectrum.00565-24] [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: 04/02/2024] [Accepted: 05/09/2024] [Indexed: 06/25/2024] Open
Abstract
Over 2.5 million prosthetic joint implantation surgeries occur annually in the United States. Periprosthetic joint infections (PJIs), though occurring in only 1-2% of patients receiving replacement joints, are challenging to diagnose and treat and are associated with significant morbidity. The Gram-positive bacterium Enterococcus faecalis, which can be highly antibiotic-resistant and is a robust biofilm producer on indwelling medical devices, accounts for 2-11% of PJIs. E. faecalis PJIs are understudied compared to those caused by other pathogens, such as Staphylococcus aureus. This motivates the need to generate a comprehensive understanding of E. faecalis PJIs to guide future treatments for these infections. To address this, we describe a panel of E. faecalis strains isolated from the surface of prosthetic joints in a cohort of individuals treated at the Mayo Clinic in Rochester, MN. Here, we present the first complete genome assemblage of E. faecalis PJI isolates. Comparative genomics shows differences in genome size, virulence factors, antimicrobial resistance genes, plasmids, and prophages, underscoring the genetic diversity of these strains. These isolates have strain-specific differences in in vitro biofilm biomass, biofilm burden, and biofilm morphology. We measured robust changes in biofilm architecture and aggregation for all isolates when grown in simulated synovial fluid (SSF). Finally, we evaluated the antibiotic efficacy of these isolates and found strain-specific changes across all strains when grown in SSF. Results of this study highlight the existence of genetic and phenotypic heterogeneity among E. faecalis PJI isolates which will provide valuable insight and resources for future E. faecalis PJI research. IMPORTANCE Periprosthetic joint infections (PJIs) affect ~1-2% of those who undergo joint replacement surgery. Enterococcus faecalis is a Gram-positive opportunistic pathogen that causes ~10% of PJIs in the United States each year, but our understanding of how and why E. faecalis causes PJIs is limited. E. faecalis infections are typically biofilm-associated and can be difficult to clear with antibiotic therapy. Here, we provide complete genomes for four E. faecalis PJI isolates from the Mayo Clinic. These isolates have strain-specific differences in biofilm formation, aggregation, and antibiotic susceptibility in simulated synovial fluid. These results provide important insight into the genomic and phenotypic features of E. faecalis isolates from PJI.
Collapse
Affiliation(s)
- Amanda L. Haeberle
- Department of Microbiology & Immunology, University of Minnesota Medical School, Minneapolis, Minnesota, USA
| | - Kerryl E. Greenwood-Quaintance
- Division of Clinical Microbiology, Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota, USA
| | - Sarah Zar
- Department of Microbiology & Immunology, University of Minnesota Medical School, Minneapolis, Minnesota, USA
| | - Stephen Johnson
- Department of Health Sciences Research, Mayo Clinic, Rochester, Minnesota, USA
| | - Robin Patel
- Division of Clinical Microbiology, Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota, USA
- Division of Public Health, Infectious Diseases, and Occupational Medicine, Department of Medicine, Mayo Clinic, Rochester, Minnesota, USA
| | - Julia L. E. Willett
- Department of Microbiology & Immunology, University of Minnesota Medical School, Minneapolis, Minnesota, USA
| |
Collapse
|
12
|
Gupta S, Luxami V, Paul K. Bacterial cell death to overcome drug resistance with multitargeting bis-naphthalimides as potent antibacterial agents against Enterococcus faecalis. J Mater Chem B 2024; 12:5645-5660. [PMID: 38747306 DOI: 10.1039/d3tb02804f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/13/2024]
Abstract
The increasing frequency of drug-resistant pathogens poses serious health issues to humans around the globe, leading to the development of new antibacterial agents to conquer drug resistance and bacterial infections. In view of this, we have synthesized a series of bis-naphthalimides to respond to awful drug resistance. Bioactivity assay and structure-activity relationship disclosed that compounds 5d and 5o exhibit potent antibacterial activity against E. faecalis, outperforming the marketed antibiotics. These drug candidates not only inhibit the biofilm formation of E. faecalis but also display rapid bactericidal properties, thus delaying the development of drug resistance within 20 passages. To explore the mechanism of antibacterial activity against E. faecalis, biofunctional examination was carried out which unveiled that 5d and 5o effectively disrupt bacterial cell membranes, causing the leakage of cytoplasmic contents and metabolic activity loss. Concurrently, 5d and 5o effectively intercalate with DNA to block DNA replication, causing the build-up of excessive reactive oxygen species and inhibiting the glutathione activity, ultimately leading to oxidative damage of E. faecalis and cell death. In addition, these compounds readily bind with HSA with a high binding constant, indicating that these drug candidates could be easily delivered to the target site. The above finding manifested that these newly synthesized bis-naphthalimides with multitargeting antibacterial properties offer a new prospect to overcome drug resistance.
Collapse
Affiliation(s)
- Saurabh Gupta
- Department of Chemistry and Biochemistry, Thapar Institute of Engineering and Technology, Patiala 147001, India.
| | - Vijay Luxami
- Department of Chemistry and Biochemistry, Thapar Institute of Engineering and Technology, Patiala 147001, India.
| | - Kamaldeep Paul
- Department of Chemistry and Biochemistry, Thapar Institute of Engineering and Technology, Patiala 147001, India.
| |
Collapse
|
13
|
Karlo J, Vijay A, Phaneeswar MS, Singh SP. Sensing the Bactericidal and Bacteriostatic Antimicrobial Mode of Action Using Raman Deuterium Stable Isotope Probing (DSIP) in Escherichia coli. ACS OMEGA 2024; 9:23753-23760. [PMID: 38854576 PMCID: PMC11154948 DOI: 10.1021/acsomega.4c01666] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Revised: 05/10/2024] [Accepted: 05/15/2024] [Indexed: 06/11/2024]
Abstract
The mode of action of antibiotics can be broadly classified as bacteriostatic and bactericidal. The bacteriostatic mode leads to the arrested growth of the cells, while the bacteriocidal mode causes cell death. In this work, we report the applicability of deuterium stable isotope probing (DSIP) in combination with Raman spectroscopy (Raman DSIP) for discriminating the mode of action of antibiotics at the community level. Escherichia coli, a well-known model microbe, was used as an organism for the study. We optimized the concentration of deuterium oxide required for metabolic activity monitoring without compromising the microbial growth. Our findings suggest that changes in the intensity of the C-D band in the high-wavenumber region could serve as a quantifiable marker for determining the antibiotic mode of action. This can be used for early identification of the antibiotic's mode of action. Our results explore the new perspective that supports the utility of deuterium-based vibrational tags in the field of clinical spectroscopy. Understanding the antibiotic's mode of action on bacterial cells in a short and objective manner can significantly enhance the clinical management abilities of infectious diseases and may also help in personalized antimicrobial therapy.
Collapse
Affiliation(s)
- Jiro Karlo
- Department of Biosciences
and Bioengineering, Indian Institute of
Technology Dharwad, Dharwad, Karnataka 580011, India
| | - Arunsree Vijay
- Department of Biosciences
and Bioengineering, Indian Institute of
Technology Dharwad, Dharwad, Karnataka 580011, India
| | - Mahamkali Sri Phaneeswar
- Department of Biosciences
and Bioengineering, Indian Institute of
Technology Dharwad, Dharwad, Karnataka 580011, India
| | - Surya Pratap Singh
- Department of Biosciences
and Bioengineering, Indian Institute of
Technology Dharwad, Dharwad, Karnataka 580011, India
| |
Collapse
|
14
|
Juber NF, Abdulle A, Ahmad A, AlAnouti F, Loney T, Idaghdour Y, Valles Y, Ali R. Associations between Polycystic Ovary Syndrome (PCOS) and Antibiotic Use: Results from the UAEHFS. Antibiotics (Basel) 2024; 13:397. [PMID: 38786126 PMCID: PMC11117232 DOI: 10.3390/antibiotics13050397] [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: 03/30/2024] [Revised: 04/21/2024] [Accepted: 04/22/2024] [Indexed: 05/25/2024] Open
Abstract
Women with polycystic ovary syndrome (PCOS) have a higher susceptibility to infections compared to those without PCOS. Studies evaluating antibiotic use based on PCOS status are scarce. Therefore, we aimed to (i) assess the associations between self-reported PCOS and antibiotic use, and (ii) whether PCOS treatment and the age at PCOS diagnosis modified the associations above. This cross-sectional analysis used the United Arab Emirates Healthy Future Study (UAEHFS) conducted from February 2016 to March 2023 involving 2063 Emirati women aged 18-62 years. We performed ordinal logistic regressions under unadjusted and demographic-health-characteristic-adjusted models to obtain the odds ratios (ORs) and 95% confidence intervals (CIs) to analyze PCOS and antibiotic use. Subgroup analyses were performed by treatment status and age at diagnosis. We found that women with PCOS were 55% more likely to frequently take a course of antibiotics in the past year (aOR 1.55; 95% CI 1.26-1.90). Similar likelihoods were also found among those being treated for PCOS and those without treatment but with a PCOS diagnosis at ≤25 years. Our study suggests that PCOS was associated with an increased use of antibiotics among Emirati women. Understanding the frequent antibiotic use susceptibility among those with PCOS may improve antibiotic use surveillance and promote antibiotic stewardship in these at-risk individuals.
Collapse
Affiliation(s)
- Nirmin F. Juber
- Public Health Research Center, New York University Abu Dhabi, Abu Dhabi P.O. Box 129188, United Arab Emirates; (A.A.); (A.A.); (Y.I.); (Y.V.); (R.A.)
| | - Abdishakur Abdulle
- Public Health Research Center, New York University Abu Dhabi, Abu Dhabi P.O. Box 129188, United Arab Emirates; (A.A.); (A.A.); (Y.I.); (Y.V.); (R.A.)
| | - Amar Ahmad
- Public Health Research Center, New York University Abu Dhabi, Abu Dhabi P.O. Box 129188, United Arab Emirates; (A.A.); (A.A.); (Y.I.); (Y.V.); (R.A.)
| | - Fatme AlAnouti
- College of Natural and Health Sciences, Zayed University, Abu Dhabi P.O. Box 19282, United Arab Emirates;
| | - Tom Loney
- College of Medicine, Mohammed Bin Rashid University of Medicine and Health Sciences, Dubai Health, Dubai P.O. Box 505055, United Arab Emirates;
| | - Youssef Idaghdour
- Public Health Research Center, New York University Abu Dhabi, Abu Dhabi P.O. Box 129188, United Arab Emirates; (A.A.); (A.A.); (Y.I.); (Y.V.); (R.A.)
| | - Yvonne Valles
- Public Health Research Center, New York University Abu Dhabi, Abu Dhabi P.O. Box 129188, United Arab Emirates; (A.A.); (A.A.); (Y.I.); (Y.V.); (R.A.)
| | - Raghib Ali
- Public Health Research Center, New York University Abu Dhabi, Abu Dhabi P.O. Box 129188, United Arab Emirates; (A.A.); (A.A.); (Y.I.); (Y.V.); (R.A.)
- MRC Epidemiology Unit, University of Cambridge, Cambridge CB2 0SL, UK
| |
Collapse
|
15
|
Zheng EJ, Valeri JA, Andrews IW, Krishnan A, Bandyopadhyay P, Anahtar MN, Herneisen A, Schulte F, Linnehan B, Wong F, Stokes JM, Renner LD, Lourido S, Collins JJ. Discovery of antibiotics that selectively kill metabolically dormant bacteria. Cell Chem Biol 2024; 31:712-728.e9. [PMID: 38029756 PMCID: PMC11031330 DOI: 10.1016/j.chembiol.2023.10.026] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Revised: 08/13/2023] [Accepted: 10/30/2023] [Indexed: 12/01/2023]
Abstract
There is a need to discover and develop non-toxic antibiotics that are effective against metabolically dormant bacteria, which underlie chronic infections and promote antibiotic resistance. Traditional antibiotic discovery has historically favored compounds effective against actively metabolizing cells, a property that is not predictive of efficacy in metabolically inactive contexts. Here, we combine a stationary-phase screening method with deep learning-powered virtual screens and toxicity filtering to discover compounds with lethality against metabolically dormant bacteria and favorable toxicity profiles. The most potent and structurally distinct compound without any obvious mechanistic liability was semapimod, an anti-inflammatory drug effective against stationary-phase E. coli and A. baumannii. Integrating microbiological assays, biochemical measurements, and single-cell microscopy, we show that semapimod selectively disrupts and permeabilizes the bacterial outer membrane by binding lipopolysaccharide. This work illustrates the value of harnessing non-traditional screening methods and deep learning models to identify non-toxic antibacterial compounds that are effective in infection-relevant contexts.
Collapse
Affiliation(s)
- Erica J Zheng
- Program in Chemical Biology, Harvard University, Cambridge, MA 02138, USA; Infectious Disease and Microbiome Program, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02115, USA
| | - Jacqueline A Valeri
- Infectious Disease and Microbiome Program, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Institute for Medical Engineering & Science, Department of Biological Engineering, and Synthetic Biology Center, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02115, USA
| | - Ian W Andrews
- Infectious Disease and Microbiome Program, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Institute for Medical Engineering & Science, Department of Biological Engineering, and Synthetic Biology Center, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Aarti Krishnan
- Infectious Disease and Microbiome Program, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Institute for Medical Engineering & Science, Department of Biological Engineering, and Synthetic Biology Center, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Whitehead Institute for Biomedical Research, Cambridge, MA 02142, USA
| | - Parijat Bandyopadhyay
- Infectious Disease and Microbiome Program, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Institute for Medical Engineering & Science, Department of Biological Engineering, and Synthetic Biology Center, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Melis N Anahtar
- Infectious Disease and Microbiome Program, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Institute for Medical Engineering & Science, Department of Biological Engineering, and Synthetic Biology Center, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02115, USA
| | - Alice Herneisen
- Whitehead Institute for Biomedical Research, Cambridge, MA 02142, USA; Department of Biology, MIT, Cambridge, MA 02139, USA
| | - Fabian Schulte
- Whitehead Institute for Biomedical Research, Cambridge, MA 02142, USA
| | - Brooke Linnehan
- Whitehead Institute for Biomedical Research, Cambridge, MA 02142, USA
| | - Felix Wong
- Infectious Disease and Microbiome Program, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Institute for Medical Engineering & Science, Department of Biological Engineering, and Synthetic Biology Center, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Jonathan M Stokes
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario L8N 3Z5, Canada
| | - Lars D Renner
- Leibniz Institute of Polymer Research and the Max Bergmann Center of Biomaterials, 01062 Dresden, Germany
| | - Sebastian Lourido
- Whitehead Institute for Biomedical Research, Cambridge, MA 02142, USA; Department of Biology, MIT, Cambridge, MA 02139, USA
| | - James J Collins
- Infectious Disease and Microbiome Program, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Institute for Medical Engineering & Science, Department of Biological Engineering, and Synthetic Biology Center, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02115, USA; Harvard-MIT Program in Health Sciences and Technology, Cambridge, MA 02139, USA.
| |
Collapse
|
16
|
Feng Y, Bian J, Yu G, Zhao P, Yue J. Quaternary ammonium-tethered hyperbranched polyurea nanoassembly synergized with antibiotics for enhanced antimicrobial efficacy. Biomater Sci 2024; 12:1185-1196. [PMID: 38226542 DOI: 10.1039/d3bm01519j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2024]
Abstract
The effective transportation of antibiotics to bacteria embedded within a biofilm consisting of a dense matrix of extracellular polymeric substances is still a challenge in the treatment of bacterial biofilm associated infections. Here, we developed an antibiotic nanocarrier constructed from quaternary ammonium-tethered hyperbranched polyureas (HPUs-QA), which showed high loading capacity for a model antibiotic, rifampicin, and high efficacy in the transportation of rifampicin to biofilms. The rifampicin-loaded HPUs-QA nanoassembly (HPUs-Rif/QA) demonstrated a synergistic antimicrobial effect in killing planktonic bacteria and eradicating the corresponding biofilms. Compared to the treatment of bacteria-infected chronic wounds by either HPUs-QA or rifampicin alone, HPUs-Rif/QA showed superior efficacy in promoting wound healing by more effectively inhibiting bacteria colonization. This study highlights the potential of the HPUs-QA nanoassembly in synergistic action with antibiotics for the treatment of biofilm associated infections.
Collapse
Affiliation(s)
- Yanwen Feng
- School of Biomedical Engineering, Shenzhen Campus of Sun Yat-sen University, Shenzhen, Guangdong 518107, P. R. China.
| | - Jiang Bian
- School of Biomedical Engineering, Shenzhen Campus of Sun Yat-sen University, Shenzhen, Guangdong 518107, P. R. China.
| | - Guoyi Yu
- School of Biomedical Engineering, Shenzhen Campus of Sun Yat-sen University, Shenzhen, Guangdong 518107, P. R. China.
| | - Pei Zhao
- Laboratory Animal Center, Shenzhen Campus of Sun Yat-sen University, Shenzhen, Guangdong 518107, P. R. China.
| | - Jun Yue
- School of Biomedical Engineering, Shenzhen Campus of Sun Yat-sen University, Shenzhen, Guangdong 518107, P. R. China.
| |
Collapse
|
17
|
Haeberle A, Greenwood-Quaintance K, Zar S, Johnson S, Patel R, Willett JLE. Genotypic and phenotypic characterization of Enterococcus faecalis isolates from periprosthetic joint infections. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.02.06.579140. [PMID: 38370742 PMCID: PMC10871183 DOI: 10.1101/2024.02.06.579140] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/20/2024]
Abstract
Over 2.5 million prosthetic joint implantation surgeries occur annually in the United States. Periprosthetic joint infections (PJIs), though occurring in only 1-2% of patients receiving replacement joints, are challenging to diagnose and treat and are associated with significant morbidity. The Gram-positive bacterium Enterococcus faecalis, which can be highly antibiotic resistant and is a robust biofilm producer on indwelling medical devices, accounts for 2-11% of PJIs. E. faecalis PJIs are understudied compared to those caused by other pathogens, such as Staphylococcus aureus. This motivates the need to generate a comprehensive understanding of E. faecalis PJIs to guide future treatments for these infections. To address this, we describe a panel of E. faecalis strains isolated from the surface of prosthetic joints in a cohort of individuals treated at Mayo Clinic in Rochester, MN. Here, we present the first complete genome assemblage of E. faecalis PJI isolates. Comparative genomics shows differences in genome size, virulence factors, antimicrobial resistance genes, plasmids, and prophages, underscoring the genetic diversity of these strains. These isolates have strain-specific differences in in vitro biofilm biomass, biofilm burden, and biofilm morphology. We measured robust changes in biofilm architecture and aggregation for all isolates when grown in simulated synovial fluid (SSF). Lastly, we evaluated antibiotic efficacy of these isolates and found strain specific changes across all strains when grown in SSF. Results of this study highlight the existence of genetic and phenotypic heterogeneity among E. faecalis PJI isolates which will provide valuable insight and resources for future E. faecalis PJI research.
Collapse
Affiliation(s)
- Amanda Haeberle
- Department of Microbiology & Immunology, University of Minnesota Medical School, Minneapolis, Minnesota USA
| | - Kerryl Greenwood-Quaintance
- Division of Clinical Microbiology, Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota, USA
| | - Sarah Zar
- Department of Microbiology & Immunology, University of Minnesota Medical School, Minneapolis, Minnesota USA
| | - Stephen Johnson
- Department of Health Sciences Research, Mayo Clinic, Rochester, Minnesota, USA
| | - Robin Patel
- Division of Clinical Microbiology, Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota, USA
- Division of Public Health, Infectious Diseases, and Occupational Medicine, Department of Medicine, Mayo Clinic, Rochester, Minnesota, USA
| | - Julia L. E. Willett
- Department of Microbiology & Immunology, University of Minnesota Medical School, Minneapolis, Minnesota USA
| |
Collapse
|
18
|
Shah R, Jankiewicz O, Johnson C, Livingston B, Dahl JU. Pseudomonas aeruginosa kills Staphylococcus aureus in a polyphosphate-dependent manner. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.12.05.570291. [PMID: 38106195 PMCID: PMC10723280 DOI: 10.1101/2023.12.05.570291] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2023]
Abstract
Due to their frequent coexistence in many polymicrobial infections, including in patients with burn or chronic wounds or cystic fibrosis, recent studies have started to investigate the mechanistic details of the interaction between the opportunistic pathogens Pseudomonas aeruginosa and Staphylococcus aureus. P. aeruginosa rapidly outcompetes S. aureus under in vitro co-cultivation conditions, which is mediated by several of P. aeruginosa's virulence factors. Here, we report that polyphosphate (polyP), an efficient stress defense system and virulence factor in P. aeruginosa, plays a role for the pathogen's ability to inhibit and kill S. aureus in a contact-independent manner. We show that P. aeruginosa cells characterized by low polyP level are less detrimental to S. aureus growth and survival while the gram-positive pathogen is significantly more compromised by the presence of P. aeruginosa cells that produce high level of polyP. We show that the polyP-dependent phenotype could be a direct effect by the biopolymer, as polyP is present in the spent media and causes significant damage to the S. aureus cell envelope. However, more likely is that polyP's effects are indirect through the regulation of one of P. aeruginosa's virulence factors, pyocyanin. We show that pyocyanin production in P. aeruginosa occurs polyP-dependent and harms S. aureus through membrane damage and the generation of reactive oxygen species, resulting in increased expression of antioxidant enzymes. In summary, our study adds a new component to the list of biomolecules that the gram-negative pathogen P. aeruginosa generates to compete with S. aureus for resources.
Collapse
Affiliation(s)
- Ritika Shah
- School of Biological Sciences, Illinois State University, Microbiology, Normal, IL, USA
| | - Olivia Jankiewicz
- School of Biological Sciences, Illinois State University, Microbiology, Normal, IL, USA
| | - Colton Johnson
- School of Biological Sciences, Illinois State University, Microbiology, Normal, IL, USA
| | - Barry Livingston
- School of Biological Sciences, Illinois State University, Microbiology, Normal, IL, USA
| | - Jan-Ulrik Dahl
- School of Biological Sciences, Illinois State University, Microbiology, Normal, IL, USA
| |
Collapse
|
19
|
Ashrafi R, Bruneaux M, Sundberg LR, Hoikkala V, Karvonen A. Multispecies coinfections and presence of antibiotics shape resistance and fitness costs in a pathogenic bacterium. Mol Ecol 2023; 32:4447-4460. [PMID: 37303030 DOI: 10.1111/mec.17040] [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: 04/28/2022] [Accepted: 05/24/2023] [Indexed: 06/13/2023]
Abstract
Increasing antimicrobial resistance (AMR) poses a challenge for treatment of bacterial diseases. In real life, bacterial infections are typically embedded within complex multispecies communities and influenced by the environment, which can shape costs and benefits of AMR. However, knowledge of such interactions and their implications for AMR in vivo is limited. To address this knowledge gap, we investigated fitness-related traits of a pathogenic bacterium (Flavobacterium columnare) in its fish host, capturing the effects of bacterial antibiotic resistance, coinfections between bacterial strains and metazoan parasites (fluke Diplostomum pseudospathaceum) and antibiotic exposure. We quantified real-time replication and virulence of sensitive and resistant bacteria and demonstrate that both bacteria can benefit from coinfection in terms of persistence and replication, depending on the coinfecting partner and antibiotic presence. We also show that antibiotics can benefit resistant bacteria by increasing bacterial replication under coinfection with flukes. These results emphasize the importance of diverse, inter-kingdom coinfection interactions and antibiotic exposure in shaping costs and benefits of AMR, supporting their role as significant contributors to spread and long-term persistence of resistance.
Collapse
Affiliation(s)
- Roghaieh Ashrafi
- Department of Biological and Environmental Science, University of Jyväskylä, Jyväskylä, Finland
| | - Matthieu Bruneaux
- Department of Biological and Environmental Science, University of Jyväskylä, Jyväskylä, Finland
| | - Lotta-Riina Sundberg
- Department of Biological and Environmental Science, University of Jyväskylä, Jyväskylä, Finland
- Nanoscience Center, University of Jyväskylä, Jyväskylä, Finland
| | - Ville Hoikkala
- Department of Biological and Environmental Science, University of Jyväskylä, Jyväskylä, Finland
- Nanoscience Center, University of Jyväskylä, Jyväskylä, Finland
| | - Anssi Karvonen
- Department of Biological and Environmental Science, University of Jyväskylä, Jyväskylä, Finland
| |
Collapse
|
20
|
Alexander HK. Quantifying stochastic establishment of mutants in microbial adaptation. MICROBIOLOGY (READING, ENGLAND) 2023; 169:001365. [PMID: 37561015 PMCID: PMC10482372 DOI: 10.1099/mic.0.001365] [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: 03/21/2023] [Accepted: 07/10/2023] [Indexed: 08/11/2023]
Abstract
Studies of microbial evolution, especially in applied contexts, have focused on the role of selection in shaping predictable, adaptive responses to the environment. However, chance events - the appearance of novel genetic variants and their establishment, i.e. outgrowth from a single cell to a sizeable population - also play critical initiating roles in adaptation. Stochasticity in establishment has received little attention in microbiology, potentially due to lack of awareness as well as practical challenges in quantification. However, methods for high-replicate culturing, mutant labelling and detection, and statistical inference now make it feasible to experimentally quantify the establishment probability of specific adaptive genotypes. I review methods that have emerged over the past decade, including experimental design and mathematical formulas to estimate establishment probability from data. Quantifying establishment in further biological settings and comparing empirical estimates to theoretical predictions represent exciting future directions. More broadly, recognition that adaptive genotypes may be stochastically lost while rare is significant both for interpreting common lab assays and for designing interventions to promote or inhibit microbial evolution.
Collapse
Affiliation(s)
- Helen K. Alexander
- Institute of Ecology & Evolution, University of Edinburgh, Edinburgh, UK
| |
Collapse
|
21
|
Ren S, Xu F, Wang H, Zhang Z. Colloidal antibiotic mimics: selective capture and killing of microorganisms by shape-anisotropic colloids. SOFT MATTER 2023; 19:3253-3256. [PMID: 37128986 DOI: 10.1039/d3sm00336a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/03/2023]
Abstract
The development of targeted and efficient antimicrobials for the selective killing of pathogenic bacteria is of great importance, yet remains challenging. Here, we propose a targeted approach to selectively capture and kill microorganisms with colloidal antibiotic mimics that are readily prepared by common chemical syntheses. The mimics are shape-anisotropic colloids, which can selectively capture shape-matching microorganisms due to lock-key depletion attractions. Furthermore, after being modified with gold nanoparticles (AuNPs) and irradiated with near-infrared light, the colloidal mimics can kill the selectively captured microorganisms due to the localized photothermal effect of the AuNPs. The work demonstrates the important ability of anisotropic colloids to selectively capture and precisely kill microorganisms, which holds considerable promise for safe and adaptive antibacterial therapies without the risk of antibiotic resistance.
Collapse
Affiliation(s)
- Sihua Ren
- College of Chemistry, Chemical Engineering and Materials Science, Soochow University, Suzhou 215123, China.
| | - Fei Xu
- College of Chemistry, Chemical Engineering and Materials Science, Soochow University, Suzhou 215123, China.
| | - Huaguang Wang
- College of Chemistry, Chemical Engineering and Materials Science, Soochow University, Suzhou 215123, China.
| | - Zexin Zhang
- College of Chemistry, Chemical Engineering and Materials Science, Soochow University, Suzhou 215123, China.
- Center for Soft Condensed Matter Physics and Interdisciplinary Research, and Institute for Advanced Study, Soochow University, Suzhou 215123, China
| |
Collapse
|
22
|
Sun Y, Hong S, Chen H, Yin Y, Wang C. Production of Helvolic Acid in Metarhizium Contributes to Fungal Infection of Insects by Bacteriostatic Inhibition of the Host Cuticular Microbiomes. Microbiol Spectr 2022; 10:e0262022. [PMID: 36047778 PMCID: PMC9602595 DOI: 10.1128/spectrum.02620-22] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2022] [Accepted: 08/15/2022] [Indexed: 01/04/2023] Open
Abstract
The nortriterpenoid helvolic acid (HA) has potent antibiotic activities and can be produced by different fungi, yet HA function remains elusive. Here, we report the chemical biology of HA production in the insect pathogen Metarhizium robertsii. After deletion of the core oxidosqualene cyclase gene in Metarhizium, insect survival rates were significantly increased compared to those of insects treated with the wild type and the gene-rescued strain during topical infections but not during injection assays to bypass insect cuticles. Further gnotobiotic infection of axenic Drosophila adults confirmed the HA contribution to fungal infection by inhibiting bacterial competitors in an inoculum-dependent manner. Loss of HA production substantially impaired fungal spore germination and membrane penetration abilities relative to the WT and gene-complemented strains during challenge with different Gram-positive bacteria. Quantitative microbiome analysis revealed that HA production could assist the fungus to suppress the Drosophila cuticular microbiomes by exerting a bacteriostatic rather than bactericidal effect. Our data unveil the chemical ecology of HA and highlight the fact that fungal pathogens have to cope with the host cuticular microbiomes prior to successful infection of hosts. IMPORTANCE Emerging evidence has shown that the plant and animal surface microbiomes can defend hosts against fungal parasite infections. The strategies employed by fungal pathogens to combat the antagonistic inhibition of insect surface bacteria are still elusive. In this study, we found that the potent antibiotic helvolic acid (HA) produced by the insect pathogen Metarhizium robertsii contributes to natural fungal infection of insect hosts. Antibiotic and gnotobiotic infection assays confirmed that HA could facilitate fungal infection of insects by suppression of the host cuticular microbiomes through its bacteriostatic instead of bactericidal activities. The data from this study provide insights into the novel chemical biology of fungal secondary metabolisms.
Collapse
Affiliation(s)
- Yanlei Sun
- Key Laboratory of Insect Developmental and Evolutionary Biology, CAS Center for Excellence in Molecular Plant Sciences, Shanghai Institute of Plant Physiology and Ecology, Chinese Academy of Sciences, Shanghai, China
- CAS Center for Excellence in Biotic Interactions, University of Chinese Academy of Sciences, Beijing, China
| | - Song Hong
- Key Laboratory of Insect Developmental and Evolutionary Biology, CAS Center for Excellence in Molecular Plant Sciences, Shanghai Institute of Plant Physiology and Ecology, Chinese Academy of Sciences, Shanghai, China
| | - Haimin Chen
- Key Laboratory of Insect Developmental and Evolutionary Biology, CAS Center for Excellence in Molecular Plant Sciences, Shanghai Institute of Plant Physiology and Ecology, Chinese Academy of Sciences, Shanghai, China
- School of Life Science and Technology, ShanghaiTech University, Shanghai, China
| | - Ying Yin
- Key Laboratory of Insect Developmental and Evolutionary Biology, CAS Center for Excellence in Molecular Plant Sciences, Shanghai Institute of Plant Physiology and Ecology, Chinese Academy of Sciences, Shanghai, China
| | - Chengshu Wang
- Key Laboratory of Insect Developmental and Evolutionary Biology, CAS Center for Excellence in Molecular Plant Sciences, Shanghai Institute of Plant Physiology and Ecology, Chinese Academy of Sciences, Shanghai, China
- CAS Center for Excellence in Biotic Interactions, University of Chinese Academy of Sciences, Beijing, China
- School of Life Science and Technology, ShanghaiTech University, Shanghai, China
| |
Collapse
|
23
|
Riazimontazer E, Heiran R, Jarrahpour A, Gholami A, Hashemi Z, Kazemi A. Molecular Docking and Antibacterial Assessment of Monocyclic
β
‐Lactams against Broad‐Spectrum and Nosocomial Multidrug‐Resistant Pathogens. ChemistrySelect 2022. [DOI: 10.1002/slct.202203373] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Affiliation(s)
- Elham Riazimontazer
- Biotechnology Research Center Shiraz University of Medical Sciences Shiraz Iran
- Department of Medicinal Chemistry School of Pharmacy Shiraz University of Medical Sciences Shiraz Iran
- Pharmaceutical Sciences Research Center Shiraz University of Medical Science Shiraz Iran
| | - Roghayeh Heiran
- Department of Chemistry Estahban Higher Education Center Estahban 74519 44655
| | - Aliasghar Jarrahpour
- Department of Chemistry College of Sciences Shiraz University Shiraz 71946-84795 Iran
| | - Ahmad Gholami
- Biotechnology Research Center Shiraz University of Medical Sciences Shiraz Iran
- Pharmaceutical Sciences Research Center Shiraz University of Medical Science Shiraz Iran
| | - Zahra Hashemi
- Pharmaceutical Sciences Research Center Shiraz University of Medical Science Shiraz Iran
| | - Aboozar Kazemi
- Pharmaceutical Sciences Research Center Shiraz University of Medical Science Shiraz Iran
| |
Collapse
|
24
|
Liu J, Li H, He Q, Chen K, Chen Y, Zhong R, Li H, Fang S, Liu S, Lin S. Design, synthesis, and biological evaluation of tetrahydroquinoline amphiphiles as membrane-targeting antimicrobials against pathogenic bacteria and fungi. Eur J Med Chem 2022; 243:114734. [PMID: 36088756 DOI: 10.1016/j.ejmech.2022.114734] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Revised: 08/27/2022] [Accepted: 08/29/2022] [Indexed: 11/15/2022]
Abstract
The rising prevalence of drug-resistant pathogens is one of the biggest threats to human health. The development of new antibiotics that can overcome drug resistance is in urgent need. Herein, we designed and synthesized a series of amphiphilic tetrahydroquinoline derivatives as small-molecule-based antimicrobial peptidomimetics. Two lead compounds 36 and 52 which contained the tetrahydroquinoline core, hydrophobic alkyl chains (n-nonyl or isoprenyl group), different spacer lengths (n = 4 or 8), and cationic guanidine moiety, showed poor hemolytic activity, low cytotoxicity, and potent broad-spectrum antimicrobial activity against Gram-positive and Gram-negative bacteria, as well as fungi. The further biological evaluation revealed that compounds 36 and 52 can kill bacteria and fungi rapidly via membrane-targeting action and avoid drug resistance development. More importantly, compounds 36 and 52 exhibited similarly potent in vivo antimicrobial activities in a murine corneal infection caused by Staphylococcus aureus ATCC29213 or Pseudomonas aeruginosa ATCC9027, as compared to vancomycin or gatifloxacin. These results suggest that compounds 36 and 52 have great potential as new broad-spectrum antimicrobial agents to combat microbial resistance.
Collapse
Affiliation(s)
- Jiayong Liu
- Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, The NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences and the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, 511436, China
| | - Hongxia Li
- School of Biological Sciences, Nanyang Technological University, 60 Nanyang Drive, Singapore, 637551, Singapore
| | - Qile He
- Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, The NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences and the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, 511436, China
| | - Kaiting Chen
- Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, The NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences and the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, 511436, China
| | - Yongzhi Chen
- Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, The NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences and the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, 511436, China
| | - Rongcui Zhong
- Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, The NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences and the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, 511436, China
| | - Haizhou Li
- Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, The NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences and the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, 511436, China
| | - Shanfang Fang
- Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, The NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences and the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, 511436, China
| | - Shouping Liu
- Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, The NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences and the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, 511436, China.
| | - Shuimu Lin
- Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, The NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences and the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, 511436, China.
| |
Collapse
|
25
|
Lee DH, Cha JH, Kim DW, Lee K, Kim YS, Oh HY, Cho YH, Cha CJ. Colistin-degrading proteases confer collective resistance to microbial communities during polymicrobial infections. MICROBIOME 2022; 10:129. [PMID: 35982474 PMCID: PMC9389796 DOI: 10.1186/s40168-022-01315-x] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Accepted: 06/19/2022] [Indexed: 06/15/2023]
Abstract
BACKGROUND The increasing prevalence of resistance against the last-resort antibiotic colistin is a significant threat to global public health. Here, we discovered a novel colistin resistance mechanism via enzymatic inactivation of the drug and proposed its clinical importance in microbial communities during polymicrobial infections. RESULTS A bacterial strain of the Gram-negative opportunistic pathogen Stenotrophomonas maltophilia capable of degrading colistin and exhibiting a high-level colistin resistance was isolated from the soil environment. A colistin-degrading protease (Cdp) was identified in this strain, and its contribution to colistin resistance was demonstrated by growth inhibition experiments using knock-out (Δcdp) and complemented (Δcdp::cdp) mutants. Coculture and coinfection experiments revealed that S. maltophilia carrying the cdp gene could inactivate colistin and protect otherwise susceptible Pseudomonas aeruginosa, which may seriously affect the clinical efficacy of the drug for the treatment of cystic fibrosis patients with polymicrobial infection. CONCLUSIONS Our results suggest that Cdp should be recognized as a colistin resistance determinant that confers collective resistance at the microbial community level. Our study will provide vital information for successful clinical outcomes during the treatment of complex polymicrobial infections, particularly including S. maltophilia and other colistin-susceptible Gram-negative pathogens such as P. aeruginosa. Video abstract.
Collapse
Affiliation(s)
- Do-Hoon Lee
- Department of Systems Biotechnology and Center for Antibiotic Resistome, Chung-Ang University, Anseong, 17456, Republic of Korea
| | - Ju-Hee Cha
- Department of Systems Biotechnology and Center for Antibiotic Resistome, Chung-Ang University, Anseong, 17456, Republic of Korea
| | - Dae-Wi Kim
- Department of Systems Biotechnology and Center for Antibiotic Resistome, Chung-Ang University, Anseong, 17456, Republic of Korea
- Division of Life Sciences, Jeonbuk National University, Jeonju, 54896, Republic of Korea
| | - Kihyun Lee
- Department of Systems Biotechnology and Center for Antibiotic Resistome, Chung-Ang University, Anseong, 17456, Republic of Korea
| | - Yong-Seok Kim
- Department of Systems Biotechnology and Center for Antibiotic Resistome, Chung-Ang University, Anseong, 17456, Republic of Korea
| | - Hyo-Young Oh
- Department of Pharmacy, College of Pharmacy and Institute of Pharmaceutical Sciences, CHA University, Seongnam, 13488, Republic of Korea
| | - You-Hee Cho
- Department of Pharmacy, College of Pharmacy and Institute of Pharmaceutical Sciences, CHA University, Seongnam, 13488, Republic of Korea
| | - Chang-Jun Cha
- Department of Systems Biotechnology and Center for Antibiotic Resistome, Chung-Ang University, Anseong, 17456, Republic of Korea.
| |
Collapse
|
26
|
Zeng BS, Zeng BY, Hung CM, Kuo HC, Chen YW, Suen MW, Shiue YL, Tseng PT, Chen CH. The efficacy and tolerability of antibiotics in scrub typhus: An updated network meta-analysis of randomized controlled trials. Int J Infect Dis 2022; 122:461-468. [PMID: 35724826 DOI: 10.1016/j.ijid.2022.06.024] [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: 03/30/2022] [Revised: 06/06/2022] [Accepted: 06/14/2022] [Indexed: 10/18/2022] Open
Abstract
INTRODUCTION Inadequate treatment of scrub typhus results in severe complications such as septic shock and is also associated with a high median mortality rate of 6%. However, there has been no conclusive evidence about the superiority of different antibiotics in managing scrub typhus in terms of efficacy and tolerability. METHODS The current network meta-analysis (NMA) was conducted using the frequentist method. The included participants were pediatric and adult patients infected with scrub typhus. The primary outcome was the cure rate in the patients included. The subgroup analysis was done according to pediatric or adult patients. RESULTS Overall, 14 randomized controlled trials (RCTs) with 1264 participants were included in this study. The NMA revealed that all the investigated antibiotics were associated with cure rates similar to those of doxycycline. The chloramphenicol and minocycline were ranked to be associated with the highest cure rate in the pediatric subgroup and adult subgroup, respectively. Second-generation quinolones, including ofloxacin and ciprofloxacin and chloramphenicol were associated with significantly lower adverse event rates than doxycycline. CONCLUSION The current update NMA provides evidence for the efficacy of chloramphenicol and minocycline in scrub typhus management. However, future large-scale RCTs with longer follow-up times are warranted. TRIAL REGISTRATION PROSPERO CRD42020222410. The current study was approved by the Institutional Review Board of the Tri-Service General Hospital, National Defense Medical Center (TSGHIRB No. B-109-29).
Collapse
Affiliation(s)
- Bing-Syuan Zeng
- Department of Internal Medicine, E-DA Cancer Hospital, Kaohsiung, Taiwan; Institute of Biomedical Sciences, National Sun Yat-sen University, Kaohsiung, Taiwan
| | - Bing-Yan Zeng
- Institute of Biomedical Sciences, National Sun Yat-sen University, Kaohsiung, Taiwan; Department of Internal Medicine, E-DA Dachang Hospital, Kaohsiung, Taiwan
| | - Chao-Ming Hung
- Division of General Surgery, Department of Surgery, E-Da Cancer Hospital, Kaohsiung, Taiwan; School of Medicine, College of Medicine, I-Shou University, Kaohsiung, Taiwan
| | - Hung-Chang Kuo
- Department of Neurology, E-Da Cancer Hospital/School of Medicine, I-Shou University, Kaohsiung, Taiwan
| | - Yen-Wen Chen
- Prospect Clinic for Otorhinolaryngology & Neurology, Kaohsiung, Taiwan
| | - Mein-Woei Suen
- Department of Psychology, College of Medical and Health Science, Asia University, Taichung, Taiwan; Gender Equality Education and Research Center, Asia University, Taichung, Taiwan; Department of Medical Research, Asia University Hospital, Asia University, Taichung, Taiwan; Department of Medical Research, China Medical University Hospital, China Medical University, Taichung, Taiwan
| | - Yow-Ling Shiue
- Institute of Biomedical Sciences, National Sun Yat-sen University, Kaohsiung, Taiwan.
| | - Ping-Tao Tseng
- Institute of Biomedical Sciences, National Sun Yat-sen University, Kaohsiung, Taiwan; Prospect Clinic for Otorhinolaryngology & Neurology, Kaohsiung, Taiwan; Department of Psychology, College of Medical and Health Science, Asia University, Taichung, Taiwan.
| | - Chang-Hua Chen
- Program in Translational Medicine, National Chung Hsing University, Taichung City, Taiwan; Rong Hsing Research Center For Translational Medicine, National Chung Hsing University, Taichung City, Taiwan; Department of Internal Medicine, Changhua Christian Hospital, Changhua, Taiwan.
| |
Collapse
|
27
|
Yue T, Xia L, Tian J, Huang B, Chen C, Cao H, Zhang W. A carboxylatopillar[5]arene-based pH-triggering supramolecular photosensitizer for enhanced photodynamic antibacterial efficacy. Chem Commun (Camb) 2022; 58:2991-2994. [PMID: 35147152 DOI: 10.1039/d1cc06116j] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
A pH-triggering supramolecular antibacterial photosensitizer was constructed by host-guest interaction between a water-soluble porphyrin photosensitizer and carboxylatopillar[5]arene (P[5]). The formation of the supramolecular complex not only improves the biocompatibility of the photosensitizer, but also enhances antibacterial efficacy by pH-triggering dissociation under the low pH bacterial microenvironment.
Collapse
Affiliation(s)
- Tao Yue
- Shanghai Key Laboratory of Functional Materials Chemistry, East China University of Science and Technology, 130 Meilong Road, Shanghai 200237, China.
| | - Lei Xia
- Shanghai Key Laboratory of Functional Materials Chemistry, East China University of Science and Technology, 130 Meilong Road, Shanghai 200237, China.
| | - Jia Tian
- Shanghai Key Laboratory of Functional Materials Chemistry, East China University of Science and Technology, 130 Meilong Road, Shanghai 200237, China.
| | - Baoxuan Huang
- Shanghai Key Laboratory of Functional Materials Chemistry, East China University of Science and Technology, 130 Meilong Road, Shanghai 200237, China.
| | - Chao Chen
- Shanghai Key Laboratory of Functional Materials Chemistry, East China University of Science and Technology, 130 Meilong Road, Shanghai 200237, China.
| | - Hongliang Cao
- Shanghai Key Laboratory of Functional Materials Chemistry, East China University of Science and Technology, 130 Meilong Road, Shanghai 200237, China.
| | - Weian Zhang
- Shanghai Key Laboratory of Functional Materials Chemistry, East China University of Science and Technology, 130 Meilong Road, Shanghai 200237, China.
| |
Collapse
|
28
|
Inter-species interactions alter antibiotic efficacy in bacterial communities. THE ISME JOURNAL 2022; 16:812-821. [PMID: 34628478 PMCID: PMC8857223 DOI: 10.1038/s41396-021-01130-6] [Citation(s) in RCA: 58] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Revised: 09/16/2021] [Accepted: 09/23/2021] [Indexed: 11/14/2022]
Abstract
The efficacy of antibiotic treatments targeting polymicrobial communities is not well predicted by conventional in vitro susceptibility testing based on determining minimum inhibitory concentration (MIC) in monocultures. One reason for this is that inter-species interactions can alter the community members' susceptibility to antibiotics. Here we quantify, and identify mechanisms for, community-modulated changes of efficacy for clinically relevant antibiotics against the pathogen Pseudomonas aeruginosa in model cystic fibrosis (CF) lung communities derived from clinical samples. We demonstrate that multi-drug resistant Stenotrophomonas maltophilia can provide high levels of antibiotic protection to otherwise sensitive P. aeruginosa. Exposure protection to imipenem was provided by chromosomally encoded metallo-β-lactamase that detoxified the environment; protection was dependent upon S. maltophilia cell density and was provided by S. maltophilia strains isolated from CF sputum, increasing the MIC of P. aeruginosa by up to 16-fold. In contrast, the presence of S. maltophilia provided no protection against meropenem, another routinely used carbapenem. Mathematical ordinary differential equation modelling shows that the level of exposure protection provided against different carbapenems can be explained by differences in antibiotic efficacy and inactivation rate. Together, these findings reveal that exploitation of pre-occurring antimicrobial resistance, and inter-specific competition, can have large impacts on pathogen antibiotic susceptibility, highlighting the importance of microbial ecology for designing successful antibiotic treatments for multispecies communities.
Collapse
|
29
|
Dhanavath R, Dharavath R, Kothula D, Bitla S, Yaku G, Birdaraju S, Puchakayala MR, Atcha KR. Synthesis and Biological Evaluation of Novel 2‐Arylquinoline‐3‐Fused Thiazolo
[2,3‐c]1,2,
4‐Triazole Heterocycles as Potential Antiproliferative and Antimicrobial Agents. J Heterocycl Chem 2022. [DOI: 10.1002/jhet.4460] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Affiliation(s)
- Ramulu Dhanavath
- Department of Chemistry, Nizam College Osmania University Hyderabad India
| | - Ravinder Dharavath
- Green & Medicinal Chemistry Lab, Department of Chemistry Osmania University Hyderabad India
| | - Devender Kothula
- Department of Chemistry, Nizam College Osmania University Hyderabad India
| | - Sampath Bitla
- Department of Chemistry, Nizam College Osmania University Hyderabad India
| | - Gugulothu Yaku
- Green & Medicinal Chemistry Lab, Department of Chemistry Osmania University Hyderabad India
| | - Saritha Birdaraju
- Department of Chemistry, Nizam College Osmania University Hyderabad India
| | | | | |
Collapse
|
30
|
Li J, Claudi B, Fanous J, Chicherova N, Cianfanelli FR, Campbell RAA, Bumann D. Tissue compartmentalization enables Salmonella persistence during chemotherapy. Proc Natl Acad Sci U S A 2021; 118:e2113951118. [PMID: 34911764 PMCID: PMC8713819 DOI: 10.1073/pnas.2113951118] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/11/2021] [Indexed: 12/14/2022] Open
Abstract
Antimicrobial chemotherapy can fail to eradicate the pathogen, even in the absence of antimicrobial resistance. Persisting pathogens can subsequently cause relapsing diseases. In vitro studies suggest various mechanisms of antibiotic persistence, but their in vivo relevance remains unclear because of the difficulty of studying scarce pathogen survivors in complex host tissues. Here, we localized and characterized rare surviving Salmonella in mouse spleen using high-resolution whole-organ tomography. Chemotherapy cleared >99.5% of the Salmonella but was inefficient against a small Salmonella subset in the white pulp. Previous models could not explain these findings: drug exposure was adequate, Salmonella continued to replicate, and host stresses induced only limited Salmonella drug tolerance. Instead, antimicrobial clearance required support of Salmonella-killing neutrophils and monocytes, and the density of such cells was lower in the white pulp than in other spleen compartments containing higher Salmonella loads. Neutrophil densities declined further during treatment in response to receding Salmonella loads, resulting in insufficient support for Salmonella clearance from the white pulp and eradication failure. However, adjunctive therapies sustaining inflammatory support enabled effective clearance. These results identify uneven Salmonella tissue colonization and spatiotemporal inflammation dynamics as main causes of Salmonella persistence and establish a powerful approach to investigate scarce but impactful pathogen subsets in complex host environments.
Collapse
Affiliation(s)
- Jiagui Li
- Biozentrum, University of Basel, CH-4056 Basel, Switzerland
| | | | - Joseph Fanous
- Biozentrum, University of Basel, CH-4056 Basel, Switzerland
| | | | | | | | - Dirk Bumann
- Biozentrum, University of Basel, CH-4056 Basel, Switzerland
| |
Collapse
|
31
|
Maclura tinctoria Extracts: In Vitro Antibacterial Activity against Aeromonas hydrophila and Sedative Effect in Rhamdia quelen. FISHES 2021. [DOI: 10.3390/fishes6030025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Maclura tinctoria is a tree species native from Brazil and rich in phenolic compounds. Since plant antibacterial activity is highly associated with phenolic compound concentration, we aim to evaluate the in vitro antimicrobial activity of different extracts against fish pathogenic bacteria. In addition, some phenolic compounds have central depressant effects and can be useful in aquaculture due to possible sedative and/or anesthetic effects. Four M. tinctoria extracts were extracted separately with ethanol; leaves (LE), bark (BE), heartwood (HE), and the sapwood (SE). In vitro antimicrobial activity was tested against Aeromonas strains at concentrations of 6400 to 3.125 μg/mL. The sedative effect was evaluated for 24 h with 30 and 100 mg/L concentrations. Chemical composition was analyzed by HPLC-DAD-MS. The HE extract had the best MIC (400 µg/mL) and MBC (800 µg/mL) compared to the LE, BE, and SE extracts. LE extract induced deep sedation and the BE, SE, and HE extracts induced light sedation. Additionally, BE, SE, and HE induced a normal behavior without side effects. Polyphenolic compounds with antimicrobial activity and sedative effects were identified mainly in HE. Thus, HE extract is safe and can be used as a sedative for silver catfish.
Collapse
|
32
|
Dörr T. Understanding tolerance to cell wall-active antibiotics. Ann N Y Acad Sci 2021; 1496:35-58. [PMID: 33274447 PMCID: PMC8359209 DOI: 10.1111/nyas.14541] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Revised: 11/02/2020] [Accepted: 11/04/2020] [Indexed: 12/19/2022]
Abstract
Antibiotic tolerance-the ability of bacteria to survive for an extended time in the presence of bactericidal antibiotics-is an understudied contributor to antibiotic treatment failure. Herein, I review the manifestations, mechanisms, and clinical relevance of tolerance to cell wall-active (CWA) antibiotics, one of the most important groups of antibiotics at the forefront of clinical use. I discuss definitions of tolerance and assays for tolerance detection, comprehensively discuss the mechanism of action of β-lactams and other CWA antibiotics, and then provide an overview of how cells mitigate the potentially lethal effects of CWA antibiotic-induced cell damage to become tolerant. Lastly, I discuss evidence for a role of CWA antibiotic tolerance in clinical antibiotic treatment failure.
Collapse
Affiliation(s)
- Tobias Dörr
- Weill Institute for Cell and Molecular Biology, Department of Microbiology, and Cornell Institute of Host–Pathogen Interactions and DiseaseCornell UniversityIthacaNew York
| |
Collapse
|
33
|
Zahid MSH, Varma DM, Johnson MM, Landavazo A, Bachelder EM, Blough BE, Ainslie KM. Overcoming reduced antibiotic susceptibility in intracellular Salmonella enterica serovar Typhimurium using AR-12. FEMS Microbiol Lett 2021; 368:6293843. [PMID: 34089315 DOI: 10.1093/femsle/fnab062] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2021] [Accepted: 06/02/2021] [Indexed: 11/14/2022] Open
Abstract
Host-directed therapies (HDTs) could enhance the activity of traditional antibiotics. AR-12 is a promising HDT against intracellular pathogens including Salmonella enterica serovar Typhimurium, and has been shown to act through modulation of autophagy and the Akt kinase pathway. Since AR-12 does not inhibit the growth of planktonic bacteria but only works in conjunction with the infected host-cell, we hypothesized that AR-12 could enhance the activity of antibiotics in less-susceptible strains in the intracellular host environment. We found that repetitive passaging of S. typhimurium in macrophages in the absence of antibiotics led to a 4-fold reduction in their intracellular susceptibility to streptomycin (STR), but had no effect on the bacteria's sensitivity to AR-12. Moreover, when the host-passaged strains were treated with a combined therapy of AR-12 and STR, there was a significant reduction of intracellular bacterial burden compared to STR monotherapy. Additionally, co-treatment of macrophages infected with multi-drug resistant S. typhimurium with AR-12 and STR or ampicillin showed enhanced clearance of the intracellular bacteria. The drug combination did not elicit this effect on planktonic bacteria. Overall, AR-12 enhanced the clearance of less susceptible S. typhimurium in an intracellular environment.
Collapse
Affiliation(s)
- M Shamim Hasan Zahid
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Devika M Varma
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Monica M Johnson
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Antonio Landavazo
- Center for Drug Discovery, RTI International, Research Triangle Park, Durham, NC 27709, USA
| | - Eric M Bachelder
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Bruce E Blough
- Center for Drug Discovery, RTI International, Research Triangle Park, Durham, NC 27709, USA
| | - Kristy M Ainslie
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA.,School of Medicine, Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA.,Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, NC, USA
| |
Collapse
|
34
|
Ecology and evolution of antimicrobial resistance in bacterial communities. THE ISME JOURNAL 2021; 15:939-948. [PMID: 33219299 PMCID: PMC8115348 DOI: 10.1038/s41396-020-00832-7] [Citation(s) in RCA: 149] [Impact Index Per Article: 37.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Revised: 10/29/2020] [Accepted: 11/03/2020] [Indexed: 02/07/2023]
Abstract
Accumulating evidence suggests that the response of bacteria to antibiotics is significantly affected by the presence of other interacting microbes. These interactions are not typically accounted for when determining pathogen sensitivity to antibiotics. In this perspective, we argue that resistance and evolutionary responses to antibiotic treatments should not be considered only a trait of an individual bacteria species but also an emergent property of the microbial community in which pathogens are embedded. We outline how interspecies interactions can affect the responses of individual species and communities to antibiotic treatment, and how these responses could affect the strength of selection, potentially changing the trajectory of resistance evolution. Finally, we identify key areas of future research which will allow for a more complete understanding of antibiotic resistance in bacterial communities. We emphasise that acknowledging the ecological context, i.e. the interactions that occur between pathogens and within communities, could help the development of more efficient and effective antibiotic treatments.
Collapse
|
35
|
Abstract
Nucleotide metabolism plays a central role in bacterial physiology, producing the nucleic acids necessary for DNA replication and RNA transcription. Recent studies demonstrate that nucleotide metabolism also proactively contributes to antibiotic-induced lethality in bacterial pathogens and that disruptions to nucleotide metabolism contributes to antibiotic treatment failure in the clinic. As antimicrobial resistance continues to grow unchecked, new approaches are needed to study the molecular mechanisms responsible for antibiotic efficacy. Here we review emerging technologies poised to transform understanding into why antibiotics may fail in the clinic. We discuss how these technologies led to the discovery that nucleotide metabolism regulates antibiotic drug responses and why these are relevant to human infections. We highlight opportunities for how studies into nucleotide metabolism may enhance understanding of antibiotic failure mechanisms.
Collapse
Affiliation(s)
- Allison J Lopatkin
- Department of Biology, Barnard College, New York, NY, United States.,Department of Ecology, Evolution, and Environmental Biology, Columbia University, New York, NY, United States.,Data Science Institute, Columbia University, New York, NY, United States
| | - Jason H Yang
- Ruy V. Lourenço Center for Emerging and Re-emerging Pathogens, Rutgers New Jersey Medical School, Newark, NJ, United States.,Department of Microbiology, Biochemistry and Molecular Genetics, Rutgers New Jersey Medical School, Newark, NJ, United States
| |
Collapse
|
36
|
Shin J, Choe D, Ransegnola B, Hong H, Onyekwere I, Cross T, Shi Q, Cho B, Westblade LF, Brito IL, Dörr T. A multifaceted cellular damage repair and prevention pathway promotes high-level tolerance to β-lactam antibiotics. EMBO Rep 2021; 22:e51790. [PMID: 33463026 PMCID: PMC7857431 DOI: 10.15252/embr.202051790] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Revised: 11/17/2020] [Accepted: 12/02/2020] [Indexed: 12/27/2022] Open
Abstract
Bactericidal antibiotics are powerful agents due to their ability to convert essential bacterial functions into lethal processes. However, many important bacterial pathogens are remarkably tolerant against bactericidal antibiotics due to inducible damage repair responses. The cell wall damage response two-component system VxrAB of the gastrointestinal pathogen Vibrio cholerae promotes high-level β-lactam tolerance and controls a gene network encoding highly diverse functions, including negative control over multiple iron uptake systems. How this system contributes to tolerance is poorly understood. Here, we show that β-lactam antibiotics cause an increase in intracellular free iron levels and collateral oxidative damage, which is exacerbated in the ∆vxrAB mutant. Mutating major iron uptake systems dramatically increases ∆vxrAB tolerance to β-lactams. We propose that VxrAB reduces antibiotic-induced toxic iron and concomitant metabolic perturbations by downregulating iron uptake transporters and show that iron sequestration enhances tolerance against β-lactam therapy in a mouse model of cholera infection. Our results suggest that a microorganism's ability to counteract diverse antibiotic-induced stresses promotes high-level antibiotic tolerance and highlights the complex secondary responses elicited by antibiotics.
Collapse
Affiliation(s)
- Jung‐Ho Shin
- Weill Institute for Cell and Molecular BiologyCornell, UniversityIthacaNYUSA
- Department of MicrobiologyCornell UniversityIthacaNYUSA
| | - Donghui Choe
- Department of Biological SciencesKorea Advanced Institute of Science and TechnologyDaejeonKorea
- KI for the BioCenturyKorea Advanced Institute of Science and TechnologyDaejeonKorea
| | - Brett Ransegnola
- Weill Institute for Cell and Molecular BiologyCornell, UniversityIthacaNYUSA
- Department of MicrobiologyCornell UniversityIthacaNYUSA
| | - Hye‐Rim Hong
- Weill Institute for Cell and Molecular BiologyCornell, UniversityIthacaNYUSA
- Department of MicrobiologyCornell UniversityIthacaNYUSA
| | - Ikenna Onyekwere
- Weill Institute for Cell and Molecular BiologyCornell, UniversityIthacaNYUSA
- Department of MicrobiologyCornell UniversityIthacaNYUSA
| | - Trevor Cross
- Weill Institute for Cell and Molecular BiologyCornell, UniversityIthacaNYUSA
- Department of MicrobiologyCornell UniversityIthacaNYUSA
| | - Qiaojuan Shi
- Meinig School of Biomedical EngineeringCornell UniversityIthacaNYUSA
| | - Byung‐Kwan Cho
- Department of Biological SciencesKorea Advanced Institute of Science and TechnologyDaejeonKorea
- KI for the BioCenturyKorea Advanced Institute of Science and TechnologyDaejeonKorea
- Intelligent Synthetic Biology CenterDaejeonKorea
| | - Lars F Westblade
- Department of Pathology and Laboratory MedicineWeill Cornell MedicineNew YorkNYUSA
- Division of Infectious DiseasesDepartment of MedicineWeill Cornell MedicineNew YorkNYUSA
| | - Ilana L Brito
- Meinig School of Biomedical EngineeringCornell UniversityIthacaNYUSA
| | - Tobias Dörr
- Weill Institute for Cell and Molecular BiologyCornell, UniversityIthacaNYUSA
- Department of MicrobiologyCornell UniversityIthacaNYUSA
- Cornell Institute of Host‐Microbe Interactions and DiseaseCornell UniversityIthacaNYUSA
| |
Collapse
|
37
|
Sidders AE, Radlinski LC, Rowe SE, Conlon BP. Stimulating Aminoglycoside Uptake to Kill Staphylococcus aureus Persisters. Methods Mol Biol 2021; 2357:223-236. [PMID: 34590262 DOI: 10.1007/978-1-0716-1621-5_15] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Aminoglycosides are bactericidal drugs which require a proton motive force (PMF) for uptake into the bacterial cell. Low energy cells, such as persisters, maintain a PMF below the threshold for drug uptake and are tolerant to aminoglycosides. In this chapter, we discuss mechanisms to target the bacterial membrane and stimulate aminoglycoside uptake to kill Staphylococcus aureus persisters.
Collapse
Affiliation(s)
- Ashelyn E Sidders
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Lauren C Radlinski
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Sarah E Rowe
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Brian P Conlon
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.
- Marsico Lung Institute, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.
| |
Collapse
|
38
|
Chaturvedi P, Giri BS, Shukla P, Gupta P. Recent advancement in remediation of synthetic organic antibiotics from environmental matrices: Challenges and perspective. BIORESOURCE TECHNOLOGY 2021; 319:124161. [PMID: 33007697 DOI: 10.1016/j.biortech.2020.124161] [Citation(s) in RCA: 58] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/15/2020] [Revised: 09/17/2020] [Accepted: 09/18/2020] [Indexed: 06/11/2023]
Abstract
Continuous discharge and persistence of antibiotics in aquatic ecosystem is identified as emerging environment health hazard. Partial degradation and inappropriate disposal induce appearance of diverse antibiotic resistant genes (ARGs) and bacteria, hence their execution is imperative. Conventional methods including waste water treatment plants (WWTPs) are found ineffective for the removal of recalcitrant antibiotics. Therefore, constructive removal of antibiotics from environmental matrices and other alternatives have been discussed. This review summarizes present scenario and removal of micro-pollutants, antibiotics from environment. Various strategies including physicochemical, bioremediation, use of bioreactor, and biocatalysts are recognized as potent antibiotic removal strategies. Microbial Fuel Cells (MFCs) and biochar have emerged as promising biodegradation processes due to low cost, energy efficient and environmental benignity. With higher removal rate (20-50%) combined/ hybrid processes seems to be more efficient for permanent and sustainable elimination of reluctant antibiotics.
Collapse
Affiliation(s)
- Preeti Chaturvedi
- Aquatic Toxicology Laboratory, Environmental Toxicology Group, Council of Scientific and Industrial Research-Indian Institute of Toxicology Research (CSIR-IITR), Vishvigyan Bhawan, 31, M.G. Marg, Lucknow 226001, Uttar Pradesh, India; Department of Biotechnology, National Institute of Technology-Raipur, G.E. Road, Raipur 492010, Chhattisgarh, India.
| | - Balendu Shekher Giri
- Aquatic Toxicology Laboratory, Environmental Toxicology Group, Council of Scientific and Industrial Research-Indian Institute of Toxicology Research (CSIR-IITR), Vishvigyan Bhawan, 31, M.G. Marg, Lucknow 226001, Uttar Pradesh, India
| | - Parul Shukla
- Aquatic Toxicology Laboratory, Environmental Toxicology Group, Council of Scientific and Industrial Research-Indian Institute of Toxicology Research (CSIR-IITR), Vishvigyan Bhawan, 31, M.G. Marg, Lucknow 226001, Uttar Pradesh, India
| | - Pratima Gupta
- Department of Biotechnology, National Institute of Technology-Raipur, G.E. Road, Raipur 492010, Chhattisgarh, India
| |
Collapse
|
39
|
Identification of novel targets of azithromycin activity against Pseudomonas aeruginosa grown in physiologically relevant media. Proc Natl Acad Sci U S A 2020; 117:33519-33529. [PMID: 33318204 DOI: 10.1073/pnas.2007626117] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Pseudomonas aeruginosa causes severe multidrug-resistant infections that often lead to bacteremia and sepsis. Physiologically relevant conditions can increase the susceptibility of pathogens to antibiotics, such as azithromycin (AZM). When compared to minimal-inhibitory concentrations (MICs) in laboratory media, AZM had a 16-fold lower MIC in tissue culture medium with 5% Mueller Hinton broth (MHB) and a 64-fold lower MIC in this tissue culture medium with 20% human serum. AZM also demonstrated increased synergy in combination with synthetic host-defense peptides DJK-5 and IDR-1018 under host-like conditions and in a murine abscess model. To mechanistically study the altered effects of AZM under physiologically relevant conditions, global transcriptional analysis was performed on P. aeruginosa with and without effective concentrations of AZM. This revealed that the arn operon, mediating arabinosaminylation of lipopolysaccharides and related regulatory systems, was down-regulated in host-like media when compared to MHB. Inactivation of genes within the arn operon led to increased susceptibility of P. aeruginosa to AZM and great increases in synergy between AZM and other antimicrobial agents, indicating that dysregulation of the arn operon might explain increased AZM uptake and synergy in host-like media. Furthermore, genes involved in central and energy metabolism and ribosome biogenesis were dysregulated more in physiologically relevant conditions treated with AZM, likely due to general changes in cell physiology as a result of the increased effectiveness of AZM in these conditions. These data suggest that, in addition to the arn operon, there are multiple factors in host-like environments that are responsible for observed changes in susceptibility.
Collapse
|
40
|
Antimicrobial Susceptibility Testing in Pseudomonas aeruginosa Biofilms: One Step Closer to a Standardized Method. Antibiotics (Basel) 2020; 9:antibiotics9120880. [PMID: 33316877 PMCID: PMC7763878 DOI: 10.3390/antibiotics9120880] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Revised: 12/04/2020] [Accepted: 12/07/2020] [Indexed: 11/30/2022] Open
Abstract
The ability of Pseudomonas aeruginosa to form biofilm during a long-term infection makes it difficult to treat patients correctly. The current clinical antimicrobial susceptibility testing methods are based on the study of planktonic strains. A standardized protocol to analyze the antimicrobial susceptibility in biofilms is necessary for routine laboratories. The aims of this study were to develop a simple biofilm model and to study the antimicrobial susceptibility of P. aeruginosa strains in biofilm growth. Different artificial sputum media, and aerobiosis and microaerobiosis conditions were analyzed using a microtiter plate method and P. aeruginosa PAO1 as reference strain. Planktonic and biofilm antimicrobial susceptibility to cefepime, imipenem, azithromycin, gentamicin, tobramycin, and ciprofloxacin were determined in clinical and non-clinical P. aeruginosa strains. The Synthetic Cystic Fibrosis Medium was proposed as a good medium. The biofilm greatly increased the resistance to tested antimicrobials, except for azithromycin. Cefepime and imipenem showed poor anti-biofilm effect while tobramycin, gentamicin, and ciprofloxacin showed good activity in some strains. Azithromycin showed a better activity in biofilm than in planktonic state when aerobic conditions were used. This study establishes useful information to test antimicrobial susceptibility in P. aeruginosa biofilms, and includes possible antimicrobial options to treat long-term infected patients.
Collapse
|
41
|
Ceylan Ş, Cebeci YU, Demirbaş N, Batur ÖÖ, Özakpınar ÖB. Antimicrobial, Antioxidant and Antiproliferative Activities of Novel Quinolones. ChemistrySelect 2020; 5:11340-11346. [DOI: 10.1002/slct.202002779] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Accepted: 09/14/2020] [Indexed: 02/05/2023]
Abstract
AbstractThe compound (2) formed by esterification of dimethyl morpholine (1) was converted to acetohydrazide (3). Subsequently Schiff bases (4 a–d) and carboxy(thio)amide derivatives (5 a–e) were synthesized. Then 1,2,4‐triazole (6 a–e), thia(oxa)zolidine (7 c,e) and thia(oxa)zol (8 c,e) derivatives were obtained by ring closure from carboxy(thio)amides. Mannich bases, which are containing quinolone were synthesized from 1,2,4‐triazoles. The structures of newly synthesized compounds were illuminated by spectroscopic methods. Their antimicrobial (MIC method), antioxidant (DPPH, FRAP, and CUPRAC methods), and anticancer activities (MTT method) were examined. Results showed that most of the compounds exhibited good antimicrobial (<0.03–31.25 μg/mL with MIC values) and antioxidant activities (IC50=0.001–0.004 with DPPH values). Also, some of the compounds have been found to have antiproliferative effects on the prostate (PC‐3), liver (Hep3B), and breast (MCF‐7) human cancer cells, and also these compounds did not have a cytotoxic effect on a normal cell.
Collapse
Affiliation(s)
- Şule Ceylan
- Artvin Çoruh University Department of Occupational Health and Safety 08000 Artvin TURKEY
| | | | - Neslihan Demirbaş
- Karadeniz Technical University Department of Chemistry 61080 Trabzon Turkey
| | - Özge Özşen Batur
- Eskisehir Osmangazi University Department of Chemistry 26480 Eskisehir Turkey
| | - Özlem Bingöl Özakpınar
- Marmara University Faculty of Pharmacy Department of Biochemistry, Haydarpasa 34668 Istanbul Turkey
| |
Collapse
|
42
|
Maisetta G, Batoni G. Editorial: Interspecies Interactions: Effects on Virulence and Antimicrobial Susceptibility of Bacterial and Fungal Pathogens. Front Microbiol 2020; 11:1922. [PMID: 32983002 PMCID: PMC7479088 DOI: 10.3389/fmicb.2020.01922] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Accepted: 07/21/2020] [Indexed: 11/24/2022] Open
Affiliation(s)
- Giuseppantonio Maisetta
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy
| | - Giovanna Batoni
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy
| |
Collapse
|
43
|
Mercer DK, Torres MDT, Duay SS, Lovie E, Simpson L, von Köckritz-Blickwede M, de la Fuente-Nunez C, O'Neil DA, Angeles-Boza AM. Antimicrobial Susceptibility Testing of Antimicrobial Peptides to Better Predict Efficacy. Front Cell Infect Microbiol 2020; 10:326. [PMID: 32733816 PMCID: PMC7358464 DOI: 10.3389/fcimb.2020.00326] [Citation(s) in RCA: 77] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2020] [Accepted: 05/29/2020] [Indexed: 12/11/2022] Open
Abstract
During the development of antimicrobial peptides (AMP) as potential therapeutics, antimicrobial susceptibility testing (AST) stands as an essential part of the process in identification and optimisation of candidate AMP. Standard methods for AST, developed almost 60 years ago for testing conventional antibiotics, are not necessarily fit for purpose when it comes to determining the susceptibility of microorganisms to AMP. Without careful consideration of the parameters comprising AST there is a risk of failing to identify novel antimicrobials at a time when antimicrobial resistance (AMR) is leading the planet toward a post-antibiotic era. More physiologically/clinically relevant AST will allow better determination of the preclinical activity of drug candidates and allow the identification of lead compounds. An important consideration is the efficacy of AMP in biological matrices replicating sites of infection, e.g., blood/plasma/serum, lung bronchiolar lavage fluid/sputum, urine, biofilms, etc., as this will likely be more predictive of clinical efficacy. Additionally, specific AST for different target microorganisms may help to better predict efficacy of AMP in specific infections. In this manuscript, we describe what we believe are the key considerations for AST of AMP and hope that this information can better guide the preclinical development of AMP toward becoming a new generation of urgently needed antimicrobials.
Collapse
Affiliation(s)
| | - Marcelo D. T. Torres
- Machine Biology Group, Departments of Psychiatry and Microbiology, Institute for Biomedical Informatics, Institute for Translational Medicine and Therapeutics, Perelman School of Medicine, Penn Institute for Computational Science, and Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, United States
| | - Searle S. Duay
- Department of Chemistry, Institute of Materials Science, University of Connecticut, Storrs, CT, United States
| | - Emma Lovie
- NovaBiotics Ltd, Aberdeen, United Kingdom
| | | | | | - Cesar de la Fuente-Nunez
- Machine Biology Group, Departments of Psychiatry and Microbiology, Institute for Biomedical Informatics, Institute for Translational Medicine and Therapeutics, Perelman School of Medicine, Penn Institute for Computational Science, and Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, United States
| | | | - Alfredo M. Angeles-Boza
- Department of Chemistry, Institute of Materials Science, University of Connecticut, Storrs, CT, United States
| |
Collapse
|
44
|
Hui A, Yan R, Wang W, Wang Q, Zhou Y, Wang A. Incorporation of quaternary ammonium chitooligosaccharides on ZnO/palygorskite nanocomposites for enhancing antibacterial activities. Carbohydr Polym 2020; 247:116685. [PMID: 32829813 DOI: 10.1016/j.carbpol.2020.116685] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Revised: 06/05/2020] [Accepted: 06/22/2020] [Indexed: 02/06/2023]
Abstract
Quaternary ammonium chitooligosaccharides (QACOS) was incorporated onto the ZnO/palygorskite (ZnO/PAL) nanocomposite by a simple electrostatic self-assembly process to produce a new organic-inorganic nanocomposite (QACOS/ZnO/PAL) with excellent antibacterial activity. After loading QACOS, the Zeta potential of ZnO/PAL was changed from -26.7 to +30.3 mV, which facilitates to improve the targeting behavior of ZnO/PAL towards bacteria and its contact with bacteria, resulting in a significant improvement of antibacterial capability. The MIC values of QACOS/ZnO/PAL for inhibiting bacteria (0.5 mg/mL for E. coli and 1 mg/L for S. aureus) were superior to ZnO/PAL and QACOS, demonstrated an expected synergistic antibacterial effect between QACOS and ZnO/PAL. The improved contact and interface interaction between QACOS/ZnO/PAL and bacteria makes it easier to destroy the structural integrity of bacteria. As a whole, the incorporation of polysaccharide as regulators of surface charge opens up a new way to further enhance the antibacterial activity of inorganic antibacterial materials.
Collapse
Affiliation(s)
- Aiping Hui
- Key Laboratory of Clay Mineral Applied Research of Gansu Province, Center of Eco-material and Green Chemistry, Lanzhou Institute of Chemical Physics, Chinese Academy of Sciences, Lanzhou, 730000, PR China; Center of Xuyi Palygorskite Applied Technology, Lanzhou Institute of Chemical Physics, Chinese Academy of Sciences, Xuyi, 211700, PR China
| | - Rui Yan
- Key Laboratory of Clay Mineral Applied Research of Gansu Province, Center of Eco-material and Green Chemistry, Lanzhou Institute of Chemical Physics, Chinese Academy of Sciences, Lanzhou, 730000, PR China; Center of Xuyi Palygorskite Applied Technology, Lanzhou Institute of Chemical Physics, Chinese Academy of Sciences, Xuyi, 211700, PR China
| | - Wenbo Wang
- Key Laboratory of Clay Mineral Applied Research of Gansu Province, Center of Eco-material and Green Chemistry, Lanzhou Institute of Chemical Physics, Chinese Academy of Sciences, Lanzhou, 730000, PR China; Center of Xuyi Palygorskite Applied Technology, Lanzhou Institute of Chemical Physics, Chinese Academy of Sciences, Xuyi, 211700, PR China
| | - Qin Wang
- Key Laboratory of Clay Mineral Applied Research of Gansu Province, Center of Eco-material and Green Chemistry, Lanzhou Institute of Chemical Physics, Chinese Academy of Sciences, Lanzhou, 730000, PR China; Center of Xuyi Palygorskite Applied Technology, Lanzhou Institute of Chemical Physics, Chinese Academy of Sciences, Xuyi, 211700, PR China
| | - Yanmin Zhou
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, 210095, PR China
| | - Aiqin Wang
- Key Laboratory of Clay Mineral Applied Research of Gansu Province, Center of Eco-material and Green Chemistry, Lanzhou Institute of Chemical Physics, Chinese Academy of Sciences, Lanzhou, 730000, PR China; Center of Xuyi Palygorskite Applied Technology, Lanzhou Institute of Chemical Physics, Chinese Academy of Sciences, Xuyi, 211700, PR China.
| |
Collapse
|
45
|
Abstract
Osteomyelitis, or inflammation of bone, is most commonly caused by invasion of bacterial pathogens into the skeleton. Bacterial osteomyelitis is notoriously difficult to treat, in part because of the widespread antimicrobial resistance in the preeminent etiologic agent, the Gram-positive bacterium Staphylococcus aureus Bacterial osteomyelitis triggers pathological bone remodeling, which in turn leads to sequestration of infectious foci from innate immune effectors and systemically delivered antimicrobials. Treatment of osteomyelitis therefore typically consists of long courses of antibiotics in conjunction with surgical debridement of necrotic infected tissues. Even with these extreme measures, many patients go on to develop chronic infection or sustain disease comorbidities. A better mechanistic understanding of how bacteria invade, survive within, and trigger pathological remodeling of bone could therefore lead to new therapies aimed at prevention or treatment of osteomyelitis as well as amelioration of disease morbidity. In this minireview, we highlight recent developments in our understanding of how pathogens invade and survive within bone, how bacterial infection or resulting innate immune responses trigger changes in bone remodeling, and how model systems can be leveraged to identify new therapeutic targets. We review the current state of osteomyelitis epidemiology, diagnostics, and therapeutic guidelines to help direct future research in bacterial pathogenesis.
Collapse
|
46
|
Yarlagadda V, Wright GD. Membrane-Active Rhamnolipids Overcome Aminoglycoside Resistance. Cell Chem Biol 2020; 26:1333-1334. [PMID: 31626780 DOI: 10.1016/j.chembiol.2019.09.015] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
In this issue of Cell Chemical Biology, Radlinski et al. (2019) identify Pseudomonas-derived rhamnolipids that potentiate aminoglycoside antibiotics in the eradication of antibiotic-tolerant bacterial phenotypes. Microbial physiological and mechanistic studies indicate that rhamnolipids permeabilize S. aureus membrane and drive the uptake of aminoglycosides that is independent of proton-motive force.
Collapse
Affiliation(s)
- Venkateswarlu Yarlagadda
- David Braley Centre for Antibiotic Discovery, M.G. DeGroote Institute for Infectious Disease Research, Department of Biochemistry and Biomedical Sciences, DeGroote School of Medicine, McMaster University, 1280 Main Street West, Hamilton, ON L8S 4K1, Canada
| | - Gerard D Wright
- David Braley Centre for Antibiotic Discovery, M.G. DeGroote Institute for Infectious Disease Research, Department of Biochemistry and Biomedical Sciences, DeGroote School of Medicine, McMaster University, 1280 Main Street West, Hamilton, ON L8S 4K1, Canada.
| |
Collapse
|
47
|
Hůlková M, Soukupová J, Carlson RP, Maršálek B. Interspecies interactions can enhance Pseudomonas aeruginosa tolerance to surfaces functionalized with silver nanoparticles. Colloids Surf B Biointerfaces 2020; 192:111027. [PMID: 32387859 DOI: 10.1016/j.colsurfb.2020.111027] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2019] [Revised: 04/03/2020] [Accepted: 04/06/2020] [Indexed: 12/19/2022]
Abstract
Development of anti-fouling surfaces is a major challenge in materials research. Microorganisms growing as biofilms have enhanced tolerance to antimicrobial strategies including antibiotics and antiseptics complicating the design of anti-fouling surfaces. Silver nanoparticles (AgNPs) are a promising antimicrobial technology with broad spectrum efficacy with a reduced likelihood of microorganisms developing resistance to the technology. This study tested the efficacy of new immobilized AgNP-modified surface technology against three common opportunistic pathogens grown either as monocultures or as cocultures. The presented study fills a gap in the literature by quantifying the efficacy of immobilized AgNP particles against multispecies biofilms. Polyethylene (PE) surfaces functionalized with the AgNPs were highly effective against Pseudomonas aeruginosa biofilms reducing viable cell counts by 99.8 % as compared to controls. However, the efficacy of the AgNP-modified PE surface was compromised when P. aeruginosa was cocultured with Candida albicans. Interspecies interactions can strongly influence the efficacy of anti-fouling AgNP coatings highlighting the need to test surfaces not only against biofilm phenotypes but under conditions representative of applications including the presence of multispecies consortia.
Collapse
Affiliation(s)
- Markéta Hůlková
- Research Centre for Toxic Compounds in the Environment, Masaryk University Brnob, Kamenice, Brno, Czech Republic; Institute of Botany, Academy of Sciences of the Czech Republic, Lidická 25/27, 602 00, Brno, Czech Republic; Department of Chemical and Biological Engineering, Center for Biofilm Engineering and Thermal Biology Institute, Montana State University, Bozeman, MT, 59717, USA.
| | - Jana Soukupová
- Regional Centre of Advanced Technologies and Materials, Department of Physical Chemistry, Faculty of Science, Palacky University in Olomouc, Šlechtitelů 27, 783 71, Olomouc, Czech Republic.
| | - Ross P Carlson
- Department of Chemical and Biological Engineering, Center for Biofilm Engineering and Thermal Biology Institute, Montana State University, Bozeman, MT, 59717, USA.
| | - Blahoslav Maršálek
- Research Centre for Toxic Compounds in the Environment, Masaryk University Brnob, Kamenice, Brno, Czech Republic; Institute of Botany, Academy of Sciences of the Czech Republic, Lidická 25/27, 602 00, Brno, Czech Republic.
| |
Collapse
|
48
|
The Small Protein CydX Is Required for Cytochrome bd Quinol Oxidase Stability and Function in Salmonella enterica Serovar Typhimurium: a Phenotypic Study. J Bacteriol 2020; 202:JB.00348-19. [PMID: 31659011 DOI: 10.1128/jb.00348-19] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2019] [Accepted: 10/21/2019] [Indexed: 01/12/2023] Open
Abstract
Cytochrome bd quinol oxidases, which have a greater affinity for oxygen than heme-copper cytochrome oxidases (HCOs), promote bacterial respiration and fitness in low-oxygen environments, such as host tissues. Here, we show that, in addition to the CydA and CydB subunits, the small protein CydX is required for the assembly and function of the cytochrome bd complex in the enteric pathogen Salmonella enterica serovar Typhimurium. Mutant S Typhimurium lacking CydX showed a loss of proper heme arrangement and impaired oxidase activity comparable to that of a ΔcydABX mutant lacking all cytochrome bd subunits. Moreover, both the ΔcydX mutant and the ΔcydABX mutant showed increased sensitivity to β-mercaptoethanol and nitric oxide (NO). Cytochrome bd-mediated protection from β-mercaptoethanol was not a result of resistance to reducing damage but, rather, was due to cytochrome bd oxidase managing Salmonella respiration, while β-mercaptoethanol interacted with the copper ions necessary for the HCO activity of the cytochrome bo-type quinol oxidase. Interactions between NO and hemes in cytochrome bd and cytochrome bd-dependent respiration during nitrosative stress indicated a direct role for cytochrome bd in mediating Salmonella resistance to NO. Additionally, CydX was required for S Typhimurium proliferation inside macrophages. Mutants deficient in cytochrome bd, however, showed a significant increase in resistance to antibiotics, including aminoglycosides, d-cycloserine, and ampicillin. The essential role of CydX in cytochrome bd assembly and function suggests that targeting this small protein could be a useful antimicrobial strategy, but potential drug tolerance responses should also be considered.IMPORTANCE Cytochrome bd quinol oxidases, which are found only in bacteria, govern the fitness of many facultative anaerobic pathogens by promoting respiration in low-oxygen environments and by conferring resistance to antimicrobial radicals. Thus, cytochrome bd complex assembly and activity are considered potential therapeutic targets. Here we report that the small protein CydX is required for the assembly and function of the cytochrome bd complex in S Typhimurium under stress conditions, including exposure to β-mercaptoethanol, nitric oxide, or the phagocytic intracellular environment, demonstrating its crucial function for Salmonella fitness. However, cytochrome bd inactivation also leads to increased resistance to some antibiotics, so considerable caution should be taken when developing therapeutic strategies targeting the CydX-dependent cytochrome bd.
Collapse
|
49
|
Chalmers SJ, Wylam ME. Methicillin-Resistant Staphylococcus aureus Infection and Treatment Options. Methods Mol Biol 2020; 2069:229-251. [PMID: 31523777 DOI: 10.1007/978-1-4939-9849-4_16] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Methicillin-resistant Staphylococcus aureus (MRSA) is a leading cause of infection worldwide, including a wide array of both hospital- and community-acquired infections-most commonly bacteremia, upper and lower respiratory tract infection, skin and soft-tissue infection, osteomyelitis, and septic arthritis. This chapter describes the epidemiology of MRSA infection, its ability to confer antibiotic resistance and produce a wide array of virulence factors, and its pivotal role in human infection, especially cystic fibrosis. It also provides an introduction to the strategies for treatment of both chronic and acute MRSA infections.
Collapse
Affiliation(s)
- Sarah J Chalmers
- Division of Pulmonary and Critical Care Medicine, Mayo Clinic College of Medicine, Rochester, MN, USA
| | - Mark E Wylam
- Division of Pulmonary and Critical Care Medicine, Mayo Clinic College of Medicine, Rochester, MN, USA.
| |
Collapse
|
50
|
Orazi G, O'Toole GA. "It Takes a Village": Mechanisms Underlying Antimicrobial Recalcitrance of Polymicrobial Biofilms. J Bacteriol 2019; 202:e00530-19. [PMID: 31548277 PMCID: PMC6932244 DOI: 10.1128/jb.00530-19] [Citation(s) in RCA: 106] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Chronic infections are frequently caused by polymicrobial biofilms. Importantly, these infections are often difficult to treat effectively in part due to the recalcitrance of biofilms to antimicrobial therapy. Emerging evidence suggests that polymicrobial interactions can lead to dramatic and unexpected changes in the ability of antibiotics to eradicate biofilms and often result in decreased antimicrobial efficacy in vitro In this review, we discuss the influence of polymicrobial interactions on the antibiotic susceptibility of biofilms, and we highlight the studies that first documented the shifted antimicrobial susceptibilities of mixed-species cultures. Recent studies have identified several mechanisms underlying the recalcitrance of polymicrobial biofilm communities, including interspecies exchange of antibiotic resistance genes, β-lactamase-mediated inactivation of antibiotics, changes in gene expression induced by metabolites and quorum sensing signals, inhibition of the electron transport chain, and changes in properties of the cell membrane. In addition to elucidating multiple mechanisms that contribute to the altered drug susceptibility of polymicrobial biofilms, these studies have uncovered novel ways in which polymicrobial interactions can impact microbial physiology. The diversity of findings discussed highlights the importance of continuing to investigate the efficacy of antibiotics against biofilm communities composed of different combinations of microbial species. Together, the data presented here illustrate the importance of studying microbes as part of mixed-species communities rather than in isolation. In light of our greater understanding of how interspecies interactions alter the efficacy of antimicrobial agents, we propose that the methods for measuring the drug susceptibility of polymicrobial infections should be revisited.
Collapse
Affiliation(s)
- Giulia Orazi
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire, USA
| | - George A O'Toole
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire, USA
| |
Collapse
|