1
|
Chakraborty A, Bhakta K, Ghosh A, Manna D, Maity AR, Sikder K, Chakraborti S, Basu A. Artesunate Perturbs GTP Binding of the Conserved GTPase Obg Thereby Alleviating Antibiotic Resistance in Methicillin-Resistant Staphylococcus aureus. ACS Infect Dis 2025; 11:1190-1202. [PMID: 40278541 DOI: 10.1021/acsinfecdis.4c01027] [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] [Indexed: 04/26/2025]
Abstract
Methicillin-resistant Staphylococcus aureus (MRSA) is an important nosocomial pathogen that causes various secondary infections among hospital-associated patients. The pathogen is challenging to treat due to its resistance to a wide spectrum of antibiotics, including the last-resort antibiotic vancomycin and newly developed drugs, such as linezolid and daptomycin. While the invention of entirely new drugs to combat MRSA infection seems almost impossible, potentiating the efficacy of conventional antibiotics is critical. Our article explores the novel application of the antimalarial drug artesunate, which enhances the efficacy of vancomycin and cefoxitin in treating MRSA infections. We focused on ObgSa, a conserved GTPase in MRSA, and found that artesunate selectively binds to its GTP-binding pocket. We further evaluated the GTP-binding activity and metal dependence (specifically, Mg2+) of this conserved GTPase. In silico analysis identified several threonine residues essential for GTP binding, which were subsequently mutated to assess their role in GTP binding. As shown in the analysis, these mutations significantly impacted both the GTP binding and hydrolysis functions of ObgSa. Notably, these threonine residues were also crucial for artesunate binding within the GTP-binding domain. When the effect of artesunate was assessed, the drug competitively inhibited GTP binding and hydrolysis of the GTPase. This result was manifested as reduced antibiotic tolerance, disruption of biofilms, and a decrease in persister cells─critical factors in chronic infections. In summary, our research presents an innovative strategy to combat antimicrobial resistance through artesunate, highlighting its potential effectiveness in eradicating infections.
Collapse
Affiliation(s)
- Asmita Chakraborty
- Department of Biomedical Science and Technology, The School of Biological Sciences, Ramakrishna Mission Vivekananda Educational Research Institute, Howrah, West Bengal 711202, India
| | - Koustav Bhakta
- Biological Sciences Unified Academic Campus Bose Institute EN-80, Sector V Bidhan Nagar, Kolkata 700 091, India
| | - Abhrajyoti Ghosh
- Biological Sciences Unified Academic Campus Bose Institute EN-80, Sector V Bidhan Nagar, Kolkata 700 091, India
| | - Dipak Manna
- Department of Biomedical Science and Technology, The School of Biological Sciences, Ramakrishna Mission Vivekananda Educational Research Institute, Howrah, West Bengal 711202, India
| | - Amit Ranjan Maity
- Institute of Biotechnology, Amity University Kolkata Campus, Major Arterial Road, Action Area II, Newtown, Kadampukur, Kolkata, West Bengal 700135, India
| | - Kunal Sikder
- Department of Biomedical Science and Technology, The School of Biological Sciences, Ramakrishna Mission Vivekananda Educational Research Institute, Howrah, West Bengal 711202, India
| | - Soumyananda Chakraborti
- Department of Biological Science, Birla Institute of Technology and Science, Pilani, Hyderabad Campus, Jawahar Nagar, Kapra Mandal, Medchal District, Hyderabad, Telangana 500078, India
| | - Arnab Basu
- Department of Biomedical Science and Technology, The School of Biological Sciences, Ramakrishna Mission Vivekananda Educational Research Institute, Howrah, West Bengal 711202, India
| |
Collapse
|
2
|
Alimu A, Wu X, Huang D, Chu C, Pan B, Xing Y, Chen W, Long L, Sheng P. Application value of baicalein in the management of periprosthetic joint infection: a preliminary in vitro study. BMC Complement Med Ther 2025; 25:152. [PMID: 40269827 PMCID: PMC12020321 DOI: 10.1186/s12906-025-04888-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Accepted: 04/09/2025] [Indexed: 04/25/2025] Open
Abstract
BACKGROUND This study aims to evaluate the efficacy of baicalein, a flavonoid derived from Scutellaria baicalensis, against Staphylococcus aureus (S. aureus), focusing on its inhibitory and eradicative effects on biofilms, as well as its cellular cytotoxicity. The goal is to provide preliminary evidence for its potential application in the management of periprosthetic joint infection (PJI). METHODS The minimum inhibitory concentration (MIC) and minimum bactericidal concentration (MBC) of baicalein against the standard strain of S. aureus ATCC 29213, a clinical strain of methicillin-sensitive S. aureus 115 (MSSA 115), and a clinical strain of methicillin-resistant S. aureus 49 (MRSA 49) were determined using broth microdilution and colony counting methods. Bactericidal kinetics over a 24-h period were evaluated using a time-kill assay. Biofilm inhibition and eradication were assessed on 96-well and titanium alloy plates, while the cellular cytotoxicity of baicalein was examined using the cell counting kit-8 (CCK-8) assay on human primary synovial fibroblasts and chondrocytes. RESULTS The MIC of baicalein was 32 μg/mL for the ATCC 29213, and 64 μg/mL for both MSSA115 and MRSA49. Meanwhile, the MBC for all three strains was 128 μg/mL. Baicalein exhibited a time-dependent bactericidal activity, with maximum efficacy at 24 h. Biofilm inhibition was evident at concentrations equal to or exceeding the MIC, as confirmed by biofilm biomass and metabolic activity assays, along with scanning electron and confocal laser microscope. However, baicalein was unable to completely eradicate preformed biofilms. Baicalein demonstrated significant cytotoxic effects on both synovial fibroblasts and chondrocytes after exposure for 16 and 24 h. CONCLUSIONS Baicalein shows significant bactericidal effects and effectively inhibits S. aureus biofilm formation. These findings suggest its potential as a promising local antibacterial agent for postoperative continuous intra-articular lavage in the treatment of S. aureus-related early postoperative or acute hematogenous PJIs.
Collapse
Affiliation(s)
- Aerman Alimu
- Department of Joint Surgery, The First Affiliated Hospital of Sun Yat-Sen University, NO.58, Zhongshan 2nd Road, Guangzhou, 510080, China
- Guangdong Provincial Key Laboratory of Orthopaedics and Traumatology, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, 510080, China
| | - Xiaoyu Wu
- Department of Joint Surgery, The First Affiliated Hospital of Sun Yat-Sen University, NO.58, Zhongshan 2nd Road, Guangzhou, 510080, China
- Guangdong Provincial Key Laboratory of Orthopaedics and Traumatology, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, 510080, China
| | - Dongwei Huang
- Department of Joint Surgery, The First Affiliated Hospital of Sun Yat-Sen University, NO.58, Zhongshan 2nd Road, Guangzhou, 510080, China
- Guangdong Provincial Key Laboratory of Orthopaedics and Traumatology, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, 510080, China
| | - Chenghan Chu
- Department of Joint Surgery, The First Affiliated Hospital of Sun Yat-Sen University, NO.58, Zhongshan 2nd Road, Guangzhou, 510080, China
- Guangdong Provincial Key Laboratory of Orthopaedics and Traumatology, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, 510080, China
| | - Baiqi Pan
- Department of Joint Surgery, The First Affiliated Hospital of Sun Yat-Sen University, NO.58, Zhongshan 2nd Road, Guangzhou, 510080, China
- Guangdong Provincial Key Laboratory of Orthopaedics and Traumatology, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, 510080, China
| | - Yang Xing
- Department of Joint Surgery, The First Affiliated Hospital of Sun Yat-Sen University, NO.58, Zhongshan 2nd Road, Guangzhou, 510080, China
- Guangdong Provincial Key Laboratory of Orthopaedics and Traumatology, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, 510080, China
| | - Weishen Chen
- Department of Joint Surgery, The First Affiliated Hospital of Sun Yat-Sen University, NO.58, Zhongshan 2nd Road, Guangzhou, 510080, China.
- Guangdong Provincial Key Laboratory of Orthopaedics and Traumatology, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, 510080, China.
| | - Lingli Long
- Research Center of Translational Medicine, The First Affiliated Hospital of Sun Yat-Sen University, NO.58, Zhongshan 2nd Road, Guangzhou, 510080, China.
| | - Puyi Sheng
- Department of Joint Surgery, The First Affiliated Hospital of Sun Yat-Sen University, NO.58, Zhongshan 2nd Road, Guangzhou, 510080, China.
- Guangdong Provincial Key Laboratory of Orthopaedics and Traumatology, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, 510080, China.
| |
Collapse
|
3
|
Beenken KE, Smeltzer MS. Staphylococcus aureus Biofilm-Associated Infections: Have We Found a Clinically Relevant Target? Microorganisms 2025; 13:852. [PMID: 40284688 PMCID: PMC12029350 DOI: 10.3390/microorganisms13040852] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2025] [Revised: 03/26/2025] [Accepted: 04/02/2025] [Indexed: 04/29/2025] Open
Abstract
Staphylococcus aureus is one of the most diverse bacterial pathogens. This is reflected in its ability to cause a wide array of infections and in genotypic and phenotypic differences between clinical isolates that extend beyond their antibiotic resistance status. Many S. aureus infections, including those involving indwelling medical devices, are therapeutically defined by the formation of a biofilm. This is reflected in the number of reports focusing on S. aureus biofilm formation and biofilm-associated infections. These infections are characterized by a level of intrinsic resistance that compromises conventional antibiotic therapy irrespective of acquired resistance, suggesting that an inhibitor of biofilm formation would have tremendous clinical value. Many reports have described large-scale screens aimed at identifying compounds that limit S. aureus biofilm formation, but relatively few examined whether the limitation was sufficient to overcome this intrinsic resistance. Similarly, while many of these reports examined the impact of putative inhibitors on S. aureus phenotypes, very few took a focused approach to identify and optimize an effective inhibitor of specific biofilm-associated targets. Such approaches are dependent on validating a target, hopefully one that is not restricted by the diversity of S. aureus as a bacterial pathogen. Rigorous biological validation of such a target would allow investigators to virtually screen vast chemical libraries to identify potential inhibitors that warrant further investigation based on their predicted function. Here, we summarize reports describing S. aureus regulatory loci implicated in biofilm formation to assess whether they are viable targets for the development of an anti-biofilm therapeutic strategy with an emphasis on whether sarA has been sufficiently validated to warrant consideration in this important clinical context.
Collapse
Affiliation(s)
- Karen E. Beenken
- Department of Microbiology and Immunology, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA;
- Department of Biochemistry and Molecular Biology, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Mark S. Smeltzer
- Department of Microbiology and Immunology, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA;
- Department of Orthopaedic Surgery, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| |
Collapse
|
4
|
Ismail AS, Berryhill BA, Gil-Gil T, Manuel JA, Smith AP, Baquero F, Levin BR. The tradeoffs between persistence and mutation rates at sub-inhibitory antibiotic concentrations in Staphylococcus aureus. Microbiol Spectr 2025; 13:e0247924. [PMID: 40035534 PMCID: PMC11960066 DOI: 10.1128/spectrum.02479-24] [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: 10/03/2024] [Accepted: 12/19/2024] [Indexed: 03/05/2025] Open
Abstract
The rational design of the antibiotic treatment of bacterial infections employs these drugs to reach concentrations that exceed the minimum needed to prevent the replication of the target bacteria. However, within a treated patient, spatial and physiological heterogeneity promotes antibiotic gradients such that the concentration of antibiotics at specific sites is below the minimum needed to inhibit bacterial growth. Here, we investigate the effects of sub-inhibitory antibiotic concentrations on three parameters central to bacterial infection and the success of antibiotic treatment, using in vitro experiments with Staphylococcus aureus and mathematical and computer-simulation models. Our results, using drugs of six different classes, demonstrate that exposure to sub-inhibitory antibiotic concentrations alters bacterial growth dynamics, increases the mutation rate to antibiotic resistance, and decreases the production of persister cells thereby reducing persistence levels. Understanding this trade-off between mutation rates and persistence levels resulting from sub-inhibitory antibiotic exposure is crucial for optimizing, and mitigating the failure of, antibiotic therapy. IMPORTANCE Much of the research on antibiotics and antibiotic treatment has focused on drug concentrations sufficient to prevent the growth of bacteria. These concentrations, however, are not always reached everywhere in the body. Here, we look at the effects of exposure to these low concentrations of antibiotics on the common clinically important pathogen Staphylococcus aureus. We confirm a previous finding that sub-inhibitory antibiotic exposure decreases the total growth and the growth rate of the bacteria. Moreover, we demonstrate that the level of persistence, an important mechanism for bacteria to survive antibiotics, is decreased due to sub-inhibitory exposure. However, we find that the rate of generation of resistant mutants is substantially increased. Taken together, these results reveal an important trade-off that emerges as a consequence of bacteria being exposed to sub-inhibitory concentrations of antibiotics.
Collapse
Affiliation(s)
| | - Brandon A. Berryhill
- Department of Biology, Emory University, Atlanta, Georgia, USA
- Program in Microbiology and Molecular Genetics, Graduate Division of Biological and Biomedical Sciences, Laney Graduate School, Emory University, Atlanta, Georgia, USA
| | - Teresa Gil-Gil
- Department of Biology, Emory University, Atlanta, Georgia, USA
| | | | - Andrew P. Smith
- Department of Biology, Emory University, Atlanta, Georgia, USA
| | - Fernando Baquero
- Servicio de Microbiología, Hospital Universitario Ramón y Cajal, Instituto Ramón y Cajal de Investigación Sanitaria, and Centro de Investigación Médica en Red de Epidemiología y Salud Pública (CIBERESP), Madrid, Spain
| | - Bruce R. Levin
- Department of Biology, Emory University, Atlanta, Georgia, USA
- Emory Antibiotic Resistance Center, Emory University, Atlanta, Georgia, USA
| |
Collapse
|
5
|
Higashihira S, Simpson SJ, Arnold CJ, Deckard ER, Meneghini RM, Greenfield EM, Buller LT. Biofilm Formation is Durably Prevented on Pre-Fabricated Antibiotic Cement Spacers Compared to Cobalt Chrome and Polyethylene. J Arthroplasty 2025; 40:779-785. [PMID: 39233097 DOI: 10.1016/j.arth.2024.08.046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Revised: 08/26/2024] [Accepted: 08/28/2024] [Indexed: 09/06/2024] Open
Abstract
BACKGROUND A 2-stage revision remains the standard for managing chronic periprosthetic joint infection. Despite multiple spacer options, whether a particular one better resists biofilm formation remains unclear. Prefabricated polymethylmethacrylate (PMMA) articulating spacers containing antibiotics and a proprietary pore structure were developed to increase antibiotic elution characterized by a rapid burst phase for the initial one to two days and an extended slow-release phase for > 28 days. This in vitro study determined whether biofilm formation is prevented during the initial rapid burst phase and/or the slow-release phase. METHODS S. aureus-Xen36 was incubated in 1.5 mL of Luria-Bertani broth with PMMA discs with the proprietary pore structure either with or without gentamycin and vancomycin or with 'Hoffman style' positive-control discs (ultra-high molecular weight polyethylene or cobalt-chrome). Nonadherent bacteria were removed by three phosphate buffered saline rinses every 20 to 24 hours. Planktonic bacterial growth in the culture broth and biofilm formation on the discs were measured by colony forming unit (CFU) counting and resazurin reduction assays. Experiments were repeated > four times. RESULTS No detectable planktonic bacterial growth or biofilm formation occurred in cultures containing PMMA with antibiotics (≤ 15 CFUs/disc), whereas biofilms formed on PMMA without antibiotics, ultra-high molecular weight polyethylene, and cobalt-chrome (1 × 107 to 4 × 108 CFUs/disc, P < 0.0001). Biofilm formation was confirmed by a 100-fold decrease in sensitivity to vancomycin. To determine whether the antibiotic slow-release phase is sufficient to block biofilm formation, PMMA discs with antibiotics were preeluted for 14 days with multiple saline changes prior to bacterial inoculation. After antibiotic elution, still no detectable biofilms formed on PMMA discs with antibiotics (≤ 15 CFUs/disc, P < 0.0001). CONCLUSIONS Antibiotic release during both the initial and slow-release phases prevented biofilm formation on PMMA with the proprietary pore structure. This may translate into improved infection eradication rates clinically.
Collapse
Affiliation(s)
- Shota Higashihira
- Department of Orthopaedic Surgery, Indiana University School of Medicine, Indianapolis, Indiana; Indiana Center for Musculoskeletal Health, Indiana University School of Medicine, Indianapolis, Indiana; Department of Orthopaedic Surgery, Yokohama City University Medical Center, Yokohama, Kanagawa, Japan
| | - Stefanie J Simpson
- Department of Orthopaedic Surgery, Indiana University School of Medicine, Indianapolis, Indiana; Indiana Center for Musculoskeletal Health, Indiana University School of Medicine, Indianapolis, Indiana
| | - Christopher J Arnold
- Department of Orthopaedic Surgery, Indiana University School of Medicine, Indianapolis, Indiana
| | - Evan R Deckard
- Indiana Joint Replacement Institute, Indianapolis, Indiana
| | | | - Edward M Greenfield
- Department of Orthopaedic Surgery, Indiana University School of Medicine, Indianapolis, Indiana; Indiana Center for Musculoskeletal Health, Indiana University School of Medicine, Indianapolis, Indiana
| | - Leonard T Buller
- Department of Orthopaedic Surgery, Indiana University School of Medicine, Indianapolis, Indiana
| |
Collapse
|
6
|
Xia Y, Hu Z, Jin Q, Chen Q, Zhao C, Qiang R, Xie Z, Li L, Zhang H. Structural characteristics, functions, and counteracting strategies of biofilms in Staphylococcus aureus. Comput Struct Biotechnol J 2025; 27:488-500. [PMID: 39916696 PMCID: PMC11799891 DOI: 10.1016/j.csbj.2025.01.021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2024] [Revised: 01/20/2025] [Accepted: 01/21/2025] [Indexed: 02/09/2025] Open
Abstract
Background Staphylococcus aureus (S. aureus) is a prevalent pathogen associated with a wide range of infections, exhibiting significant antibiotic resistance and posing therapeutic challenges in clinical settings. The formation of biofilms contributes to the emergence of resistant strains, further diminishing the efficacy of antibiotics. This, in turn, leads to chronic and recurrent infections, ultimately increasing the healthcare burden. Consequently, preventing and eliminating biofilms has become a critical focus in clinical management and research. Aim of review This review systematically examines the mechanisms underlying biofilm formation in S. aureus and its contribution to antibiotic resistance, emphasizing the essential roles biofilms play in maintaining structural integrity and enhancing resistance. It also analyses the protective mechanisms that fortify S. aureus biofilms against antimicrobial treatments. Furthermore, the review provides a comprehensive overview of recent therapeutic innovations, including enzymatic therapy, nanotechnology, gene editing, and phage therapy. Key scientific concepts of review Emerging therapeutic strategies present novel approaches to combat S. aureus biofilm-associated infections through various mechanisms. This review discusses recent advancements in these therapies, their practical challenges in clinical application, and provides an in-depth analysis of each strategy's mechanisms and therapeutic potential. By mapping future research directions, this review aims to refine anti-biofilm strategies to control infection progression and effectively mitigate recurrence.
Collapse
Affiliation(s)
- Yanze Xia
- Department of Orthopedics, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Zhenghui Hu
- Department of Orthopedics, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Qiyuan Jin
- Department of Clinical Laboratory, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Qi Chen
- Department of Clinical Laboratory, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Chenhao Zhao
- Department of Clinical Laboratory, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Rui Qiang
- Department of Clinical Laboratory, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Zonggang Xie
- Department of Orthopedics, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Liubing Li
- Department of Orthopedics, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Haifang Zhang
- Department of Clinical Laboratory, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
- MOE Key Laboratory of Geriatric Diseases and Immunology, Soochow University, Suzhou, China
| |
Collapse
|
7
|
Liu JD, VanTreeck KE, Marston WA, Papadopoulou V, Rowe SE. Ultrasound-Mediated Antibiotic Delivery to In Vivo Biofilm Infections: A Review. Chembiochem 2024; 25:e202400181. [PMID: 38924307 PMCID: PMC11483220 DOI: 10.1002/cbic.202400181] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Revised: 06/21/2024] [Accepted: 06/24/2024] [Indexed: 06/28/2024]
Abstract
Bacterial biofilms are a significant concern in various medical contexts due to their resilience to our immune system as well as antibiotic therapy. Biofilms often require surgical removal and frequently lead to recurrent or chronic infections. Therefore, there is an urgent need for improved strategies to treat biofilm infections. Ultrasound-mediated drug delivery is a technique that combines ultrasound application, often with the administration of acoustically-active agents, to enhance drug delivery to specific target tissues or cells within the body. This method involves using ultrasound waves to assist in the transportation or activation of medications, improving their penetration, distribution, and efficacy at the desired site. The advantages of ultrasound-mediated drug delivery include targeted and localized delivery, reduced systemic side effects, and improved efficacy of the drug at lower doses. This review scrutinizes recent advances in the application of ultrasound-mediated drug delivery for treating biofilm infections, focusing on in vivo studies. We examine the strengths and limitations of this technology in the context of wound infections, device-associated infections, lung infections and abscesses, and discuss current gaps in knowledge and clinical translation considerations.
Collapse
Affiliation(s)
- Jamie D. Liu
- Department of Microbiology and Immunology, University of North Carolina-Chapel Hill, Chapel Hill, North Carolina 27599, USA
| | - Kelly E. VanTreeck
- Joint Department of Biomedical Engineering, The University of North Carolina and North Carolina State University, Chapel Hill, North Carolina 27599, USA
- Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, NC 27599, USA
| | - William A. Marston
- Department of Surgery, University of North Carolina-Chapel Hill, Chapel Hill, North Carolina 27599, USA
| | - Virginie Papadopoulou
- Joint Department of Biomedical Engineering, The University of North Carolina and North Carolina State University, Chapel Hill, North Carolina 27599, USA
- Department of Radiology, The University of North Carolina at Chapel Hill, NC, USA
| | - Sarah E. Rowe
- Department of Microbiology and Immunology, University of North Carolina-Chapel Hill, Chapel Hill, North Carolina 27599, USA
| |
Collapse
|
8
|
Liao H, Yan X, Wang C, Huang C, Zhang W, Xiao L, Jiang J, Bao Y, Huang T, Zhang H, Guo C, Zhang Y, Pu Y. Cyclic di-GMP as an antitoxin regulates bacterial genome stability and antibiotic persistence in biofilms. eLife 2024; 13:RP99194. [PMID: 39365286 PMCID: PMC11452175 DOI: 10.7554/elife.99194] [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] [Indexed: 10/05/2024] Open
Abstract
Biofilms are complex bacterial communities characterized by a high persister prevalence, which contributes to chronic and relapsing infections. Historically, persister formation in biofilms has been linked to constraints imposed by their dense structures. However, we observed an elevated persister frequency accompanying the stage of cell adhesion, marking the onset of biofilm development. Subsequent mechanistic studies uncovered a comparable type of toxin-antitoxin (TA) module (TA-like system) triggered by cell adhesion, which is responsible for this elevation. In this module, the toxin HipH acts as a genotoxic deoxyribonuclease, inducing DNA double strand breaks and genome instability. While the second messenger c-di-GMP functions as the antitoxin, exerting control over HipH expression and activity. The dynamic interplay between c-di-GMP and HipH levels emerges as a crucial determinant governing genome stability and persister generation within biofilms. These findings unveil a unique TA system, where small molecules act as the antitoxin, outlining a biofilm-specific molecular mechanism influencing genome stability and antibiotic persistence, with potential implications for treating biofilm infections.
Collapse
Affiliation(s)
- Hebin Liao
- The State Key Laboratory Breeding Base of Basic Science of Stomatology & Key Laboratory of Oral Biomedicine Ministry of Education, School & Hospital of Stomatology, Medical Research Institute, Wuhan UniversityWuhanChina
- Frontier Science Center for Immunology and Metabolism, Wuhan UniversityWuhanChina
- Translational Medicine Research Center, North Sichuan Medical CollegeNanchongChina
| | - Xiaodan Yan
- The State Key Laboratory Breeding Base of Basic Science of Stomatology & Key Laboratory of Oral Biomedicine Ministry of Education, School & Hospital of Stomatology, Medical Research Institute, Wuhan UniversityWuhanChina
- Frontier Science Center for Immunology and Metabolism, Wuhan UniversityWuhanChina
| | - Chenyi Wang
- The State Key Laboratory Breeding Base of Basic Science of Stomatology & Key Laboratory of Oral Biomedicine Ministry of Education, School & Hospital of Stomatology, Medical Research Institute, Wuhan UniversityWuhanChina
- Frontier Science Center for Immunology and Metabolism, Wuhan UniversityWuhanChina
| | - Chun Huang
- The State Key Laboratory Breeding Base of Basic Science of Stomatology & Key Laboratory of Oral Biomedicine Ministry of Education, School & Hospital of Stomatology, Medical Research Institute, Wuhan UniversityWuhanChina
- Frontier Science Center for Immunology and Metabolism, Wuhan UniversityWuhanChina
| | - Wei Zhang
- The State Key Laboratory Breeding Base of Basic Science of Stomatology & Key Laboratory of Oral Biomedicine Ministry of Education, School & Hospital of Stomatology, Medical Research Institute, Wuhan UniversityWuhanChina
- Frontier Science Center for Immunology and Metabolism, Wuhan UniversityWuhanChina
| | - Leyi Xiao
- The State Key Laboratory Breeding Base of Basic Science of Stomatology & Key Laboratory of Oral Biomedicine Ministry of Education, School & Hospital of Stomatology, Medical Research Institute, Wuhan UniversityWuhanChina
- Frontier Science Center for Immunology and Metabolism, Wuhan UniversityWuhanChina
| | - Jun Jiang
- Center for Life Sciences, School of Life Sciences, Yunnan UniversityKunmingChina
| | - Yongjia Bao
- Center for Life Sciences, School of Life Sciences, Yunnan UniversityKunmingChina
| | - Tao Huang
- Center for Life Sciences, School of Life Sciences, Yunnan UniversityKunmingChina
| | - Hanbo Zhang
- Center for Life Sciences, School of Life Sciences, Yunnan UniversityKunmingChina
| | - Chunming Guo
- Center for Life Sciences, School of Life Sciences, Yunnan UniversityKunmingChina
| | - Yufeng Zhang
- The State Key Laboratory Breeding Base of Basic Science of Stomatology & Key Laboratory of Oral Biomedicine Ministry of Education, School & Hospital of Stomatology, Medical Research Institute, Wuhan UniversityWuhanChina
- Frontier Science Center for Immunology and Metabolism, Wuhan UniversityWuhanChina
- Taikang Center for Life and Medical Sciences, Wuhan UniversityWuhanChina
| | - Yingying Pu
- The State Key Laboratory Breeding Base of Basic Science of Stomatology & Key Laboratory of Oral Biomedicine Ministry of Education, School & Hospital of Stomatology, Medical Research Institute, Wuhan UniversityWuhanChina
- Frontier Science Center for Immunology and Metabolism, Wuhan UniversityWuhanChina
- Department of Immunology, Hubei Province Key Laboratory of Allergy and Immunology, State Key Laboratory of Virology and Medical Research Institute, Wuhan University School of Basic Medical SciencesWuhanChina
| |
Collapse
|
9
|
Mannala GK, Rupp M, Walter N, Youf R, Bärtl S, Riool M, Alt V. Repetitive combined doses of bacteriophages and gentamicin protect against Staphylococcus aureus implant-related infections in Galleria mellonella. Bone Joint Res 2024; 13:383-391. [PMID: 39089687 PMCID: PMC11293943 DOI: 10.1302/2046-3758.138.bjr-2023-0340.r1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 08/04/2024] Open
Abstract
Aims Bacteriophages infect, replicate inside bacteria, and are released from the host through lysis. Here, we evaluate the effects of repetitive doses of the Staphylococcus aureus phage 191219 and gentamicin against haematogenous and early-stage biofilm implant-related infections in Galleria mellonella. Methods For the haematogenous infection, G. mellonella larvae were implanted with a Kirschner wire (K-wire), infected with S. aureus, and subsequently phages and/or gentamicin were administered. For the early-stage biofilm implant infection, the K-wires were pre-incubated with S. aureus suspension before implantation. After 24 hours, the larvae received phages and/or gentamicin. In both models, the larvae also received daily doses of phages and/or gentamicin for up to five days. The effect was determined by survival analysis for five days and quantitative culture of bacteria after two days of repetitive doses. Results In the haematogenous infection, a single combined dose of phages and gentamicin, and repetitive injections with gentamicin or in combination with phages, resulted in significantly improved survival rates. In the early-stage biofilm infection, only repetitive combined administration of phages and gentamicin led to a significantly increased survival. Additionally, a significant reduction in number of bacteria was observed in the larvae after receiving repetitive doses of phages and/or gentamicin in both infection models. Conclusion Based on our results, a single dose of the combination of phages and gentamicin is sufficient to prevent a haematogenous S. aureus implant-related infection, whereas gentamicin needs to be administered daily for the same effect. To treat early-stage S. aureus implant-related infection, repetitive doses of the combination of phages and gentamicin are required.
Collapse
Affiliation(s)
- Gopala K. Mannala
- Department of Trauma Surgery, University Hospital Regensburg, Regensburg, Germany
| | - Markus Rupp
- Department of Trauma Surgery, University Hospital Regensburg, Regensburg, Germany
| | - Nike Walter
- Department of Trauma Surgery, University Hospital Regensburg, Regensburg, Germany
- Department for Psychosomatic Medicine, University Hospital Regensburg, Regensburg, Germany
| | - Raphaelle Youf
- Department of Trauma Surgery, University Hospital Regensburg, Regensburg, Germany
| | - Susanne Bärtl
- Department of Trauma Surgery, University Hospital Regensburg, Regensburg, Germany
| | - Martijn Riool
- Department of Trauma Surgery, University Hospital Regensburg, Regensburg, Germany
| | - Volker Alt
- Department of Trauma Surgery, University Hospital Regensburg, Regensburg, Germany
| |
Collapse
|
10
|
Sekar A, Fan Y, Tierney P, McCanne M, Jones P, Malick F, Kannambadi D, Wannomae KK, Inverardi N, Muratoglu O, Oral E. Investigating the translational value of Periprosthetic Joint Infection (PJI) models to determine the risk and severity of Staphylococcal biofilms. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.29.591689. [PMID: 38746179 PMCID: PMC11092509 DOI: 10.1101/2024.04.29.591689] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/16/2024]
Abstract
With the advent of antibiotic-eluting polymeric materials for targeting recalcitrant infections, using preclinical models to study biofilm is crucial for improving the treatment efficacy in periprosthetic joint infections. The stratification of risk and severity of infections is needed to develop an effective clinical dosing framework with better outcomes. Here, using in-vivo and in-vitro implant-associated infection models, we demonstrate that methicillin-sensitive and resistant Staphylococcus aureus (MSSA and MRSA) have model-dependent distinct implant and peri-implant tissue colonization patterns. The maturity of biofilms and the location (implant vs tissue) were found to influence the antibiotic susceptibility evolution profiles of MSSA and MRSA and the models could capture the differing host-microbe interactions in vivo. Gene expression studies revealed the molecular heterogeneity of colonizing bacterial populations. The comparison and stratification of the risk and severity of infection across different preclinical models provided in this study can guide clinical dosing to effectively prevent or treat PJI.
Collapse
Affiliation(s)
- Amita Sekar
- Harris Orthopaedics laboratory, Massachusetts General Hospital, Boston, USA
- Department of Orthopaedic Surgery, Harvard Medical School, Boston USA
| | - Yingfang Fan
- Harris Orthopaedics laboratory, Massachusetts General Hospital, Boston, USA
- Department of Orthopaedic Surgery, Harvard Medical School, Boston USA
| | - Peyton Tierney
- Harris Orthopaedics laboratory, Massachusetts General Hospital, Boston, USA
| | - Madeline McCanne
- Harris Orthopaedics laboratory, Massachusetts General Hospital, Boston, USA
| | - Parker Jones
- Harris Orthopaedics laboratory, Massachusetts General Hospital, Boston, USA
| | - Fawaz Malick
- Harris Orthopaedics laboratory, Massachusetts General Hospital, Boston, USA
| | - Devika Kannambadi
- Harris Orthopaedics laboratory, Massachusetts General Hospital, Boston, USA
| | - Keith K Wannomae
- Harris Orthopaedics laboratory, Massachusetts General Hospital, Boston, USA
| | - Nicoletta Inverardi
- Harris Orthopaedics laboratory, Massachusetts General Hospital, Boston, USA
- Department of Orthopaedic Surgery, Harvard Medical School, Boston USA
| | - Orhun Muratoglu
- Harris Orthopaedics laboratory, Massachusetts General Hospital, Boston, USA
- Department of Orthopaedic Surgery, Harvard Medical School, Boston USA
| | - Ebru Oral
- Harris Orthopaedics laboratory, Massachusetts General Hospital, Boston, USA
- Department of Orthopaedic Surgery, Harvard Medical School, Boston USA
| |
Collapse
|
11
|
Mishra AK, Thakare RP, Santani BG, Yabaji SM, Dixit SK, Srivastava KK. Unlocking the enigma of phenotypic drug tolerance: Mechanisms and emerging therapeutic strategies. Biochimie 2024; 220:67-83. [PMID: 38168626 DOI: 10.1016/j.biochi.2023.12.009] [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: 10/11/2023] [Revised: 12/09/2023] [Accepted: 12/27/2023] [Indexed: 01/05/2024]
Abstract
In the ongoing battle against antimicrobial resistance, phenotypic drug tolerance poses a formidable challenge. This adaptive ability of microorganisms to withstand drug pressure without genetic alterations further complicating global healthcare challenges. Microbial populations employ an array of persistence mechanisms, including dormancy, biofilm formation, adaptation to intracellular environments, and the adoption of L-forms, to develop drug tolerance. Moreover, molecular mechanisms like toxin-antitoxin modules, oxidative stress responses, energy metabolism, and (p)ppGpp signaling contribute to this phenomenon. Understanding these persistence mechanisms is crucial for predicting drug efficacy, developing strategies for chronic bacterial infections, and exploring innovative therapies for refractory infections. In this comprehensive review, we dissect the intricacies of drug tolerance and persister formation, explore their role in acquired drug resistance, and highlight emerging therapeutic approaches to combat phenotypic drug tolerance. Furthermore, we outline the future landscape of interventions for persistent bacterial infections.
Collapse
Affiliation(s)
- Alok K Mishra
- Division of Microbiology, CSIR-Central Drug Research Institute (CDRI), Jankipuram Extension, Lucknow, Uttar Pradesh, 226031, India; Department of Molecular Cell and Cancer Biology, UMass Chan Medical School, Worcester, MA, 01605, USA.
| | - Ritesh P Thakare
- Division of Microbiology, CSIR-Central Drug Research Institute (CDRI), Jankipuram Extension, Lucknow, Uttar Pradesh, 226031, India; Department of Molecular Cell and Cancer Biology, UMass Chan Medical School, Worcester, MA, 01605, USA
| | - Bela G Santani
- Department of Microbiology, Sant Gadge Baba Amravati University (SGBAU), Amravati, Maharashtra, India
| | - Shivraj M Yabaji
- Division of Microbiology, CSIR-Central Drug Research Institute (CDRI), Jankipuram Extension, Lucknow, Uttar Pradesh, 226031, India; National Emerging Infectious Diseases Laboratories (NEIDL), Boston University, Boston, MA, USA
| | - Shivendra K Dixit
- Division of Medicine ICAR-Indian Veterinary Research Institute (IVRI), Izatnagar Bareilly, Uttar Pradesh, 243122, India.
| | - Kishore K Srivastava
- Division of Microbiology, CSIR-Central Drug Research Institute (CDRI), Jankipuram Extension, Lucknow, Uttar Pradesh, 226031, India.
| |
Collapse
|
12
|
Obando MC, Serra DO. Dissecting cell heterogeneities in bacterial biofilms and their implications for antibiotic tolerance. Curr Opin Microbiol 2024; 78:102450. [PMID: 38422558 DOI: 10.1016/j.mib.2024.102450] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Revised: 01/12/2024] [Accepted: 02/07/2024] [Indexed: 03/02/2024]
Abstract
Bacterial biofilms consist of large, self-formed aggregates where resident bacteria can exhibit very different physiological states and phenotypes. This heterogeneity of cell types is crucial for many structural and functional emergent properties of biofilms. Consequently, it becomes essential to understand what drives cells to differentiate and how they achieve it within the three-dimensional landscape of the biofilms. Here, we discuss recent advances in comprehending two forms of cell heterogeneity that, while recognized to coexist within biofilms, have proven challenging to distinguish. These two forms include cell heterogeneity arising as a consequence of bacteria physiologically responding to resource gradients formed across the biofilms and cell-to-cell phenotypic heterogeneity, which emerges locally within biofilm subzones among neighboring bacteria due to stochastic variations in gene expression. We describe the defining features and concepts related to both forms of cell heterogeneity and discuss their implications, with a particular focus on antibiotic tolerance.
Collapse
Affiliation(s)
- Mayra C Obando
- Laboratorio de Estructura y Fisiología de Biofilms Microbianos, Instituto de Biología Molecular y Celular de Rosario (IBR, CONICET-UNR), Predio CONICET Rosario, Ocampo y Esmeralda, 2000 Rosario, Argentina
| | - Diego O Serra
- Laboratorio de Estructura y Fisiología de Biofilms Microbianos, Instituto de Biología Molecular y Celular de Rosario (IBR, CONICET-UNR), Predio CONICET Rosario, Ocampo y Esmeralda, 2000 Rosario, Argentina.
| |
Collapse
|
13
|
Ismail AS, Berryhill BA, Gil-Gil T, Manuel JA, Smith AP, Baquero F, Levin BR. The Tradeoffs Between Persistence and Mutation Rates at Sub-Inhibitory Antibiotic Concentrations in Staphylococcus aureus. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.01.587561. [PMID: 38617265 PMCID: PMC11014548 DOI: 10.1101/2024.04.01.587561] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/16/2024]
Abstract
The rational design of the antibiotic treatment of bacterial infections employs these drugs to reach concentrations that exceed the minimum needed to prevent the replication of the target bacteria. However, within a treated patient, spatial and physiological heterogeneity promotes antibiotic gradients such that the concentration of antibiotics at specific sites is below the minimum needed to inhibit bacterial growth. Here, we investigate the effects of sub-inhibitory antibiotic concentrations on three parameters central to bacterial infection and the success of antibiotic treatment, using in vitro experiments with Staphylococcus aureus and mathematical-computer simulation models. Our results, using drugs of six different classes, demonstrate that exposure to sub-inhibitory antibiotic concentrations not only alters the dynamics of bacterial growth but also increases the mutation rate to antibiotic resistance and decreases the rate of production of persister cells thereby reducing the persistence level. Understanding this trade-off between mutation rates and persistence levels resulting from sub-inhibitory antibiotic exposure is crucial for optimizing, and mitigating the failure of, antibiotic therapy.
Collapse
Affiliation(s)
| | - Brandon A. Berryhill
- Department of Biology, Emory University, Atlanta, Georgia, USA
- Program in Microbiology and Molecular Genetics, Graduate Division of Biological and Biomedical Sciences, Laney Graduate School, Emory University, Atlanta, Georgia, USA
| | - Teresa Gil-Gil
- Department of Biology, Emory University, Atlanta, Georgia, USA
| | | | - Andrew P. Smith
- Department of Biology, Emory University, Atlanta, Georgia, USA
| | - Fernando Baquero
- Servicio de Microbiología, Hospital Universitario Ramón y Cajal, Instituto Ramón y Cajal de Investigación Sanitaria, and Centro de Investigación Médica en Red, Epidemiología y Salud Pública (CIBERESP) Madrid, Spain
| | - Bruce R. Levin
- Department of Biology, Emory University, Atlanta, Georgia, USA
- Emory Antibiotic Resistance Center, Emory University, Atlanta, Georgia, USA
| |
Collapse
|
14
|
Dombach JL, Christensen GL, Allgood SC, Quintana JLJ, Detweiler CS. Inhibition of multiple staphylococcal growth states by a small molecule that disrupts membrane fluidity and voltage. mSphere 2024; 9:e0077223. [PMID: 38445864 PMCID: PMC10964410 DOI: 10.1128/msphere.00772-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Accepted: 01/26/2024] [Indexed: 03/07/2024] Open
Abstract
New molecular approaches to disrupting bacterial infections are needed. The bacterial cell membrane is an essential structure with diverse potential lipid and protein targets for antimicrobials. While rapid lysis of the bacterial cell membrane kills bacteria, lytic compounds are generally toxic to whole animals. In contrast, compounds that subtly damage the bacterial cell membrane could disable a microbe, facilitating pathogen clearance by the immune system with limited compound toxicity. A previously described small molecule, D66, terminates Salmonella enterica serotype Typhimurium (S. Typhimurium) infection of macrophages and reduces tissue colonization in mice. The compound dissipates bacterial inner membrane voltage without rapid cell lysis under broth conditions that permeabilize the outer membrane or disable efflux pumps. In standard media, the cell envelope protects Gram-negative bacteria from D66. We evaluated the activity of D66 in Gram-positive bacteria because their distinct envelope structure, specifically the absence of an outer membrane, could facilitate mechanism of action studies. We observed that D66 inhibited Gram-positive bacterial cell growth, rapidly increased Staphylococcus aureus membrane fluidity, and disrupted membrane voltage while barrier function remained intact. The compound also prevented planktonic staphylococcus from forming biofilms and a disturbed three-dimensional structure in 1-day-old biofilms. D66 furthermore reduced the survival of staphylococcal persister cells and of intracellular S. aureus. These data indicate that staphylococcal cells in multiple growth states germane to infection are susceptible to changes in lipid packing and membrane conductivity. Thus, agents that subtly damage bacterial cell membranes could have utility in preventing or treating disease.IMPORTANCEAn underutilized potential antibacterial target is the cell membrane, which supports or associates with approximately half of bacterial proteins and has a phospholipid makeup distinct from mammalian cell membranes. Previously, an experimental small molecule, D66, was shown to subtly damage Gram-negative bacterial cell membranes and to disrupt infection of mammalian cells. Here, we show that D66 increases the fluidity of Gram-positive bacterial cell membranes, dissipates membrane voltage, and inhibits the human pathogen Staphylococcus aureus in several infection-relevant growth states. Thus, compounds that cause membrane damage without lysing cells could be useful for mitigating infections caused by S. aureus.
Collapse
Affiliation(s)
- Jamie L. Dombach
- Department of Molecular, Cellular, and Developmental Biology, University of Colorado Boulder, Boulder, Colorado, USA
| | - Grace L. Christensen
- Department of Molecular, Cellular, and Developmental Biology, University of Colorado Boulder, Boulder, Colorado, USA
| | - Samual C. Allgood
- Department of Molecular, Cellular, and Developmental Biology, University of Colorado Boulder, Boulder, Colorado, USA
| | - Joaquin L. J. Quintana
- Department of Molecular, Cellular, and Developmental Biology, University of Colorado Boulder, Boulder, Colorado, USA
| | - Corrella S. Detweiler
- Department of Molecular, Cellular, and Developmental Biology, University of Colorado Boulder, Boulder, Colorado, USA
| |
Collapse
|
15
|
Higashihira S, Simpson SJ, Morita A, Suryavanshi JR, Arnold CJ, Natoli RM, Greenfield EM. Halicin remains active against Staphylococcus aureus in biofilms grown on orthopaedically relevant substrates. Bone Joint Res 2024; 13:101-109. [PMID: 38432258 PMCID: PMC10909403 DOI: 10.1302/2046-3758.133.bjr-2023-0038.r2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 03/05/2024] Open
Abstract
Aims Biofilm infections are among the most challenging complications in orthopaedics, as bacteria within the biofilms are protected from the host immune system and many antibiotics. Halicin exhibits broad-spectrum activity against many planktonic bacteria, and previous studies have demonstrated that halicin is also effective against Staphylococcus aureus biofilms grown on polystyrene or polypropylene substrates. However, the effectiveness of many antibiotics can be substantially altered depending on which orthopaedically relevant substrates the biofilms grow. This study, therefore, evaluated the activity of halicin against less mature and more mature S. aureus biofilms grown on titanium alloy, cobalt-chrome, ultra-high molecular weight polyethylene (UHMWPE), devitalized muscle, or devitalized bone. Methods S. aureus-Xen36 biofilms were grown on the various substrates for 24 hours or seven days. Biofilms were incubated with various concentrations of halicin or vancomycin and then allowed to recover without antibiotics. Minimal biofilm eradication concentrations (MBECs) were defined by CFU counting and resazurin reduction assays, and were compared with the planktonic minimal inhibitory concentrations (MICs). Results Halicin continued to exert significantly (p < 0.01) more antibacterial activity against biofilms grown on all tested orthopaedically relevant substrates than vancomycin, an antibiotic known to be affected by biofilm maturity. For example, halicin MBECs against both less mature and more mature biofilms were ten-fold to 40-fold higher than its MIC. In contrast, vancomycin MBECs against the less mature biofilms were 50-fold to 200-fold higher than its MIC, and 100-fold to 400-fold higher against the more mature biofilms. Conclusion Halicin is a promising antibiotic that should be tested in animal models of orthopaedic infection.
Collapse
Affiliation(s)
- Shota Higashihira
- Department of Orthopaedic Surgery, Indiana University School of Medicine, Indianapolis, Indiana, USA
- Indiana Center for Musculoskeletal Health, Indiana University School of Medicine, Indianapolis, Indiana, USA
- Department of Orthopaedic Surgery, Yokohama City University, Yokohama, Japan
- Department of Orthopaedic Surgery, Yokohama City University Medical Center, Yokohama, Japan
| | - Stefanie J. Simpson
- Department of Orthopaedic Surgery, Indiana University School of Medicine, Indianapolis, Indiana, USA
- Indiana Center for Musculoskeletal Health, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Akira Morita
- Department of Orthopaedic Surgery, Indiana University School of Medicine, Indianapolis, Indiana, USA
- Indiana Center for Musculoskeletal Health, Indiana University School of Medicine, Indianapolis, Indiana, USA
- Department of Orthopaedic Surgery, Yokohama City University, Yokohama, Japan
| | - Joash R. Suryavanshi
- Department of Orthopaedic Surgery, Indiana University School of Medicine, Indianapolis, Indiana, USA
- Indiana Center for Musculoskeletal Health, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Christopher J. Arnold
- Department of Orthopaedic Surgery, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Roman M. Natoli
- Department of Orthopaedic Surgery, Indiana University School of Medicine, Indianapolis, Indiana, USA
- Indiana Center for Musculoskeletal Health, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Edward M. Greenfield
- Department of Orthopaedic Surgery, Indiana University School of Medicine, Indianapolis, Indiana, USA
- Indiana Center for Musculoskeletal Health, Indiana University School of Medicine, Indianapolis, Indiana, USA
| |
Collapse
|
16
|
Chosy MB, Sun J, Rahn HP, Liu X, Brčić J, Wender PA, Cegelski L. Vancomycin-Polyguanidino Dendrimer Conjugates Inhibit Growth of Antibiotic-Resistant Gram-Positive and Gram-Negative Bacteria and Eradicate Biofilm-Associated S. aureus. ACS Infect Dis 2024; 10:384-397. [PMID: 38252999 PMCID: PMC11646489 DOI: 10.1021/acsinfecdis.3c00168] [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] [Indexed: 01/24/2024]
Abstract
The global challenge of antibiotic resistance necessitates the introduction of more effective antibiotics. Here we report a potentially general design strategy, exemplified with vancomycin, that improves and expands antibiotic performance. Vancomycin is one of the most important antibiotics in use today for the treatment of Gram-positive infections. However, it fails to eradicate difficult-to-treat biofilm populations. Vancomycin is also ineffective in killing Gram-negative bacteria due to its inability to breach the outer membrane. Inspired by our seminal studies on cell penetrating guanidinium-rich transporters (e.g., octaarginine), we recently introduced vancomycin conjugates that effectively eradicate Gram-positive biofilm bacteria, persister cells and vancomycin-resistant enterococci (with V-r8, vancomycin-octaarginine), and Gram-negative pathogens (with V-R, vancomycin-arginine). Having shown previously that the spatial array (linear versus dendrimeric) of multiple guanidinium groups affects cell permeation, we report here for the first time vancomycin conjugates with dendrimerically displayed guanidinium groups that exhibit superior efficacy and breadth, presenting the best activity of V-r8 and V-R in single broad-spectrum compounds active against ESKAPE pathogens. Mode-of-action studies reveal cell-surface activity and enhanced vancomycin-like killing. The vancomycin-polyguanidino dendrimer conjugates exhibit no acute mammalian cell toxicity or hemolytic activity. Our study introduces a new class of broad-spectrum vancomycin derivatives and a general strategy to improve or expand antibiotic performance through combined mode-of-action and function-oriented design studies.
Collapse
Affiliation(s)
- Madeline B. Chosy
- Department of Chemistry, Stanford University, Stanford, CA 94305, USA
| | - Jiuzhi Sun
- Department of Chemistry, Stanford University, Stanford, CA 94305, USA
| | - Harrison P. Rahn
- Department of Chemistry, Stanford University, Stanford, CA 94305, USA
| | - Xinyu Liu
- Department of Chemistry, Stanford University, Stanford, CA 94305, USA
| | - Jasna Brčić
- Department of Chemistry, Stanford University, Stanford, CA 94305, USA
| | - Paul A. Wender
- Department of Chemistry, Stanford University, Stanford, CA 94305, USA
- Department of Chemical and Systems Biology, Stanford University, Stanford, CA 94305, USA
| | - Lynette Cegelski
- Department of Chemistry, Stanford University, Stanford, CA 94305, USA
| |
Collapse
|
17
|
Okolo EA, de Alencar ER, Machado SG, Faroni LRD, Silva MVDA, Costa NADS, Falqueto A. Ozonation for Pseudomonas paracarnis control: biofilm removal and preservation of chicken meat during refrigerated storage. Braz J Microbiol 2023; 54:3051-3060. [PMID: 37910305 PMCID: PMC10689663 DOI: 10.1007/s42770-023-01157-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2023] [Accepted: 10/16/2023] [Indexed: 11/03/2023] Open
Abstract
Ozone has been studied to control microorganisms in food, as well as to control biofilm. In this context, the goals of this work were to determine the effect of ozonated water in the removal of Pseudomonas paracarnis biofilm and the effect of ozone gas and ozonated water on inactivating P. paracarnis in deboned chicken breast meat and its effect on product color. AISI 304 coupons were used as a surface for biofilm formation. The coupons were immerged into minimal medium for Pseudomonas inoculated with the P. paracarnis overnight culture (1% w/v) followed by incubation at 25 °C for 7 days. To obtain ozonized water, two different systems were used: system with microbubble generator (MB) and system with porous stone diffuser (PSD). The inlet ozone concentration was 19 mg/L and flow rate of 1 L/min. The coupons were subjected to ozonized water for 10 and 20 min. The chicken breast meat was exposed to gaseous ozone and ozonized water for 40 min. After the ozonation process, chicken meat samples were stored at 8 °C, for 5 days. More expressive removals of biofilm were obtained when using ozonized water obtained in the system with microbubble generator (MB for 20 min-reduction of 2.3 log cycles) and system with porous stone diffuser (PSD for 10 min-reduction of 2.7 log cycles; PSD for 20 min-reduction of 2.6 log cycles). The treatment of chicken meat with ozone gas resulted in lower counting of Pseudomonas, when compared with the control treatments and with ozonized water, both immediately after ozonation (day 1) and after 5 days of storage. The luminosity in the chicken meat samples treated with ozonized water was higher than that verified in the control treatments and with ozone gas, immediately after ozonation (day 1). A similar trend was observed in hue angle and color difference, in which the highest values were obtained for treatment with ozonized water. Based on the results obtained in this study, it was concluded that ozonated water can be used to remove P. paracarnis biofilm from stainless steel under static conditions and gaseous ozone is more efficient in the inactivation of P. paracarnis from chicken breast meat, when compared to ozonated water.
Collapse
Affiliation(s)
- Ejima Akogwu Okolo
- Department of Agricultural Engineering, Universidade Federal de Viçosa, Viçosa, MG, 36570900, Brazil
- Agricultural and Bio-Environmental Engineering Department, Federal Polytechnic Nekede, Imo State, P.M.B. 1036, Owerri, Nigeria
| | | | - Solimar Goncalves Machado
- Department of Food Technology, INOVALEITE, Universidade Federal de Viçosa, Viçosa, MG, 36570900, Brazil
| | | | | | | | - Andressa Falqueto
- Department of Food Technology, INOVALEITE, Universidade Federal de Viçosa, Viçosa, MG, 36570900, Brazil
| |
Collapse
|
18
|
Francis D, Veeramanickathadathil Hari G, Koonthanmala Subash A, Bhairaddy A, Joy A. The biofilm proteome of Staphylococcus aureus and its implications for therapeutic interventions to biofilm-associated infections. ADVANCES IN PROTEIN CHEMISTRY AND STRUCTURAL BIOLOGY 2023; 138:327-400. [PMID: 38220430 DOI: 10.1016/bs.apcsb.2023.08.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/16/2024]
Abstract
Staphylococcus aureus is a major healthcare concern due to its ability to inflict life-threatening infections and evolve antibiotic resistance at an alarming pace. It is frequently associated with hospital-acquired infections, especially device-associated infections. Systemic infections due to S. aureus are difficult to treat and are associated with significant mortality and morbidity. The situation is worsened by the ability of S. aureus to form social associations called biofilms. Biofilms embed a community of cells with the ability to communicate with each other and share resources within a polysaccharide or protein matrix. S. aureus establish biofilms on tissues and conditioned abiotic surfaces. Biofilms are hyper-tolerant to antibiotics and help evade host immune responses. Biofilms exacerbate the severity and recalcitrance of device-associated infections. The development of a biofilm involves various biomolecules, such as polysaccharides, proteins and nucleic acids, contributing to different structural and functional roles. Interconnected signaling pathways and regulatory molecules modulate the expression of these molecules. A comprehensive understanding of the molecular biology of biofilm development would help to devise effective anti-biofilm therapeutics. Although bactericidal agents, antimicrobial peptides, bacteriophages and nano-conjugated anti-biofilm agents have been employed with varying levels of success, there is still a requirement for effective and clinically viable anti-biofilm therapeutics. Proteins that are expressed and utilized during biofilm formation, constituting the biofilm proteome, are a particularly attractive target for anti-biofilm strategies. The proteome can be explored to identify potential anti-biofilm drug targets and utilized for rational drug discovery. With the aim of uncovering the biofilm proteome, this chapter explores the mechanism of biofilm formation and its regulation. Furthermore, it explores the antibiofilm therapeutics targeted against the biofilm proteome.
Collapse
Affiliation(s)
- Dileep Francis
- Department of Life Sciences, Kristu Jayanti College (Autonomous), Bengaluru, India.
| | | | | | - Anusha Bhairaddy
- Department of Life Sciences, Kristu Jayanti College (Autonomous), Bengaluru, India
| | - Atheene Joy
- Department of Life Sciences, Kristu Jayanti College (Autonomous), Bengaluru, India
| |
Collapse
|
19
|
Jia J, Zheng M, Zhang C, Li B, Lu C, Bai Y, Tong Q, Hang X, Ge Y, Zeng L, Zhao M, Song F, Zhang H, Zhang L, Hong K, Bi H. Killing of Staphylococcus aureus persisters by a multitarget natural product chrysomycin A. SCIENCE ADVANCES 2023; 9:eadg5995. [PMID: 37540745 PMCID: PMC10403215 DOI: 10.1126/sciadv.adg5995] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Accepted: 07/05/2023] [Indexed: 08/06/2023]
Abstract
Staphylococcus aureus poses a severe public health problem as one of the vital causative agents of healthcare- and community-acquired infections. There is a globally urgent need for new drugs with a novel mode of action (MoA) to combat S. aureus biofilms and persisters that tolerate antibiotic treatment. We demonstrate that a benzonaphthopyranone glycoside, chrysomycin A (ChryA), is a rapid bactericide that is highly active against S. aureus persisters, robustly eradicates biofilms in vitro, and shows a sustainable killing efficacy in vivo. ChryA was suggested to target multiple critical cellular processes. A wide range of genetic and biochemical approaches showed that ChryA directly binds to GlmU and DapD, involved in the biosynthetic pathways for the cell wall peptidoglycan and lysine precursors, respectively, and inhibits the acetyltransferase activities by competition with their mutual substrate acetyl-CoA. Our study provides an effective antimicrobial strategy combining multiple MoAs onto a single small molecule for treatments of S. aureus persistent infections.
Collapse
Affiliation(s)
- Jia Jia
- Department of Pathogen Biology, Jiangsu Key Laboratory of Pathogen Biology, Nanjing Medical University, Nanjing 211166, China
| | - Mingxin Zheng
- Department of Pathogen Biology, Jiangsu Key Laboratory of Pathogen Biology, Nanjing Medical University, Nanjing 211166, China
| | - Chongwen Zhang
- Department of Pathogen Biology, Jiangsu Key Laboratory of Pathogen Biology, Nanjing Medical University, Nanjing 211166, China
| | - Binglei Li
- Department of Pathogen Biology, Jiangsu Key Laboratory of Pathogen Biology, Nanjing Medical University, Nanjing 211166, China
| | - Cai Lu
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Yuefan Bai
- Department of Pathogen Biology, Jiangsu Key Laboratory of Pathogen Biology, Nanjing Medical University, Nanjing 211166, China
| | - Qian Tong
- Department of Pathogen Biology, Jiangsu Key Laboratory of Pathogen Biology, Nanjing Medical University, Nanjing 211166, China
| | - Xudong Hang
- Department of Pathogen Biology, Jiangsu Key Laboratory of Pathogen Biology, Nanjing Medical University, Nanjing 211166, China
| | - Yixin Ge
- Department of Pathogen Biology, Jiangsu Key Laboratory of Pathogen Biology, Nanjing Medical University, Nanjing 211166, China
| | - Liping Zeng
- Department of Pathogen Biology, Jiangsu Key Laboratory of Pathogen Biology, Nanjing Medical University, Nanjing 211166, China
| | - Ming Zhao
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Fuhang Song
- School of Light Industry, Beijing Technology and Business University, Beijing 100048, China
| | - Huawei Zhang
- School of Pharmaceutical Sciences, Zhejiang University of Technology, Hangzhou 310014, China
| | - Liang Zhang
- Department of Pharmacology and Chemical Biology, State Key Laboratory of Oncogenes and Related Genes, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Kui Hong
- Key Laboratory of Combinatorial Biosynthesis and Drug Discovery, Ministry of Education and School of Pharmaceutical Sciences, Wuhan University, Wuhan 430071, China
| | - Hongkai Bi
- Department of Pathogen Biology, Jiangsu Key Laboratory of Pathogen Biology, Nanjing Medical University, Nanjing 211166, China
| |
Collapse
|
20
|
Tran NN, Morrisette T, Jorgensen SCJ, Orench-Benvenutti JM, Kebriaei R. Current therapies and challenges for the treatment of Staphylococcus aureus biofilm-related infections. Pharmacotherapy 2023; 43:816-832. [PMID: 37133439 DOI: 10.1002/phar.2806] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Revised: 03/10/2023] [Accepted: 03/16/2023] [Indexed: 05/04/2023]
Abstract
Staphylococcus aureus is a major cause of nosocomial and community-acquired infections and contributes to significant increase in morbidity and mortality especially when associated with medical devices and in biofilm form. Biofilm structure provides a pathway for the enrichment of resistant and persistent phenotypes of S. aureus leading to relapse and recurrence of infection. Minimal diffusion of antibiotics inside biofilm structure leads to heterogeneity and distinct physiological activity. Additionally, horizontal gene transfer between cells in proximity adds to the challenges associated with eradication of biofilms. This narrative review focuses on biofilm-associated infections caused by S. aureus, the impact of environmental conditions on biofilm formation, interactions inside biofilm communities, and the clinical challenges that they present. Conclusively, potential solutions, novel treatment strategies, combination therapies, and reported alternatives are discussed.
Collapse
Affiliation(s)
- Nikki N Tran
- Department of Pharmacy, The Ohio State University Wexner Medical Center - The James Cancer Hospital and Solove Research Institute, Columbus, Ohio, USA
| | - Taylor Morrisette
- Department of Clinical Pharmacy and Outcomes Sciences, Medical University of South Carolina College of Pharmacy, Charleston, South Carolina, USA
- Department of Pharmacy Services, Medical University of South Carolina Shawn Jenkins Children's Hospital, Charleston, South Carolina, USA
| | - Sarah C J Jorgensen
- Institute of Medical Science, Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - José M Orench-Benvenutti
- P3 Research Laboratory, Division of Outcomes and Translational Sciences, College of Pharmacy, The Ohio State University, Columbus, Ohio, USA
| | - Razieh Kebriaei
- P3 Research Laboratory, Division of Outcomes and Translational Sciences, College of Pharmacy, The Ohio State University, Columbus, Ohio, USA
| |
Collapse
|
21
|
Ganesan N, Mishra B, Felix L, Mylonakis E. Antimicrobial Peptides and Small Molecules Targeting the Cell Membrane of Staphylococcus aureus. Microbiol Mol Biol Rev 2023; 87:e0003722. [PMID: 37129495 PMCID: PMC10304793 DOI: 10.1128/mmbr.00037-22] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/03/2023] Open
Abstract
Clinical management of Staphylococcus aureus infections presents a challenge due to the high incidence, considerable virulence, and emergence of drug resistance mechanisms. The treatment of drug-resistant strains, such as methicillin-resistant S. aureus (MRSA), is further complicated by the development of tolerance and persistence to antimicrobial agents in clinical use. To address these challenges, membrane disruptors, that are not generally considered during drug discovery for agents against S. aureus, should be explored. The cell membrane protects S. aureus from external stresses and antimicrobial agents, but membrane-targeting antimicrobial agents are probably less likely to promote bacterial resistance. Nontypical linear cationic antimicrobial peptides (AMPs), highly modified AMPs such as daptomycin (lipopeptide), bacitracin (cyclic peptide), and gramicidin S (cyclic peptide), are currently in clinical use. Recent studies have demonstrated that AMPs and small molecules can penetrate the cell membrane of S. aureus, inhibit phospholipid biosynthesis, or block the passage of solutes between the periplasm and the exterior of the cell. In addition to their primary mechanism of action (MOA) that targets the bacterial membrane, AMPs and small molecules may also impact bacteria through secondary mechanisms such as targeting the biofilm, and downregulating virulence genes of S. aureus. In this review, we discuss the current state of research into cell membrane-targeting AMPs and small molecules and their potential mechanisms of action against drug-resistant physiological forms of S. aureus, including persister cells and biofilms.
Collapse
Affiliation(s)
- Narchonai Ganesan
- Infectious Diseases Division, Alpert Medical School, Brown University, Providence, Rhode Island, USA
| | - Biswajit Mishra
- Infectious Diseases Division, Alpert Medical School, Brown University, Providence, Rhode Island, USA
- Department of Medicine, The Miriam Hospital, Providence, Rhode Island, USA
| | - LewisOscar Felix
- Infectious Diseases Division, Alpert Medical School, Brown University, Providence, Rhode Island, USA
| | - Eleftherios Mylonakis
- Infectious Diseases Division, Alpert Medical School, Brown University, Providence, Rhode Island, USA
- Department of Medicine, Houston Methodist Hospital, Houston, Texas, USA
| |
Collapse
|
22
|
Patel H, Rawat S. A genetic regulatory see-saw of biofilm and virulence in MRSA pathogenesis. Front Microbiol 2023; 14:1204428. [PMID: 37434702 PMCID: PMC10332168 DOI: 10.3389/fmicb.2023.1204428] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Accepted: 05/30/2023] [Indexed: 07/13/2023] Open
Abstract
Staphylococcus aureus is one of the most common opportunistic human pathogens causing several infectious diseases. Ever since the emergence of the first methicillin-resistant Staphylococcus aureus (MRSA) strain decades back, the organism has been a major cause of hospital-acquired infections (HA-MRSA). The spread of this pathogen across the community led to the emergence of a more virulent subtype of the strain, i.e., Community acquired Methicillin resistant Staphylococcus aureus (CA-MRSA). Hence, WHO has declared Staphylococcus aureus as a high-priority pathogen. MRSA pathogenesis is remarkable because of the ability of this "superbug" to form robust biofilm both in vivo and in vitro by the formation of polysaccharide intercellular adhesin (PIA), extracellular DNA (eDNA), wall teichoic acids (WTAs), and capsule (CP), which are major components that impart stability to a biofilm. On the other hand, secretion of a diverse array of virulence factors such as hemolysins, leukotoxins, enterotoxins, and Protein A regulated by agr and sae two-component systems (TCS) aids in combating host immune response. The up- and downregulation of adhesion genes involved in biofilm formation and genes responsible for synthesizing virulence factors during different stages of infection act as a genetic regulatory see-saw in the pathogenesis of MRSA. This review provides insight into the evolution and pathogenesis of MRSA infections with a focus on genetic regulation of biofilm formation and virulence factors secretion.
Collapse
Affiliation(s)
| | - Seema Rawat
- Microbiology Laboratory, School of Life Sciences, Central University of Gujarat, Gandhinagar, Gujarat, India
| |
Collapse
|
23
|
Papadopoulou V, Sidders AE, Lu KY, Velez AZ, Durham PG, Bui DT, Angeles-Solano M, Dayton PA, Rowe SE. Overcoming biological barriers to improve treatment of a Staphylococcus aureus wound infection. Cell Chem Biol 2023; 30:513-526.e5. [PMID: 37148883 PMCID: PMC10198964 DOI: 10.1016/j.chembiol.2023.04.009] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Revised: 01/19/2023] [Accepted: 04/17/2023] [Indexed: 05/08/2023]
Abstract
Chronic wounds frequently become infected with bacterial biofilms which respond poorly to antibiotic therapy. Aminoglycoside antibiotics are ineffective at treating deep-seated wound infections due to poor drug penetration, poor drug uptake into persister cells, and widespread antibiotic resistance. In this study, we combat the two major barriers to successful aminoglycoside treatment against a biofilm-infected wound: limited antibiotic uptake and limited biofilm penetration. To combat the limited antibiotic uptake, we employ palmitoleic acid, a host-produced monounsaturated fatty acid that perturbs the membrane of gram-positive pathogens and induces gentamicin uptake. This novel drug combination overcomes gentamicin tolerance and resistance in multiple gram-positive wound pathogens. To combat biofilm penetration, we examined the ability of sonobactericide, a non-invasive ultrasound-mediated-drug delivery technology to improve antibiotic efficacy using an in vivo biofilm model. This dual approach dramatically improved antibiotic efficacy against a methicillin-resistant Staphylococcus aureus (MRSA) wound infection in diabetic mice.
Collapse
Affiliation(s)
- Virginie Papadopoulou
- Joint Department of Biomedical Engineering, The University of North Carolina and North Carolina State University, Chapel Hill, NC 27599, USA.
| | - Ashelyn E Sidders
- Department of Microbiology and Immunology, University of North Carolina-Chapel Hill, Chapel Hill, NC 27599, USA
| | - Kuan-Yi Lu
- Department of Microbiology and Immunology, University of North Carolina-Chapel Hill, Chapel Hill, NC 27599, USA
| | - Amanda Z Velez
- Department of Microbiology and Immunology, University of North Carolina-Chapel Hill, Chapel Hill, NC 27599, USA
| | - Phillip G Durham
- Joint Department of Biomedical Engineering, The University of North Carolina and North Carolina State University, Chapel Hill, NC 27599, USA; Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Duyen T Bui
- Department of Microbiology and Immunology, University of North Carolina-Chapel Hill, Chapel Hill, NC 27599, USA
| | - Michelle Angeles-Solano
- Department of Microbiology and Immunology, University of North Carolina-Chapel Hill, Chapel Hill, NC 27599, USA
| | - Paul A Dayton
- Joint Department of Biomedical Engineering, The University of North Carolina and North Carolina State University, Chapel Hill, NC 27599, USA; Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Sarah E Rowe
- Department of Microbiology and Immunology, University of North Carolina-Chapel Hill, Chapel Hill, NC 27599, USA.
| |
Collapse
|
24
|
Theis TJ, Daubert TA, Kluthe KE, Brodd KL, Nuxoll AS. Staphylococcus aureus persisters are associated with reduced clearance in a catheter-associated biofilm infection. Front Cell Infect Microbiol 2023; 13:1178526. [PMID: 37228667 PMCID: PMC10203555 DOI: 10.3389/fcimb.2023.1178526] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Accepted: 04/24/2023] [Indexed: 05/27/2023] Open
Abstract
Background Staphylococcus aureus causes a wide variety of infections, many of which are chronic or relapsing in nature. Antibiotic therapy is often ineffective against S. aureus biofilm-mediated infections. Biofilms are difficult to treat partly due to their tolerance to antibiotics, however the underlying mechanism responsible for this remains unknown. One possible explanation is the presence of persister cells-dormant-like cells that exhibit tolerance to antibiotics. Recent studies have shown a connection between a fumC (fumarase C, a gene in the tricarboxylic acid cycle) knockout strain and increased survival to antibiotics, antimicrobial peptides, and in a Drosophila melanogaster model. Objective It remained unclear whether a S. aureus high persister strain would have a survival advantage in the presence of innate and adaptive immunity. To further investigate this, a fumC knockout and wild type strains were examined in a murine catheter-associated biofilm model. Results Interestingly, mice struggled to clear both S. aureus wild type and the fumC knockout strains. We reasoned both biofilm-mediated infections predominantly consisted of persister cells. To determine the persister cell population within biofilms, expression of a persister cell marker (Pcap5A::dsRED) in a biofilm was examined. Cell sorting of biofilms challenged with antibiotics revealed cells with intermediate and high expression of cap5A had 5.9-and 4.5-fold higher percent survival compared to cells with low cap5A expression. Based on previous findings that persisters are associated with reduced membrane potential, flow cytometry analysis was used to examine the metabolic state of cells within a biofilm. We confirmed cells within biofilms had reduced membrane potential compared to both stationary phase cultures (2.5-fold) and exponential phase cultures (22.4-fold). Supporting these findings, cells within a biofilm still exhibited tolerance to antibiotic challenge following dispersal of the matrix through proteinase K. Conclusion Collectively, these data show that biofilms are largely comprised of persister cells, and this may explain why biofilm infections are often chronic and/or relapsing in clinical settings.
Collapse
|
25
|
Thornton CS, Parkins MD. Microbial Epidemiology of the Cystic Fibrosis Airways: Past, Present, and Future. Semin Respir Crit Care Med 2023; 44:269-286. [PMID: 36623820 DOI: 10.1055/s-0042-1758732] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Progressive obstructive lung disease secondary to chronic airway infection, coupled with impaired host immunity, is the leading cause of morbidity and mortality in cystic fibrosis (CF). Classical pathogens found in the airways of persons with CF (pwCF) include Pseudomonas aeruginosa, Staphylococcus aureus, the Burkholderia cepacia complex, Achromobacter species, and Haemophilus influenzae. While traditional respiratory-tract surveillance culturing has focused on this limited range of pathogens, the use of both comprehensive culture and culture-independent molecular approaches have demonstrated complex highly personalized microbial communities. Loss of bacterial community diversity and richness, counteracted with relative increases in dominant taxa by traditional CF pathogens such as Burkholderia or Pseudomonas, have long been considered the hallmark of disease progression. Acquisition of these classic pathogens is viewed as a harbinger of advanced disease and postulated to be driven in part by recurrent and frequent antibiotic exposure driven by frequent acute pulmonary exacerbations. Recently, CF transmembrane conductance regulator (CFTR) modulators, small molecules designed to potentiate or restore diminished protein levels/function, have been successfully developed and have profoundly influenced disease course. Despite the multitude of clinical benefits, structural lung damage and consequent chronic airway infection persist in pwCF. In this article, we review the microbial epidemiology of pwCF, focus on our evolving understanding of these infections in the era of modulators, and identify future challenges in infection surveillance and clinical management.
Collapse
Affiliation(s)
- Christina S Thornton
- Department of Medicine, Cumming School of Medicine, University of Calgary, Alberta, Canada
| | - Michael D Parkins
- Department of Medicine, Cumming School of Medicine, University of Calgary, Alberta, Canada.,Department of Microbiology, Immunology and Infectious Diseases, University of Calgary, Alberta, Canada
| |
Collapse
|
26
|
Wang L, Wang H, Zhang H, Wu H. Formation of a biofilm matrix network shapes polymicrobial interactions. THE ISME JOURNAL 2023; 17:467-477. [PMID: 36639539 PMCID: PMC9938193 DOI: 10.1038/s41396-023-01362-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Revised: 12/30/2022] [Accepted: 01/09/2023] [Indexed: 01/15/2023]
Abstract
Staphylococcus aureus colonizes the same ecological niche as many commensals. However, little is known about how such commensals modulate staphylococcal fitness and persistence. Here we report a new mechanism that mediates dynamic interactions between a commensal streptococcus and S. aureus. Commensal Streptococcus parasanguinis significantly increased the staphylococcal biofilm formation in vitro and enhanced its colonization in vivo. A streptococcal biofilm-associated protein BapA1, not fimbriae-associated protein Fap1, is essential for dual-species biofilm formation. On the other side, three staphylococcal virulence determinants responsible for the BapA1-dependent dual-species biofilm formation were identified by screening a staphylococcal transposon mutant library. The corresponding staphylococcal mutants lacked binding to recombinant BapA1 (rBapA1) due to lower amounts of eDNA in their culture supernatants and were defective in biofilm formation with streptococcus. The rBapA1 selectively colocalized with eDNA within the dual-species biofilm and bound to eDNA in vitro, highlighting the contributions of the biofilm matrix formed between streptococcal BapA1 and staphylococcal eDNA to dual-species biofilm formation. These findings have revealed an additional new mechanism through which an interspecies biofilm matrix network mediates polymicrobial interactions.
Collapse
Affiliation(s)
- Lijun Wang
- Departments of Pediatric Dentistry and Microbiology, University of Alabama at Birmingham Schools of Dentistry and Medicine, Birmingham, Alabama, 35294, USA
- Department of Laboratory Medicine, Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua University, 102218, Beijing, China
| | - Hongxia Wang
- Departments of Pediatric Dentistry and Microbiology, University of Alabama at Birmingham Schools of Dentistry and Medicine, Birmingham, Alabama, 35294, USA
| | - Hua Zhang
- Departments of Pediatric Dentistry and Microbiology, University of Alabama at Birmingham Schools of Dentistry and Medicine, Birmingham, Alabama, 35294, USA
- Department of Integrative Biomedical and Diagnostic Sciences, Oregon Health and Science University School of Dentistry, Portland, OR, 97239, USA
| | - Hui Wu
- Departments of Pediatric Dentistry and Microbiology, University of Alabama at Birmingham Schools of Dentistry and Medicine, Birmingham, Alabama, 35294, USA.
- Department of Integrative Biomedical and Diagnostic Sciences, Oregon Health and Science University School of Dentistry, Portland, OR, 97239, USA.
| |
Collapse
|
27
|
Soliman MKY, Salem SS, Abu-Elghait M, Azab MS. Biosynthesis of Silver and Gold Nanoparticles and Their Efficacy Towards Antibacterial, Antibiofilm, Cytotoxicity, and Antioxidant Activities. Appl Biochem Biotechnol 2023; 195:1158-1183. [PMID: 36342621 PMCID: PMC9852169 DOI: 10.1007/s12010-022-04199-7] [Citation(s) in RCA: 56] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/21/2022] [Indexed: 11/09/2022]
Abstract
The World Health Organization (WHO) reports that the emergence of multidrug-resistant and the slow advent of novel and more potent antitumor and antimicrobial chemotherapeutics continue to be of the highest concern for human health. Additionally, the stability, low solubility, and negative effects of existing drugs make them ineffective. Studies into alternative tactics to tackle such tenacious diseases was sparked by anticancer and antibacterial. Silver (Ag) and gold (Au) nanoparticles (NPs) were created from Trichoderma saturnisporum, the much more productive fungal strain. Functional fungal extracellular enzymes and proteins carried out the activities of synthesis and capping of the generated nano-metals. Characterization was done on the obtained Ag-NPs and Au-NPs through UV-vis, FTIR, XRD, TEM, and SEM. Additionally, versus methicillin-sensitive Staphylococcus aureus (MSSA) and methicillin-resistant Staphylococcus aureus (MRSA), Pseudomonas aeruginosa, and Klebsiella pneumoniae, the antibacterial activities of Ag-NPs and Au-NPs were assessed. In particular, the Ag-NPs were more effective against pathogenic bacteria than Au-NPs. Furthermore, antibiofilm study that shown Au-NPs had activity more than Ag-NPs. Interestingly, applying the DPPH procedure, these noble metallic NPs had antioxidant activity, in which the IC50 for Ag-NPs and Au-NPs was 73.5 μg/mL and 190.0 μg/mL, respectively. According to the cytotoxicity evaluation results, the alteration in the cells was shown as loss of their typical shape, partial or complete loss of monolayer, granulation, shrinking, or cell rounding with IC50 for normal Vero cell were 693.68 μg/mL and 661.24 μg/mL, for Ag-NPs and Au-NPs, respectively. While IC50 for cancer cell (Mcf7) was 370.56 μg/mL and 394.79 μg/mL for Ag-NPs and Au-NPs, respectively. Ag-NPs and Au-NPs produced via green synthesis have the potential to be employed in the medical industry as beneficial nanocompounds.
Collapse
Affiliation(s)
- Mohamed K Y Soliman
- Botany and Microbiology Department, Faculty of Science, Al-Azhar University, 11884, Nasr City, Cairo, Egypt
| | - Salem S Salem
- Botany and Microbiology Department, Faculty of Science, Al-Azhar University, 11884, Nasr City, Cairo, Egypt.
| | - Mohammed Abu-Elghait
- Botany and Microbiology Department, Faculty of Science, Al-Azhar University, 11884, Nasr City, Cairo, Egypt
| | - Mohamed Salah Azab
- Botany and Microbiology Department, Faculty of Science, Al-Azhar University, 11884, Nasr City, Cairo, Egypt
| |
Collapse
|
28
|
Zhu X, Hong A, Sun X, Wang W, He G, Luo H, Wu Z, Xu Q, Hu Z, Wu X, Huang D, Li L, Zhao X, Deng X. Nigericin is effective against multidrug resistant gram-positive bacteria, persisters, and biofilms. Front Cell Infect Microbiol 2022; 12:1055929. [PMID: 36605124 PMCID: PMC9807916 DOI: 10.3389/fcimb.2022.1055929] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Accepted: 12/05/2022] [Indexed: 12/24/2022] Open
Abstract
Multidrug-resistant (MDR) bacteria pose a significant clinical threat to human health, but the development of antibiotics cannot meet the urgent need for effective agents, especially those that can kill persisters and biofilms. Here, we reported that nigericin showed potent bactericidal activity against various clinical MDR Gram-positive bacteria, persisters and biofilms, with low frequencies of resistance development. Moreover, nigericin exhibited favorable in vivo efficacy in deep-seated mouse biofilm, murine skin and bloodstream infection models. With Staphylococcus aureus, nigericin disrupted ATP production and electron transport chain; cell death was associated with altered membrane structure and permeability. Obtaining nigericin-resistant/tolerant mutants required multiple rounds of challenge, and, cross-resistance to members of several antimicrobial classes was absent, probably due to distinct nigericin action with the GraSR two-component regulatory system. Thus, our work reveals that nigericin is a promising antibiotic candidate for the treatment of chronic or recurrent infections caused by Gram-positive bacteria.
Collapse
Affiliation(s)
- Xiaoli Zhu
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, Fujian, China
- State-province Joint Engineering Laboratory of Targeted Drugs from Natural Products, Xiamen University, Xiamen, Fujian, China
| | - Anjin Hong
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, Fujian, China
- State-province Joint Engineering Laboratory of Targeted Drugs from Natural Products, Xiamen University, Xiamen, Fujian, China
| | - Xihuan Sun
- Clinical Microbiology Laboratory, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian, China
| | - Weijie Wang
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, School of Public Health, Xiamen University, Xiamen, Fujian, China
| | - Guanghui He
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, Fujian, China
- State-province Joint Engineering Laboratory of Targeted Drugs from Natural Products, Xiamen University, Xiamen, Fujian, China
| | - Huan Luo
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, School of Public Health, Xiamen University, Xiamen, Fujian, China
| | - Zhenhua Wu
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, Fujian, China
- State-province Joint Engineering Laboratory of Targeted Drugs from Natural Products, Xiamen University, Xiamen, Fujian, China
| | - Qingyan Xu
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, Fujian, China
- State-province Joint Engineering Laboratory of Targeted Drugs from Natural Products, Xiamen University, Xiamen, Fujian, China
| | - Zhiyu Hu
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, Fujian, China
- State-province Joint Engineering Laboratory of Targeted Drugs from Natural Products, Xiamen University, Xiamen, Fujian, China
| | - Xiaobing Wu
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, Fujian, China
- State-province Joint Engineering Laboratory of Targeted Drugs from Natural Products, Xiamen University, Xiamen, Fujian, China
| | - Donghong Huang
- Clinical Microbiology Laboratory, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian, China
| | - Li Li
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, Fujian, China
- State-province Joint Engineering Laboratory of Targeted Drugs from Natural Products, Xiamen University, Xiamen, Fujian, China
| | - Xilin Zhao
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, School of Public Health, Xiamen University, Xiamen, Fujian, China
| | - Xianming Deng
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, Fujian, China
- State-province Joint Engineering Laboratory of Targeted Drugs from Natural Products, Xiamen University, Xiamen, Fujian, China
| |
Collapse
|
29
|
González JF, Hitt R, Laipply B, Gunn JS. The Effect of the Gallbladder Environment during Chronic Infection on Salmonella Persister Cell Formation. Microorganisms 2022; 10:microorganisms10112276. [PMID: 36422346 PMCID: PMC9698170 DOI: 10.3390/microorganisms10112276] [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: 09/30/2022] [Revised: 11/07/2022] [Accepted: 11/12/2022] [Indexed: 11/18/2022] Open
Abstract
Typhoid fever is caused by Salmonella enterica serovar Typhi (S. Typhi). Around 3-5% of individuals infected become chronic carriers, with the gallbladder (GB) as the predominant site of persistence. Gallstones (GS) aid in the development and maintenance of GB carriage, serving as a substrate to which Salmonellae attach and form a biofilm. This biofilm matrix protects bacteria from the host immune system and environmental stress. This shielded environment is an ideal place for the development of persister cells, a transient phenotype of a subset of cells within a population that allows survival after antibiotic treatment. Persisters can also arise in response to harsh environments such as the GB. Here we investigate if GB conditions affect the number of persisters in a Salmonella population. To simulate the chronic GB environment, we cultured biofilms in cholesterol-coated 96-well plates in the presence of ox or human bile. We then treated planktonic or biofilm Salmonella cultures with high concentrations of different antibiotics. This study suggests that biofilms provide a niche for persister cells, but GB conditions either play no role or have a negative influence on persister formation, especially after kanamycin treatment. The antibiotic target was important, as antimicrobials directed against DNA replication or the cell wall had no effect on persister cell formation. Interestingly, repeated treatment with ciprofloxacin increased the percentage of S. Typhimurium persisters in a biofilm, but this increase was abolished by GB conditions. On the other hand, repeated ciprofloxacin treatment of S. Typhi biofilms in GB conditions slightly increased the fraction of persisters. Thus, while the harsh conditions in the GB would be thought to give rise to increased persisters, therefore contributing to the development of chronic carriage, these data suggest persister cell formation is dampened in this environment.
Collapse
Affiliation(s)
- Juan F. González
- Center for Microbial Pathogenesis, Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus, OH 43215, USA
- Infectious Diseases Institute, The Ohio State University, Columbus, OH 43210, USA
| | - Regan Hitt
- Center for Microbial Pathogenesis, Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus, OH 43215, USA
- Infectious Diseases Institute, The Ohio State University, Columbus, OH 43210, USA
| | - Baileigh Laipply
- Center for Microbial Pathogenesis, Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus, OH 43215, USA
- Infectious Diseases Institute, The Ohio State University, Columbus, OH 43210, USA
| | - John S. Gunn
- Center for Microbial Pathogenesis, Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus, OH 43215, USA
- Infectious Diseases Institute, The Ohio State University, Columbus, OH 43210, USA
- Department of Pediatrics, College of Medicine, The Ohio State University, Columbus, OH 43210, USA
- Correspondence: ; Tel.: +1-(614)-355-3403
| |
Collapse
|
30
|
Menousek J, Horn CM, Heim CE, Van Roy Z, Korshoj LE, Kielian T. Transcriptional Profiling of Phagocytic Leukocytes and Microglia Reveals a Critical Role for Reactive Oxygen Species in Biofilm Containment during Staphylococcus aureus Craniotomy Infection. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2022; 209:1973-1986. [PMID: 36426943 PMCID: PMC9643635 DOI: 10.4049/jimmunol.2200503] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Accepted: 09/13/2022] [Indexed: 12/31/2022]
Abstract
Craniotomies are performed to treat a variety of intracranial pathology. Surgical site infection remains a complication of craniotomy despite the use of prophylactic antibiotics and universal sterile precautions. Infections occur in 1-3% of procedures, with approximately half caused by Staphylococcus aureus that forms a biofilm on the bone flap and is recalcitrant to systemic antibiotic therapy. We used an S. aureus-dsRed construct to compare the phagocytic capacity of leukocytes and microglia in vitro and in vivo using a mouse model of craniotomy infection. In addition, single-cell RNA sequencing (scRNA-seq) was applied to determine whether a transcriptional signature could be identified for phagocytic versus nonphagocytic cells in vivo. S. aureus was phagocytosed to equivalent extents in microglia, macrophages, neutrophils, and granulocytic myeloid-derived suppressor cells in vitro; however, microglial uptake of S. aureus was limited in vivo, whereas the other leukocyte populations exhibited phagocytic activity. scRNA-seq comparing the transcriptional signatures of phagocytic (S. aureus-dsRed+) versus nonphagocytic (S. aureus-dsRed-) leukocytes identified classical pathways enriched in phagocytic cells (i.e., reactive oxygen species [ROS]/reactive nitrogen species, lysosome, iron uptake, and transport), whereas nonphagocytic populations had increased ribosomal, IFN, and hypoxia signatures. scRNA-seq also revealed a robust ROS profile, which led to the exploration of craniotomy infection in NADPH oxidase 2 knockout mice. S. aureus burden, leukocyte recruitment, and intracellular bacterial load were significantly increased in NADPH oxidase 2 KO compared with wild-type animals. Collectively, these results highlight the importance of ROS generation in phagocytes for S. aureus biofilm containment, but not clearance, during craniotomy infection.
Collapse
Affiliation(s)
- Joseph Menousek
- Department of Neurosurgery, University of Nebraska Medical Center, Omaha, NE 68198
| | - Christopher M. Horn
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE 68198
| | - Cortney E. Heim
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE 68198
| | - Zachary Van Roy
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE 68198
| | - Lee E. Korshoj
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE 68198
| | - Tammy Kielian
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE 68198
| |
Collapse
|
31
|
Hussan JR, Irwin SG, Mathews B, Swift S, Williams DL, Cornish J. Optimal dose of lactoferrin reduces the resilience of in vitro Staphylococcus aureus colonies. PLoS One 2022; 17:e0273088. [PMID: 35960734 PMCID: PMC9374217 DOI: 10.1371/journal.pone.0273088] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Accepted: 08/02/2022] [Indexed: 11/19/2022] Open
Abstract
The rise in antibiotic resistance has stimulated research into adjuvants that can improve the efficacy of broad-spectrum antibiotics. Lactoferrin is a candidate adjuvant; it is a multifunctional iron-binding protein with antimicrobial properties. It is known to show dose-dependent antimicrobial activity against Staphylococcus aureus through iron sequestration and repression of β-lactamase expression. However, S. aureus can extract iron from lactoferrin through siderophores for their growth, which confounds the resolution of lactoferrin's method of action. We measured the minimum inhibitory concentration (MIC) for a range of lactoferrin/ β-lactam antibiotic dose combinations and observed that at low doses (< 0.39 μM), lactoferrin contributes to increased S. aureus growth, but at higher doses (> 6.25 μM), iron-depleted native lactoferrin reduced bacterial growth and reduced the MIC of the β-lactam-antibiotic cefazolin. This differential behaviour points to a bacterial population response to the lactoferrin/ β-lactam dose combination. Here, with the aid of a mathematical model, we show that lactoferrin stratifies the bacterial population, and the resulting population heterogeneity is at the basis of the dose dependent response seen. Further, lactoferrin disables a sub-population from β-lactam-induced production of β-lactamase, which when sufficiently large reduces the population's ability to recover after being treated by an antibiotic. Our analysis shows that an optimal dose of lactoferrin acts as a suitable adjuvant to eliminate S. aureus colonies using β-lactams, but sub-inhibitory doses of lactoferrin reduces the efficacy of β-lactams.
Collapse
Affiliation(s)
- Jagir R. Hussan
- Auckland Bioengineering Institute, University of Auckland, Auckland, NZ
- Department of Molecular Medicine and Pathology, University of Auckland, Auckland, NZ
| | - Stuart G. Irwin
- Department of Molecular Medicine and Pathology, University of Auckland, Auckland, NZ
| | - Brya Mathews
- Department of Molecular Medicine and Pathology, University of Auckland, Auckland, NZ
| | - Simon Swift
- Department of Molecular Medicine and Pathology, University of Auckland, Auckland, NZ
| | - Dustin L. Williams
- Department of Microbiology and Immunology, School of Medicine, University of Utah, Salt Lake City, Utah, United States of America
| | - Jillian Cornish
- Department of Medicine, University of Auckland, Auckland, NZ
| |
Collapse
|
32
|
Daphnia magna and Gammarus pulex, novel promising agents for biomedical and agricultural applications. Sci Rep 2022; 12:13690. [PMID: 35953507 PMCID: PMC9372163 DOI: 10.1038/s41598-022-17790-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2022] [Accepted: 07/31/2022] [Indexed: 12/03/2022] Open
Abstract
Various studies have shown the importance of using different types of Zooplankton biomasses as an additional substance in the diet of fish. In addition, the drainage water of the fish cultures could be used in plant irrigation. In this study, biomasses of water flea Daphnia magna and Gammarus pulex collected and tested, for the first time, their effect against pathogenic microorganisms and on plant germination. The results showed significant antibacterial activity of D. magna and G. pulex against Staphylococcus aureus and Pseudomonas aeruginosa bacteria, as well as antifungal activity against Alternaria solani and Penicillium expansum, which gives the possibility to be used as biocontrol against these bacteria and plant pathogenic fungi. Furthermore, both animals showed positive activity in the germination rate of Vicia faba seed, reaching 83.0 ± 3.5 and 86.0 ± 3.8%, respectively. In conclusion, the biomasses of D. magna and G. pulex are promising and effective agents for their use in the medical field against some pathogenic microbes and as stimulators of plant growth.
Collapse
|
33
|
Li Y, Wuermanbieke S, Zhang X, Mu W, Ma H, Qi F, Sun X, Amat A, Cao L. Effects of intra-articular D-amino acids combined with systemic vancomycin on an experimental Staphylococcus aureus-induced periprosthetic joint infection. JOURNAL OF MICROBIOLOGY, IMMUNOLOGY, AND INFECTION = WEI MIAN YU GAN RAN ZA ZHI 2022; 55:716-727. [PMID: 35346597 DOI: 10.1016/j.jmii.2022.01.005] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Revised: 12/25/2021] [Accepted: 01/20/2022] [Indexed: 06/14/2023]
Abstract
BACKGROUND The D-isoforms of amino acids (D-AAs) exhibit anti-biofilm potential against a diverse range of bacterial species in vitro, while its role in vivo remains unclear. The aim of this study was to investigate the effects of a combination of D-AAs and vancomycin on a PJI rat model. METHODS Eight-week-old male SD rats were randomized to the control group, sham group, vancomycin group, D-AAs-vancomycin group. After treatment for 6 weeks, we analysed the levels of inflammatory factors in serum, behavioural change, imaging manifestations. The anti-biofilm ability of D-AAs was detected by crystal violet staining and scanning electron microscope observation, and its ability to assist antibiotics in killing bacteria was assessed by culture of bacteria. Additionally, micro-CT and histological analysis were used to evaluate the impact of D-AAs combined with vancomycin on the bone remodelling around the prosthesis. RESULTS The group treated with a D-AAs-vancomycin combination sustained normal weight gain and exhibited reduced the serum levels of α2M, IL-1β, IL-6, IL-10, TNF-α and PGE2. Moreover, treated with D-AAs in combination with vancomycin improved the weight-bearing activity performance, increased the sizes and widths of distal femurs, and improved Rissing scale scoring. In particular, treatment using D-AAs enhanced the ability of vancomycin to eradicate Staphylococcus aureus, as demonstrated by the dispersion of existing biofilms and the inhibition of biofilm formation that occurred in a concentration-dependent manner. This treatment combination also resulted in a reduction in bacterial burden with in the soft tissues, bones, and implants. Furthermore, D-AAs-vancomycin combination treatment attenuated abnormal bone remodelling around the implant, as evidenced by an observed increase in BMD, BV/TV, and Tb.Th and the presence of reduced Trap+ osteoclasts and elevated osterix+ osteo-progenitors. CONCLUSIONS Combining D-AAs with vancomycin provides an effective therapeutic strategy for the treatment of PJI by promoting biofilm dispersion to enhance antimicrobial activity.
Collapse
Affiliation(s)
- Yicheng Li
- Department of Orthopaedics, First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| | | | - Xiaogang Zhang
- Department of Orthopaedics, First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| | - Wenbo Mu
- Department of Orthopaedics, First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| | - Hairong Ma
- Department of Orthopaedics, First Affiliated Hospital of Xinjiang Medical University, Urumqi, China; Xinjiang Uygur Autonomous Region Clinical Research Center for Orthopedic Diseases, Urumqi, China
| | - Fei Qi
- Department of Orthopaedics, First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| | - Xiaoyue Sun
- Department of Orthopaedics, First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| | - Abdusami Amat
- Department of Orthopaedics, First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| | - Li Cao
- Department of Orthopaedics, First Affiliated Hospital of Xinjiang Medical University, Urumqi, China.
| |
Collapse
|
34
|
Higashihira S, Simpson SJ, Collier CD, Natoli RM, Kittaka M, Greenfield EM. Halicin Is Effective Against Staphylococcus aureus Biofilms In Vitro. Clin Orthop Relat Res 2022; 480:1476-1487. [PMID: 35583504 PMCID: PMC9278916 DOI: 10.1097/corr.0000000000002251] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Accepted: 04/28/2022] [Indexed: 01/31/2023]
Abstract
BACKGROUND Biofilms protect bacteria from the host immune system and many antibiotics, making the treatment of orthopaedic infections difficult. Halicin, a recently discovered antibiotic, has potent activity against nonorthopaedic infections in mice and the planktonic, free-living forms of many bacterial species, including Staphylococcus aureus , a common cause of orthopaedic infections. Importantly, halicin did not induce resistance in vitro and was effective against drug-resistant bacteria and proliferating and quiescent bacteria. Quiescence is an important cause of antibiotic tolerance in biofilms. However, whether halicin acts on biofilms has not been tested. QUESTIONS/PURPOSES (1) Does halicin reduce the viability of S. aureus in less mature and more mature biofilms as it does in planktonic cultures? (2) How do the relative effects of halicin on S. aureus biofilms and planktonic cultures compare with those of conventional antibiotics (tobramycin, cefazolin, vancomycin, or rifampicin) that are commonly used in clinical orthopaedic infections? METHODS To measure minimal biofilm eradication concentrations (MBECs) with less mature 3-day and more mature 7-day biofilms, we used 96-well peg plates that provided high throughput and excellent reproducibility. After S. aureus -Xen36 biofilm formation, planktonic bacteria were removed from the cultures, and the biofilms were exposed to various concentrations of halicin, tobramycin, cefazolin, vancomycin, or rifampicin for 20 hours. Biofilm viability was determined by measuring resazurin reduction or by counting colony-forming units after sonication. To determine effects of halicin and the conventional antibiotics on biofilm viability, we defined MBEC 75 as the lowest concentration that decreased viability by 75% or more. To determine effects on bacterial viability in planktonic cultures, minimum inhibitory concentrations (MICs) were determined with the broth dilution method. Each result was measured in four to 10 independent experiments. RESULTS We found no differences between halicin's effectiveness against planktonic S. aureus and 3-day biofilms (MIC and MBEC 75 for 3-day biofilms was 25 μM [interquartile range 25 to 25 and 25 to 25, respectively]; p > 0.99). Halicin was eightfold less effective against more mature 7-day biofilms (MBEC 75 = 200 μM [100 to 200]; p < 0.001). Similarly, tobramycin was equally effective against planktonic culture and 3-day biofilms (MIC and MBEC 75 for 3-day biofilms was 20 μM [20 to 20 and 10 to 20, respectively]; p > 0.99). Tobramycin's MBEC 75 against more mature 7-day biofilms was 320 μM (320 to 480), which is 16-fold greater than its planktonic MIC (p = 0.03). In contrast, the MBEC 75 for cefazolin, vancomycin, and rifampicin against more mature 7-day biofilms were more than 1000-fold (> 1000; p < 0.001), 500-fold (500 to 875; p < 0.001), and 3125-fold (3125 to 5469; p = 0.004) greater than their planktonic MICs, respectively, consistent with those antibiotics' relative inactivity against biofilms. CONCLUSION Halicin was as effective against S. aureus in less mature 3-day biofilms as those in planktonic cultures, but eightfold higher concentrations were needed for more mature 7-day biofilms. Tobramycin, an antibiotic whose effectiveness depends on biofilm maturity, was also as effective against S. aureus in less mature 3-day biofilms as those in planktonic cultures, but 16-fold higher concentrations were needed for more mature 7-day biofilms. In contrast, cefazolin, vancomycin, and rifampicin were substantially less active against both less and more mature biofilms than against planktonic cultures. CLINICAL RELEVANCE Halicin is a promising antibiotic that may be effective against S. aureus osteomyelitis and infections on orthopaedic implants. Future studies should assess the translational value of halicin by testing its effects in animal models of orthopaedic infections; on the biofilms of other bacterial species, including multidrug-resistant bacteria; and in combination therapy with conventional antibiotics.
Collapse
Affiliation(s)
- Shota Higashihira
- Department of Orthopaedic Surgery, Indiana University School of Medicine, Indianapolis, IN, USA
- Indiana Center for Musculoskeletal Health, Indiana University School of Medicine, Indianapolis, IN, USA
- Department of Orthopaedic Surgery, Yokohama City University, Yokohama, Japan
| | - Stefanie Jan Simpson
- Department of Orthopaedic Surgery, Indiana University School of Medicine, Indianapolis, IN, USA
- Indiana Center for Musculoskeletal Health, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Christopher David Collier
- Department of Orthopaedic Surgery, Indiana University School of Medicine, Indianapolis, IN, USA
- Indiana Center for Musculoskeletal Health, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Roman Michael Natoli
- Department of Orthopaedic Surgery, Indiana University School of Medicine, Indianapolis, IN, USA
- Indiana Center for Musculoskeletal Health, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Mizuho Kittaka
- Indiana Center for Musculoskeletal Health, Indiana University School of Medicine, Indianapolis, IN, USA
- Department of Biomedical Sciences and Comprehensive Care, Indiana University School of Dentistry, Indianapolis, IN, USA
| | - Edward Michael Greenfield
- Department of Orthopaedic Surgery, Indiana University School of Medicine, Indianapolis, IN, USA
- Indiana Center for Musculoskeletal Health, Indiana University School of Medicine, Indianapolis, IN, USA
| |
Collapse
|
35
|
Karau M, Schmidt-Malan S, Mandrekar J, Lehoux D, Schuch R, Cassino C, Patel R. Locally delivered antistaphylococcal lysin exebacase or CF-296 is active in methicillin-resistant Staphylococcus aureus implant-associated osteomyelitis. J Bone Jt Infect 2022; 7:169-175. [PMID: 36032801 PMCID: PMC9399932 DOI: 10.5194/jbji-7-169-2022] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Accepted: 07/07/2022] [Indexed: 11/11/2022] Open
Abstract
Abstract. Introduction: Staphylococcus aureus is the most common cause of orthopedic infections and can be
challenging to treat, especially in the presence of a foreign body. The
antistaphylococcal lysins exebacase and CF-296 have rapid bactericidal
activity, a low propensity for resistance development, and synergize with
some antibiotics.
Methods: Rabbit implant-associated osteomyelitis was induced by drilling
into the medial tibia followed by locally delivering exebacase, CF-296, or
lysin carrier. A titanium screw colonized with methicillin-resistant S. aureus (MRSA) IDRL-6169 was inserted.
Intravenous daptomycin or saline was administered and continued daily for
4 d. On day 5, rabbits were euthanized, and the tibiae and implants were
collected for culture. Results were reported as log10 colony forming units (cfu) per gram of bone or log10 cfu per implant, and comparisons among the six groups were performed using the
Wilcoxon rank sum test.
Results: Based on implant and bone cultures, all treatments resulted in
significantly lower bacterial counts than those of controls (P≤0.0025).
Exebacase alone or with daptomycin as well as CF-296 with daptomycin were more
active than daptomycin alone (P≤0.0098) or CF-296 alone (P≤0.0154)
based on implant cultures. CF-296 with daptomycin was more active than
either CF-296 alone (P=0.0040) or daptomycin alone (P=0.0098) based on
bone cultures.
Conclusion: Local delivery of either exebacase or CF-296 offers a promising
complement to conventional antibiotics in implant-associated infections.
Collapse
Affiliation(s)
- Melissa Karau
- Division of Clinical Microbiology, Department of Laboratory Medicine
and Pathology, Mayo Clinic, Rochester, MN, USA
| | - Suzannah Schmidt-Malan
- Division of Clinical Microbiology, Department of Laboratory Medicine
and Pathology, Mayo Clinic, Rochester, MN, USA
| | - Jay Mandrekar
- Division of Biomedical Statistics and Informatics, Department of
Health Sciences Research, Mayo Clinic, Rochester, MN, USA
| | | | | | | | - Robin Patel
- Division of Clinical Microbiology, Department of Laboratory Medicine
and Pathology, Mayo Clinic, Rochester, MN, USA
- Division of Public Health, Infectious Diseases, and Occupational
Medicine, Department of Medicine, Mayo Clinic, Rochester, MN, USA
| |
Collapse
|
36
|
Felix L, Mishra B, Khader R, Ganesan N, Mylonakis E. In Vitro and In Vivo Bactericidal and Antibiofilm Efficacy of Alpha Mangostin Against Staphylococcus aureus Persister Cells. Front Cell Infect Microbiol 2022; 12:898794. [PMID: 35937701 PMCID: PMC9353584 DOI: 10.3389/fcimb.2022.898794] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Accepted: 06/23/2022] [Indexed: 11/23/2022] Open
Abstract
The formation of persister cells is associated with recalcitrance and infections. In this study, we examined the antimicrobial property of alpha mangostin, a natural xanthone molecule, against methicillin-resistant Staphylococcus aureus (MRSA) persisters and biofilm. The MIC of alpha mangostin against MRSA persisters was 2 µg/ml, and activity was mediated by causing membrane permeabilization within 30 min of exposure. The membrane activity of alpha mangostin was further studied by fast-killing kinetics of MRSA persiste r cells and found that the compound exhibited 99.99% bactericidal activity within 30 min. Furthermore, alpha mangostin disrupted established MRSA biofilms and inhibited bacterial attachment as biofilm formation. Alpha mangostin down-regulated genes associated with the formation of persister cells and biofilms, such as norA, norB, dnaK, groE, and mepR, ranging from 2 to 4-folds. Alpha mangostin at 16 μg/ml was non-toxic (> 95% cell survival) to liver-derived HepG2 and lung-derived A549 cells, similarly. Still, alpha mangostin exhibited 50% cell lysis of human RBC at 16 μg/ml. Interestingly, alpha mangostin was effective in vivo at increasing the survival up to 75% (p<0.0001) of Galleria mellonella larvae infected with MRSA persister for 120 h. In conclusion, we report that alpha mangostin is active against MRSA persisters and biofilms, and these data further our understanding of the antistaphylococcal activity and toxicity of this natural compound.
Collapse
|
37
|
Small-Molecule-Induced Activation of Cellular Respiration Inhibits Biofilm Formation and Triggers Metabolic Remodeling in Staphylococcus aureus. mBio 2022; 13:e0084522. [PMID: 35852317 PMCID: PMC9426486 DOI: 10.1128/mbio.00845-22] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Staphylococcus aureus, a major pathogen of community-acquired and nosocomial-associated infections, forms biofilms consisting of extracellular matrix-embedded cell aggregates. S. aureus biofilm formation on implanted medical devices can cause local and systemic infections due to the dispersion of cells from the biofilms. Usually, conventional antibiotic treatments are not effective against biofilm-related infections, and there is no effective treatment other than removing the contaminated devices. Therefore, the development of new therapeutic agents to combat biofilm-related infections is urgently needed. We conducted high-throughput screening of S. aureus biofilm inhibitors and obtained a small compound, JBD1. JBD1 strongly inhibits biofilm formation of S. aureus, including methicillin-resistant strains. In addition, JBD1 activated the respiratory activity of S. aureus cells and increased the sensitivity to aminoglycosides. Furthermore, it was shown that the metabolic profile of S. aureus was significantly altered in the presence of JBD1 and that metabolic remodeling was induced. Surprisingly, these JBD1-induced phenotypes were blocked by adding an excess amount of the electron carrier menaquinone to suppress respiratory activation. These results indicate that JBD1 induces biofilm inhibition and metabolic remodeling through respiratory activation. This study demonstrates that compounds that enhance the respiratory activity of S. aureus may be potential leads in the development of therapeutic agents for chronic S. aureus-biofilm-related infections.
Collapse
|
38
|
Sionov RV, Steinberg D. Targeting the Holy Triangle of Quorum Sensing, Biofilm Formation, and Antibiotic Resistance in Pathogenic Bacteria. Microorganisms 2022; 10:1239. [PMID: 35744757 PMCID: PMC9228545 DOI: 10.3390/microorganisms10061239] [Citation(s) in RCA: 75] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2022] [Revised: 06/12/2022] [Accepted: 06/14/2022] [Indexed: 12/12/2022] Open
Abstract
Chronic and recurrent bacterial infections are frequently associated with the formation of biofilms on biotic or abiotic materials that are composed of mono- or multi-species cultures of bacteria/fungi embedded in an extracellular matrix produced by the microorganisms. Biofilm formation is, among others, regulated by quorum sensing (QS) which is an interbacterial communication system usually composed of two-component systems (TCSs) of secreted autoinducer compounds that activate signal transduction pathways through interaction with their respective receptors. Embedded in the biofilms, the bacteria are protected from environmental stress stimuli, and they often show reduced responses to antibiotics, making it difficult to eradicate the bacterial infection. Besides reduced penetration of antibiotics through the intricate structure of the biofilms, the sessile biofilm-embedded bacteria show reduced metabolic activity making them intrinsically less sensitive to antibiotics. Moreover, they frequently express elevated levels of efflux pumps that extrude antibiotics, thereby reducing their intracellular levels. Some efflux pumps are involved in the secretion of QS compounds and biofilm-related materials, besides being important for removing toxic substances from the bacteria. Some efflux pump inhibitors (EPIs) have been shown to both prevent biofilm formation and sensitize the bacteria to antibiotics, suggesting a relationship between these processes. Additionally, QS inhibitors or quenchers may affect antibiotic susceptibility. Thus, targeting elements that regulate QS and biofilm formation might be a promising approach to combat antibiotic-resistant biofilm-related bacterial infections.
Collapse
Affiliation(s)
- Ronit Vogt Sionov
- The Biofilm Research Laboratory, The Institute of Biomedical and Oral Research, The Faculty of Dental Medicine, Hadassah Medical School, The Hebrew University, Jerusalem 9112102, Israel;
| | | |
Collapse
|
39
|
Kim SM, Zou G, Kim H, Kang M, Ahn S, Heo HY, Kim JS, Lim KM, Ausubel FM, Mylonakis E, Gao H, Kim W. Antimicrobial activity of the membrane-active compound nTZDpa is enhanced at low pH. Biomed Pharmacother 2022; 150:112977. [PMID: 35447554 DOI: 10.1016/j.biopha.2022.112977] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Revised: 04/03/2022] [Accepted: 04/12/2022] [Indexed: 11/30/2022] Open
Abstract
The opportunistic human pathogen Staphylococcus aureus can evade antibiotics by acquiring antibiotic resistance genes or by entering into a non-growing dormant state. Moreover, the particular circumstances of a specific infection site, such as acidity or anaerobicity, often weaken antibiotic potency. Decreased bacterial susceptibility combined with diminished antibiotic potency is responsible for high failure rates when treating S. aureus infections. Here, we report that the membrane-active antimicrobial agent nTZDpa does not only exhibit enhanced antibiotic activity against multidrug-resistant Gram-positive pathogens in acidic pH, but also retains antimicrobial potency under anaerobic conditions. This agent completely eradicated highly antibiotic-tolerant cells and biofilms formed by methicillin-resistant S. aureus at pH 5.5 at concentrations at which it was not potent at pH 7.4. Furthermore, nTZDpa was more potent at synergistically potentiating gentamicin killing against antibiotic-tolerant MRSA cells at low pH than at high pH. All-atom molecular dynamics simulations combined with membrane-permeabilization assays revealed that the neutral form of nTZDpa, which contains carboxylic acid, is more effective than the deprotonated form at penetrating the bacterial membrane and plays an essential role in membrane activity. An acidic pH increases the proportion of the neutrally charged nTZDpa, which results in antimicrobial enhancement. Our results provide key insights into rational design of pH-sensitive membrane-active antimicrobials and antibiotic adjuvants that are effective in an infection environment. These findings demonstrate that nTZDpa is a promising lead compound for developing new therapeutics against hard-to-cure infections caused by drug-resistant and -tolerant S. aureus.
Collapse
Affiliation(s)
- Soo Min Kim
- College of Pharmacy, Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul 03760, Republic of Korea
| | - Guijin Zou
- Institute of High Performance Computing, A⁎STAR, Singapore 138632, Singapore
| | - Hyerim Kim
- College of Pharmacy, Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul 03760, Republic of Korea
| | - Minjeong Kang
- College of Pharmacy, Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul 03760, Republic of Korea
| | - Soyeon Ahn
- College of Pharmacy, Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul 03760, Republic of Korea
| | - Hee Young Heo
- College of Pharmacy, Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul 03760, Republic of Korea
| | - Jae-Seok Kim
- Department of Laboratory Medicine, Kangdong Sacred Heart Hospital, Hallym University College of Medicine, Seoul 05355, Republic of Korea
| | - Kyung-Min Lim
- College of Pharmacy, Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul 03760, Republic of Korea
| | - Frederick M Ausubel
- Department of Molecular Biology, Massachusetts General Hospital, Boston, MA 02114, USA; Department of Genetics, Harvard Medical School, Boston, MA 02115, USA
| | - Eleftherios Mylonakis
- Division of Infectious Diseases, Rhode Island Hospital, Warren Alpert Medical School of Brown University, Providence, RI 02903, USA
| | - Huajian Gao
- Institute of High Performance Computing, A⁎STAR, Singapore 138632, Singapore; School of Mechanical and Aerospace Engineering, College of Engineering, Nanyang Technological University, Singapore 639789, Singapore
| | - Wooseong Kim
- College of Pharmacy, Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul 03760, Republic of Korea.
| |
Collapse
|
40
|
Low Concentration of the Neutrophil Proteases Cathepsin G, Cathepsin B, Proteinase-3 and Metalloproteinase-9 Induce Biofilm Formation in Non-Biofilm-Forming Staphylococcus epidermidis Isolates. Int J Mol Sci 2022; 23:ijms23094992. [PMID: 35563384 PMCID: PMC9102557 DOI: 10.3390/ijms23094992] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Revised: 04/26/2022] [Accepted: 04/28/2022] [Indexed: 12/04/2022] Open
Abstract
Neutrophils play a crucial role in eliminating bacteria that invade the human body; however, cathepsin G can induce biofilm formation in a non-biofilm-forming Staphylococcus epidermidis 1457 strain, suggesting that neutrophil proteases may be involved in biofilm formation. Cathepsin G, cathepsin B, proteinase-3, and metalloproteinase-9 (MMP-9) from neutrophils were tested on the biofilm induction in commensal (skin isolated) and clinical non-biofilm-forming S. epidermidis isolates. From 81 isolates, 53 (74%) were aap+, icaA−, icaD− genotype, and without the capacity of biofilm formation under conditions of 1% glucose, 4% ethanol or 4% NaCl, but these 53 non-biofilm-forming isolates induced biofilm by the use of different neutrophil proteases. Of these, 62.3% induced biofilm with proteinase-3, 15% with cathepsin G, 10% with cathepsin B and 5% with MMP -9, where most of the protease-induced biofilm isolates were commensal strains (skin). In the biofilm formation kinetics analysis, the addition of phenylmethylsulfonyl fluoride (PMSF; a proteinase-3 inhibitor) showed that proteinase-3 participates in the cell aggregation stage of biofilm formation. A biofilm induced with proteinase-3 and DNAse-treated significantly reduced biofilm formation at an early time (initial adhesion stage of biofilm formation) compared to untreated proteinase-3-induced biofilm (p < 0.05). A catheter inoculated with a commensal (skin) non-biofilm-forming S. epidermidis isolate treated with proteinase-3 and another one without the enzyme were inserted into the back of a mouse. After 7 days of incubation period, the catheters were recovered and the number of grown bacteria was quantified, finding a higher amount of adhered proteinase-3-treated bacteria in the catheter than non-proteinase-3-treated bacteria (p < 0.05). Commensal non-biofilm-forming S. epidermidis in the presence of neutrophil cells significantly induced the biofilm formation when multiplicity of infection (MOI) 1:0.01 (neutrophil:bacteria) was used, but the addition of a cocktail of protease inhibitors impeded biofilm formation. A neutrophil:bacteria assay did not induce neutrophil extracellular traps (NETs). Our results suggest that neutrophils, in the presence of commensal non-biofilm-forming S. epidermidis, do not generate NETs formation. The effect of neutrophils is the production of proteases, and proteinase-3 releases bacterial DNA at the initial adhesion, favoring cell aggregation and subsequently leading to biofilm formation.
Collapse
|
41
|
Manandhar S, Singh A, Varma A, Pandey S, Shrivastava N. High level of persister frequency in clinical staphylococcal isolates. BMC Microbiol 2022; 22:109. [PMID: 35448965 PMCID: PMC10124895 DOI: 10.1186/s12866-022-02529-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Accepted: 04/12/2022] [Indexed: 01/11/2023] Open
Abstract
BACKGROUND Staphylococcus aureus is a notorious human pathogen that causes often lethal systemic conditions that are mostly medical device associated biofilm infections. Similarly, coagulase negative staphylococci are emerging as leading pathogen for nosocomial infections owing to their ability to form biofilm on implanted medical equipment. Chronic in nature, these infections are difficult to treat. Such recalcitrance of these infections is caused mainly due to the presence of persister cells, which exhibit transient yet extreme tolerance to antibiotics. Despite tremendous clinical significance, there is lack of studies on persister cells formation among clinical bacterial isolates. Considering the importance of factors influencing persister formation, in this study, we evaluate the association of antibiotic tolerance with biofilm production, antibiotic stress, growth phase, specimen type, and dependency on staphylococcal species. Biofilm formation was detected among 375 clinical staphylococcal isolates by quantitative tissue culture plate method (TCP) and icaAD genes by genotypic method. The antibiotic susceptibility was determined by Kirby Bauer disc diffusion method while minimum inhibitory concentration values were obtained by agar dilution method. Persister cells were measured in the susceptible staphylococcal isolates in the presence of clinically relevant antibiotics. RESULTS In the study, 161 (43%) S. aureus and 214 (57%) coagulase negative staphylococci (CNS) were isolated from different clinical samples. TCP method detected biofilm production in 84 (52.2%) S. aureus and 90 (42.1%) CNS isolates. The genotypic method detected icaAD genes in 86 (22.9%) isolates. Majority (> 90%) of both the biofilm producers and non-producers were sensitive to chloramphenicol and tetracycline but were resistant to penicillin. Interestingly, all isolates were sensitive to vancomycin irrespective of biofilm production. While high persister frequency was observed among all staphylococci isolates in the stationary growth phase, the persister frequency in exponential growth phase was statistically high among isolates possessing icaAD genes compared to icaAD negative isolates. CONCLUSION The research findings provide strong evidence that the clinical staphylococcal isolates exhibit extreme antibiotic tolerance suggesting their causal link with treatment failures. Understanding the factors influencing the formation and maintenance of persister cells are of utmost important aspect to design therapeutics and control recalcitrant bacterial infections.
Collapse
Affiliation(s)
- Sarita Manandhar
- Tri-Chandra Multiple College, Tribhuvan University, Kathmandu, Nepal.
| | - Anjana Singh
- Central Department of Microbiology, Tribhuvan University, Kathmandu, Nepal
| | - Ajit Varma
- Amity Institute of Microbial Technology, Amity University, Uttar Pradesh, Noida, UP, 201303, India
| | - Shanti Pandey
- The University of Southern Mississippi, Hattiesburg, MS-39406, USA
| | - Neeraj Shrivastava
- Amity Institute of Microbial Technology, Amity University, Uttar Pradesh, Noida, UP, 201303, India
| |
Collapse
|
42
|
Hou H, Li Y, Jin Y, Chen S, Long J, Duan G, Yang H. The crafty opponent: the defense systems of Staphylococcus aureus and response measures. Folia Microbiol (Praha) 2022; 67:233-243. [PMID: 35149955 DOI: 10.1007/s12223-022-00954-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Accepted: 01/29/2022] [Indexed: 11/29/2022]
Abstract
Staphylococcus aureus is a serious threat to public health. S. aureus infection can cause acute or long-term persistent infections that are often resistant to antibiotics and are associated with high morbidity and death. Understanding the defensive systems of S. aureus can help clinicians make the best use of antimicrobial drugs and can also help with antimicrobial stewardship. The mechanisms and clinical implications of S. aureus defense systems, as well as potential response systems, were discussed in this study. Because resistance to all currently available antibiotics is unavoidable, new medicines are always being developed. Alternative techniques, such as anti-virulence and bacteriophage therapies, are being researched and may become major tools in the fight against staphylococcal infections in the future, in addition to the development of new small compounds that affect cell viability.
Collapse
Affiliation(s)
- Hongjie Hou
- Department of Epidemiology, School of Public Health, Zhengzhou University, No. 100 of Science Avenue, Zhengzhou, 450001, China
| | - Yang Li
- Department of Epidemiology, School of Public Health, Zhengzhou University, No. 100 of Science Avenue, Zhengzhou, 450001, China
| | - Yuefei Jin
- Department of Epidemiology, School of Public Health, Zhengzhou University, No. 100 of Science Avenue, Zhengzhou, 450001, China
| | - Shuaiyin Chen
- Department of Epidemiology, School of Public Health, Zhengzhou University, No. 100 of Science Avenue, Zhengzhou, 450001, China
| | - Jinzhao Long
- Department of Epidemiology, School of Public Health, Zhengzhou University, No. 100 of Science Avenue, Zhengzhou, 450001, China
| | - Guangcai Duan
- Department of Epidemiology, School of Public Health, Zhengzhou University, No. 100 of Science Avenue, Zhengzhou, 450001, China
| | - Haiyan Yang
- Department of Epidemiology, School of Public Health, Zhengzhou University, No. 100 of Science Avenue, Zhengzhou, 450001, China.
| |
Collapse
|
43
|
Guo H, Tong Y, Cheng J, Abbas Z, Li Z, Wang J, Zhou Y, Si D, Zhang R. Biofilm and Small Colony Variants-An Update on Staphylococcus aureus Strategies toward Drug Resistance. Int J Mol Sci 2022; 23:ijms23031241. [PMID: 35163165 PMCID: PMC8835882 DOI: 10.3390/ijms23031241] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Revised: 01/16/2022] [Accepted: 01/20/2022] [Indexed: 02/07/2023] Open
Abstract
Recently, the drawbacks arising from the overuse of antibiotics have drawn growing public attention. Among them, drug-resistance (DR) and even multidrug-resistance (MDR) pose significant challenges in clinical practice. As a representative of a DR or MDR pathogen, Staphylococcus aureus can cause diversity of infections related to different organs, and can survive or adapt to the diverse hostile environments by switching into other phenotypes, including biofilm and small colony variants (SCVs), with altered physiologic or metabolic characteristics. In this review, we briefly describe the development of the DR/MDR as well as the classical mechanisms (accumulation of the resistant genes). Moreover, we use multidimensional scaling analysis to evaluate the MDR relevant hotspots in the recent published reports. Furthermore, we mainly focus on the possible non-classical resistance mechanisms triggered by the two important alternative phenotypes of the S. aureus, biofilm and SCVs, which are fundamentally caused by the different global regulation of the S. aureus population, such as the main quorum-sensing (QS) and agr system and its coordinated regulated factors, such as the SarA family proteins and the alternative sigma factor σB (SigB). Both the biofilm and the SCVs are able to escape from the host immune response, and resist the therapeutic effects of antibiotics through the physical or the biological barriers, and become less sensitive to some antibiotics by the dormant state with the limited metabolisms.
Collapse
|
44
|
Lohan S, Mandal D, Choi W, Konshina AG, Tiwari RK, Efremov RG, Maslennikov I, Parang K. Small Amphiphilic Peptides: Activity Against a Broad Range of Drug-Resistant Bacteria and Structural Insight into Membranolytic Properties. J Med Chem 2022; 65:665-687. [PMID: 34978443 DOI: 10.1021/acs.jmedchem.1c01782] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
We report the synthesis and antibacterial activities of a series of amphiphilic membrane-active peptides composed, in part, of various nongenetically coded hydrophobic amino acids. The lead cyclic peptides, 8C and 9C, showed broad-spectrum activity against drug-resistant Gram-positive (minimum inhibitory concentration (MIC) = 1.5-6.2 μg/mL) and Gram-negative (MIC = 12.5-25 μg/mL) bacteria. The cytotoxicity study showed the predominant lethal action of the peptides against bacteria as compared with mammalian cells. A plasma stability study revealed approximately 2-fold higher stability of lead cyclic peptides as compared to their linear counterparts after 24 h of incubation. A calcein dye leakage experiment revealed the membranolytic effect of the cyclic peptides. Nuclear magnetic resonance spectroscopy and molecular dynamics simulation studies of the interaction of the peptides with the phospholipid bilayer provided a solid structural basis to explain the membranolytic action of the peptides with atomistic details. These results highlight the potential of newly designed amphiphilic peptides as the next generation of peptide-based antibiotics.
Collapse
Affiliation(s)
- Sandeep Lohan
- Center for Targeted Drug Delivery, Department of Biomedical and Pharmaceutical Sciences, Chapman University School of Pharmacy, Harry and Diane Rinker Health Science Campus, 9401 Jeronimo Road, Irvine, California 92618, United States
- AJK Biopharmaceutical, 5270 California Avenue, Irvine, California 92617, United States
| | - Dindyal Mandal
- Center for Targeted Drug Delivery, Department of Biomedical and Pharmaceutical Sciences, Chapman University School of Pharmacy, Harry and Diane Rinker Health Science Campus, 9401 Jeronimo Road, Irvine, California 92618, United States
- AJK Biopharmaceutical, 5270 California Avenue, Irvine, California 92617, United States
- School of Biotechnology, KIIT Deemed to be University, Bhubaneswar 751024, India
| | - Wonsuk Choi
- Structural Biology Research Center, Department of Biomedical and Pharmaceutical Sciences, Chapman University School of Pharmacy, Harry and Diane Rinker Health Science Campus, 9401 Jeronimo Road, Irvine, California 92618, United States
| | - Anastasia G Konshina
- M.M. Shemyakin & Yu.A. Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Miklukho-Maklaya Street, 16/10, Moscow 117997, Russia
| | - Rakesh K Tiwari
- Center for Targeted Drug Delivery, Department of Biomedical and Pharmaceutical Sciences, Chapman University School of Pharmacy, Harry and Diane Rinker Health Science Campus, 9401 Jeronimo Road, Irvine, California 92618, United States
| | - Roman G Efremov
- M.M. Shemyakin & Yu.A. Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Miklukho-Maklaya Street, 16/10, Moscow 117997, Russia
- National Research University Higher School of Economics, Myasnitskaya ul. 20, Moscow 101000, Russia
- Moscow Institute of Physics and Technology (State University), Dolgoprudny, 141701 Moscow, Oblast, Russia
| | - Innokentiy Maslennikov
- Structural Biology Research Center, Department of Biomedical and Pharmaceutical Sciences, Chapman University School of Pharmacy, Harry and Diane Rinker Health Science Campus, 9401 Jeronimo Road, Irvine, California 92618, United States
| | - Keykavous Parang
- Center for Targeted Drug Delivery, Department of Biomedical and Pharmaceutical Sciences, Chapman University School of Pharmacy, Harry and Diane Rinker Health Science Campus, 9401 Jeronimo Road, Irvine, California 92618, United States
| |
Collapse
|
45
|
Wu S, Zhang J, Peng Q, Liu Y, Lei L, Zhang H. The Role of Staphylococcus aureus YycFG in Gene Regulation, Biofilm Organization and Drug Resistance. Antibiotics (Basel) 2021; 10:antibiotics10121555. [PMID: 34943766 PMCID: PMC8698359 DOI: 10.3390/antibiotics10121555] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Revised: 12/09/2021] [Accepted: 12/16/2021] [Indexed: 02/05/2023] Open
Abstract
Antibiotic resistance is a serious global health concern that may have significant social and financial consequences. Methicillin-resistant Staphylococcus aureus (MRSA) infection is responsible for substantial morbidity and leads to the death of 21.8% of infected patients annually. A lack of novel antibiotics has prompted the exploration of therapies targeting bacterial virulence mechanisms. The two-component signal transduction system (TCS) enables microbial cells to regulate gene expression and the subsequent metabolic processes that occur due to environmental changes. The YycFG TCS in S. aureus is essential for bacterial viability, the regulation of cell membrane metabolism, cell wall synthesis and biofilm formation. However, the role of YycFG-associated biofilm organization in S. aureus antimicrobial drug resistance and gene regulation has not been discussed in detail. We reviewed the main molecules involved in YycFG-associated cell wall biosynthesis, biofilm development and polysaccharide intercellular adhesin (PIA) accumulation. Two YycFG-associated regulatory mechanisms, accessory gene regulator (agr) and staphylococcal accessory regulator (SarA), were also discussed. We highlighted the importance of biofilm formation in the development of antimicrobial drug resistance in S. aureus infections. Data revealed that inhibition of the YycFG pathway reduced PIA production, biofilm formation and bacterial pathogenicity, which provides a potential target for the management of MRSA-induced infections.
Collapse
Affiliation(s)
- Shizhou Wu
- Department of Orthopedics, West China Hospital, Sichuan University, Chengdu 610041, China; (S.W.); (J.Z.); (Q.P.)
| | - Junqi Zhang
- Department of Orthopedics, West China Hospital, Sichuan University, Chengdu 610041, China; (S.W.); (J.Z.); (Q.P.)
| | - Qi Peng
- Department of Orthopedics, West China Hospital, Sichuan University, Chengdu 610041, China; (S.W.); (J.Z.); (Q.P.)
| | - Yunjie Liu
- West China School of Public Health, Sichuan University, Chengdu 610041, China;
| | - Lei Lei
- West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
- Correspondence: (L.L.); (H.Z.)
| | - Hui Zhang
- Department of Orthopedics, West China Hospital, Sichuan University, Chengdu 610041, China; (S.W.); (J.Z.); (Q.P.)
- Correspondence: (L.L.); (H.Z.)
| |
Collapse
|
46
|
Nguyen TK, Peyrusson F, Siala W, Pham NH, Nguyen HA, Tulkens PM, Van Bambeke F. Activity of Moxifloxacin Against Biofilms Formed by Clinical Isolates of Staphylococcus aureus Differing by Their Resistant or Persister Character to Fluoroquinolones. Front Microbiol 2021; 12:785573. [PMID: 34975808 PMCID: PMC8715871 DOI: 10.3389/fmicb.2021.785573] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Accepted: 11/19/2021] [Indexed: 11/13/2022] Open
Abstract
Staphylococcus aureus biofilms are poorly responsive to antibiotics. Underlying reasons include a matrix effect preventing drug access to embedded bacteria, or the presence of dormant bacteria with reduced growth rate. Using 18 clinical isolates previously characterized for their moxifloxacin-resistant and moxifloxacin-persister character in stationary-phase culture, we studied their biofilm production and matrix composition and the anti-biofilm activity of moxifloxacin. Biofilms were grown in microtiter plates and their abundance quantified by crystal violet staining and colony counting; their content in polysaccharides, extracellular DNA and proteins was measured. Moxifloxacin activity was assessed after 24 h of incubation with a broad range of concentrations to establish full concentration-response curves. All clinical isolates produced more biofilm biomass than the reference strain ATCC 25923, the difference being more important for those with high relative persister fractions to moxifloxacin, most of which being also resistant. High biofilm producers expressed icaA to higher levels, enriching the matrix in polysaccharides. Moxifloxacin was less potent against biofilms from clinical isolates than from ATCC 25923, especially against moxifloxacin-resistant isolates with high persister fractions, which was ascribed to a lower concentration of moxifloxacin in these biofilms. Time-kill curves in biofilms revealed the presence of a moxifloxacin-tolerant subpopulation, with low multiplication capacity, whatever the persister character of the isolate. Thus, moxifloxacin activity depends on its local concentration in biofilm, which is reduced in most isolates with high-relative persister fractions due to matrix effects, and insufficient to kill resistant isolates due to their high MIC.
Collapse
Affiliation(s)
- Tiep K. Nguyen
- Pharmacologie cellulaire et moléculaire, Louvain Drug Research Institute, Université catholique de Louvain, Brussels, Belgium
- Department of Pharmaceutical Industry, Hanoi University of Pharmacy, Hanoi, Vietnam
| | - Frédéric Peyrusson
- Pharmacologie cellulaire et moléculaire, Louvain Drug Research Institute, Université catholique de Louvain, Brussels, Belgium
| | - Wafi Siala
- Pharmacologie cellulaire et moléculaire, Louvain Drug Research Institute, Université catholique de Louvain, Brussels, Belgium
| | - Nhung H. Pham
- Department of Microbiology, Bach Mai Hospital, Hanoi, Vietnam
| | - Hoang A. Nguyen
- The National Center for Drug Information and Adverse Drug Reactions Monitoring, Hanoi University of Pharmacy, Hanoi, Vietnam
| | - Paul M. Tulkens
- Pharmacologie cellulaire et moléculaire, Louvain Drug Research Institute, Université catholique de Louvain, Brussels, Belgium
| | - Françoise Van Bambeke
- Pharmacologie cellulaire et moléculaire, Louvain Drug Research Institute, Université catholique de Louvain, Brussels, Belgium
- *Correspondence: Françoise Van Bambeke,
| |
Collapse
|
47
|
Ivory JD, Vellinga A, O'Gara J, Gethin G. A scoping review protocol to identify clinical signs, symptoms and biomarkers indicative of biofilm presence in chronic wounds. HRB Open Res 2021; 4:71. [PMID: 35224441 PMCID: PMC8847728 DOI: 10.12688/hrbopenres.13300.2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/12/2021] [Indexed: 11/20/2022] Open
Abstract
Introduction:
Wound healing is characterised by haemostatic, inflammatory, proliferative and remodelling phases. In the presence of comorbidities such as diabetes, healing can stall and chronic wounds may result. Infection is detrimental to these wounds and associated with poor outcomes. Wounds are contaminated with microbes and debris, and factors such as host resistance, bacterial virulence, species synergy and bioburden determine whether a wound will deteriorate to critically colonised/infected states. Biofilms are sessile microbial communities, exhibiting high-level antibiotic tolerance and resistance to host defences. Biofilm in critically colonised wounds can contribute to delayed healing. Little is known about clinical presentation and diagnosis of wound biofilms. Objective:
To
identify from the literature clinical signs, symptoms and biomarkers that may indicate biofilm presence in chronic wounds. Methods:
This review will be guided by the Preferred Reporting Items for Systematic Reviews extension for Scoping Reviews (PRISMA-ScR), and the Joanna Briggs Institute Manual for Evidence Synthesis. Studies of any design in any language recruiting adult patients with venous, diabetic, pressure or mixed arterial-venous ulcers and reporting data on clinical signs/symptoms of biofilm are eligible. Searches of Medline, Embase, CINAHL, Cochrane Central and BASE will be conducted from inception to present. Reference scanning and contact with content experts will be employed. Title/abstract screening and full text selection will be executed by two reviewers independently. Discrepancies will be resolved by discussion between reviewers or through third party intervention. Data will be extracted by a single reviewer and verified by a second. Clinical signs and symptoms data will be presented in terms of study design, setting and participant demographic data. Discussion:
Understanding biofilm impact on chronic wounds is inconsistent and based largely on
in vitro research. This work will consolidate clinical signs, symptoms and biomarkers of biofilm in chronic wounds reported in the literature.
Collapse
Affiliation(s)
- John D. Ivory
- School of Nursing & Midwifery, National University of Ireland, Galway, Galway, Galway, H91TK33, Ireland
- Alliance for Research & Innovation in Wounds (ARIW), National University of Ireland, Galway, Galway, Galway, H91TK33, Ireland
- CDA Diabetic Foot Disease: from PRevention to IMproved patient Outcomes (CDA DFD-PRIMO) Program, National University of Ireland, Galway, Galway, Galway, H91TK33, Ireland
- Irish Research Council, 3 Shelbourne Buildings, Crampton Avenue, Ballsbridge, D04 C2Y6, Ireland
| | - Akke Vellinga
- Alliance for Research & Innovation in Wounds (ARIW), National University of Ireland, Galway, Galway, Galway, H91TK33, Ireland
- School of Medicine, National University of Ireland, Galway, Galway, Galway, H91TK33, Ireland
| | - James O'Gara
- Alliance for Research & Innovation in Wounds (ARIW), National University of Ireland, Galway, Galway, Galway, H91TK33, Ireland
- CDA Diabetic Foot Disease: from PRevention to IMproved patient Outcomes (CDA DFD-PRIMO) Program, National University of Ireland, Galway, Galway, Galway, H91TK33, Ireland
- Microbiology, School of Natural Sciences, National University of Ireland, Galway, Galway, Galway, H91TK33, Ireland
| | - Georgina Gethin
- School of Nursing & Midwifery, National University of Ireland, Galway, Galway, Galway, H91TK33, Ireland
- Alliance for Research & Innovation in Wounds (ARIW), National University of Ireland, Galway, Galway, Galway, H91TK33, Ireland
- CDA Diabetic Foot Disease: from PRevention to IMproved patient Outcomes (CDA DFD-PRIMO) Program, National University of Ireland, Galway, Galway, Galway, H91TK33, Ireland
- School of Nursing and Midwifery, Monash University, Melbourne, Australia
| |
Collapse
|
48
|
Vancomycin-decorated microbubbles as a theranostic agent for Staphylococcus aureus biofilms. Int J Pharm 2021; 609:121154. [PMID: 34624449 DOI: 10.1016/j.ijpharm.2021.121154] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2021] [Revised: 09/28/2021] [Accepted: 09/30/2021] [Indexed: 12/20/2022]
Abstract
Bacterial biofilms are a huge burden on our healthcare systems worldwide. The lack of specificity in diagnostic and treatment possibilities result in difficult-to-treat and persistent infections. The aim of this in vitro study was to investigate if microbubbles targeted specifically to bacteria in biofilms could be used both for diagnosis as well for sonobactericide treatment and demonstrate their theranostic potential for biofilm infection management. The antibiotic vancomycin was chemically coupled to the lipid shell of microbubbles and validated using mass spectrometry and high-axial resolution 4Pi confocal microscopy. Theranostic proof-of-principle was investigated by demonstrating the specific binding of vancomycin-decorated microbubbles (vMB) to statically and flow grown Staphylococcus aureus (S. aureus) biofilms under increasing shear stress flow conditions (0-12 dyn/cm2), as well as confirmation of microbubble oscillation and biofilm disruption upon ultrasound exposure (2 MHz, 250 kPa, and 5,000 or 10,000 cycles) during flow shear stress of 5 dyn/cm2 using time-lapse confocal microscopy combined with the Brandaris 128 ultra-high-speed camera. Vancomycin was successfully incorporated into the microbubble lipid shell. vMB bound significantly more often than control microbubbles to biofilms, also in the presence of free vancomycin (up to 1000 µg/mL) and remained bound under increasing shear stress flow conditions (up to 12 dyn/cm2). Upon ultrasound insonification biofilm area was reduced of up to 28%, as confirmed by confocal microscopy. Our results confirm the successful production of vMB and support their potential as a new theranostic tool for S. aureus biofilm infections by allowing for specific bacterial detection and biofilm disruption.
Collapse
|
49
|
Survey on phenotypic resistance in Enterococcus faecalis: comparison between the expression of biofilm-associated genes in Enterococcus faecalis persister and non-persister cells. Mol Biol Rep 2021; 49:971-979. [PMID: 34751916 DOI: 10.1007/s11033-021-06915-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Accepted: 10/28/2021] [Indexed: 10/19/2022]
Abstract
BACKGROUND Phenotypic resistance is considered as a serious therapeutic challenge for which a definitive remedy has not been discovered yet. Biofilm and persister cell formation are two well-studied phenotypic resistance phenomena, leading to the recalcitrance and relapse of different types of chronic infections. The presence of persister cells in biofilm structures seems to be one of the main factors contributing to the relapse of infections and treatment failure. Given the dormant and inert nature of persister cells, they can be easy targets for the immune system factors. Biofilm formation can be a survival strategy for the defenseless persister cells. Thus, this study was aimed to evaluate the expression of biofilm-associated genes in Enterococcus faecalis persister and non-persister cells. METHODS Vancomycin susceptibility and biofilm formation ability were investigated among 95 E. faecalis clinical isolates using microtiter broth dilution and microtiter plate assays, respectively. PCR was used to determine the presence of biofilm-related genes (gelE, esp, and agg) among the vancomycin-susceptible, biofilm producer E. faecalis isolates (91 isolates). Minimum bactericidal concentration for biofilms (MBCB) were determined for vancomycin using the MTP assay. Bacterial persister assay was performed using an enzymatic lysis assay. Finally, the expression of biofilm-related genes was compared between the persister and non-persister isolates of E. faecalis using real-time qPCR. RESULTS E. faecalis isolates showed a high level of susceptibility (95.8%) to vancomycin (MIC < 1 µg/mL). The gelE, esp, and agg genes were found in 91 (100%), 72 (79.12), and 74 (81.32) of the isolates, respectively. All the E. faecalis isolates were tolerant to vancomycin in the biofilm condition, showing a MBCB of > 2500 µg/mL. Based on the enzymatic lysis assay, only 3 isolates, out of the 91, had the ability to form persister cells. The expression of biofilm-associated genes was higher among the persister compared to non-persister E. faecalis isolates. CONCLUSIONS Biofilm-associated persister cells indicated a high vancomycin tolerance compared to non-persister cells. Moreover, persister isolates showed a higher tendency for biofilm formation and a higher expression level of the biofilm-associated genes, compared to non-persister isolates.
Collapse
|
50
|
Bio-evaluation of fluoro and trifluoromethyl-substituted salicylanilides against multidrug-resistant S. aureus. Med Chem Res 2021; 30:2301-2315. [PMID: 34720564 PMCID: PMC8548355 DOI: 10.1007/s00044-021-02808-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Accepted: 10/06/2021] [Indexed: 11/20/2022]
Abstract
Methicillin-resistant Staphylococcus aureus (MRSA) and vancomycin-resistant Staphylococcus aureus (VRSA) are primary causes of skin and soft tissue infections worldwide. To address the emergency caused due to increasing multidrug-resistant (MDR) bacterial infections, a series of novel fluoro and trifluoromethyl-substituted salicylanilide derivatives were synthesized and their antimicrobial activity was investigated. MIC data reveal that the compounds inhibited S. aureus specifically (MIC 0.25–64 µg/mL). The in vitro cytotoxicity of compounds with MIC < 1 µg/mL against Vero cells led to identification of four compounds (20, 22, 24 and 25) with selectivity index above 10. These four compounds were tested against MDR S. aureus panel. Remarkably, 5-chloro-N-(4’-bromo-3’-trifluoromethylphenyl)-2-hydroxybenzamide (22) demonstrated excellent activity against nine MRSA and three VRSA strains with MIC 0.031–0.062 µg/mL, which is significantly better than the control drugs methicillin and vancomycin. The comparative time–kill kinetic experiment revealed that the effect of bacterial killing of 22 is comparable with vancomycin. Compound 22 did not synergize with or antagonize any FDA-approved antibiotic and reduced pre-formed S. aureus biofilm better than vancomycin. Overall, study suggested that 22 could be further developed as a potent anti-staphylococcal therapeutic. ![]()
Collapse
|