1
|
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
|
2
|
Uddin MJ, Julin K, Overkleeft HS, Johannessen M, Lentz CS. Activity-Based Protein Profiling Identifies an α-Amylase Family Protein Contributing to the Virulence of Methicillin-Resistant Staphylococcus aureus. ACS Infect Dis 2025; 11:573-583. [PMID: 39916318 PMCID: PMC11915364 DOI: 10.1021/acsinfecdis.4c00638] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Revised: 01/28/2025] [Accepted: 02/03/2025] [Indexed: 03/15/2025]
Abstract
In search of new putative antimicrobial drug targets in methicillin-resistant Staphylococcus aureus, we aimed to identify and characterize retaining glycosidase activities in this bacterial pathogen. Using activity-based protein profiling (ABPP), a panel of 7 fluorescent probes was screened to detect activities of diverse retaining glycosidase families. Based on this, a cocktail of 3 biotinylated probes (targeting α-glucosidases, β-galactosidases and α-fucosidases) was used for target enrichment and three glycoside hydrolase family proteins were identified by mass-spectrometry: 6-phospho-β-glucosidase (BglA), α-amylase family protein trehalase C (TreC), and autolysin (Atl). The physiological relevance of previously uncharacterized BglA and TreC was addressed in CRISPRi and inhibitor studies with the putative TreC inhibitor α-cyclophellitol-aziridine. Silencing of treC did not affect bacterial growth in rich media, but reduced biofilm formation in vitro, and attenuated virulence during Galleria mellonella infection, warranting future investigations into the biochemical function of this enzyme.
Collapse
Affiliation(s)
- Md Jalal Uddin
- Centre
for New Antibacterial Strategies (CANS) and Research Group for Host-Microbe
Interactions, Department of Medical Biology (IMB), UiT—The Arctic University of Norway, 9019 Tromsø, Norway
| | - Kjersti Julin
- Centre
for New Antibacterial Strategies (CANS) and Research Group for Host-Microbe
Interactions, Department of Medical Biology (IMB), UiT—The Arctic University of Norway, 9019 Tromsø, Norway
| | - Herman S. Overkleeft
- Leiden
Institute of Chemistry, Leiden University, Einsteinweg 55, 2333 CC, Leiden, The Netherlands
| | - Mona Johannessen
- Centre
for New Antibacterial Strategies (CANS) and Research Group for Host-Microbe
Interactions, Department of Medical Biology (IMB), UiT—The Arctic University of Norway, 9019 Tromsø, Norway
| | - Christian S. Lentz
- Centre
for New Antibacterial Strategies (CANS) and Research Group for Host-Microbe
Interactions, Department of Medical Biology (IMB), UiT—The Arctic University of Norway, 9019 Tromsø, Norway
| |
Collapse
|
3
|
Yang Y, Liu X, Sun C, Fang Y, Qu D, Tang Z, Sun Z, Zhou X, Wang D. Synthesis and Evaluation of Colchicine C-Cyclic AmineDerivatives as Potent Anti-Biofilms Agents AgainstMethicillin-Resistant Staphylococcus aureus. Antibiotics (Basel) 2025; 14:173. [PMID: 40001417 PMCID: PMC11851440 DOI: 10.3390/antibiotics14020173] [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: 12/31/2024] [Revised: 02/01/2025] [Accepted: 02/07/2025] [Indexed: 02/27/2025] Open
Abstract
BACKGROUND/OBJECTIVES The elimination of bacterial biofilm formation is an effective strategy against bacterial infections. The objective was to design 27 colchicine C-ring modified amine derivatives and evaluate their inhibitory activities against the biofilms of MRSA USA300. METHODS Design 27 colchicine C-ring modified amine derivatives. Evaluate their inhibitory activities against MRSA USA300 biofilms. Conduct antibacterial or synergistic antibacterial experiments. Research the phenotypic mechanisms related to biofilm-related genes icaA and agrA. RESULTS The experiments showed that most compounds in this series exhibited varying degrees of biofilm inhibitory activity (with inhibition rates ranging from 7.72% to 40.79%). Further verification through antibacterial or synergistic antibacterial experiments revealed that the compounds with biofilm-inhibiting effects (compounds 7b-11b) generally had certain antibacterial activities (MICs = 16-32 μg/mL) or synergistic antibacterial effects (FICIs < 0.5). Furthermore, through in-depth research on their phenotypic mechanisms (i.e., research on biofilm-related mechanisms), it was found that the compounds with antibacterial or synergistic antibacterial properties could inhibit the formation of biofilms by affecting the regulation of the biofilm-related genes icaA and agrA. CONCLUSIONS The designed colchicine C-ring modified amine derivatives showed potential in inhibiting MRSA biofilms, and their antibacterial or synergistic antibacterial properties are related to the regulation of biofilm-related genes icaA and agrA, demonstrating inhibitory activity against MRSA.
Collapse
Affiliation(s)
- Yuxin Yang
- College of Animal Science, Jilin University, Changchun 130062, China; (Y.Y.); (X.L.); (C.S.); (D.Q.); (Z.S.)
- School of Pharmaceutical Science, Jilin University, Changchun 130021, China; (Y.F.); (Z.T.)
| | - Xin Liu
- College of Animal Science, Jilin University, Changchun 130062, China; (Y.Y.); (X.L.); (C.S.); (D.Q.); (Z.S.)
| | - Can Sun
- College of Animal Science, Jilin University, Changchun 130062, China; (Y.Y.); (X.L.); (C.S.); (D.Q.); (Z.S.)
| | - Yuan Fang
- School of Pharmaceutical Science, Jilin University, Changchun 130021, China; (Y.F.); (Z.T.)
| | - Danyang Qu
- College of Animal Science, Jilin University, Changchun 130062, China; (Y.Y.); (X.L.); (C.S.); (D.Q.); (Z.S.)
| | - Zhengbin Tang
- School of Pharmaceutical Science, Jilin University, Changchun 130021, China; (Y.F.); (Z.T.)
| | - Zetao Sun
- College of Animal Science, Jilin University, Changchun 130062, China; (Y.Y.); (X.L.); (C.S.); (D.Q.); (Z.S.)
| | - Xiaoping Zhou
- School of Pharmaceutical Science, Jilin University, Changchun 130021, China; (Y.F.); (Z.T.)
| | - Dacheng Wang
- College of Animal Science, Jilin University, Changchun 130062, China; (Y.Y.); (X.L.); (C.S.); (D.Q.); (Z.S.)
| |
Collapse
|
4
|
Spiegel C, Ünalan B, Kaserbacher A, Arora R, Coraça-Huber DC. Eicosapentaenoic Acid and Docosahexaenoic Acid as an Antimicrobial Agent in Orthopedics-An In Vitro Study About the Race for Surface. Pathogens 2025; 14:57. [PMID: 39861018 PMCID: PMC11768219 DOI: 10.3390/pathogens14010057] [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: 10/24/2024] [Revised: 01/03/2025] [Accepted: 01/09/2025] [Indexed: 01/27/2025] Open
Abstract
BACKGROUND The burden of prosthetic joint infection in combination with antibiotic-resistant bacterial strains is a rising dilemma for patients experiencing total joint replacements. Around 0.8-2% of patients experience prosthetic joint infections, while up to 21% of patients are considered fatal cases after 5 years. Staphylococcus aureus is one of the main reasons for prosthetic joint infections. Its capability of forming biofilms and developing mechanisms against antibiotics is one of the most dangerous clinical topics being currently discussed. Previous studies have shown the promising results of omega-3 fatty acids as an antimicrobial agent against Staphylococcus aureus. Though an antimicrobial effect has been examined, the influence of polyunsaturated fatty acids on Staphylococcus aureus in the presence of human osteoblasts has not been reported yet. In this study, we aimed to investigate the influence of omega-3 fatty acids on the biofilm formation of Staphylococcus aureus ATCC 29213 in the presence of hFOB 1.19 cells. The co-culture setup helped to examine the influence of omega-3 fatty acids on the race for surface to simulate prosthetic joint infections. METHODS In this study, we tested Staphylococcus aureus ATCC 29213 co-cultured with human fetal osteoblasts hFOB 1.19 in the presence of sub-MIC and MIC concentrations of docosahexaenoic acid (1.25 mg/L, 2.5 mg/L) and eicosapentaenoic acid (0.15 mg/L, 0.3 mg/L) after 1, 6 and 24 h of incubation. After establishing the co-culture, cell culture and biofilm, we performed colony-forming unit counting and cell counting to examine cell survivability. In addition, we carried out scanning electron microscopy to study the race for surface behaviour of the cells. RESULTS We found a protective influence of omega-3 fatty acids on osteoblasts when present in co-culture with Staphylococcus aureus after 6 h of incubation. Omega-3 fatty acids increase the cell survival of osteoblasts after 6 h in a co-culture with bacteria and are able to influence the race for surface. In this study, the strain of Staphylcoccus aureus ATCC 29213 showed signs of growth inhibition within the first 6 h. CONCLUSIONS Omega-3 fatty acids can be a valuable antimicrobial agent in terms of decreasing the risk of on-site infection during surgery. Omega-3 fatty acids were shown to decrease the bacterial load within the first 6 h of incubation and increase the survivability of osteoblasts.
Collapse
Affiliation(s)
- Christopher Spiegel
- Research Laboratory for Biofilms and Implant Associated Infections (BIOFILM LAB), University Hospital for Orthopaedics and Traumatology, Medical University of Innsbruck, Müllerstraße 44, 6020 Innsbruck, Austria; (B.Ü.); (A.K.); (D.C.C.-H.)
| | - Burak Ünalan
- Research Laboratory for Biofilms and Implant Associated Infections (BIOFILM LAB), University Hospital for Orthopaedics and Traumatology, Medical University of Innsbruck, Müllerstraße 44, 6020 Innsbruck, Austria; (B.Ü.); (A.K.); (D.C.C.-H.)
| | - Andreas Kaserbacher
- Research Laboratory for Biofilms and Implant Associated Infections (BIOFILM LAB), University Hospital for Orthopaedics and Traumatology, Medical University of Innsbruck, Müllerstraße 44, 6020 Innsbruck, Austria; (B.Ü.); (A.K.); (D.C.C.-H.)
| | - Rohit Arora
- Department of Orthopaedics and Traumatology, Medical University of Innsbruck, Anichstraße 35, 6020 Innsbruck, Austria;
| | - Débora C. Coraça-Huber
- Research Laboratory for Biofilms and Implant Associated Infections (BIOFILM LAB), University Hospital for Orthopaedics and Traumatology, Medical University of Innsbruck, Müllerstraße 44, 6020 Innsbruck, Austria; (B.Ü.); (A.K.); (D.C.C.-H.)
| |
Collapse
|
5
|
Salikin NH, Keong LC, Azemin WA, Philip N, Yusuf N, Daud SA, Rashid SA. Combating multidrug-resistant (MDR) Staphylococcus aureus infection using terpene and its derivative. World J Microbiol Biotechnol 2024; 40:402. [PMID: 39627623 DOI: 10.1007/s11274-024-04190-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: 07/24/2024] [Accepted: 11/01/2024] [Indexed: 12/13/2024]
Abstract
Multidrug-resistant (MDR) Staphylococcus aureus represents a major global health issue resulting in a wide range of debilitating infections and fatalities. The slow progression of new antibiotics, limited choices for treatment, and scarcity of new drug approvals create immense obstacles in new drug line development. S. aureus poses a significant public health risk, due to the emergence of methicillin-resistant (MRSA) and vancomycin-resistant strains (VRSA), necessitating novel antibiotics for effective control management. Current studies are delving into the terpenes' potential as an antimicrobial agent, indicating positive prospects as promising substitutes or complementary to conventional antibiotics. Concurrent reactions of terpenes with conventional antibiotics create synergistic effects that significantly enhance antibiotic efficacy. Accumulated evidence has shown that while efflux pump (e.g., NorA, TetK, and MepA) is revealed as an essential defense of S. aureus against antibiotics, terpene and its derivative act as its potent inhibitor, suggesting the promising potential of terpenes in combating those infectious pathogens. Furthermore, pronounced cell membrane disruptive activity and antibiofilm properties by terpenes have been exerted, signifying their significance as promising prevention against microbial pathogenesis and antimicrobial resistance. This review provides an overview of the potential of terpenes and their derivatives in combating S. aureus infections, highlighting their potential mechanisms of action (MOA), synergistic effects with conventional antibiotics, and challenges in clinical translation. The unique properties of terpenes offer an opportunity for their use in developing an exceptional defense strategy against antibiotic-resistant S. aureus.
Collapse
Affiliation(s)
- Nor Hawani Salikin
- School of Industrial Technology, Universiti Sains Malaysia, 11800, Minden Pulau Pinang, Malaysia
| | - Lee Chee Keong
- School of Industrial Technology, Universiti Sains Malaysia, 11800, Minden Pulau Pinang, Malaysia
| | - Wan-Atirah Azemin
- School of Biological Sciences, Universiti Sains Malaysia, 11800, Minden Pulau Pinang, Malaysia
| | - Noraini Philip
- School of Biological Sciences, Universiti Sains Malaysia, 11800, Minden Pulau Pinang, Malaysia
| | - Nurhaida Yusuf
- Department of Chemistry, Faculty of Mathematics and Natural Sciences, Syiah Kuala University, Aceh, Indonesia
| | - Siti Aisyah Daud
- School of Biological Sciences, Universiti Sains Malaysia, 11800, Minden Pulau Pinang, Malaysia
| | - Syarifah Ab Rashid
- School of Biological Sciences, Universiti Sains Malaysia, 11800, Minden Pulau Pinang, Malaysia.
| |
Collapse
|
6
|
Korshoj LE, Kielian T. Bacterial single-cell RNA sequencing captures biofilm transcriptional heterogeneity and differential responses to immune pressure. Nat Commun 2024; 15:10184. [PMID: 39580490 PMCID: PMC11585574 DOI: 10.1038/s41467-024-54581-8] [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: 09/18/2024] [Accepted: 11/14/2024] [Indexed: 11/25/2024] Open
Abstract
Biofilm formation is an important mechanism of survival and persistence for many bacterial pathogens. These multicellular communities contain subpopulations of cells that display metabolic and transcriptional diversity along with recalcitrance to antibiotics and host immune defenses. Here, we present an optimized bacterial single-cell RNA sequencing method, BaSSSh-seq, to study Staphylococcus aureus diversity during biofilm growth and transcriptional adaptations following immune cell exposure. BaSSSh-seq captures extensive transcriptional heterogeneity during biofilm compared to planktonic growth. We quantify and visualize transcriptional regulatory networks across heterogeneous biofilm subpopulations and identify gene sets that are associated with a trajectory from planktonic to biofilm growth. BaSSSh-seq also detects alterations in biofilm metabolism, stress response, and virulence induced by distinct immune cell populations. This work facilitates the exploration of biofilm dynamics at single-cell resolution, unlocking the potential for identifying biofilm adaptations to environmental signals and immune pressure.
Collapse
Affiliation(s)
- Lee E Korshoj
- Department of Pathology, Microbiology, and Immunology, University of Nebraska Medical Center, Omaha, NE, USA.
| | - Tammy Kielian
- Department of Pathology, Microbiology, and Immunology, University of Nebraska Medical Center, Omaha, NE, USA.
| |
Collapse
|
7
|
Cheng X, Shi Y, Liu Y, Xu Y, Ma J, Ma L, Wang Z, Guo S, Su J. Adaptive physiological and metabolic alterations in Staphylococcus aureus evolution under vancomycin exposure. World J Microbiol Biotechnol 2024; 40:322. [PMID: 39283509 DOI: 10.1007/s11274-024-04128-2] [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: 05/02/2024] [Accepted: 08/31/2024] [Indexed: 10/17/2024]
Abstract
Staphylococcus aureus can develop antibiotic resistance and evade immune responses, causing infections in different body sites. However, the metabolic changes underlying this process are poorly understood. A variant strain, C1V, was derived from the parental strain C1 by exposing it to increasing concentrations of vancomycin in vitro. C1V exhibited a vancomycin-intermediate phenotype and physiological changes compared to C1. It showed higher survival rates than C1 when phagocytosed by Raw264.7 cells. Metabolomics analysis identified significant metabolic differences pre- and post-induction (C1 + SC1 vs. C1V + SC1V: 201 metabolites) as well as pre- and post-phagocytosis (C1 vs. SC1: 50 metabolites; C1V vs. SC1V: 95 metabolites). The variant strain had distinct morphological characteristics, decreased adhesion ability, impaired virulence, and enhanced resistance to phagocytosis compared to the parental strain. Differential metabolites may contribute to S. aureus ' resistance to antibiotics and phagocytosis, offering insights into potential strategies for altering vancomycin nonsusceptibility and enhancing phagocyte killing by manipulating bacterial metabolism.
Collapse
Affiliation(s)
- Xin Cheng
- Clinical Laboratory Center, Beijing Friendship Hospital, Capital Medical University, Beijing, 100050, China
| | - Yue Shi
- Clinical Laboratory Center, Beijing Friendship Hospital, Capital Medical University, Beijing, 100050, China
| | - Yadong Liu
- State Key Laboratory of Microbial Resources, Institute of Microbiology, Chinese Academy of Sciences, Beijing, 100101, China
- University of Chinese Academy of Sciences, Beijing, 101408, China
| | - Yibin Xu
- State Key Laboratory of Microbial Resources, Institute of Microbiology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Jingxin Ma
- Clinical Laboratory Center, Beijing Friendship Hospital, Capital Medical University, Beijing, 100050, China
| | - Liyan Ma
- Clinical Laboratory Center, Beijing Friendship Hospital, Capital Medical University, Beijing, 100050, China
| | - Zerui Wang
- Biomedical Sciences College & Shandong Medical Biotechnology Centre, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, 250117, Shandong, China
| | - Shuilong Guo
- Department of Gastroenterology, Beijing Friendship Hospital, Capital Medical University, Beijing, 100050, China.
| | - Jianrong Su
- Clinical Laboratory Center, Beijing Friendship Hospital, Capital Medical University, Beijing, 100050, China.
| |
Collapse
|
8
|
Tian LL, Li Y, Yang R, Jiang Y, He JJ, Wang H, Chen LQ, Zhu WY, Xue T, Li BB. Low concentrations of tetrabromobisphenol A promote the biofilm formation of methicillin-resistant Staphylococcus aureus. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2024; 283:116853. [PMID: 39137468 DOI: 10.1016/j.ecoenv.2024.116853] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/31/2024] [Revised: 08/02/2024] [Accepted: 08/06/2024] [Indexed: 08/15/2024]
Abstract
The effect and underlying mechanism of tetrabromobisphenol A (TBBPA), a plastic additive, on biofilm formation of methicillin-resistant Staphylococcus aureus (MRSA USA300) remain unknown. This study first investigated the impact of different concentrations of TBBPA on the growth and biofilm formation of USA300. The results indicated that a low concentration (0.5 mg/L) of TBBPA promoted the growth and biofilm formation of USA300, whereas high concentrations (5 mg/L and 10 mg/L) of TBBPA had inhibitory effects. Further exploration revealed that the low concentration of TBBPA enhance biofilm formation by promoting the synthesis of extracellular proteins, release of extracellular DNA (eDNA), and production of staphyloxanthin. RTqPCR analysis demonstrated that the low concentration of TBBPA upregulated genes associated with extracellular protein synthesis (sarA, fnbA, fnbB, aur) and eDNA formation (atlA) and increased the expression of genes involved in staphyloxanthin biosynthesis (crtM), suggesting a potential mechanism for enhanced resistance of USA300 to adverse conditions. These findings shed light on how low concentrations of TBBPA facilitate biofilm formation in USA300 and highlight the indirect impact of plastic additives on pathogenic bacteria in terms of human health. In the future, in-depth studies about effects of plastic additives on pathogenicity of pathogenic bacteria should be conducted. CAPSULE: The protein and eDNA contents in biofilms of methicillin-resistant Staphylococcus aureus are increased by low concentrations of TBBPA.
Collapse
Affiliation(s)
- Lin-Lin Tian
- School of Life Sciences, Anhui Agricultural University, Hefei, Anhui 230036, China
| | - Yun Li
- School of Life Sciences, Anhui Agricultural University, Hefei, Anhui 230036, China
| | - Rui Yang
- School of Life Sciences, Anhui Agricultural University, Hefei, Anhui 230036, China
| | - Ying Jiang
- School of Life Sciences, Anhui Agricultural University, Hefei, Anhui 230036, China
| | - Jiao-Jiao He
- School of Life Sciences, Anhui Agricultural University, Hefei, Anhui 230036, China
| | - Hui Wang
- School of Life Sciences, Anhui Agricultural University, Hefei, Anhui 230036, China
| | - Li-Qi Chen
- School of Life Sciences, Anhui Agricultural University, Hefei, Anhui 230036, China
| | - Wen-Ya Zhu
- School of Life Sciences, Anhui Agricultural University, Hefei, Anhui 230036, China
| | - Ting Xue
- School of Life Sciences, Anhui Agricultural University, Hefei, Anhui 230036, China; Food Procession Research Institute, Anhui Agricultural University, Hefei, Anhui 230036, China.
| | - Bing-Bing Li
- School of Life Sciences, Anhui Agricultural University, Hefei, Anhui 230036, China.
| |
Collapse
|
9
|
Rimal B, Chang JD, Liu C, Kim H, Aderotoye O, Zechmann B, Kim SJ. Scanning Electron Microscopy and Energy-Dispersive X-ray Spectroscopy of Staphylococcus aureus Biofilms. ACS OMEGA 2024; 9:37610-37620. [PMID: 39281927 PMCID: PMC11391442 DOI: 10.1021/acsomega.4c01168] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Revised: 06/06/2024] [Accepted: 07/01/2024] [Indexed: 09/18/2024]
Abstract
Understanding the dynamics of biofilm formation and its elemental composition is crucial for developing effective strategies against biofilm-associated infections. In this study, we employed scanning electron microscopy (SEM) and energy-dispersive X-ray spectroscopy (EDS) to investigate the morphological changes and elemental compositions of Staphylococcus aureus biofilms. SEM images revealed distinct stages of biofilm development, from initial aggregation to the formation of mature and aged biofilms. EDS analysis consistently showed elevated levels of sodium (Na), oxygen (O), and phosphorus (P) in the biofilm matrix, indicating its high negative charge and the presence of anionic biopolymers. The incorporation of extracellular DNA (eDNA) into the biofilm matrix, leading to significant retention of sodium ions, underscored the importance of electrostatic interactions in biofilm formation and stability. Our findings highlight the potential of EDS analysis in quantifying elemental compositions and elucidating the role of anionic biopolymers in biofilm development.
Collapse
Affiliation(s)
- Binayak Rimal
- Institute of Biomedical Studies, Baylor University, Waco, Texas 76798, United States
| | - James D Chang
- Department of Chemistry and Biochemistry, Baylor University, Waco, Texas 76798, United States
| | - Chengyin Liu
- Department of Chemistry, Howard University, Washington, District of Columbia 20059, United States
| | - Haley Kim
- Department of Chemistry, University of Maryland, College Park, Maryland 20742, United States
| | - Oluwatobi Aderotoye
- Department of Chemistry, Howard University, Washington, District of Columbia 20059, United States
| | - Bernd Zechmann
- Center for Microscopy and Imaging, Baylor University, Waco, Texas 76798, United States
| | - Sung Joon Kim
- Department of Chemistry, Howard University, Washington, District of Columbia 20059, United States
| |
Collapse
|
10
|
de Oliveira LD, Ribeiro ALM, Dias SDO, da Cruz GM, de Menezes RT, de Carvalho LS, Diamantino MGG, Pereira TC, Marcucci MC, Abu Hasna A. Phytochemical Composition and Antimicrobial and Antibiofilm Effect of Myrciaria cauliflora Hydroethanolic Extract against Staphylococcus aureus and Acinetobacter baumannii. Methods Protoc 2024; 7:60. [PMID: 39195438 PMCID: PMC11357044 DOI: 10.3390/mps7040060] [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: 07/12/2024] [Revised: 07/29/2024] [Accepted: 07/30/2024] [Indexed: 08/29/2024] Open
Abstract
Staphylococcus aureus and Acinetobacter baumannii are opportunistic pathogens, and both are involved in different oral infections. This work aimed to analyze the phytochemical composition of Myrciaria cauliflora hydroethanolic extract and to evaluate its antimicrobial and antibiofilm action against Staphylococcus aureus (ATCC 6538) and Acinetobacter baumannii (ATCC 19606; multi-resistant clinical strains 58004, 50098, 566006, and H557). Myrciaria cauliflora hydroethanolic extract was prepared, and the content of soluble solids, flavonoids, and phenols was quantified. High-performance liquid chromatography (HPLC) was performed later. The minimum inhibitory concentration was determined using the broth microdilution method according to the Clinical and Laboratory Standards Institute, standard M7-A6, and subsequently, its minimum bactericidal concentration was determined. Then, the most effective concentrations were analyzed against biofilms. Statistical analysis was performed using the ANOVA method with Tukey's test. The soluble solids content in the prepared hydroethanolic extract of M. cauliflora was 2.22%. Additionally, the total flavonoid content, measured using the quercetin standard curve, was 0.040 mg/mL. Furthermore, the total phenol content, determined using the gallic acid standard curve, was 0.729 mg/mL. HPLC analysis presented peaks of gallic acid (11.80 m), p-coumaric acid (12.09 m), cinnamic acid derivative (19.02 m), and ellagic acid (29.83 m). The extract demonstrated antimicrobial and antibiofilm action against all tested strains. However, the most effective antibacterial concentration against all the tested bacteria was 5.55 mg/mL. Therefore, these chemical components justify that M. cauliflora hydroethanolic extract is effective in reducing biofilm formation in S. aureus (standard strain) and A. baumannii (standard and clinical strains).
Collapse
Affiliation(s)
- Luciane Dias de Oliveira
- Department of Bioscience and Oral Diagnosis, Institute of Science and Technology, São Paulo State University, Campus of São José dos Campos, São Paulo 12245-000, Brazil; (L.D.d.O.); (S.d.O.D.); (G.M.d.C.); (R.T.d.M.); (L.S.d.C.); (T.C.P.); (M.C.M.)
| | - Ana Luisa Monteiro Ribeiro
- Campus São José dos Campos, Universidade Paulista—UNIP, Highway Presidente Dutra, km 157.5, South Lane, São José dos Campos, São Paulo 12240-420, Brazil;
| | - Sthéfani de Oliveira Dias
- Department of Bioscience and Oral Diagnosis, Institute of Science and Technology, São Paulo State University, Campus of São José dos Campos, São Paulo 12245-000, Brazil; (L.D.d.O.); (S.d.O.D.); (G.M.d.C.); (R.T.d.M.); (L.S.d.C.); (T.C.P.); (M.C.M.)
| | - Geovani Moreira da Cruz
- Department of Bioscience and Oral Diagnosis, Institute of Science and Technology, São Paulo State University, Campus of São José dos Campos, São Paulo 12245-000, Brazil; (L.D.d.O.); (S.d.O.D.); (G.M.d.C.); (R.T.d.M.); (L.S.d.C.); (T.C.P.); (M.C.M.)
| | - Raquel Teles de Menezes
- Department of Bioscience and Oral Diagnosis, Institute of Science and Technology, São Paulo State University, Campus of São José dos Campos, São Paulo 12245-000, Brazil; (L.D.d.O.); (S.d.O.D.); (G.M.d.C.); (R.T.d.M.); (L.S.d.C.); (T.C.P.); (M.C.M.)
| | - Lara Steffany de Carvalho
- Department of Bioscience and Oral Diagnosis, Institute of Science and Technology, São Paulo State University, Campus of São José dos Campos, São Paulo 12245-000, Brazil; (L.D.d.O.); (S.d.O.D.); (G.M.d.C.); (R.T.d.M.); (L.S.d.C.); (T.C.P.); (M.C.M.)
| | - Mariana Gadelho Gimenez Diamantino
- Department of Restorative Dentistry, Endodontics Division, Institute of Science and Technology, São Paulo State University, Campus of São José dos Campos, São Paulo 12245-000, Brazil;
| | - Thaís Cristine Pereira
- Department of Bioscience and Oral Diagnosis, Institute of Science and Technology, São Paulo State University, Campus of São José dos Campos, São Paulo 12245-000, Brazil; (L.D.d.O.); (S.d.O.D.); (G.M.d.C.); (R.T.d.M.); (L.S.d.C.); (T.C.P.); (M.C.M.)
| | - Maria Cristina Marcucci
- Department of Bioscience and Oral Diagnosis, Institute of Science and Technology, São Paulo State University, Campus of São José dos Campos, São Paulo 12245-000, Brazil; (L.D.d.O.); (S.d.O.D.); (G.M.d.C.); (R.T.d.M.); (L.S.d.C.); (T.C.P.); (M.C.M.)
| | - Amjad Abu Hasna
- Department of Restorative Dentistry, Endodontics Division, Institute of Science and Technology, São Paulo State University, Campus of São José dos Campos, São Paulo 12245-000, Brazil;
- School of Dentistry, Universidad Espíritu Santo, Samborondón 092301, Ecuador
| |
Collapse
|
11
|
Xu Z, Li Y, Xu A, Soteyome T, Yuan L, Ma Q, Seneviratne G, Li X, Liu J. Cell-wall-anchored proteins affect invasive host colonization and biofilm formation in Staphylococcus aureus. Microbiol Res 2024; 285:127782. [PMID: 38833832 DOI: 10.1016/j.micres.2024.127782] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2023] [Revised: 05/21/2024] [Accepted: 05/21/2024] [Indexed: 06/06/2024]
Abstract
As a major human and animal pathogen, Staphylococcus aureus can attach to medical implants (abiotic surface) or host tissues (biotic surface), and further establish robust biofilms which enhances resistance and persistence to host immune system and antibiotics. Cell-wall-anchored proteins (CWAPs) covalently link to peptidoglycan, and largely facilitate the colonization of S. aureus on various surfaces (including adhesion and biofilm formation) and invasion into host cells (including adhesion, immune evasion, iron acquisition and biofilm formation). During biofilm formation, CWAPs function in adhesion, aggregation, collagen-like fiber network formation, and consortia formation. In this review, we firstly focus on the structural features of CWAPs, including their intracellular function and interactions with host cells, as well as the functions and ligand binding of CWAPs in different stages of S. aureus biofilm formation. Then, the roles of CWAPs in different biofilm processes with regards in development of therapeutic approaches are clarified, followed by the association between CWAPs genes and clonal lineages. By touching upon these aspects, we hope to provide comprehensive knowledge and clearer understanding on the CWAPs of S. aureus and their roles in biofilm formation, which may further aid in prevention and treatment infection and vaccine development.
Collapse
Affiliation(s)
- Zhenbo Xu
- School of Food Science and Engineering, Guangdong Province Key Laboratory for Green Processing of Natural Products and Product Safety, Engineering Research Center of Starch and Vegetable Protein Processing Ministry of Education, South China University of Technology, Guangzhou 510640, China; Department of Laboratory Medicine, the Second Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong, China.
| | - Yaqin Li
- School of Food Science and Engineering, Guangdong Province Key Laboratory for Green Processing of Natural Products and Product Safety, Engineering Research Center of Starch and Vegetable Protein Processing Ministry of Education, South China University of Technology, Guangzhou 510640, China
| | - Aijuan Xu
- Guangzhou Hybribio Medical Laboratory, Guangzhou 510730, China
| | - Thanapop Soteyome
- Home Economics Technology, Rajamangala University of Technology Phra Nakhon, Bangkok, Thailand
| | - Lei Yuan
- School of Food Science and Engineering, Yangzhou University, Yangzhou, Jiangsu 225127, China
| | - Qin Ma
- Sericultural & Agri-Food Research Institute Guangdong Academy of Agricultural Sciences/Key Laboratory of Functional Foods, Ministry of Agriculture /Guangdong Key Laboratory of Agricultural Products Processing, Guangzhou 510610, China
| | - Gamini Seneviratne
- National Institute of Fundamental Studies, Hantana road, Kandy, Sri Lanka
| | - Xuejie Li
- School of Food Science and Engineering, Guangdong Province Key Laboratory for Green Processing of Natural Products and Product Safety, Engineering Research Center of Starch and Vegetable Protein Processing Ministry of Education, South China University of Technology, Guangzhou 510640, China.
| | - Junyan Liu
- College of Light Industry and Food Science, Guangdong Provincial Key Laboratory of Lingnan Specialty Food Science and Technology, Academy of Contemporary Agricultural Engineering Innovations, Zhongkai University of Agriculture and Engineering, Guangzhou 510225, China; Key Laboratory of Green Processing and Intelligent Manufacturing of Lingnan Specialty Food, Ministry of Agriculture, Guangzhou 510225, China.
| |
Collapse
|
12
|
Korshoj LE, Kielian T. Bacterial single-cell RNA sequencing captures biofilm transcriptional heterogeneity and differential responses to immune pressure. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.28.601229. [PMID: 38979200 PMCID: PMC11230364 DOI: 10.1101/2024.06.28.601229] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/10/2024]
Abstract
Biofilm formation is an important mechanism of survival and persistence for many bacterial pathogens. These multicellular communities contain subpopulations of cells that display vast metabolic and transcriptional diversity along with high recalcitrance to antibiotics and host immune defenses. Investigating the complex heterogeneity within biofilm has been hindered by the lack of a sensitive and high-throughput method to assess stochastic transcriptional activity and regulation between bacterial subpopulations, which requires single-cell resolution. We have developed an optimized bacterial single-cell RNA sequencing method, BaSSSh-seq, to study Staphylococcus aureus diversity during biofilm growth and transcriptional adaptations following immune cell exposure. We validated the ability of BaSSSh-seq to capture extensive transcriptional heterogeneity during biofilm compared to planktonic growth. Application of new computational tools revealed transcriptional regulatory networks across the heterogeneous biofilm subpopulations and identification of gene sets that were associated with a trajectory from planktonic to biofilm growth. BaSSSh-seq also detected alterations in biofilm metabolism, stress response, and virulence that were tailored to distinct immune cell populations. This work provides an innovative platform to explore biofilm dynamics at single-cell resolution, unlocking the potential for identifying biofilm adaptations to environmental signals and immune pressure.
Collapse
|
13
|
Bowden LC, Finlinson J, Jones B, Berges BK. Beyond the double helix: the multifaceted landscape of extracellular DNA in Staphylococcus aureus biofilms. Front Cell Infect Microbiol 2024; 14:1400648. [PMID: 38903938 PMCID: PMC11188362 DOI: 10.3389/fcimb.2024.1400648] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Accepted: 05/17/2024] [Indexed: 06/22/2024] Open
Abstract
Staphylococcus aureus forms biofilms consisting of cells embedded in a matrix made of proteins, polysaccharides, lipids, and extracellular DNA (eDNA). Biofilm-associated infections are difficult to treat and can promote antibiotic resistance, resulting in negative healthcare outcomes. eDNA within the matrix contributes to the stability, growth, and immune-evasive properties of S. aureus biofilms. eDNA is released by autolysis, which is mediated by murein hydrolases that access the cell wall via membrane pores formed by holin-like proteins. The eDNA content of S. aureus biofilms varies among individual strains and is influenced by environmental conditions, including the presence of antibiotics. eDNA plays an important role in biofilm development and structure by acting as an electrostatic net that facilitates protein-cell and cell-cell interactions. Because of eDNA's structural importance in biofilms and its ubiquitous presence among S. aureus isolates, it is a potential target for therapeutics. Treatment of biofilms with DNase can eradicate or drastically reduce them in size. Additionally, antibodies that target DNABII proteins, which bind to and stabilize eDNA, can also disperse biofilms. This review discusses the recent literature on the release, structure, and function of eDNA in S. aureus biofilms, in addition to a discussion of potential avenues for targeting eDNA for biofilm eradication.
Collapse
Affiliation(s)
| | | | | | - Bradford K. Berges
- Department of Microbiology and Molecular Biology, Brigham Young University, Provo, UT, United States
| |
Collapse
|
14
|
Yang Y, Yao Z, Zhang J, Shao W, Li B, Wu H, Tang W, Zhang J. Inhibiting Peptidoglycan Hydrolase Alleviates MRSA Pneumonia Through Autolysin-Mediated MDP-NOD2 Pathway. Infect Drug Resist 2024; 17:1231-1242. [PMID: 38560705 PMCID: PMC10981453 DOI: 10.2147/idr.s455339] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2023] [Accepted: 03/15/2024] [Indexed: 04/04/2024] Open
Abstract
Background Methicillin-resistant Staphylococcus aureus (MRSA) is a cause of staph infection that is difficult to treat because of resistance to some antibiotics. A recent study indicated that diarylurea ZJ-2 is a novel antibacterial agent against multi-drug resistant Enterococcus faecium. In this work, we refined the bactericidal mechanism of ZJ-2 as a peptidoglycan (PG) hydrolase by affecting AtlA-mediated PG homeostasis. Methods A wild-type strain (WT) and a mutant strain (ΔatlA) were used to investigate the effects of ZJ-2 on the cell wall, PG, and autolysin regulatory system by antimicrobial susceptibility testing, hemolytic toxin assay, microanalysis, autolysis assay, qRT-PCR, ELISA and mouse model of pneumonia. Results The results revealed that ZJ-2 down-regulated the expression of genes related to peptidoglycan hydrolase (PGH) (sprX, walR, atlA, and lytM), and reduced the levels of PG, muramyl dipeptide (MDP), cytokines, and hemolytic toxin, while ΔatlA interfered with the genes regulation and PG homeostasis. In the mouse MRSA pneumonia model, the same trend was observed in the nucleotide oligomerization domain protein 2 (NOD2) and relative proinflammatory factors. Conclusion ZJ-2 may act as a novel inhibitor of PG hydrolyse, disrupting autolysin-mediated PG homeostasis, and reducing inflammation by down-regulating the MDP-NOD2 pathway.
Collapse
Affiliation(s)
- Yang Yang
- School of Medicine, Anhui University of Science and Technology, Huainan, People’s Republic of China
- Anhui Province Key Laboratory of Occupational Health, Anhui No.2 Provincial People’s Hospital, Hefei, People’s Republic of China
| | - Zongze Yao
- School of Medicine, Anhui University of Science and Technology, Huainan, People’s Republic of China
| | - Jiazhen Zhang
- School of Medicine, Anhui University of Science and Technology, Huainan, People’s Republic of China
| | - Wei Shao
- School of Pharmacy, Anhui Medical University, Hefei, People’s Republic of China
| | - Bo Li
- Anhui Province Key Laboratory of Occupational Health, Anhui No.2 Provincial People’s Hospital, Hefei, People’s Republic of China
| | - Huihui Wu
- Anhui Province Key Laboratory of Occupational Health, Anhui No.2 Provincial People’s Hospital, Hefei, People’s Republic of China
| | - Wenjian Tang
- School of Pharmacy, Anhui Medical University, Hefei, People’s Republic of China
| | - Jing Zhang
- Anhui Province Key Laboratory of Occupational Health, Anhui No.2 Provincial People’s Hospital, Hefei, People’s Republic of China
| |
Collapse
|
15
|
Aboelnaga N, Elsayed SW, Abdelsalam NA, Salem S, Saif NA, Elsayed M, Ayman S, Nasr M, Elhadidy M. Deciphering the dynamics of methicillin-resistant Staphylococcus aureus biofilm formation: from molecular signaling to nanotherapeutic advances. Cell Commun Signal 2024; 22:188. [PMID: 38519959 PMCID: PMC10958940 DOI: 10.1186/s12964-024-01511-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Accepted: 02/01/2024] [Indexed: 03/25/2024] Open
Abstract
Methicillin-resistant Staphylococcus aureus (MRSA) represents a global threat, necessitating the development of effective solutions to combat this emerging superbug. In response to selective pressures within healthcare, community, and livestock settings, MRSA has evolved increased biofilm formation as a multifaceted virulence and defensive mechanism, enabling the bacterium to thrive in harsh conditions. This review discusses the molecular mechanisms contributing to biofilm formation across its developmental stages, hence representing a step forward in developing promising strategies for impeding or eradicating biofilms. During staphylococcal biofilm development, cell wall-anchored proteins attach bacterial cells to biotic or abiotic surfaces; extracellular polymeric substances build scaffolds for biofilm formation; the cidABC operon controls cell lysis within the biofilm, and proteases facilitate dispersal. Beside the three main sequential stages of biofilm formation (attachment, maturation, and dispersal), this review unveils two unique developmental stages in the biofilm formation process for MRSA; multiplication and exodus. We also highlighted the quorum sensing as a cell-to-cell communication process, allowing distant bacterial cells to adapt to the conditions surrounding the bacterial biofilm. In S. aureus, the quorum sensing process is mediated by autoinducing peptides (AIPs) as signaling molecules, with the accessory gene regulator system playing a pivotal role in orchestrating the production of AIPs and various virulence factors. Several quorum inhibitors showed promising anti-virulence and antibiofilm effects that vary in type and function according to the targeted molecule. Disrupting the biofilm architecture and eradicating sessile bacterial cells are crucial steps to prevent colonization on other surfaces or organs. In this context, nanoparticles emerge as efficient carriers for delivering antimicrobial and antibiofilm agents throughout the biofilm architecture. Although metal-based nanoparticles have been previously used in combatting biofilms, its non-degradability and toxicity within the human body presents a real challenge. Therefore, organic nanoparticles in conjunction with quorum inhibitors have been proposed as a promising strategy against biofilms. As nanotherapeutics continue to gain recognition as an antibiofilm strategy, the development of more antibiofilm nanotherapeutics could offer a promising solution to combat biofilm-mediated resistance.
Collapse
Affiliation(s)
- Nirmeen Aboelnaga
- Center for Genomics, Helmy Institute for Medical Sciences, Zewail City of Science and Technology, Giza, Egypt
- Biomedical Sciences Program, University of Science and Technology, Zewail City of Science and Technology, Giza, Egypt
| | - Salma W Elsayed
- Center for Genomics, Helmy Institute for Medical Sciences, Zewail City of Science and Technology, Giza, Egypt
- Biomedical Sciences Program, University of Science and Technology, Zewail City of Science and Technology, Giza, Egypt
- Department of Microbiology & Immunology, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt
| | - Nehal Adel Abdelsalam
- Center for Genomics, Helmy Institute for Medical Sciences, Zewail City of Science and Technology, Giza, Egypt
- Biomedical Sciences Program, University of Science and Technology, Zewail City of Science and Technology, Giza, Egypt
- Department of Microbiology and Immunology, Faculty of Pharmacy, Cairo University, Cairo, Egypt
| | - Salma Salem
- Center for Genomics, Helmy Institute for Medical Sciences, Zewail City of Science and Technology, Giza, Egypt
- Biomedical Sciences Program, University of Science and Technology, Zewail City of Science and Technology, Giza, Egypt
| | - Nehal A Saif
- Center for Genomics, Helmy Institute for Medical Sciences, Zewail City of Science and Technology, Giza, Egypt
- Biomedical Sciences Program, University of Science and Technology, Zewail City of Science and Technology, Giza, Egypt
| | - Manar Elsayed
- Center for Genomics, Helmy Institute for Medical Sciences, Zewail City of Science and Technology, Giza, Egypt
- Biomedical Sciences Program, University of Science and Technology, Zewail City of Science and Technology, Giza, Egypt
| | - Shehab Ayman
- Center for Genomics, Helmy Institute for Medical Sciences, Zewail City of Science and Technology, Giza, Egypt
- Biomedical Sciences Program, University of Science and Technology, Zewail City of Science and Technology, Giza, Egypt
| | - Maha Nasr
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt
| | - Mohamed Elhadidy
- Center for Genomics, Helmy Institute for Medical Sciences, Zewail City of Science and Technology, Giza, Egypt.
- Biomedical Sciences Program, University of Science and Technology, Zewail City of Science and Technology, Giza, Egypt.
- Department of Bacteriology, Mycology and Immunology, Faculty of Veterinary Medicine, Mansoura University, Mansoura, Egypt.
| |
Collapse
|
16
|
Crozier D, Gray JM, Maltas JA, Bonomo RA, Burke ZDC, Card KJ, Scott JG. The evolution of diverse antimicrobial responses in vancomycin-intermediate Staphylococcus aureus and its therapeutic implications. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.11.30.569373. [PMID: 38077036 PMCID: PMC10705500 DOI: 10.1101/2023.11.30.569373] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/24/2023]
Abstract
Staphylococcus aureus causes endocarditis, osteomyelitis, and bacteremia. Clinicians often prescribe vancomycin as an empiric therapy to account for methicillin-resistant S. aureus (MRSA) and narrow treatment based on culture susceptibility results. However, these results reflect a single time point before empiric treatment and represent a limited subset of the total bacterial population within the patient. Thus, while they may indicate that the infection is susceptible to a particular drug, this recommendation may no longer be accurate during therapy. Here, we addressed how antibiotic susceptibility changes over time by accounting for evolution. We evolved 18 methicillin-susceptible S. aureus (MSSA) populations under increasing vancomycin concentrations until they reached intermediate resistance levels. Sequencing revealed parallel mutations that affect cell membrane stress response and cell-wall biosynthesis. The populations exhibited repeated cross-resistance to daptomycin and varied responses to meropenem, gentamicin, and nafcillin. We accounted for this variability by deriving likelihood estimates that express a population's probability of exhibiting a drug response following vancomycin treatment. Our results suggest antistaphylococcal penicillins are preferable first-line treatments for MSSA infections but also highlight the inherent uncertainty that evolution poses to effective therapies. Infections may take varied evolutionary paths; therefore, considering evolution as a probabilistic process should inform our therapeutic choices.
Collapse
|
17
|
Ul Haq I, Khan TA, Krukiewicz K. Etiology, pathology, and host-impaired immunity in medical implant-associated infections. J Infect Public Health 2024; 17:189-203. [PMID: 38113816 DOI: 10.1016/j.jiph.2023.11.024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Revised: 10/20/2023] [Accepted: 11/21/2023] [Indexed: 12/21/2023] Open
Abstract
Host impaired immunity and pathogens adhesion factors are the key elements in analyzing medical implant-associated infections (MIAI). The infection chances are further influenced by surface properties of implants. This review addresses the medical implant-associated pathogens and summarizes the etiology, pathology, and host-impaired immunity in MIAI. Several bacterial and fungal pathogens have been isolated from MIAI; together, they form cross-kingdom species biofilms and support each other in different ways. The adhesion factors initiate the pathogen's adherence on the implant's surface; however, implant-induced impaired immunity promotes the pathogen's colonization and biofilm formation. Depending on the implant's surface properties, immune cell functions get slow or get exaggerated and cause immunity-induced secondary complications resulting in resistant depression and immuno-incompetent fibro-inflammatory zone that compromise implant's performance. Such consequences lead to the unavoidable and straightforward conclusion for the downstream transformation of new ideas, such as the development of multifunctional implant coatings.
Collapse
Affiliation(s)
- Ihtisham Ul Haq
- Department of Physical Chemistry and Technology of Polymers, Silesian University of Technology, M. Strzody 9, 44-100 Gliwice, Poland; Joint Doctoral School, Silesian University of Technology, Akademicka 2A, 44-100 Gliwice, Poland; Programa de Pós-graduação em Inovação Tecnológica, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, MG, Brazil.
| | - Taj Ali Khan
- Division of Infectious Diseases & Global Medicine, Department of Medicine, University of Florida, Gainesville, FL, United States; Institute of Pathology and Diagnostic Medicine, Khyber Medical University, Peshawar, Pakistan.
| | - Katarzyna Krukiewicz
- Department of Physical Chemistry and Technology of Polymers, Silesian University of Technology, M. Strzody 9, 44-100 Gliwice, Poland; Centre for Organic and Nanohybrid Electronics, Silesian University of Technology, Konarskiego 22B, 44-100 Gliwice, Poland.
| |
Collapse
|
18
|
Hernández-Cuellar E, Tsuchiya K, Valle-Ríos R, Medina-Contreras O. Differences in Biofilm Formation by Methicillin-Resistant and Methicillin-Susceptible Staphylococcus aureus Strains. Diseases 2023; 11:160. [PMID: 37987271 PMCID: PMC10660471 DOI: 10.3390/diseases11040160] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2023] [Revised: 10/27/2023] [Accepted: 11/02/2023] [Indexed: 11/22/2023] Open
Abstract
Staphylococcus aureus (S. aureus) is a common pathogen involved in community- and hospital-acquired infections. Its biofilm formation ability predisposes it to device-related infections. Methicillin-resistant S. aureus (MRSA) strains are associated with more serious infections and higher mortality rates and are more complex in terms of antibiotic resistance. It is still controversial whether MRSA are indeed more virulent than methicillin-susceptible S. aureus (MSSA) strains. A difference in biofilm formation by both types of bacteria has been suggested, but how only the presence of the SCCmec cassette or mecA influences this phenotype remains unclear. In this review, we have searched for literature studying the difference in biofilm formation by MRSA and MSSA. We highlighted the relevance of the icaADBC operon in the PIA-dependent biofilms generated by MSSA under osmotic stress conditions, and the role of extracellular DNA and surface proteins in the PIA-independent biofilms generated by MRSA. We described the prominent role of surface proteins with the LPXTG motif and hydrolases for the release of extracellular DNA in the MRSA biofilm formation. Finally, we explained the main regulatory systems in S. aureus involved in virulence and biofilm formation, such as the SarA and Agr systems. As most of the studies were in vitro using inert surfaces, it will be necessary in the future to focus on biofilm formation on extracellular matrix components and its relevance in the pathogenesis of infection by both types of strains using in vivo animal models.
Collapse
Affiliation(s)
- Eduardo Hernández-Cuellar
- Laboratorio de Biología Celular y Tisular, Departamento de Morfología, Universidad Autónoma de Aguascalientes, Aguascalientes 20100, C.P., México
| | - Kohsuke Tsuchiya
- Division of Immunology and Molecular Biology, Cancer Research Institute, Kanazawa University, Kakuma-machi, Kanazawa 920-1192, Japan;
| | - Ricardo Valle-Ríos
- Research Division, Faculty of Medicine, Universidad Nacional Autónoma de México (UNAM), Mexico City 04360, C.P., México;
- Laboratory of Research in Immunology and Proteomics, Federico Gómez Children’s Hospital of Mexico, Mexico City 06720, C.P., México
| | - Oscar Medina-Contreras
- Epidemiology, Endocrinology & Nutrition Research Unit, Mexico Children’s Hospital (HIMFG), Mexico City 06720, C.P., México;
| |
Collapse
|
19
|
Wang B, Zhan Q, Xiao Y, Xu Y, Zhao H, Rao L, Wang X, Zhang J, Shen L, Zhou Y, Guo Y, Wu X, Yu J, Yu F. Mupirocin enhances the biofilm formation of Staphylococcus epidermidis in an atlE-dependent manner. Int J Antimicrob Agents 2023; 62:106904. [PMID: 37385560 DOI: 10.1016/j.ijantimicag.2023.106904] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 05/27/2023] [Accepted: 06/25/2023] [Indexed: 07/01/2023]
Abstract
The pathogenicity of Staphylococcus epidermidis is largely attributed to its exceptional ability to form biofilms. Here, we report that mupirocin, an antimicrobial agent widely used for staphylococcal decolonization and anti-infection, strongly stimulates the biofilm formation of S. epidermidis. Although the polysaccharide intercellular adhesin (PIA) production was unaffected, mupirocin significantly facilitated extracellular DNA (eDNA) release by accelerating autolysis, thereby positively triggering cell surface attachment and intercellular agglomeration during biofilm development. Mechanistically, mupirocin regulated the expression of genes encoding for the autolysin AtlE as well as the programmed cell death system CidA-LrgAB. Critically, through gene knockout, we found out that deletion of atlE, but not cidA or lrgA, abolished the enhancement of biofilm formation and eDNA release in response to mupirocin treatment, indicating that atlE is required for this effect. In Triton X-100 induced autolysis assay, mupirocin treated atlE mutant displayed a slower autolysis rate compared with the wild-type strain and complementary strain. Therefore, we concluded that subinhibitory concentrations of mupirocin enhance the biofilm formation of S. epidermidis in an atlE dependent manner. This induction effect could conceivably be responsible for some of the more unfavourable outcomes of infectious diseases.
Collapse
Affiliation(s)
- Bingjie Wang
- Department of Clinical Laboratory, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Qing Zhan
- Department of Clinical Laboratory, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China; Infection Control Department, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yanghua Xiao
- Department of Clinical Laboratory, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Yanlei Xu
- Department of Clinical Laboratory, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Huilin Zhao
- Department of Clinical Laboratory, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Lulin Rao
- Department of Clinical Laboratory, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Xinyi Wang
- Department of Clinical Laboratory, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Jiao Zhang
- Department of Clinical Laboratory, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Li Shen
- Department of Clinical Laboratory, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Ying Zhou
- Department of Clinical Laboratory, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Yinjuan Guo
- Department of Clinical Laboratory, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Xiaocui Wu
- Department of Clinical Laboratory, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Jingyi Yu
- Department of Clinical Laboratory, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Fangyou Yu
- Department of Clinical Laboratory, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China; Department of Laboratory Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China.
| |
Collapse
|
20
|
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
|
21
|
Domingues N, Ramos LDP, Pereira LM, do Rosário Estevam Dos Santos PB, Scorzoni L, Pereira TC, Abu Hasna A, Carvalho CAT, de Oliveira LD. Antimicrobial action of four herbal plants over mixed-species biofilms of Candida albicans with four different microorganisms. AUST ENDOD J 2023; 49:262-271. [PMID: 36057926 DOI: 10.1111/aej.12681] [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: 01/07/2022] [Revised: 07/18/2022] [Accepted: 08/17/2022] [Indexed: 11/02/2022]
Abstract
This study aimed to evaluate the antimicrobial effect of four herbal plants glycolic extracts over mixed-species biofilm composed of Candida albicans (C. albicans) and another pathogenic bacterium as alternative therapy to be investigated. Four plants extract of Pfaffia paniculata roots; Hamamelis virginiana leaf, Stryphnodendron barbatiman tree bark and Gymnema sylvestre stem and leaves were tested over multi-species biofilm of C. albicans (ATCC 18804) and Streptococcus mutans (ATCC 35688), Staphylococcus aureus (ATCC 6538), Enterococcus faecalis (ATCC 4083) or Pseudomonas aeruginosa (ATCC 15442) for 5 min and 24 h and colony forming units per millilitre was calculated. The data were analysed using Kruskal-Wallis with Dunn's test (p ≤ 0.05). All tested extracts showed antimicrobial action over the mixed-species biofilms after 24 h. Some extracts eliminated totally the biofilms. The glycolic extract of P. paniculata, H. virginiana, S. barbatiman and G. sylvestre are effective over mixed-species biofilms and may be indicated as endodontic irrigant or intracanal medication.
Collapse
Affiliation(s)
- Nádia Domingues
- Department of Bioscience and Oral Diagnosis, Institute of Science and Technology, São Paulo State University - UNESP, São José dos Campos, Brazil
| | - Lucas de Paula Ramos
- Department of Bioscience and Oral Diagnosis, Institute of Science and Technology, São Paulo State University - UNESP, São José dos Campos, Brazil
| | - Larissa Marques Pereira
- Department of Bioscience and Oral Diagnosis, Institute of Science and Technology, São Paulo State University - UNESP, São José dos Campos, Brazil
| | - Pâmela Beatriz do Rosário Estevam Dos Santos
- Department of Bioscience and Oral Diagnosis, Institute of Science and Technology, São Paulo State University - UNESP, São José dos Campos, Brazil
- Universidade Paulista (UNIP), Health Sciences Institute, São José dos Campos, Brazil
| | - Liliana Scorzoni
- Department of Bioscience and Oral Diagnosis, Institute of Science and Technology, São Paulo State University - UNESP, São José dos Campos, Brazil
| | - Thaís Cristine Pereira
- Department of Bioscience and Oral Diagnosis, Institute of Science and Technology, São Paulo State University - UNESP, São José dos Campos, Brazil
| | - Amjad Abu Hasna
- Department of Restorative Dentistry, Endodontics Division, Institute of Science and Technology, São Paulo State University (Unesp), São José dos Campos, SP, Brazil
| | - Cláudio Antonio Talge Carvalho
- Department of Restorative Dentistry, Endodontics Division, Institute of Science and Technology, São Paulo State University (Unesp), São José dos Campos, SP, Brazil
| | - Luciane Dias de Oliveira
- Department of Bioscience and Oral Diagnosis, Institute of Science and Technology, São Paulo State University - UNESP, São José dos Campos, Brazil
| |
Collapse
|
22
|
Wu Y, Chen T, Wang Y, Huang M, Wang Y, Luo Z. New insight into the virulence and inflammatory response of Staphylococcus aureus strains isolated from diabetic foot ulcers. Front Cell Infect Microbiol 2023; 13:1234994. [PMID: 37577369 PMCID: PMC10416727 DOI: 10.3389/fcimb.2023.1234994] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Accepted: 07/14/2023] [Indexed: 08/15/2023] Open
Abstract
Staphylococcus aureus strains isolated from diabetic foot ulcers (DFUs) have less virulence, but still cause severe infections. Furthermore, hypovirulent S. aureus strains appear to be localized in the deep tissues of diabetic foot osteomyelitis, indicating that the unique environment within DFUs affects the pathogenicity of S. aureus. In this study, the cell-free culture medium (CFCM) of S. aureus strains isolated from DFUs exhibited higher cytotoxicity to human erythrocytes than those isolated from non-diabetic patients with sepsis or wounds. Among these S. aureus strains isolated from DFUs, β-toxin negative strains have less virulence than β-toxin positive strains, but induced a higher expression of inflammatory cytokines. Our study and previous studies have shown that the synergistic effect of phenol-soluble modulin α and β-toxin contributes to the higher hemolytic activity of β-toxin positive strains. However, lysis of human erythrocytes by the CFCM of β-toxin negative strains was greatly inhibited by an autolysin inhibitor, sodium polyanethole sulfonate (SPS). A high level of glucose greatly reduced the hemolytic activity of S. aureus, but promoted the expression of interleukin-6 (IL-6) in human neutrophils. However, 5 mM glucose or glucose-6-phosphate (G6P) increased the hemolytic activity of SA118 (a β-toxin negative strain) isolated from DFUs. Additionally, patients with DFUs with growth of S. aureus had lower level of serum IL-6 than those with other bacteria, and the CFCM of S. aureus strains significantly reduced lipopolysaccharide-induced IL-6 expression in human neutrophils. Therefore, the virulence and inflammatory response of S. aureus strains isolated from DFUs are determined by the levels of glucose and its metabolites, which may explain why it is the predominant bacteria isolated from DFUs.
Collapse
Affiliation(s)
- Yuan Wu
- Department of Laboratory Medicine, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Ti Chen
- Department of Laboratory Medicine, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Yanle Wang
- Department of Laboratory Medicine, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Mao Huang
- Department of Laboratory Medicine, Xiangya School of Medicine, Central South University, Changsha, China
| | - Yurong Wang
- Department of Laboratory Medicine, Xiangya School of Medicine, Central South University, Changsha, China
| | - Zhen Luo
- Department of Laboratory Medicine, The Third Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
23
|
Lacey KA, Serpas L, Makita S, Wang Y, Rashidfarrokhi A, Soni C, Gonzalez S, Moreira A, Torres VJ, Reizis B. Secreted mammalian DNases protect against systemic bacterial infection by digesting biofilms. J Exp Med 2023; 220:e20221086. [PMID: 36928522 PMCID: PMC10037111 DOI: 10.1084/jem.20221086] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Revised: 01/18/2023] [Accepted: 03/01/2023] [Indexed: 03/18/2023] Open
Abstract
Extracellular DNase DNASE1L3 maintains tolerance to self-DNA in humans and mice, whereas the role of its homolog DNASE1 remains controversial, and the overall function of secreted DNases in immunity is unclear. We report that deletion of murine DNASE1 neither caused autoreactivity in isolation nor exacerbated lupus-like disease in DNASE1L3-deficient mice. However, combined deficiency of DNASE1 and DNASE1L3 rendered mice susceptible to bloodstream infection with Staphylococcus aureus. DNASE1/DNASE1L3 double-deficient mice mounted a normal innate response to S. aureus and did not accumulate neutrophil extracellular traps (NETs). However, their kidneys manifested severe pathology, increased bacterial burden, and biofilm-like bacterial lesions that contained bacterial DNA and excluded neutrophils. Furthermore, systemic administration of recombinant DNASE1 protein during S. aureus infection rescued the mortality of DNase-deficient mice and ameliorated the disease in wild-type mice. Thus, DNASE1 and DNASE1L3 jointly facilitate the control of bacterial infection by digesting extracellular microbial DNA in biofilms, suggesting the original evolutionary function of secreted DNases as antimicrobial agents.
Collapse
Affiliation(s)
- Keenan A. Lacey
- Department of Microbiology, New York University Grossman School of Medicine, New York, NY, USA
| | - Lee Serpas
- Department of Pathology, New York University Grossman School of Medicine, New York, NY, USA
| | - Sohei Makita
- Department of Pathology, New York University Grossman School of Medicine, New York, NY, USA
| | - Yueyang Wang
- Department of Pathology, New York University Grossman School of Medicine, New York, NY, USA
| | - Ali Rashidfarrokhi
- Department of Pathology, New York University Grossman School of Medicine, New York, NY, USA
| | - Chetna Soni
- Department of Pathology, New York University Grossman School of Medicine, New York, NY, USA
| | - Sandra Gonzalez
- Department of Microbiology, New York University Grossman School of Medicine, New York, NY, USA
| | - Andre Moreira
- Department of Pathology, New York University Grossman School of Medicine, New York, NY, USA
| | - Victor J. Torres
- Department of Microbiology, New York University Grossman School of Medicine, New York, NY, USA
- Antimicrobial-Resistant Pathogens Program, New York University Grossman School of Medicine, New York, NY, USA
| | - Boris Reizis
- Department of Pathology, New York University Grossman School of Medicine, New York, NY, USA
| |
Collapse
|
24
|
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
|
25
|
Linz MS, Mattappallil A, Finkel D, Parker D. Clinical Impact of Staphylococcus aureus Skin and Soft Tissue Infections. Antibiotics (Basel) 2023; 12:557. [PMID: 36978425 PMCID: PMC10044708 DOI: 10.3390/antibiotics12030557] [Citation(s) in RCA: 61] [Impact Index Per Article: 30.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2023] [Revised: 03/03/2023] [Accepted: 03/09/2023] [Indexed: 03/14/2023] Open
Abstract
The pathogenic bacterium Staphylococcus aureus is the most common pathogen isolated in skin-and-soft-tissue infections (SSTIs) in the United States. Most S. aureus SSTIs are caused by the epidemic clone USA300 in the USA. These infections can be serious; in 2019, SSTIs with S. aureus were associated with an all-cause, age-standardized mortality rate of 0.5 globally. Clinical presentations of S. aureus SSTIs vary from superficial infections with local symptoms to monomicrobial necrotizing fasciitis, which can cause systemic manifestations and may lead to serious complications or death. In order to cause skin infections, S. aureus employs a host of virulence factors including cytolytic proteins, superantigenic factors, cell wall-anchored proteins, and molecules used for immune evasion. The immune response to S. aureus SSTIs involves initial responders such as keratinocytes and neutrophils, which are supported by dendritic cells and T-lymphocytes later during infection. Treatment for S. aureus SSTIs is usually oral therapy, with parenteral therapy reserved for severe presentations; it ranges from cephalosporins and penicillin agents such as oxacillin, which is generally used for methicillin-sensitive S. aureus (MSSA), to vancomycin for methicillin-resistant S. aureus (MRSA). Treatment challenges include adverse effects, risk for Clostridioides difficile infection, and potential for antibiotic resistance.
Collapse
Affiliation(s)
- Matthew S. Linz
- Department of Pathology, Immunology and Laboratory Medicine, Center for Immunity and Inflammation, Rutgers New Jersey Medical School, Newark, NJ 07103, USA
| | - Arun Mattappallil
- Department of Pharmaceutical Services, University Hospital, Newark, NJ 07103, USA
| | - Diana Finkel
- Division of Infectious Diseases, Department of Medicine, Rutgers New Jersey Medical School, Newark, NJ 07103, USA
| | - Dane Parker
- Department of Pathology, Immunology and Laboratory Medicine, Center for Immunity and Inflammation, Rutgers New Jersey Medical School, Newark, NJ 07103, USA
| |
Collapse
|
26
|
Dehnavi M, Haghighat S, Yazdi MH, Mahdavi M. Glucomannan as a polysaccharide adjuvant improved immune responses against Staphylococcus aureus: Potency and efficacy studies. Microb Pathog 2023; 176:106007. [PMID: 36709850 DOI: 10.1016/j.micpath.2023.106007] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Revised: 01/24/2023] [Accepted: 01/25/2023] [Indexed: 01/28/2023]
Abstract
Staphylococcus aureus is a gram-positive bacterium, representing one of the most important nosocomial pathogens. The treatment of infections, caused by S. aureus, has become increasingly intricate due to the emergence of highly resistant strains. Therefore, it is obvious that an effective prevention strategy against this bacterium could significantly decrease such infections. In the present study, the protective efficacy and immunological properties of recombinant autolysin, formulated in Montanide ISA266 and Alum adjuvants with Glucomannan as a polysaccharide, were assessed in the systemic mouse model of infection. Mice were immunized with the purified recombinant protein in various formulations in different groups and, subsequently, mice were challenged with 5 × 108 CFU of bacteria for the evaluation of their survival and bacterial clearances in the internal organs. ELISA was performed to determine the type of induced immunity, cytokine secretion (IFN-γ, IL-4, IL-2, and IL-17), and isotyping (IgG1 and IgG2a). In addition, we measured the opsonophagocytic activities of the antibodies. Results showed that immunization with r-autolysin + Alum + Glucomannan and r-autolysin + MontanideISA266+Glucomannan formulations significantly increased total IgG and isotypes (IgG1 and IgG2a), as compared with other vaccinated and control groups. Furthermore, the formulation of r-autolysin in Alum and MontanideISA266 adjuvants with Glucomannan enhanced IFN-γ, IL-4, and IL-17 cytokine secretion as well as protectivity, following experimental challenge. We concluded that Glucomannan has the potential to induce immune responses and would be used as an adjuvant factor in vaccine formulation.
Collapse
Affiliation(s)
- Meghdad Dehnavi
- Department of Microbiology, Faculty of Pharmaceutical Sciences, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Setareh Haghighat
- Department of Microbiology, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran.
| | - Mohammad Hossein Yazdi
- Biotechnology Research Center, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran; Immunotherapy Group, The Institute of Pharmaceutical Sciences (TIPS), Tehran University of Medical Sciences, Tehran, Iran; Recombinant Vaccine Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Mehdi Mahdavi
- Advanced Therapy Medicinal Product (ATMP) Department, Breast Cancer Research Center, Motamed Cancer Institute, Academic Center for Education, Culture and Research (ACECR), Tehran, Iran; Department of Immunology, Pasteur Institute of Iran, Tehran, Iran
| |
Collapse
|
27
|
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
|
28
|
Regulation of Staphylococcus aureus Virulence and Application of Nanotherapeutics to Eradicate S. aureus Infection. Pharmaceutics 2023; 15:pharmaceutics15020310. [PMID: 36839634 PMCID: PMC9960757 DOI: 10.3390/pharmaceutics15020310] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Revised: 01/11/2023] [Accepted: 01/13/2023] [Indexed: 01/19/2023] Open
Abstract
Staphylococcus aureus is a versatile pathogen known to cause hospital- and community-acquired, foodborne, and zoonotic infections. The clinical infections by S. aureus cause an increase in morbidity and mortality rates and treatment costs, aggravated by the emergence of drug-resistant strains. As a multi-faceted pathogen, it is imperative to consolidate the knowledge on its pathogenesis, including the mechanisms of virulence regulation, development of antimicrobial resistance, and biofilm formation, to make it amenable to different treatment strategies. Nanomaterials provide a suitable platform to address this challenge, with the potential to control intracellular parasitism and multidrug resistance where conventional therapies show limited efficacy. In a nutshell, the first part of this review focuses on the impact of S. aureus on human health and the role of virulence factors and biofilms during pathogenesis. The second part discusses the large diversity of nanoparticles and their applications in controlling S. aureus infections, including combination with antibiotics and phytochemicals and the incorporation of antimicrobial coatings for biomaterials. Finally, the limitations and prospects using nanomaterials are highlighted, aiming to foster the development of novel nanotechnology-driven therapies against multidrug-resistant S. aureus.
Collapse
|
29
|
Costa MDOCE, do Nascimento APB, Martins YC, dos Santos MT, Figueiredo AMDS, Perez-Rueda E, Nicolás MF. The gene regulatory network of Staphylococcus aureus ST239-SCC mecIII strain Bmb9393 and assessment of genes associated with the biofilm in diverse backgrounds. Front Microbiol 2023; 13:1049819. [PMID: 36704545 PMCID: PMC9871828 DOI: 10.3389/fmicb.2022.1049819] [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: 09/21/2022] [Accepted: 12/19/2022] [Indexed: 01/12/2023] Open
Abstract
Introduction Staphylococcus aureus is one of the most prevalent and relevant pathogens responsible for a wide spectrum of hospital-associated or community-acquired infections. In addition, methicillin-resistant Staphylococcus aureus may display multidrug resistance profiles that complicate treatment and increase the mortality rate. The ability to produce biofilm, particularly in device-associated infections, promotes chronic and potentially more severe infections originating from the primary site. Understanding the complex mechanisms involved in planktonic and biofilm growth is critical to identifying regulatory connections and ways to overcome the global health problem of multidrug-resistant bacteria. Methods In this work, we apply literature-based and comparative genomics approaches to reconstruct the gene regulatory network of the high biofilm-producing strain Bmb9393, belonging to one of the highly disseminating successful clones, the Brazilian epidemic clone. To the best of our knowledge, we describe for the first time the topological properties and network motifs for the Staphylococcus aureus pathogen. We performed this analysis using the ST239-SCCmecIII Bmb9393 strain. In addition, we analyzed transcriptomes available in the literature to construct a set of genes differentially expressed in the biofilm, covering different stages of the biofilms and genetic backgrounds of the strains. Results and discussion The Bmb9393 gene regulatory network comprises 1,803 regulatory interactions between 64 transcription factors and the non-redundant set of 1,151 target genes with the inclusion of 19 new regulons compared to the N315 transcriptional regulatory network published in 2011. In the Bmb9393 network, we found 54 feed-forward loop motifs, where the most prevalent were coherent type 2 and incoherent type 2. The non-redundant set of differentially expressed genes in the biofilm consisted of 1,794 genes with functional categories relevant for adaptation to the variable microenvironments established throughout the biofilm formation process. Finally, we mapped the set of genes with altered expression in the biofilm in the Bmb9393 gene regulatory network to depict how different growth modes can alter the regulatory systems. The data revealed 45 transcription factors and 876 shared target genes. Thus, the gene regulatory network model provided represents the most up-to-date model for Staphylococcus aureus, and the set of genes altered in the biofilm provides a global view of their influence on biofilm formation from distinct experimental perspectives and different strain backgrounds.
Collapse
Affiliation(s)
| | - Ana Paula Barbosa do Nascimento
- Departamento de Análises Clínicas e Toxicológicas, Faculdade de Ciências Farmacêuticas, Universidade de São Paulo, São Paulo, Brazil
| | | | | | - Agnes Marie de Sá Figueiredo
- Instituto de Investigaciones en Matemáticas Aplicadas y en Sistemas, Universidad Nacional Autónoma de México, Unidad Académica Yucatán, Merida, Mexico
| | - Ernesto Perez-Rueda
- Laboratório de Biologia Molecular de Bactérias, Instituto de Microbiologia Paulo de Goés, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil,*Correspondence: Ernesto Perez-Rueda ✉
| | - Marisa Fabiana Nicolás
- Laboratório Nacional de Computação Científica (LNCC), Petrópolis, Brazil,Marisa Fabiana Nicolás ✉
| |
Collapse
|
30
|
Synthetic selenium nanoparticles as co-adjuvant improved immune responses against methicillin-resistant Staphylococcus aureus. World J Microbiol Biotechnol 2023; 39:16. [PMID: 36401129 PMCID: PMC9676803 DOI: 10.1007/s11274-022-03455-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Accepted: 11/01/2022] [Indexed: 11/21/2022]
Abstract
Methicillin-resistant Staphylococcus aureus (MRSA) is one of the leading causes of hospital-acquired infections worldwide, which is resistant to many antibiotics, resulting in significant mortality in societies. Vaccination is a well-known approach to preventing disease. Autolysin, a surface-associated protein in S. aureus with multiple functions, is a suitable candidate for vaccine development. As a co-adjuvant, selenium nanoparticles (SeNPs) can increase the immune system, presumably resulting in increased vaccine efficacy. The present study evaluated the immunogenicity and defense of recombinant autolysin formulated in SeNPs and Alum adjuvants against MRSA. r-Autolysin was expressed and purified by the Ni-NTA affinity chromatography. SeNPs were synthetically obtained from sodium dioxide, followed by an assessment of shape and size using SEM and DLS. Balb/c mice were injected subcutaneously with 20 mg of r-autolysin formulated in Alum and SeNps adjuvants three times with the proper control group in 2 weeks intervals. Cytokine profile and isotyping ELISA were conducted to determine the type of induced immunity. Opsonophagocytosis tests assessed the functional activity of the vaccine, and the bacterial burden from the infected tissues was determined. Results showed that mice receiving SeNps and r-Autolysin had higher levels of total IgG and isotypes (IgG1 and IgG2a) and increased cytokine levels (IFN-γ, TNF-α, IL-12, and IL-4) as compared with those only receiving autolysin and PBS as a control. More importantly, mice immunized with SeNps and r-Autolysin exhibited a decrease in mortality and bacterial burden compared to the control group. We concluded that SeNps could stimulate immune responses and can be used as an adjuvant element in vaccine formulation.
Collapse
|
31
|
Serrapeptase impairs biofilm, wall, and phospho-homeostasis of resistant and susceptible Staphylococcus aureus. Appl Microbiol Biotechnol 2023; 107:1373-1389. [PMID: 36635396 PMCID: PMC9898353 DOI: 10.1007/s00253-022-12356-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Revised: 11/25/2022] [Accepted: 12/22/2022] [Indexed: 01/14/2023]
Abstract
Staphylococcus aureus biofilms are implicated in hospital infections due to elevated antibiotic and host immune system resistance. Molecular components of cell wall including amyloid proteins, peptidoglycans (PGs), and lipoteichoic acid (LTA) are crucial for biofilm formation and tolerance of methicillin-resistant S. aureus (MRSA). Significance of alkaline phosphatases (ALPs) for biofilm formation has been recorded. Serrapeptase (SPT), a protease of Serratia marcescens, possesses antimicrobial properties similar or superior to those of many antibiotics. In the present study, SPT anti-biofilm activity was demonstrated against S. aureus (ATCC 25923, methicillin-susceptible strain, methicillin-susceptible S. aureus (MSSA)) and MRSA (ST80), with IC50 values of 0.67 μg/mL and 7.70 μg/mL, respectively. SPT affected bacterial viability, causing a maximum inhibition of - 46% and - 27%, respectively. Decreased PGs content at [SPT] ≥ 0.5 μg/mL and ≥ 8 μg/mL was verified for MSSA and MRSA, respectively. In MSSA, LTA levels decreased significantly (up to - 40%) at lower SPT doses but increased at the highest dose of 2 μg/mL, a counter to spectacularly increased cellular and secreted LTA levels in MRSA. SPT also reduced amyloids of both strains. Additionally, intracellular ALP activity decreased in both MSSA and MRSA (up to - 85% and - 89%, respectively), while extracellular activity increased up to + 482% in MSSA and + 267% in MRSA. Altered levels of DING proteins, which are involved in phosphate metabolism, in SPT-treated bacteria, were also demonstrated here, implying impaired phosphorus homeostasis. The differential alterations in the studied molecular aspects underline the differences between MSSA and MRSA and offer new insights in the treatment of resistant bacterial biofilms. KEY POINTS: • SPT inhibits biofilm formation in methicillin-resistant and methicillin-susceptible S. aureus. • SPT treatment decreases bacterial viability, ALP activity, and cell wall composition. • SPT-treated bacteria present altered levels of phosphate-related DING proteins.
Collapse
|
32
|
Liang J, Huang TY, Mao Y, Li X. Biofilm formation of two genetically diverse Staphylococcus aureus isolates under beta-lactam antibiotics. Front Microbiol 2023; 14:1139753. [PMID: 36950159 PMCID: PMC10025342 DOI: 10.3389/fmicb.2023.1139753] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2023] [Accepted: 01/30/2023] [Indexed: 03/08/2023] Open
Abstract
PURPOSE Our aim was to evaluate the biofilm formation of 2 genetically diverse Staphylococcus aureus isolates, 10379 and 121940, under different concentrations of beta-lactam antibiotics on biomass content and biofilm viability. METHODS Biofilm formation and methicillin resistance genes were tested using PCR and multiplex PCR. PCR was combined with bioinformatics analysis to detect multilocal sequence typing (MLST) and SCCmec types, to study the genetical correlation between the tested strains. Then, the crystal violet (CV) test and XTT were used to detect biomass content and biofilm activity. Antibiotic susceptibility was tested using a broth dilution method. According to their specific MIC, different concentrations of beta-lactam antibiotics were used to study its effect on biomass content and biofilm viability. RESULTS Strain 10379 carried the icaD, icaBC, and MRSA genes, not the icaA, atl, app, and agr genes, and MLST and SCCmec typing was ST45 and IV, respectively. Strain 121940 carried the icaA, icaD, icaBC, atl, and agr genes, not the aap gene, and MLST and SCCmec typed as ST546 and IV, respectively. This suggested that strains 10379 and 121940 were genotypically very different. Two S. aureus isolates, 10379 and 121940, showed resistance to beta-lactam antibiotics, penicillin, ampicillin, meropenem, streptomycin and kanamycin, some of which promoted the formation of biofilm and biofilm viability at low concentrations. CONCLUSION Despite the large differences in the genetic background of S. aureus 10379 and 121940, some sub-inhibitory concentrations of beta-lactam antibiotics are able to promote biomass and biofilm viability of both two isolates.
Collapse
Affiliation(s)
- Jinglong Liang
- College of Light Industry and Food Technology, Zhongkai University of Agriculture and Engineering, Guangzhou, China
- Guangdong Provincial Key Laboratory of Lingnan Specialty Food Science and Technology, Zhongkai University of Agriculture and Engineering, Guangzhou, China
- Key Laboratory of Green Processing and Intelligent Manufacturing of Lingnan Specialty Food, Ministry of Agriculture, Zhongkai University of Agriculture and Engineering, Guangzhou, China
| | - Teng Yi Huang
- Department of Diagnostics, Second Affiliated Hospital of Shantou University Medical College, Shantou, China
- Teng Yi Huang,
| | - Yuzhu Mao
- Department of Civil and Environmental Engineering, University of Maryland, College Park, MD, United States
| | - Xuejie Li
- School of Food Science and Engineering, Guangdong Province Key Laboratory for Green Processing of Natural Products and Product Safety, Engineering Research Center of Starch and Vegetable Protein Processing Ministry of Education, South China University of Technology, Guangzhou, China
- Research Institute for Food Nutrition and Human Health, Guangzhou, China
- *Correspondence: Xuejie Li,
| |
Collapse
|
33
|
Jean-Pierre V, Boudet A, Sorlin P, Menetrey Q, Chiron R, Lavigne JP, Marchandin H. Biofilm Formation by Staphylococcus aureus in the Specific Context of Cystic Fibrosis. Int J Mol Sci 2022; 24:ijms24010597. [PMID: 36614040 PMCID: PMC9820612 DOI: 10.3390/ijms24010597] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2022] [Revised: 12/21/2022] [Accepted: 12/22/2022] [Indexed: 12/31/2022] Open
Abstract
Staphylococcus aureus is a major human pathogen whose characteristics support its success in various clinical settings including Cystic Fibrosis (CF). In CF, S. aureus is indeed the most commonly identified opportunistic pathogen in children and the overall population. S. aureus colonization/infection, either by methicillin-susceptible or methicillin-resistant strains, will become chronic in about one third of CF patients. The persistence of S. aureus in CF patients' lungs, despite various eradication strategies, is favored by several traits in both host and pathogen. Among the latter, living in biofilm is a highly protective way to survive despite deleterious environmental conditions, and is a common characteristic shared by the main pathogens identified in CF. This is why CF has earned the status of a biofilm-associated disease for several years now. Biofilm formation by S. aureus, and the molecular mechanisms governing and regulating it, have been extensively studied but have received less attention in the specific context of CF lungs. Here, we review the current knowledge on S. aureus biofilm in this very context, i.e., the importance, study methods, molecular data published on mono- and multi-species biofilm and anti-biofilm strategies. This focus on studies including clinical isolates from CF patients shows that they are still under-represented in the literature compared with studies based on reference strains, and underlines the need for such studies. Indeed, CF clinical strains display specific characteristics that may not be extrapolated from results obtained on laboratory strains.
Collapse
Affiliation(s)
- Vincent Jean-Pierre
- HSM—HydroSciences Montpellier, Université de Montpellier, CNRS, IRD, Service de Microbiologie et Hygiène Hospitalière, CHU Nîmes, 34093 Montpellier, France
| | - Agathe Boudet
- VBIC—Virulence Bactérienne et Infections Chroniques, Université de Montpellier, INSERM U1047, Service de Microbiologie et Hygiène Hospitalière, CHU Nîmes, 30900 Nîmes, France
| | - Pauline Sorlin
- HSM—HydroSciences Montpellier, Université de Montpellier, CNRS, IRD, 34093 Montpellier, France
| | - Quentin Menetrey
- INFINITE—Institute for Translational Research in Inflammation, Université de Lille, INSERM U1286, CHU Lille, 59000 Lille, France
| | - Raphaël Chiron
- HSM—HydroSciences Montpellier, Université de Montpellier, CNRS, IRD, Centre de Ressources et de Compétences de la Mucoviscidose, CHU Montpellier, 34295 Montpellier, France
| | - Jean-Philippe Lavigne
- VBIC—Virulence Bactérienne et Infections Chroniques, Université de Montpellier, INSERM U1047, Service de Microbiologie et Hygiène Hospitalière, CHU Nîmes, 30900 Nîmes, France
| | - Hélène Marchandin
- HSM—HydroSciences Montpellier, Université de Montpellier, CNRS, IRD, Service de Microbiologie et Hygiène Hospitalière, CHU Nîmes, 34093 Montpellier, France
- Correspondence:
| |
Collapse
|
34
|
He X, Zhang W, Cao Q, Li Y, Bao G, Lin T, Bao J, Chang C, Yang C, Yin Y, Xu J, Ren Z, Jin Y, Lu F. Global Downregulation of Penicillin Resistance and Biofilm Formation by MRSA Is Associated with the Interaction between Kaempferol Rhamnosides and Quercetin. Microbiol Spectr 2022; 10:e0278222. [PMID: 36354319 PMCID: PMC9769653 DOI: 10.1128/spectrum.02782-22] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Accepted: 10/24/2022] [Indexed: 11/12/2022] Open
Abstract
The rapid development of methicillin-resistant Staphylococcus aureus (MRSA) drug resistance and the formation of biofilms seriously challenge the clinical application of classic antibiotics. Extracts of the traditional herb Chenopodium ambrosioides L. were found to have strong antibiofilm activity against MRSA, but their mechanism of action remains poorly understood. This study was designed to investigate the antibacterial and antibiofilm activities against MRSA of flavonoids identified from C. ambrosioides L. in combination with classic antibiotics, including ceftazidime, erythromycin, levofloxacin, penicillin G, and vancomycin. Liquid chromatography-mass spectrometry (LC-MS) was used to analyze the nonvolatile chemical compositions. Reverse transcription (RT)-PCR was used to investigate potential multitargets of flavonoids based on global transcriptional responses of virulence and antibiotic resistance. A synergistic antibacterial and biofilm-inhibiting activity of the alcoholic extract of the ear of C. ambrosioides L. in combination with penicillin G was observed against MRSA, which proved to be closely related to the interaction of the main components of kaempferol rhamnosides with quercetin. In regard to the mechanism, the increased sensitivity of MRSA to penicillin G was shown to be related to the downregulation of penicillinase with SarA as a potential drug target, while the antibiofilm activity was mainly related to downregulation of various virulence factors involved in the initial and mature stages of biofilm development, with SarA and/or σB as drug targets. This study provides a theoretical basis for further exploration of the medicinal activity of kaempferol rhamnosides and quercetin and their application in combination with penicillin G against MRSA biofilm infection. IMPORTANCE In this study, the synergistic antibacterial and antibiofilm effects of the traditional herb C. ambrosioides L. and the classic antibiotic penicillin G on MRSA provide a potential strategy to deal with the rapid development of MRSA antibiotic resistance. This study also provides a theoretical basis for further optimizing the combined effect of kaempferol rhamnosides, quercetin, and penicillin G and exploring anti-MRSA biofilm infection research with SarA and σB as drug targets.
Collapse
Affiliation(s)
- Xinlong He
- Department of Pathogenic Biology, School of Medicine, Yangzhou University, Yangzhou, People’s Republic of China
- Affiliated Hospital of Yangzhou University, Yangzhou, People’s Republic of China
- Jiangsu Key Laboratory of Zoonosis, Yangzhou University, Yangzhou, People’s Republic of China
- Jiangsu Key Laboratory of Experimental & Translational Non-coding RNA Research, School of Medicine, Yangzhou University, Yangzhou, People’s Republic of China
- Jiangsu Co-Innovation Center for the Prevention and Control of Important Animal Infectious Diseases and Zoonoses, College of Veterinary Medicine, Yangzhou University, Yangzhou, People’s Republic of China
| | - Wenwen Zhang
- Department of Pathogenic Biology, School of Medicine, Yangzhou University, Yangzhou, People’s Republic of China
| | - Qingchao Cao
- College of Bioscience and Biotechnology, Yangzhou University, Yangzhou, People’s Republic of China
| | - Yinyue Li
- Department of Pathogenic Biology, School of Medicine, Yangzhou University, Yangzhou, People’s Republic of China
| | - Guangyu Bao
- Affiliated Hospital of Yangzhou University, Yangzhou, People’s Republic of China
| | - Tao Lin
- Affiliated Hospital of Yangzhou University, Yangzhou, People’s Republic of China
| | - Jiaojiao Bao
- Affiliated Hospital of Yangzhou University, Yangzhou, People’s Republic of China
| | - Caiwang Chang
- Affiliated Hospital of Yangzhou University, Yangzhou, People’s Republic of China
| | - Changshui Yang
- Department of Pharmacy, School of Medicine, Yangzhou University, Yangzhou, People’s Republic of China
| | - Yi Yin
- Department of Pathogenic Biology, School of Medicine, Yangzhou University, Yangzhou, People’s Republic of China
| | - Jiahui Xu
- Department of Pathogenic Biology, School of Medicine, Yangzhou University, Yangzhou, People’s Republic of China
| | - Zhenyu Ren
- Department of Pathogenic Biology, School of Medicine, Yangzhou University, Yangzhou, People’s Republic of China
| | - Yingshan Jin
- College of Bioscience and Biotechnology, Yangzhou University, Yangzhou, People’s Republic of China
- Jiangsu Key Laboratory of Zoonosis, Yangzhou University, Yangzhou, People’s Republic of China
| | - Feng Lu
- Department of Pathogenic Biology, School of Medicine, Yangzhou University, Yangzhou, People’s Republic of China
- Affiliated Hospital of Yangzhou University, Yangzhou, People’s Republic of China
- Jiangsu Key Laboratory of Zoonosis, Yangzhou University, Yangzhou, People’s Republic of China
- Jiangsu Key Laboratory of Experimental & Translational Non-coding RNA Research, School of Medicine, Yangzhou University, Yangzhou, People’s Republic of China
- Jiangsu Co-Innovation Center for the Prevention and Control of Important Animal Infectious Diseases and Zoonoses, College of Veterinary Medicine, Yangzhou University, Yangzhou, People’s Republic of China
| |
Collapse
|
35
|
Oligopeptide Sortase Inhibitor Modulates Staphylococcus aureus Cell Adhesion and Biofilm Formation. Antibiotics (Basel) 2022; 11:antibiotics11121836. [PMID: 36551492 PMCID: PMC9774600 DOI: 10.3390/antibiotics11121836] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Revised: 12/14/2022] [Accepted: 12/15/2022] [Indexed: 12/24/2022] Open
Abstract
Prevention of bacterial adhesion is one of the most important antivirulence strategies for meeting the global challenge posed by antimicrobial resistance. We aimed to investigate the influence of a peptidic S. aureus sortase A inhibitor on bacterial adhesion to eukaryotic cells and biofilm formation as a potential method for reducing S. aureus virulence. The pentapeptide LPRDA was synthesized and characterized as a pure individual organic compound. Incubation of MSSA and MRSA strains with LPRDA induced a subsequent reduction in staphylococcal adhesion to Vero cells and biofilm formation, as visualized by microscopic and spectrophotometric methods, respectively. LPRDA did not have a cytotoxic effect on eukaryotic or bacterial cells. The pentapeptide LPRDA deserves further investigation using in vitro and in vivo models of Gram-positive bacteriemia as a potential antibacterial agent with an antiadhesive mechanism of action.
Collapse
|
36
|
Study of SarA by DNA Affinity Capture Assay (DACA) Employing Three Promoters of Key Virulence and Resistance Genes in Methicillin-Resistant Staphylococcus aureus. Antibiotics (Basel) 2022; 11:antibiotics11121714. [PMID: 36551372 PMCID: PMC9774152 DOI: 10.3390/antibiotics11121714] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Revised: 11/21/2022] [Accepted: 11/25/2022] [Indexed: 11/29/2022] Open
Abstract
Methicillin-resistant Staphylococcus aureus (MRSA), one of the most well-known human pathogens, houses many virulence factors and regulatory proteins that confer resistance to diverse antibiotics. Although they have been investigated intensively, the correlations among virulence factors, regulatory proteins and antibiotic resistance are still elusive. We aimed to identify the most significant global MRSA regulator by concurrently analyzing protein-binding and several promoters under same conditions and at the same time point. DNA affinity capture assay (DACA) was performed with the promoters of mecA, sarA, and sarR, all of which significantly impact survival of MRSA. Here, we show that SarA protein binds to all three promoters. Consistent with the previous reports, ΔsarA mutant exhibited weakened antibiotic resistance to oxacillin and reduced biofilm formation. Additionally, production and activity of many virulence factors such as phenol-soluble modulins (PSM), α-hemolysin, motility, staphyloxanthin, and other related proteins were decreased. Comparing the sequence of SarA with that of clinical strains of various lineages showed that all sequences were highly conserved, in contrast to that observed for AgrA, another major regulator of virulence and resistance in MRSA. We have demonstrated that SarA regulates antibiotic resistance and the expression of various virulence factors. Our results warrant that SarA could be a leading target for developing therapeutic agents against MRSA infections.
Collapse
|
37
|
The survival of epidemic and sporadic MRSA on human skin mimics is determined by both host and bacterial factors. Epidemiol Infect 2022; 150:e203. [PMID: 36382385 PMCID: PMC9987022 DOI: 10.1017/s0950268822001765] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Bacterial survival on, and interactions with, human skin may explain the epidemiological success of MRSA strains. We evaluated the bacterial counts for 27 epidemic and 31 sporadic MRSA strains on 3D epidermal models based on N/TERT cells (NEMs) after 1, 2 and 8 days. In addition, the expression of antimicrobial peptides (hBD-2, RNase 7), inflammatory cytokines (IL-1β, IL-6) and chemokine IL-8 by NEMs was assessed using immunoassays and the expression of 43 S. aureus virulence factors was determined by a multiplex competitive Luminex assay. To explore donor variation, bacterial counts for five epidemic and seven sporadic MRSA strains were determined on 3D primary keratinocyte models (LEMs) from three human donors. Bacterial survival was comparable on NEMs between the two groups, but on LEMs, sporadic strains showed significantly lower survival numbers compared to epidemic strains. Both groups triggered the expression of immune factors. Upon interaction with NEMs, only the epidemic MRSA strains expressed pore-forming toxins, including alpha-hemolysin (Hla), gamma-hemolysin (HlgB), Panton-Valentine leucocidin (LukS) and LukED. Together, these data indicate that the outcome of the interaction between MRSA and human skin mimics, depends on the unique combination of bacterial strain and host factors.
Collapse
|
38
|
Lu Y, Cai WJ, Ren Z, Han P. The Role of Staphylococcal Biofilm on the Surface of Implants in Orthopedic Infection. Microorganisms 2022; 10:1909. [PMID: 36296183 PMCID: PMC9612000 DOI: 10.3390/microorganisms10101909] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Revised: 09/11/2022] [Accepted: 09/20/2022] [Indexed: 08/27/2023] Open
Abstract
Despite advanced implant sterilization and aseptic surgical techniques, implant-associated infection remains a major challenge for orthopedic surgeries. The subject of bacterial biofilms is receiving increasing attention, probably as a result of the wide acknowledgement of the ubiquity of biofilms in the clinical environment, as well as the extreme difficulty in eradicating them. Biofilm can be defined as a structured microbial community of cells that are attached to a substratum and embedded in a matrix of extracellular polymeric substances (EPS) that they have produced. Biofilm development has been proposed as occurring in a multi-step process: (i) attachment and adherence, (ii) accumulation/maturation due to cellular aggregation and EPS production, and (iii) biofilm detachment (also called dispersal) of bacterial cells. In all these stages, characteristic proteinaceous and non-proteinaceous compounds are expressed, and their expression is strictly controlled. Bacterial biofilm formation around implants shelters the bacteria and encourages the persistence of infection, which could lead to implant failure and osteomyelitis. These complications need to be treated by major revision surgeries and extended antibiotic therapies, which could lead to high treatment costs and even increase mortality. Effective preventive and therapeutic measures to reduce risks for implant-associated infections are thus in urgent need.
Collapse
Affiliation(s)
| | | | | | - Pei Han
- Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| |
Collapse
|
39
|
Gu K, Ouyang P, Hong Y, Dai Y, Tang T, He C, Shu G, Liang X, Tang H, Zhu L, Xu Z, Yin L. Geraniol inhibits biofilm formation of methicillin-resistant Staphylococcus aureus and increase the therapeutic effect of vancomycin in vivo. Front Microbiol 2022; 13:960728. [PMID: 36147840 PMCID: PMC9485828 DOI: 10.3389/fmicb.2022.960728] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2022] [Accepted: 08/16/2022] [Indexed: 12/04/2022] Open
Abstract
Methicillin-resistant Staphylococcus aureus (MRSA) is among the common drug resistant bacteria, which has gained worldwide attention due to its high drug resistance and infection rates. Biofilms produced by S. aureus are known to increase antibiotic resistance, making the treatment of S. aureus infections even more challenging. Hence, inhibition of biofilm formation has become an alternative strategy for controlling persistent infections. In this study, we evaluated the efficacy of geraniol as a treatment for MRSA biofilm infection. The results of crystal violet staining indicated that 256 μg/mL concentration of geraniol inhibited USA300 biofilm formation by 86.13% and removed mature biofilms by 49.87%. Geraniol exerted its anti-biofilm effect by influencing the major components of the MRSA biofilm structure. We found that geraniol inhibited the synthesis of major virulence factors, including staphyloxanthin and autolysins. The colony count revealed that geraniol inhibited staphyloxanthin and sensitized USA300 cells to hydrogen peroxide. Interestingly, geraniol not only reduced the release of extracellular nucleic acids (eDNA) but also inhibited cell autolysis. Real-time polymerase chain reaction data revealed the downregulation of genes involved in biofilm formation, which verified the results of the phenotypic analysis. Geraniol increased the effect of vancomycin in eliminating USA300 biofilms in a mouse infection model. Our findings revealed that geraniol effectively inhibits biofilm formation in vitro. Furthermore, in combination with vancomycin, geraniol can reduce the biofilm adhesion to the implant in mice. This suggests the potential of geraniol as an anti-MRSA biofilm drug and can provide a solution for the clinical treatment of biofilm infection.
Collapse
|
40
|
Long L, Sulaiman JE, Xiao Y, Cheng A, Wang R, Malit JJ, Wong WC, Liu W, Li YX, Chen F, Lam H, Qian PY. Mode of action of elasnin as biofilm formation eradicator of methicillin-resistant Staphylococcus aureus. Front Microbiol 2022; 13:967845. [PMID: 36003935 PMCID: PMC9393526 DOI: 10.3389/fmicb.2022.967845] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Accepted: 07/20/2022] [Indexed: 11/13/2022] Open
Abstract
Biofilm is made up of microbes and their extracellular matrix, making microorganisms highly tolerant, resistant, and resilient to a wide range of antimicrobials. Biofilm treatment with conventional antimicrobial agents can accelerate the evolution and spread of resistance due to the reduced efficacy and increased gene transfer and differentiation within biofilms. Therefore, effective biofilm-targeting compounds are currently highly sought after. In the present study, we identified elasnin as a potent biofilm-targeting compound against methicillin-resistant Staphylococcus aureus (MRSA). Elasnin effectively inhibited biofilm formation and especially eradicated the pre-formed biofilms of MRSA with low cytotoxicity and low risk of resistance development and retains its activity in a chronic wound biofilms model. A comprehensive mechanistic study using multi-omics and confocal and scanning electron microscopy revealed that elasnin induced the biofilm matrix destruction in a time-dependent manner and interfered with the cell division during the exponential phase, primarily by repressing the expression of virulence factors. Cells released from the elasnin-treated biofilms exhibited a defective appearance and became more sensitive to beta-lactam antibiotic penicillin G. Through gene overexpression and deletion assay, we discovered the key role of sarZ during elasnin-induced biofilm eradication. Overall, the present study identified elasnin as a potent biofilm eradicator against MRSA that harbors potential to be developed for biofilm removal and chronic wound treatment, and provided new insights into the molecular targets for biofilm eradication in MRSA.
Collapse
Affiliation(s)
- Lexin Long
- Department of Ocean Science, The Hong Kong University of Science and Technology, Kowloon, Hong Kong, China
- SZU-HKUST Joint PhD Program in Marine Environmental Science, Shenzhen University, Shenzhen, China
- Southern Marine Science and Engineering Guangdong Laboratory (Guangzhou), Guangzhou, China
| | - Jordy Evan Sulaiman
- Department of Chemical and Biological Engineering, The Hong Kong University of Science and Technology, Kowloon, Hong Kong SAR, China
| | - Yao Xiao
- Department of Ocean Science, The Hong Kong University of Science and Technology, Kowloon, Hong Kong, China
- Southern Marine Science and Engineering Guangdong Laboratory (Guangzhou), Guangzhou, China
| | - Aifang Cheng
- Department of Ocean Science, The Hong Kong University of Science and Technology, Kowloon, Hong Kong, China
- Southern Marine Science and Engineering Guangdong Laboratory (Guangzhou), Guangzhou, China
| | - Ruojun Wang
- Department of Ocean Science, The Hong Kong University of Science and Technology, Kowloon, Hong Kong, China
- Southern Marine Science and Engineering Guangdong Laboratory (Guangzhou), Guangzhou, China
| | - Jessie James Malit
- Department of Ocean Science, The Hong Kong University of Science and Technology, Kowloon, Hong Kong, China
- Southern Marine Science and Engineering Guangdong Laboratory (Guangzhou), Guangzhou, China
| | - Wai Chuen Wong
- Department of Ocean Science, The Hong Kong University of Science and Technology, Kowloon, Hong Kong, China
- Southern Marine Science and Engineering Guangdong Laboratory (Guangzhou), Guangzhou, China
| | - Wenchao Liu
- Department of Ocean Science, The Hong Kong University of Science and Technology, Kowloon, Hong Kong, China
- Southern Marine Science and Engineering Guangdong Laboratory (Guangzhou), Guangzhou, China
| | - Yong-Xin Li
- Department of Chemistry, The University of Hong Kong, Pokfulam, Hong Kong SAR, China
- The Swire Institute of Marine Science and Hong Kong Branch of Southern Marine Science and Engineering Guangdong Laboratory (Guangzhou), The University of Hong Kong, Pokfulam, Hong Kong SAR, China
| | - Feng Chen
- Institute for Advanced Study, Shenzhen University, Shenzhen, China
| | - Henry Lam
- Department of Chemical and Biological Engineering, The Hong Kong University of Science and Technology, Kowloon, Hong Kong SAR, China
- Henry Lam,
| | - Pei-Yuan Qian
- Department of Ocean Science, The Hong Kong University of Science and Technology, Kowloon, Hong Kong, China
- Southern Marine Science and Engineering Guangdong Laboratory (Guangzhou), Guangzhou, China
- *Correspondence: Pei-Yuan Qian,
| |
Collapse
|
41
|
Sionov RV, Banerjee S, Bogomolov S, Smoum R, Mechoulam R, Steinberg D. Targeting the Achilles' Heel of Multidrug-Resistant Staphylococcus aureus by the Endocannabinoid Anandamide. Int J Mol Sci 2022; 23:7798. [PMID: 35887146 PMCID: PMC9319909 DOI: 10.3390/ijms23147798] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Revised: 07/10/2022] [Accepted: 07/12/2022] [Indexed: 02/06/2023] Open
Abstract
Antibiotic-resistant Staphylococcus aureus is a major health issue that requires new therapeutic approaches. Accumulating data suggest that it is possible to sensitize these bacteria to antibiotics by combining them with inhibitors targeting efflux pumps, the low-affinity penicillin-binding protein PBP2a, cell wall teichoic acid, or the cell division protein FtsZ. We have previously shown that the endocannabinoid Anandamide (N-arachidonoylethanolamine; AEA) could sensitize drug-resistant S. aureus to a variety of antibiotics, among others, through growth arrest and inhibition of drug efflux. Here, we looked at biochemical alterations caused by AEA. We observed that AEA increased the intracellular drug concentration of a fluorescent penicillin and augmented its binding to membrane proteins with concomitant altered membrane distribution of these proteins. AEA also prevented the secretion of exopolysaccharides (EPS) and reduced the cell wall teichoic acid content, both processes known to require transporter proteins. Notably, AEA was found to inhibit membrane ATPase activity that is necessary for transmembrane transport. AEA did not affect the membrane GTPase activity, and the GTPase cell division protein FtsZ formed the Z-ring of the divisome normally in the presence of AEA. Rather, AEA caused a reduction in murein hydrolase activities involved in daughter cell separation. Altogether, this study shows that AEA affects several biochemical processes that culminate in the sensitization of the drug-resistant bacteria to antibiotics.
Collapse
Affiliation(s)
- Ronit Vogt Sionov
- Biofilm Research Laboratory, Institute of Biomedical and Oral Sciences, Faculty of Dentistry, Hadassah Medical School, The Hebrew University of Jerusalem, Jerusalem 9112102, Israel; (S.B.); (S.B.); (D.S.)
| | - Shreya Banerjee
- Biofilm Research Laboratory, Institute of Biomedical and Oral Sciences, Faculty of Dentistry, Hadassah Medical School, The Hebrew University of Jerusalem, Jerusalem 9112102, Israel; (S.B.); (S.B.); (D.S.)
| | - Sergei Bogomolov
- Biofilm Research Laboratory, Institute of Biomedical and Oral Sciences, Faculty of Dentistry, Hadassah Medical School, The Hebrew University of Jerusalem, Jerusalem 9112102, Israel; (S.B.); (S.B.); (D.S.)
| | - Reem Smoum
- Institute for Drug Research, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem 9112102, Israel; (R.S.); (R.M.)
| | - Raphael Mechoulam
- Institute for Drug Research, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem 9112102, Israel; (R.S.); (R.M.)
| | - Doron Steinberg
- Biofilm Research Laboratory, Institute of Biomedical and Oral Sciences, Faculty of Dentistry, Hadassah Medical School, The Hebrew University of Jerusalem, Jerusalem 9112102, Israel; (S.B.); (S.B.); (D.S.)
| |
Collapse
|
42
|
Spiegel C, Steixner SJM, Coraça-Huber DC. Antibiofilm Activity of Omega-3 Fatty Acids and Its Influence on the Expression of Biofilm Formation Genes on Staphylococcus aureus. Antibiotics (Basel) 2022; 11:antibiotics11070932. [PMID: 35884185 PMCID: PMC9311851 DOI: 10.3390/antibiotics11070932] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Revised: 06/29/2022] [Accepted: 07/08/2022] [Indexed: 02/05/2023] Open
Abstract
Background: Currently, 1–2% of all prosthetic joint surgeries are followed by an infection. These infections cause approximately 4% of deaths in the first year after surgery, while the 5-year mortality rate is up to 21%. Prosthetic joint infections are mainly caused by Staphylococcus aureus or Staphylococcus epidermis strains. Both species share the capability of biofilm formation and methicillin resistance. The formation of biofilm helps bacterial cells to withstand critical environmental conditions. Due to their tolerance against antibacterial substances, biofilms are a significant problem in modern medicine. Alternatives for the use of methicillin as a therapeutic are not yet widespread. The use of omega-3 fatty acids, such as docosahexaenoic acid, may help against prosthetic joint infections and lower mortality rates. The aim of this study is to evaluate if docosahexaenoic acid offers a safe anti-biofilm activity against Staphylococcus aureus and MRSA without enhancing icaADBC-dependent biofilm formation or additional stress responses, therefore enhancing antibiotic tolerance and resistance. Methods: In this study, we examined the gene expression of biofilm-associated genes and regulators. We performed RT-qPCR after RNA extraction of Staphylococcus aureus ATCC 29213 and one clinical MRSA strain. We compared gene expression of icaADBC, SarA, SigB, and agrAC under the influence of 1.25 mg /L and 0.625 mg/L of docosahexaenoic acid to their controls. Results: We found a higher expression of regulatory genes such as SarA, SigB, agrA, and agrC at 1.25 mg/L of docosahexaenoic acid in ATCC 29213 and a lower increase in gene expression levels in clinical MRSA isolates. icaADBC was not affected in both strains at both concentration levels by docosahexaenoic acid. Conclusions: Docosahexaenoic acid does not enhance icaADBC-dependent biofilm formation while still reducing bacterial CFU in biofilms. Docosahexaenoic acid can be considered an option as a therapeutic substance against biofilm formation and may be a good alternative in reducing the risk of MRSA formation.
Collapse
|
43
|
Abbas HA, Shaker GH, Mosallam FM, Gomaa SE. Novel silver metformin nano-structure to impede virulence of Staphylococcus aureus. AMB Express 2022; 12:84. [PMID: 35771288 PMCID: PMC9247137 DOI: 10.1186/s13568-022-01426-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Accepted: 06/21/2022] [Indexed: 11/25/2022] Open
Abstract
Staphylococcus aureus is a prevalent etiological agent of health care associated and community acquired infections. Antibiotic abuse resulted in developing multidrug resistance in S. aureus that complicates treatment of infections. Targeting bacterial virulence using FDA approved medication offers an alternative to the antibiotics with no stress on bacterial viability. Using nanomaterials as anti-virulence agent against S. aureus virulence factors is a valuable approach. This study aims to investigate the impact of metformin (MET), metformin nano (MET-Nano), silver metformin nano structure (Ag-MET-Ns) and silver nanoparticles (AgNPs) on S. aureus virulence and pathogenicity. The in vitro results showed a higher inhibitory activity against S. aureus virulence factors with both MET-Nano and Ag-MET-Ns treatment. However, genotypically, it was found that except for agrA and icaR genes that are upregulated, the tested agents significantly downregulated the expression of crtM, sigB, sarA and fnbA genes, with Ag-MET-Ns being the most efficient one. MET-Nano exhibited the highest protection against S. aureus infection in mice. These data indicate the promising anti-virulence activity of nanoformulations especially Ag-MET-Ns against multidrug resistant S. aureus by inhibiting quorum sensing signaling system. A new formation of silver metformin nanostructure. The in vitro inhibition of S. aureus virulence factors. Nano structure form improves the activity of anti-virulence agents.
Collapse
Affiliation(s)
- Hisham A Abbas
- Department of Microbiology and Immunology, Faculty of Pharmacy, Zagazig University,, Zagazig, Egypt
| | - Ghada H Shaker
- Department of Microbiology and Immunology, Faculty of Pharmacy, Zagazig University,, Zagazig, Egypt
| | - Farag M Mosallam
- Drug Microbiology Lab., Drug Radiation Research Department, Biotechnology Division, National Center for Radiation Research and Technology (NCRRT), Egyptian Atomic Energy Authority, Cairo, Egypt.
| | - Salwa E Gomaa
- Department of Microbiology and Immunology, Faculty of Pharmacy, Zagazig University,, Zagazig, Egypt
| |
Collapse
|
44
|
Wang M, Buist G, van Dijl JM. Staphylococcus aureus cell wall maintenance - the multifaceted roles of peptidoglycan hydrolases in bacterial growth, fitness, and virulence. FEMS Microbiol Rev 2022; 46:6604383. [PMID: 35675307 PMCID: PMC9616470 DOI: 10.1093/femsre/fuac025] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Revised: 04/22/2022] [Accepted: 05/25/2022] [Indexed: 01/07/2023] Open
Abstract
Staphylococcus aureus is an important human and livestock pathogen that is well-protected against environmental insults by a thick cell wall. Accordingly, the wall is a major target of present-day antimicrobial therapy. Unfortunately, S. aureus has mastered the art of antimicrobial resistance, as underscored by the global spread of methicillin-resistant S. aureus (MRSA). The major cell wall component is peptidoglycan. Importantly, the peptidoglycan network is not only vital for cell wall function, but it also represents a bacterial Achilles' heel. In particular, this network is continuously opened by no less than 18 different peptidoglycan hydrolases (PGHs) encoded by the S. aureus core genome, which facilitate bacterial growth and division. This focuses attention on the specific functions executed by these enzymes, their subcellular localization, their control at the transcriptional and post-transcriptional levels, their contributions to staphylococcal virulence and their overall importance in bacterial homeostasis. As highlighted in the present review, our understanding of the different aspects of PGH function in S. aureus has been substantially increased over recent years. This is important because it opens up new possibilities to exploit PGHs as innovative targets for next-generation antimicrobials, passive or active immunization strategies, or even to engineer them into effective antimicrobial agents.
Collapse
Affiliation(s)
- Min Wang
- Department of Medical Microbiology, University of Groningen, University Medical Center Groningen, Hanzeplein 1, PO Box 30001, 9700 RB Groningen, the Netherlands
| | | | - Jan Maarten van Dijl
- Corresponding author: Department of Medical Microbiology, University of Groningen, University Medical Center Groningen, Hanzeplein 1, P.O. box 30001, HPC EB80, 9700 RB Groningen, the Netherlands, Tel. +31-50-3615187; Fax. +31-50-3619105; E-mail:
| |
Collapse
|
45
|
Rahman MA, Amirkhani A, Chowdhury D, Mempin M, Molloy MP, Deva AK, Vickery K, Hu H. Proteome of Staphylococcus aureus Biofilm Changes Significantly with Aging. Int J Mol Sci 2022; 23:6415. [PMID: 35742863 PMCID: PMC9223533 DOI: 10.3390/ijms23126415] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2022] [Revised: 05/30/2022] [Accepted: 06/06/2022] [Indexed: 01/15/2023] Open
Abstract
Staphylococcus aureus is a notorious biofilm-producing pathogen that is frequently isolated from implantable medical device infections. As biofilm ages, it becomes more tolerant to antimicrobial treatment leading to treatment failure and necessitating the costly removal of infected devices. In this study, we performed in-solution digestion followed by TMT-based high-throughput mass spectrometry and investigated what changes occur in the proteome of S. aureus biofilm grown for 3-days and 12-days in comparison with 24 h planktonic. It showed that proteins associated with biosynthetic processes, ABC transporter pathway, virulence proteins, and shikimate kinase pathway were significantly upregulated in a 3-day biofilm, while proteins associated with sugar transporter, degradation, and stress response were downregulated. Interestingly, in a 3-day biofilm, we observed numerous proteins involved in the central metabolism pathways which could lead to biofilm growth under diverse environments by providing an alternative metabolic route to utilize energy. In 12-day biofilms, proteins associated with peptidoglycan biosynthesis, sugar transporters, and stress responses were upregulated, whereas proteins associated with ABC transporters, DNA replication, and adhesion proteins were downregulated. Gene Ontology analysis revealed that more proteins are involved in metabolic processes in 3dwb compared with 12dwb. Furthermore, we observed significant variations in the formation of biofilms resulting from changes in the level of metabolic activity in the different growth modes of biofilms that could be a significant factor in S. aureus biofilm maturation and persistence. Collectively, potential marker proteins were identified and further characterized to understand their exact role in S. aureus biofilm development, which may shed light on possible new therapeutic regimes in the treatment of biofilm-related implant-associated infections.
Collapse
Affiliation(s)
- Md. Arifur Rahman
- Surgical Infection Research Group, Faculty of Medicine, Health and Human Sciences, Macquarie University, Sydney 2109, Australia; (D.C.); (M.M.); (A.K.D.); (K.V.)
| | - Ardeshir Amirkhani
- Australian Proteome Analysis Facility, Macquarie University, Sydney 2109, Australia; (A.A.); (M.P.M.)
| | - Durdana Chowdhury
- Surgical Infection Research Group, Faculty of Medicine, Health and Human Sciences, Macquarie University, Sydney 2109, Australia; (D.C.); (M.M.); (A.K.D.); (K.V.)
| | - Maria Mempin
- Surgical Infection Research Group, Faculty of Medicine, Health and Human Sciences, Macquarie University, Sydney 2109, Australia; (D.C.); (M.M.); (A.K.D.); (K.V.)
| | - Mark P. Molloy
- Australian Proteome Analysis Facility, Macquarie University, Sydney 2109, Australia; (A.A.); (M.P.M.)
| | - Anand Kumar Deva
- Surgical Infection Research Group, Faculty of Medicine, Health and Human Sciences, Macquarie University, Sydney 2109, Australia; (D.C.); (M.M.); (A.K.D.); (K.V.)
| | - Karen Vickery
- Surgical Infection Research Group, Faculty of Medicine, Health and Human Sciences, Macquarie University, Sydney 2109, Australia; (D.C.); (M.M.); (A.K.D.); (K.V.)
| | - Honghua Hu
- Surgical Infection Research Group, Faculty of Medicine, Health and Human Sciences, Macquarie University, Sydney 2109, Australia; (D.C.); (M.M.); (A.K.D.); (K.V.)
| |
Collapse
|
46
|
Luo S, Yang X, Wu S, Li Y, Wu J, Liu M, Liu Z, Yu K, Wang X, Dai T, Huang X, Hu X. Understanding a defensive response of methicillin-resistant Staphylococcus aureus (MRSA) after exposure to multiple cycles of sub-lethal blue Light. FEMS Microbiol Lett 2022; 369:6604381. [PMID: 35675215 DOI: 10.1093/femsle/fnac050] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Revised: 04/04/2022] [Accepted: 06/03/2022] [Indexed: 11/14/2022] Open
Abstract
Blue light (BL) has showed bactericidal effectiveness against methicillin-resistant Staphylococcus aureus (MRSA), one of the major clinical pathogens with antibiotic resistance. Bacteria likely respond to the oxidative stress induced by BL, however, the defensive response is still unclear. This study was to reveal the phenotypic change of MRSA after exposed to 15 cycles of sub-lethal blue light illumination. The comparative transcriptomic results showed that the expression of peptidoglycan (PG) synthesis gene glmS was significantly up-regulated in the cells after the multiple cycle light treatment, and the biochemical analysis determined that the content of PG synthesized was increased by 25.86% when compared to that in control cells. Furthermore, significant thickening of the cell wall was observed under transmission electron microscope (P < 0.05). The light sensitivity of the tested MRSA strain was reduced after the multiple cycles light treatment, indicating the possibility of MRSA being more adaptive to the BL stress. The present study suggested that the multiple cycles of sub-lethal BL could change the light susceptibility of MRSA through thickening cell wall.
Collapse
Affiliation(s)
- Shuanghua Luo
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi 214122, China
| | - Xi Yang
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi 214122, China
| | - Shuyan Wu
- AgResearch Ltd., Hopkirk Research Institute, Cnr University Ave and Library Road, Massey University, Palmerston North 4442, New Zealand
| | - Yuanbu Li
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi 214122, China
| | - Jiaxin Wu
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi 214122, China
| | - Minmin Liu
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi 214122, China
| | - Zhaojun Liu
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi 214122, China
| | - Keyang Yu
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi 214122, China
| | - Xiaoyuan Wang
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi 214122, China
| | - Tianhong Dai
- Department of Dermatology, Harvard Medical School, Boston, MA 02114, USA
| | - Xiaodong Huang
- Guangzhou YueHui Cosmetics Co. Ltd., Guangzhou 514410, China
| | - Xiaoqing Hu
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi 214122, China
| |
Collapse
|
47
|
Characterization of Staphylococcus aureus biofilms via crystal violet binding and biochemical composition assays of isolates from hospitals, raw meat, and biofilm-associated gene mutants. Microb Pathog 2022; 167:105554. [DOI: 10.1016/j.micpath.2022.105554] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Revised: 04/21/2022] [Accepted: 04/22/2022] [Indexed: 11/18/2022]
|
48
|
Pulia MS, Anderson J, Ye Z, Elsayed NS, Le T, Patitucci J, Ganta K, Hall M, Singh VK, Shukla SK. Expression of Staphylococcal Virulence Genes In Situ in Human Skin and Soft Tissue Infections. Antibiotics (Basel) 2022; 11:527. [PMID: 35453277 PMCID: PMC9032627 DOI: 10.3390/antibiotics11040527] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Revised: 04/08/2022] [Accepted: 04/10/2022] [Indexed: 11/16/2022] Open
Abstract
BACKGROUND Staphylococcus aureus, the most common pathogen in skin and soft tissue infections (SSTI), harbors many well-characterized virulence genes. However, the expression of many of them in SSTIs is unknown. In this study, S. aureus virulence genes expressed in SSTI were investigated. METHODS Fifty-three subjects presenting to the outpatient's care and emergency departments with a purulent SSTI at two medical centers in Wisconsin, USA, were enrolled in the study. Total mRNA was extracted from the purulent or swab materials, made into cDNA and sequenced on MiSeq platform. The relative cDNA counts to gmk and identifications of the transcripts were carried out with respect to USA300 reference genome and using SAMTOOLS v.1.3 and BWA, respectively. RESULT A significantly higher cDNA count was observed for many of the virulence and regulatory gene transcripts in the pus samples compared to the swab samples relative to the cDNA counts for gmk, a housekeeping gene. They were for lukS-PV (18.6 vs. 14.2), isaA (13.4 vs. 8.5), ssaA (4.8 vs. 3.1), hlgC (1.4 vs. 1.33), atl (17.7 vs. 8.33), clfA (3.9 vs. 0.83), eno (6.04 vs. 3.16), fnbA (5.93 vs. 0.33), saeS (6.3 vs. 1.33), saeR (5.4 vs. 3.33) and agrC (5.6 vs. 1.5). CONCLUSIONS A relative increase in the transcripts of several toxins, adhesion and regulatory genes with respect to a gmk in purulent materials suggests their role in situ during SSTIs, perhaps in an orchestrated manner.
Collapse
Affiliation(s)
- Michael S. Pulia
- Department of Emergency Medicine, University of Wisconsin, School of Medicine and Public Health, Madison, WI 53726, USA;
| | - Jennifer Anderson
- Integrated Research Development Center, Marshfield Clinic Research Institute, Marshfield, WI 54449, USA; (J.A.); (T.L.)
| | - Zhan Ye
- Bioinformatics Research Center, Marshfield Clinic Research Institute, Marshfield, WI 54449, USA; (Z.Y.); (J.P.)
| | - Noha S. Elsayed
- Center for Precision Medicine Research, Marshfield Clinic Research Institute, Marshfield, WI 54449, USA; (N.S.E.); (K.G.)
| | - Thao Le
- Integrated Research Development Center, Marshfield Clinic Research Institute, Marshfield, WI 54449, USA; (J.A.); (T.L.)
| | - Jacob Patitucci
- Bioinformatics Research Center, Marshfield Clinic Research Institute, Marshfield, WI 54449, USA; (Z.Y.); (J.P.)
| | - Krishna Ganta
- Center for Precision Medicine Research, Marshfield Clinic Research Institute, Marshfield, WI 54449, USA; (N.S.E.); (K.G.)
| | - Matthew Hall
- Department of Infectious Diseases, Marshfield Clinic Health System, Marshfield, WI 54449, USA;
| | - Vineet K. Singh
- Department of Microbiology and Immunology, Kirksville College of Osteopathic Medicine, A.T. Still, University of Health Sciences, Kirksville, MO 63501, USA;
| | - Sanjay K. Shukla
- Center for Precision Medicine Research, Marshfield Clinic Research Institute, Marshfield, WI 54449, USA; (N.S.E.); (K.G.)
| |
Collapse
|
49
|
Hesperidin inhibits biofilm formation, virulence and staphyloxanthin synthesis in methicillin resistant Staphylococcus aureus by targeting SarA and CrtM: an in vitro and in silico approach. World J Microbiol Biotechnol 2022; 38:44. [PMID: 35064842 DOI: 10.1007/s11274-022-03232-5] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Accepted: 01/07/2022] [Indexed: 01/20/2023]
Abstract
Methicillin resistant Staphylococcus aureus is considered multidrug resistant bacterium due to developing biofilm formation associated with antimicrobial resistance mechanisms. Therefore, inhibition of biofilm formation is an alternative therapeutic action to control MRSA infections. The present study revealed the non-antibacterial biofilm inhibitory potential of hesperidin against ATCC strain and clinical isolates of S. aureus. Hesperidin is a flavanone glycoside commonly found in citrus fruit. Hesperidin has been shown to exhibits numerous pharmacological activities. The present study aimed to evaluate the antibiofilm and antivirulence potential of hesperidin against MRSA. Results showed that hesperidin treatment significantly impedes lipase, hemolysin, autolysin, autoaggregation and staphyloxanthin production. Reductions of staphyloxanthin production possibly increase the MRSA susceptibility rate to H2O2 oxidative stress condition. In gene expression study revealed that hesperidin treatment downregulated the biofilm-associated gene (sarA), polysaccharide intracellular adhesion gene (icaA and icaD), autolysin (altA), fibronectin-binding protein (fnbA and fnbB) and staphyloxanthin production (crtM). Molecular docking analysis predicted the ability of hesperidin to interact with SarA and CrtM proteins involved in biofilm formation and staphyloxanthin production in MRSA.
Collapse
|
50
|
Rattanakiat S, Taensantia A, Jaemamporn K, Khamnuanin S, Mudjupa C, Jaruchotikamol A, Pulbutr P. Antibiofilm Formation Activity of Lupinifolin Against Methicillin-Resistant Staphylococcus aureus. Pak J Biol Sci 2022; 25:961-970. [PMID: 36591926 DOI: 10.3923/pjbs.2022.961.970] [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: 11/22/2022]
Abstract
<b>Background and Objective:</b> Biofilm formation activity of Methicillin-resistant <i>Staphylococcus aureus</i> (MRSA) is one of the crucial factors rendering this pathogenic bacterium difficult to be eradicated. It has been reported that lupinifolin, is a major phytochemical agent isolated from <i>Derris reticulata</i> Craib. stem possesses antibacterial activity against MRSA. This study aimed to investigate the effects of lupinifolin and its combinations with some antibacterial drugs, including ampicillin, cloxacillin or vancomycin, on the biofilm formation activity of MRSA. <b>Materials and Methods:</b> The crystal violet biofilm formation assay was performed to evaluate the biofilm formation activity. <b>Results:</b> Lupinifolin produced a significant inhibitory activity against MRSA biofilm formation with the median inhibitory concentration (IC<sub>50</sub>) of 7.96±3.05 μg mL<sup>1</sup> (n = 6) at 24 hrs incubation. Lupinifolin at the concentrations of sub-MICs (1, 2, 4 and 8 μg mL<sup>1</sup>) combined with the antibacterial drugs at their sub-MICs also exhibited substantial antibiofilm formation activities. The maximal antibiofilm activity was found with the combination of lupinifolin (8 μg mL<sup>1</sup>) and vancomycin (1 μg mL<sup>1</sup>) by the percentage inhibition of 102.39±0.89 (n = 8). The antibiofilm formation activities of the combinations between lupinifolin and the antibacterial drugs at various concentrations tested were also significantly higher than those of lupinifolin alone. <b>Conclusion:</b> These results indicated that lupinifolin can potentially be developed as an antibacterial enhancer for the management of biofilm-associated bacterial infections caused by MRSA, in which the current pharmacological treatment is still limited.
Collapse
|