1
|
Mayyas A, Al-Samydai A, Al-Karablieh N, Zalloum WA, Al-Tawalbeh D, Al-Mamoori F, Amr RA, Al Nsairat H, Carradori S, Al-Halaseh LK, Aburjai T. A phytotherapeutic approach to hinder the resistance against clindamycin by MRSA: in vitro and in silico studies. Future Sci OA 2025; 11:2458438. [PMID: 39895160 PMCID: PMC11792796 DOI: 10.1080/20565623.2025.2458438] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Accepted: 01/09/2025] [Indexed: 02/04/2025] Open
Abstract
AIMS This study investigates the potential effects of essential oils (EOs) in enhancing the efficacy of clindamycin against Methicillin-resistant Staphylococcus aureus (MRSA) using in vitro and computer simulations. The research seeks to identify essential oils that exhibit synergistic activity with clindamycin and determine their potential key active components. MATERIALS AND METHODS Essential oils commonly used in traditional medicine were tested for their antimicrobial activity against MRSA. The minimum inhibitory concentration (MIC) was determined using in vitro microdilution assays. A synergistic test with clindamycin was performed, and molecular docking studies evaluated the interaction between a key compound (trans-cinnamaldehyde) and MRSA protein. RESULTS EOs from Cinnamomum verum, Rosmarinus officinalis, Salvia officinalis, and Thymus vulgaris demonstrated significant inhibitory and synergistic activities against MRSA, standard strain, and human clinical isolates. Gas Chromatography/Mass Spectroscopy identified trans-cinnamaldehyde, eucalyptol, and thymol as prominent antibacterial compounds. Molecular docking studies confirmed trans-cinnamaldehyde's strong binding to MRSA's AgrA protein, elucidating its enhanced efficacy. CONCLUSION The study underscores the potential of plant-based therapies to augment the effectiveness of conventional antibiotics like clindamycin in combating MRSA and addressing antibiotic resistance by integrating traditional plant remedies with modern medical approaches.
Collapse
Affiliation(s)
- Amal Mayyas
- Department of Pharmacy, Faculty of Health Sciences, American University of Madaba, Madaba, Jordan
| | - Ali Al-Samydai
- Pharmacological and Diagnostic Research Centre, Faculty of Pharmacy, Al-Ahliyya Amman University, Amman, Jordan
| | - Nehaya Al-Karablieh
- Department of Plant Protection, School of Agriculture, The University of Jordan, Amman, Jordan
- Hamdi Mango Centre for Scientific Research, The University of Jordan, Amman, Jordan
| | - Waleed A Zalloum
- Department of Pharmacy, Faculty of Health Sciences, American University of Madaba, Madaba, Jordan
| | - Deniz Al-Tawalbeh
- Department of Medicinal Chemistry and Pharmacognosy, Faculty of Pharmacy, Yarmouk University, Irbid, Jordan
| | - Farah Al-Mamoori
- Department of Pharmaceutical Sciences, Faculty of Pharmacy, Zarqa University, Zarqa, Jordan
| | - Rula A. Amr
- Department of Pharmacy, Faculty of Health Sciences, American University of Madaba, Madaba, Jordan
| | - Hamdi Al Nsairat
- Pharmacological and Diagnostic Research Centre, Faculty of Pharmacy, Al-Ahliyya Amman University, Amman, Jordan
| | - Simone Carradori
- Department of Pharmacy “G. d’Annunzio”, University of Chieti-Pescara, Chieti, SC, Italy
| | - Lidia Kamal Al-Halaseh
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Mutah University, Al-Karak, Jordan
| | - Talal Aburjai
- Faculty of Pharmacy, The University of Jordan, Amman, Jordan
| |
Collapse
|
2
|
He X, Zhang W, Liu J, Liu J, Chen Y, Luan C, Zhang J, Bao G, Lin X, Muh F, Lin T, Lu F. The global regulatory role of RsbUVW in virulence and biofilm formation in MRSA. Microb Pathog 2025; 203:107508. [PMID: 40158706 DOI: 10.1016/j.micpath.2025.107508] [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: 12/26/2024] [Revised: 03/22/2025] [Accepted: 03/23/2025] [Indexed: 04/02/2025]
Abstract
The widespread prevalence of methicillin-resistant Staphylococcus aureus (MRSA) has caused serious challenges to clinical treatment. This study was designed to explore effective targets for MRSA prevention and control. The key virulence regulator was screened through the correlation analysis between virulence and various regulatory factors in the main clinical epidemic MRSA. The potential key factors were inactivated to further evaluate the inhibitory effect on the virulence of MRSA standard strain S. aureus ATCC43300 and its influence on drug resistance and biofilm formation. Enterobacterial repetitive intergenic consensus-PCR was used to analyze the clinical epidemic genotypes of MRSA. The virulence of MRSA was evaluated mainly by measuring its adhesion and invasion ability to A549 cells, the lethality to Galleria mellonella larvae, and the transcription level of related genes. The biofilm formation was assessed by crystal violet staining on polystyrene microplates. The results showed that most virulence factors of clinical representative MRSA strain were significantly positively correlated with RsbUVW system. After knocking out the rsbV gene, a key component of the rsbUVW system, the virulence of S. aureus ATCC43300 was significantly reduced (P < 0.05), as indicated by a significant decrease in lethality against G. mellonella larvae and invasion against A549 cells, and a significant decrease in the expression of immune escape related virulence factors polysaccharide intercellular adhesin (PIA) and staphyloxanthin. The biomass and stability of protein-dependent biofilm by S. aureus ATCC43300 were significantly increased. This study will provide useful information for the effective prevention and control of MRSA.
Collapse
Affiliation(s)
- Xinlong He
- Department of Pathogenic Biology, Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, 225001, China; Institute of Translational Medicine, School of Medicine, Yangzhou University, Yangzhou, 225001, China; The Key Laboratory of the Jiangsu Higher Education Institutions for Nucleic Acid & Cell Fate Regulation (Yangzhou University), Yangzhou, 225001, China; Jiangsu Key Laboratory of Zoonosis/Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonosis, Yangzhou, 225001, China
| | - Wenwen Zhang
- Department of Pathogenic Biology, Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, 225001, China; Department of Clinical Laboratory, Changning Maternity and Infant Health Hospital, Affiliated Hospital of East China Normal University, Shanghai, 200050, China
| | - Jie Liu
- Department of Pathogenic Biology, Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, 225001, China
| | - Jiali Liu
- Department of Pathogenic Biology, Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, 225001, China
| | - Yinsong Chen
- Department of Lung, Third People's Hospital of Yangzhou, Yangzhou, China
| | - Changjiao Luan
- Department of Lung, Third People's Hospital of Yangzhou, Yangzhou, China
| | - Jun Zhang
- Department of Clinical Laboratory, First Affiliated Hospital of Yangzhou University, Yangzhou, 225000, China
| | - Guangyu Bao
- Department of Clinical Laboratory, First Affiliated Hospital of Yangzhou University, Yangzhou, 225000, China
| | - Xiangfang Lin
- Department of Clinical Laboratory, First Affiliated Hospital of Yangzhou University, Yangzhou, 225000, China
| | - Fauzi Muh
- Department of Epidemiology & Tropical Diseases, Faculty of Public Health, Universitas Diponegoro, Tembalang, Semarang, 50275, Indonesia
| | - Tao Lin
- Department of Clinical Laboratory, First Affiliated Hospital of Yangzhou University, Yangzhou, 225000, China.
| | - Feng Lu
- Department of Pathogenic Biology, Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, 225001, China.
| |
Collapse
|
3
|
Silva-Santana G, Baêta Júnior ES, Silva Conceição GM, Aguiar-Alves F, Lima Brandão ML, Lopes-Torres EJ, Mattos-Guaraldi AL. Intervention of Corynebacterium striatum in the sessile lifestyle of Staphylococcus aureus wild-type and mutants for ica genes in polymicrobial biofilms. Microb Pathog 2025; 204:107577. [PMID: 40222568 DOI: 10.1016/j.micpath.2025.107577] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2024] [Revised: 04/05/2025] [Accepted: 04/10/2025] [Indexed: 04/15/2025]
Abstract
This study investigated the interactions between Corynebacterium striatum and Staphylococcus aureus, two bacterial species commonly found in the human microbiota, particularly colonizing the skin and mucous membranes. Both organisms, however, are also capable of causing acute and chronic infections. While S. aureus is widely recognized as a clinically significant pathogen, C. striatum is frequently underestimated and often regarded as a contaminant-even when isolated in pure culture from nosocomial infections. The ability of these microorganisms to develop multidrug resistance and form biofilms complicates the management of the infections they cause. This study focused on the interaction between C. striatum and S. aureus, particularly the influence of the former on the pathogenic potential of the latter, emphasizing biofilm formation in S. aureus mutants deficient in the icaR and icaC genes. Antimicrobial susceptibility testing revealed that 85.7 % of the S. aureus strains were multidrug-resistant, with all strains resistant to β-lactam antibiotics. Additionally, 55.6 % of the strains produced strong slime on Congo Red agar, indicating a high potential for biofilm formation. In monomicrobial assays, both C. striatum and S. aureus exhibited enhanced adhesion to hydrophilic surfaces. In polymicrobial settings, C. striatum predominated in most cases: on glass surfaces, 70 % of biofilms were dominated by C. striatum, 20 % by S. aureus, and 10 % showed an even distribution. On polystyrene, 80 % of the biofilms were dominated by C. striatum, while 20 % were dominated by S. aureus. Analysis of extracellular polymeric substances (EPS) revealed distinct compositional profiles: C. striatum primarily produced proteinaceous matrices, whereas S. aureus biofilms were rich in polysaccharides. Ultrastructural examination showed that S. aureus formed dense clusters embedded in large amounts of EPS, while C. striatum biofilms exhibited lower EPS production overall. Furthermore, the effect of C. striatum-derived compounds on S. aureus biofilms was assessed. In 90 % of co-cultured strains, a progressive decrease in sessile cell populations was observed, accompanied by an increase in planktonic cells. This finding suggests that C. striatum can disrupt the biofilm integrity of S. aureus, potentially modulating its pathogenic phenotype. In conclusion, the results demonstrate that C. striatum competes effectively with S. aureus for surface colonization and, under certain conditions, may induce a transition of S. aureus from a sessile to a planktonic state. These findings highlight the complexity of interspecies interactions in polymicrobial communities and suggest that C. striatum may play a modulatory role in S. aureus virulence. Such insights have important implications for the understanding and treatment of polymicrobial infections.
Collapse
Affiliation(s)
- Giorgio Silva-Santana
- Health Science Center, Institute of Microbiology Paulo de Góes, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil; Laboratory of Diphtheria and Corynebacteria of Clinical Relevance, Faculty of Medical Sciences, University of the State of Rio de Janeiro. The Collaborating Centre for Reference and Research on Diphtheria/National Health Foundation/Ministry of Health, Rio de Janeiro, Brazil.
| | - Eustáquio Souza Baêta Júnior
- Department of Mechanical Engineering, Postgraduate Program in Mechanical Engineering, FEN, University of the State of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Greice Maria Silva Conceição
- Systems, Analytical Indicators and Data Section, Department of Quality Control, Bio- Manguinhos, Fiocruz, Rio de Janeiro, Brazil
| | - Fabio Aguiar-Alves
- Department of Pharmaceutical Sciences, Lloyd L. Gregory School of Pharmacy, Palm Beach Atlantic University, FL, 33401, USA
| | - Marcelo Luiz Lima Brandão
- Laboratory of Microbiology Control, Department of Quality Control, Bio-Manguinhos, Fiocruz, Rio de Janeiro, Brazil
| | - Eduardo José Lopes-Torres
- Laboratório de Helmintologia Romero Lascasas Porto, Departamento de Microbiologia, Imunologia e Parasitologia, Universidade do Estado do Rio de Janeiro, Brazil; Laboratório Multiusuário de Parasitologia, Departamento de Microbiologia, Imunologia e Parasitologia, Universidade do Estado do Rio de Janeiro, Brazil
| | - Ana Luiza Mattos-Guaraldi
- Health Science Center, Institute of Microbiology Paulo de Góes, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil; Laboratory of Diphtheria and Corynebacteria of Clinical Relevance, Faculty of Medical Sciences, University of the State of Rio de Janeiro. The Collaborating Centre for Reference and Research on Diphtheria/National Health Foundation/Ministry of Health, Rio de Janeiro, Brazil
| |
Collapse
|
4
|
Enroth TJ, Severn MM, Costa FG, Bovee AR, Wilkening RV, Nguyen DT, Langouët-Astrié C, Horswill AR. Global changes in Staphylococcus aureus virulence and metabolism during colonization of healthy skin. Infect Immun 2025; 93:e0002825. [PMID: 40116525 PMCID: PMC11977313 DOI: 10.1128/iai.00028-25] [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: 01/22/2025] [Accepted: 02/15/2025] [Indexed: 03/23/2025] Open
Abstract
Staphylococcus aureus and its antibiotic-resistant derivative, methicillin-resistant S. aureus (MRSA), are the leading causative agents of skin and soft tissue infections globally. S. aureus transiently colonizes the skin of healthy adults, and this transient colonization likely precedes an active infection. In recent years, there have been efforts to elucidate specific factors that help MRSA transition to an active infection, but the specific genetic determinants required for this transition following skin colonization are largely unknown. To address this question, we developed a model of asymptomatic colonization of mouse skin by MRSA. From this model, we could determine the MRSA and mouse transcriptional profiles by RNA sequencing (RNAseq) at 5- and 24-hour post-colonization. The fadXDEBA locus, required for fatty acid metabolism, was highly upregulated in our data, as were numerous virulence factors. RNAseq data were confirmed via functional in vitro and in vivo promoter-fusion assays using live bioluminescent imaging of the fadXDEBA locus promoter driving fadB transcription. We analyzed the functional capacity of members of the fadXDEBA locus, which encode crucial enzymatic components of the S. aureus β-oxidation pathway. The genes fadD and fadA modulate MRSA resistance to fosfomycin and other oxidative stressors during growth in the presence of the common skin fatty acid, palmitic acid. Overall, our data demonstrate that there are global changes to the MRSA transcriptome, priming the bacteria for survival by upregulation of known virulence factors and metabolic genes implicated in host skin-nutrient utilization.IMPORTANCEStaphylococcus aureus is a major global agent of skin and soft tissue infections. S. aureus colonizes the skin transiently, an important precursor to infection. However, little is known about how S. aureus adapts to the skin at the transcriptional level. This study provides an overview of the S. aureus transcriptome during mouse skin colonization via RNA sequencing. We identified that the most highly upregulated genes during colonization are related to fatty acid metabolism. The disruption of certain genes in the fatty acid degradation pathway altered resistance of S. aureus to the antibiotic fosfomycin. This study provides an important step in understanding the transcriptional changes that occur during S. aureus skin colonization and may reveal novel targets of therapeutic interest for preventing skin infections.
Collapse
Affiliation(s)
- Timothy J. Enroth
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, Colorado, USA
| | - Morgan M. Severn
- Department of Dermatology, Duke University, Durham, North Carolina, USA
| | - Flavia G. Costa
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, Colorado, USA
| | - Alyson R. Bovee
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, Colorado, USA
| | - Reid V. Wilkening
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, Colorado, USA
- Section of Pediatric Critical Care Medicine, Department of Pediatrics, University of Colorado School of Medicine, Aurora, Colorado, USA
| | - Dustin T. Nguyen
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, Colorado, USA
| | | | - Alexander R. Horswill
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, Colorado, USA
- Department of Veterans Affairs Eastern, Colorado Healthcare System, Grand Junction, Colorado, USA
| |
Collapse
|
5
|
Barman P, Sharma C, Joshi S, Sharma S, Maan M, Rishi P, Singla N, Saini A. In Vivo Acute Toxicity and Therapeutic Potential of a Synthetic Peptide, DP1 in a Staphylococcus aureus Infected Murine Wound Excision Model. Probiotics Antimicrob Proteins 2025; 17:843-856. [PMID: 37910332 DOI: 10.1007/s12602-023-10176-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/10/2023] [Indexed: 11/03/2023]
Abstract
Bacterial infections at the surgical sites are one of the most prevalent skin infections that impair the healing mechanism. They account for about 20% of all types of infections and lead to approximately 75% of surgical-site infection-associated mortality. Several antibiotics, such as cephalosporins, fluoroquinolones, quinolones, penicillin, sulfonamides, etc., that are used to treat such wound infections not only counter infections but also disrupt the normal flora. Moreover, antibiotics, when used for a prolonged duration, may impair the formation of new blood vessels, delay collagen production, or inhibit the migration of certain cells involved in wound repair, leading to an impaired healing process. Therefore, there is a dire need for alternate therapeutic approaches against such infections. Antimicrobial peptides have gained considerable attention as a promising strategy to counter these pathogens and prevent the spread of infection. Recently, we have reported a designed peptide, DP1, and its broad-spectrum in vitro antimicrobial activity against Gram-positive and Gram-negative bacteria. In the present study, in vivo acute toxicity of DP1 was evaluated and even at a high dose (20 mg/kg body weight) of DP1, a 100% survival of mice was observed. Subsequently, a Staphylococcus aureus-infected murine wound excision model was established to assess the wound healing efficacy of DP1. The study revealed significant wound healing vis-a-vis attenuated S. aureus bioburden at the wound site and also controlled the oxidative stress depicting anti-oxidant activity as well. Healing of the infected wounds was also verified by histopathological examination. Based on the results of this study, it can be concluded that DP1 improves wound resolution despite infections and promotes the healing mechanism. Hence, DP1 holds compelling potential as a novel antimicrobial drug that requires further explorations in clinical platforms.
Collapse
Affiliation(s)
- Panchali Barman
- Institute of Forensic Science and Criminology (UIEAST), Panjab University, Chandigarh, 160014, India
| | - Chakshu Sharma
- Department of Biophysics, Panjab University, Chandigarh, U.T, 160014, India
| | - Shubhi Joshi
- Department of Biophysics, Panjab University, Chandigarh, U.T, 160014, India
| | - Sheetal Sharma
- Department of Biophysics, Panjab University, Chandigarh, U.T, 160014, India
| | - Mayank Maan
- Department of Biophysics, Panjab University, Chandigarh, U.T, 160014, India
| | - Praveen Rishi
- Department of Microbiology, Panjab University, Chandigarh, U.T, 160014, India
| | - Neha Singla
- Department of Biophysics, Panjab University, Chandigarh, U.T, 160014, India
| | - Avneet Saini
- Department of Biophysics, Panjab University, Chandigarh, U.T, 160014, India.
| |
Collapse
|
6
|
Younes KM, Abouzied AS, Alafnan A, Huwaimel B, Khojali WMA, Alzahrani RM. Investigating the bispecific lead compounds against methicillin-resistant Staphylococcus aureus SarA and CrtM using machine learning and molecular dynamics approach. J Biomol Struct Dyn 2025; 43:3348-3365. [PMID: 38147401 DOI: 10.1080/07391102.2023.2297012] [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: 09/08/2023] [Accepted: 12/14/2023] [Indexed: 12/28/2023]
Abstract
Methicillin-resistant Staphylococcus aureus (MRSA) is a notorious pathogen that has emerged as a serious global health concern over the past few decades. Staphylococcal accessory regulator A (SarA) and 4,4'-diapophytoene synthase (CrtM) play a crucial role in biofilm formation and staphyloxanthin biosynthesis. Thus, the present study used a machine learning-based QSAR model to screen 1261 plant-derived natural organic compounds in order to identify a medication candidate with both biofilm and virulence inhibitory potential. Additionally, the in-silico molecular docking analysis has demonstrated significant binding efficacy of the identified hit compound, that is 85137543, with SarA and CrtM when compared to the control compound, hesperidin. Post-MD simulation analysis of the complexes depicted strong binding of 85137543 to both SarA and CrtM. Moreover, 85137543 showed hydrogen bonding with the key residues of both proteins during docking (ALA138 of SarA and ALA134 of CrtM) and post-MD simulation (LYS273 of CrtM and ASN212 of SarA). The RMSD of 85137543 was stable and consistent when bound to both CrtM and SarA with RMSDs of 1.3 and 1 nm, respectively. In addition, principal component analysis and the free energy landscape showed stable complex formation with both proteins. Low binding free energy (ΔGTotal) was observed by 85137543 for SarA (-47.92 kcal/mol) and CrtM (-36.43 kcal/mol), which showed strong binding. Overall, this study identified 85137543 as a potential inhibitor of both SarA and CrtM in MRSA.
Collapse
Affiliation(s)
- Kareem M Younes
- Department of Pharmaceutical Chemistry, College of Pharmacy, University of Hail, Hail, Saudi Arabia
- Department of Analytical Chemistry, Faculty of Pharmacy, Cairo University, Cairo, Egypt
| | - Amr S Abouzied
- Department of Pharmaceutical Chemistry, College of Pharmacy, University of Hail, Hail, Saudi Arabia
- Department of Pharmaceutical Chemistry, National Organization for Drug Control and Research (NODCAR), Giza, Egypt
| | - Ahmed Alafnan
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Hail, Hail, Saudi Arabia
| | - Bader Huwaimel
- Department of Pharmaceutical Chemistry, College of Pharmacy, University of Hail, Hail, Saudi Arabia
- Medical and Diagnostic Research Center, University of Ha'il, Hail, Saudi Arabia
| | - Weam M A Khojali
- Department of Pharmaceutical Chemistry, College of Pharmacy, University of Hail, Hail, Saudi Arabia
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Omdurman Islamic University, Omdurman, Sudan
| | - Rami M Alzahrani
- Department of Pharmaceutics, College of Pharmacy, Taif University, Taif, Saudi Arabia
| |
Collapse
|
7
|
de Castro IM, Antunes C, Valentim CC, Spoladori LFDA, Suzukawa HT, Correia GF, Silva-Rodrigues G, Borges PHG, Bartolomeu-Gonçalves G, Silva ML, Bispo MDLF, Machado RRB, Nakamura CV, Nakazato G, Pinge-Filho P, Tavares ER, Yamauchi LM, Yamada-Ogatta SF. Synergistic Antibacterial Interaction of Geraniol and Biogenic Silver Nanoparticles on Methicillin-Resistant Staphylococcus aureus. PLANTS (BASEL, SWITZERLAND) 2025; 14:1059. [PMID: 40219128 PMCID: PMC11991589 DOI: 10.3390/plants14071059] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/11/2025] [Revised: 03/25/2025] [Accepted: 03/27/2025] [Indexed: 04/14/2025]
Abstract
Since ancient times, plants have been used in folk medicine to treat different diseases. Plants offer exceptional chemical diversity with a wide range of biological activities, and have therefore been the most promising sources for the discovery and development of drugs, including antimicrobial agents. This study reports the antibacterial effect of geraniol (GER), alone and in combination with biogenic silver nanoparticles (bioAgNPs), produced using the aqueous extract of Trichilia catigua bark, against planktonic and sessile cells of methicillin-resistant Staphylococcus aureus (MRSA), one of the main opportunistic and potentially fatal human pathogens. GER had a time-dependent bactericidal effect on planktonic cells, impairing the cell membrane integrity. In addition, GER inhibited the staphyloxanthin production, and molecular docking analyses supported the in silico affinity of GER to dehydrosqualene synthase (CrtM) and 4,4'-diaponeurosporen-aldehyde dehydrogenase (AldH), which are key enzymes within the pigment biosynthesis pathway in S. aureus. GER treatment increased the sensitivity of MRSA to hydrogen peroxide killing. GER displayed synergism with bioAgNPs against planktonic and sessile cells, inhibiting bacterial adhesion and the viability of biofilms formed on abiotic surfaces. MRSA planktonic and sessile cells treated with GER or GER/bioAgNPs displayed severe morphological and ultrastructural alterations. Notably, neither GER nor its combination caused in vitro and in vivo toxicity in mammalian cells and Galleria mellonella larvae, respectively. These findings suggest that the combination of GER/bioAgNPs may be a promising strategy to control MRSA infections.
Collapse
Affiliation(s)
- Isabela Madeira de Castro
- Programa de Pós-graduação em Microbiologia, Universidade Estadual de Londrina, Londrina 86055-900, Paraná, Brazil; (I.M.d.C.); (L.F.d.A.S.); (H.T.S.); (G.F.C.); (G.S.-R.); (P.H.G.B.); (C.V.N.); (G.N.); (P.P.-F.); (E.R.T.); (L.M.Y.)
- Laboratório de Biologia Molecular de Microrganismos, Universidade Estadual de Londrina, Londrina 86055-900, Paraná, Brazil; (C.A.); (C.C.V.); (G.B.-G.)
| | - Camila Antunes
- Laboratório de Biologia Molecular de Microrganismos, Universidade Estadual de Londrina, Londrina 86055-900, Paraná, Brazil; (C.A.); (C.C.V.); (G.B.-G.)
| | - Camila Cristina Valentim
- Laboratório de Biologia Molecular de Microrganismos, Universidade Estadual de Londrina, Londrina 86055-900, Paraná, Brazil; (C.A.); (C.C.V.); (G.B.-G.)
| | - Laís Fernanda de Almeida Spoladori
- Programa de Pós-graduação em Microbiologia, Universidade Estadual de Londrina, Londrina 86055-900, Paraná, Brazil; (I.M.d.C.); (L.F.d.A.S.); (H.T.S.); (G.F.C.); (G.S.-R.); (P.H.G.B.); (C.V.N.); (G.N.); (P.P.-F.); (E.R.T.); (L.M.Y.)
- Laboratório de Biologia Molecular de Microrganismos, Universidade Estadual de Londrina, Londrina 86055-900, Paraná, Brazil; (C.A.); (C.C.V.); (G.B.-G.)
| | - Helena Tiemi Suzukawa
- Programa de Pós-graduação em Microbiologia, Universidade Estadual de Londrina, Londrina 86055-900, Paraná, Brazil; (I.M.d.C.); (L.F.d.A.S.); (H.T.S.); (G.F.C.); (G.S.-R.); (P.H.G.B.); (C.V.N.); (G.N.); (P.P.-F.); (E.R.T.); (L.M.Y.)
- Laboratório de Biologia Molecular de Microrganismos, Universidade Estadual de Londrina, Londrina 86055-900, Paraná, Brazil; (C.A.); (C.C.V.); (G.B.-G.)
| | - Guilherme Ferreira Correia
- Programa de Pós-graduação em Microbiologia, Universidade Estadual de Londrina, Londrina 86055-900, Paraná, Brazil; (I.M.d.C.); (L.F.d.A.S.); (H.T.S.); (G.F.C.); (G.S.-R.); (P.H.G.B.); (C.V.N.); (G.N.); (P.P.-F.); (E.R.T.); (L.M.Y.)
- Laboratório de Biologia Molecular de Microrganismos, Universidade Estadual de Londrina, Londrina 86055-900, Paraná, Brazil; (C.A.); (C.C.V.); (G.B.-G.)
| | - Gislaine Silva-Rodrigues
- Programa de Pós-graduação em Microbiologia, Universidade Estadual de Londrina, Londrina 86055-900, Paraná, Brazil; (I.M.d.C.); (L.F.d.A.S.); (H.T.S.); (G.F.C.); (G.S.-R.); (P.H.G.B.); (C.V.N.); (G.N.); (P.P.-F.); (E.R.T.); (L.M.Y.)
- Laboratório de Biologia Molecular de Microrganismos, Universidade Estadual de Londrina, Londrina 86055-900, Paraná, Brazil; (C.A.); (C.C.V.); (G.B.-G.)
| | - Paulo Henrique Guilherme Borges
- Programa de Pós-graduação em Microbiologia, Universidade Estadual de Londrina, Londrina 86055-900, Paraná, Brazil; (I.M.d.C.); (L.F.d.A.S.); (H.T.S.); (G.F.C.); (G.S.-R.); (P.H.G.B.); (C.V.N.); (G.N.); (P.P.-F.); (E.R.T.); (L.M.Y.)
- Laboratório de Biologia Molecular de Microrganismos, Universidade Estadual de Londrina, Londrina 86055-900, Paraná, Brazil; (C.A.); (C.C.V.); (G.B.-G.)
| | - Guilherme Bartolomeu-Gonçalves
- Laboratório de Biologia Molecular de Microrganismos, Universidade Estadual de Londrina, Londrina 86055-900, Paraná, Brazil; (C.A.); (C.C.V.); (G.B.-G.)
- Programa de Pós-graduação em Fisiopatologia Clínica e Laboratorial, Universidade Estadual de Londrina, Londrina 86038-350, Paraná, Brazil
| | - Mariana Luiza Silva
- Laboratório de Síntese de Moléculas Medicinais, Departamento de Química, Centro de Ciências Exatas e da Terra, Universidade Estadual de Londrina, Londrina 86055-900, Paraná, Brazil; (M.L.S.); (M.d.L.F.B.)
| | - Marcelle de Lima Ferreira Bispo
- Laboratório de Síntese de Moléculas Medicinais, Departamento de Química, Centro de Ciências Exatas e da Terra, Universidade Estadual de Londrina, Londrina 86055-900, Paraná, Brazil; (M.L.S.); (M.d.L.F.B.)
| | - Rayanne Regina Beltrame Machado
- Laboratório de Inovação Tecnológica no Desenvolvimento de Fármacos e Cosméticos, Universidade Estadual de Maringá, Maringá 87020-900, Paraná, Brazil;
| | - Celso Vataru Nakamura
- Programa de Pós-graduação em Microbiologia, Universidade Estadual de Londrina, Londrina 86055-900, Paraná, Brazil; (I.M.d.C.); (L.F.d.A.S.); (H.T.S.); (G.F.C.); (G.S.-R.); (P.H.G.B.); (C.V.N.); (G.N.); (P.P.-F.); (E.R.T.); (L.M.Y.)
- Laboratório de Inovação Tecnológica no Desenvolvimento de Fármacos e Cosméticos, Universidade Estadual de Maringá, Maringá 87020-900, Paraná, Brazil;
| | - Gerson Nakazato
- Programa de Pós-graduação em Microbiologia, Universidade Estadual de Londrina, Londrina 86055-900, Paraná, Brazil; (I.M.d.C.); (L.F.d.A.S.); (H.T.S.); (G.F.C.); (G.S.-R.); (P.H.G.B.); (C.V.N.); (G.N.); (P.P.-F.); (E.R.T.); (L.M.Y.)
| | - Phileno Pinge-Filho
- Programa de Pós-graduação em Microbiologia, Universidade Estadual de Londrina, Londrina 86055-900, Paraná, Brazil; (I.M.d.C.); (L.F.d.A.S.); (H.T.S.); (G.F.C.); (G.S.-R.); (P.H.G.B.); (C.V.N.); (G.N.); (P.P.-F.); (E.R.T.); (L.M.Y.)
- Laboratório de Imunopatologia Experimental, Departmento of Imunologia, Parasitologia e Patologia Geral, Universidade Estadual de Londrina, Londrina 86055-900, Paraná, Brazil
| | - Eliandro Reis Tavares
- Programa de Pós-graduação em Microbiologia, Universidade Estadual de Londrina, Londrina 86055-900, Paraná, Brazil; (I.M.d.C.); (L.F.d.A.S.); (H.T.S.); (G.F.C.); (G.S.-R.); (P.H.G.B.); (C.V.N.); (G.N.); (P.P.-F.); (E.R.T.); (L.M.Y.)
- Laboratório de Biologia Molecular de Microrganismos, Universidade Estadual de Londrina, Londrina 86055-900, Paraná, Brazil; (C.A.); (C.C.V.); (G.B.-G.)
- Departamento de Medicina, Pontifícia Universidade Católica do Paraná, Londrina 86067-000, Paraná, Brazil
| | - Lucy Megumi Yamauchi
- Programa de Pós-graduação em Microbiologia, Universidade Estadual de Londrina, Londrina 86055-900, Paraná, Brazil; (I.M.d.C.); (L.F.d.A.S.); (H.T.S.); (G.F.C.); (G.S.-R.); (P.H.G.B.); (C.V.N.); (G.N.); (P.P.-F.); (E.R.T.); (L.M.Y.)
- Laboratório de Biologia Molecular de Microrganismos, Universidade Estadual de Londrina, Londrina 86055-900, Paraná, Brazil; (C.A.); (C.C.V.); (G.B.-G.)
| | - Sueli Fumie Yamada-Ogatta
- Programa de Pós-graduação em Microbiologia, Universidade Estadual de Londrina, Londrina 86055-900, Paraná, Brazil; (I.M.d.C.); (L.F.d.A.S.); (H.T.S.); (G.F.C.); (G.S.-R.); (P.H.G.B.); (C.V.N.); (G.N.); (P.P.-F.); (E.R.T.); (L.M.Y.)
- Laboratório de Biologia Molecular de Microrganismos, Universidade Estadual de Londrina, Londrina 86055-900, Paraná, Brazil; (C.A.); (C.C.V.); (G.B.-G.)
- Programa de Pós-graduação em Fisiopatologia Clínica e Laboratorial, Universidade Estadual de Londrina, Londrina 86038-350, Paraná, Brazil
| |
Collapse
|
8
|
Nair SS, Kleffmann T, Smith B, Morris V, Göbl C, Pletzer D, Fellner M. Comparative lipidomics profiles of planktonic and biofilms of methicillin-resistant and -susceptible Staphylococcus aureus. Anal Biochem 2025; 698:115746. [PMID: 39672221 DOI: 10.1016/j.ab.2024.115746] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2024] [Revised: 11/27/2024] [Accepted: 12/06/2024] [Indexed: 12/15/2024]
Abstract
Staphylococcus aureus is a significant human pathogen causing acute life-threatening, and chronic infections often linked to biofilms. This study conducted a comparative lipidomic analysis of a methicillin-resistant (MRSA) and a methicillin-susceptible (MSSA) S. aureus strain in both planktonic and biofilm cultures using liquid chromatography-mass spectrometry (LC-MS) and nuclear magnetic resonance (NMR) spectroscopy. The developed protocol successfully differentiates between the strains in various living states (planktonic and biofilm) and growth media (Tryptic Soy Broth and Brain Heart Infusion) using S. aureus USA300 LAC (MRSA) and S. aureus Newman (MSSA). LC-MS and NMR lipidomics profiles revealed global differences and particular ones among the following classes of bacterial lipids: phosphatidylglycerols, diacylglycerols, monoglycosyldiacylglycerols, diglycosyldiacylglycerols, and cardiolipins. Lipid content was higher in the biofilm states for most of these classes. Growth media differences were significant, while differences between MRSA and MSSA were less pronounced but still detectable. Additionally, we provide data on hundreds of unknown compounds that differ based on living state, strain background, or growth media. This study offer insights into the dynamic nature of S. aureus lipid composition and the used methods are adaptable to other organisms.
Collapse
Affiliation(s)
- Shilpa Saseendran Nair
- Biochemistry Department, University of Otago, Dunedin, New Zealand; Department of Microbiology and Immunology, University of Otago, Dunedin, New Zealand
| | - Torsten Kleffmann
- Division of Health Sciences, Research Infrastructure Centre, University of Otago, Dunedin, New Zealand
| | - Briana Smith
- Mātai Hāora - Centre for Redox Biology and Medicine, Department of Pathology and Biomedical Science, University of Otago, Christchurch, New Zealand
| | - Vanessa Morris
- School of Biological Sciences, University of Canterbury, Christchurch, New Zealand; Biomolecular Interaction Centre, University of Canterbury, Christchurch, New Zealand
| | - Christoph Göbl
- Mātai Hāora - Centre for Redox Biology and Medicine, Department of Pathology and Biomedical Science, University of Otago, Christchurch, New Zealand; Biomolecular Interaction Centre, University of Canterbury, Christchurch, New Zealand
| | - Daniel Pletzer
- Department of Microbiology and Immunology, University of Otago, Dunedin, New Zealand
| | - Matthias Fellner
- Biochemistry Department, University of Otago, Dunedin, New Zealand.
| |
Collapse
|
9
|
Silva-Santana G. Staphylococcus aureus: Dynamics of pathogenicity and antimicrobial-resistance in hospital and community environments - Comprehensive overview. Res Microbiol 2025; 176:104267. [PMID: 39805330 DOI: 10.1016/j.resmic.2025.104267] [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: 01/08/2025] [Accepted: 01/09/2025] [Indexed: 01/16/2025]
Abstract
This study reviews Staphylococcus aureus, a significant pathogen in both hospital and community-acquired infections, addressing its epidemiology, pathogenesis, and antimicrobial resistance. It highlights virulence mechanisms, such as adhesion factors, toxins, enzymes, and biofilms, which contribute to survival and immune evasion. The spread of resistance occurs through the transfer of mobile genetic elements like SCCmec and genetic mutations. The analysis also compares hospital and community strains, including multidrug-resistant lineages like MRSA, VISA, and VRSA. The study concludes that S. aureus presents a major public health challenge, requiring new therapeutic approaches and preventive strategies.
Collapse
Affiliation(s)
- Giorgio Silva-Santana
- Health Science Center, Institute of Microbiology Paulo de Góes, Federal University of Rio de Janeiro, Rio de Janeiro (RJ), Brazil.
| |
Collapse
|
10
|
Rasquel-Oliveira FS, Ribeiro JM, Martelossi-Cebinelli G, Costa FB, Nakazato G, Casagrande R, Verri WA. Staphylococcus aureus in Inflammation and Pain: Update on Pathologic Mechanisms. Pathogens 2025; 14:185. [PMID: 40005560 PMCID: PMC11858194 DOI: 10.3390/pathogens14020185] [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: 01/03/2025] [Revised: 01/23/2025] [Accepted: 02/10/2025] [Indexed: 02/27/2025] Open
Abstract
Staphylococcus aureus (S. aureus) is a Gram-positive bacterium of significant clinical importance, known for its versatility and ability to cause a wide array of infections, such as osteoarticular, pulmonary, cardiovascular, device-related, and hospital-acquired infections. This review describes the most recent evidence of the pathogenic potential of S. aureus, which is commonly part of the human microbiota but can lead to severe infections. The prevalence of pathogenic S. aureus in hospital and community settings contributes to substantial morbidity and mortality, particularly in individuals with compromised immune systems. The immunopathogenesis of S. aureus infections involves intricate interactions with the host immune and non-immune cells, characterized by various virulence factors that facilitate adherence, invasion, and evasion of the host's defenses. This review highlights the complexity of S. aureus infections, ranging from mild to life-threatening conditions, and underscores the growing public health concern posed by multidrug-resistant strains, including methicillin-resistant S. aureus (MRSA). This article aims to provide an updated perspective on S. aureus-related infections, highlighting the main diseases linked to this pathogen, how the different cell types, virulence factors, and signaling molecules are involved in the immunopathogenesis, and the future perspectives to overcome the current challenges to treat the affected individuals.
Collapse
Affiliation(s)
- Fernanda S. Rasquel-Oliveira
- Laboratory of Pain, Inflammation, Neuropathy, and Cancer, Department of Immunology, Parasitology and General Pathology, Center of Biological Sciences, Londrina State University, Londrina 86057-970, PR, Brazil; (F.S.R.-O.)
| | - Jhonatan Macedo Ribeiro
- Department of Microbiology, Center of Biological Sciences, Londrina State University, Londrina 86057-970, PR, Brazil (G.N.)
| | - Geovana Martelossi-Cebinelli
- Laboratory of Pain, Inflammation, Neuropathy, and Cancer, Department of Immunology, Parasitology and General Pathology, Center of Biological Sciences, Londrina State University, Londrina 86057-970, PR, Brazil; (F.S.R.-O.)
| | - Fernanda Barbosa Costa
- Laboratory of Pain, Inflammation, Neuropathy, and Cancer, Department of Immunology, Parasitology and General Pathology, Center of Biological Sciences, Londrina State University, Londrina 86057-970, PR, Brazil; (F.S.R.-O.)
| | - Gerson Nakazato
- Department of Microbiology, Center of Biological Sciences, Londrina State University, Londrina 86057-970, PR, Brazil (G.N.)
| | - Rubia Casagrande
- Department of Pharmaceutical Sciences, Center of Health Science, Londrina State University, Londrina 86038-440, PR, Brazil
| | - Waldiceu A. Verri
- Laboratory of Pain, Inflammation, Neuropathy, and Cancer, Department of Immunology, Parasitology and General Pathology, Center of Biological Sciences, Londrina State University, Londrina 86057-970, PR, Brazil; (F.S.R.-O.)
| |
Collapse
|
11
|
Ahmed H, Böhmdorfer M, Jäger W, Zeitlinger M. Insights into interspecies protein binding variability using clindamycin as an example. J Antimicrob Chemother 2025; 80:363-371. [PMID: 39556193 PMCID: PMC11787889 DOI: 10.1093/jac/dkae412] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Accepted: 10/28/2024] [Indexed: 11/19/2024] Open
Abstract
BACKGROUND In the preclinical development of new drugs, animal models are often employed to predict their efficacy in humans, relying on translational pharmacokinetic/pharmacodynamic (PK/PD) studies. We performed in vitro experiments focusing on the comparison of plasma protein binding (PPB) and bacterial growth dynamics of clindamycin, a commonly used antimicrobial agent, across a range of drug concentrations and plasma environments. METHODS Human, bovine and rat plasma were used for determining PPB of clindamycin at various antibiotic concentrations in buffer and media containing 20% to 70% plasma or pure plasma using ultrafiltration (UF) and equilibrium dialysis (ED). Also bacterial growth and time-kill assays were performed in Mueller-Hinton broth (MHB) containing various percentages of plasma. RESULTS Protein binding of clindamycin correlated well between UF and ED. Notably, clindamycin exhibited substantially lower protein binding to rat plasma compared with human and bovine plasma. Staphylococcus aureus growth was significantly reduced in 70% human, bovine, and rat plasma after 4, 8 and 24 h compared with standard MHB. Time-kill data demonstrated that bacterial counts at both 20% and 70% plasmas were less when compared with MHB at drug concentrations lower than MIC after 4 and 8 h of incubation. For rat plasma, the difference was maintained over 24 h of incubation. Furthermore, a complete bacterial killing at 16 mg/L was observed after 24 h in 20% and 70% human and bovine plasma, but not for rat plasma. CONCLUSIONS Recognizing interspecies differences in PB might be essential for optimizing the translational relevance of preclinical studies.
Collapse
Affiliation(s)
- Hifza Ahmed
- Department of Clinical Pharmacology, Medical University of Vienna, Vienna, Austria
| | | | - Walter Jäger
- Department of Clinical Pharmacy, University of Vienna, Vienna, Austria
| | - Markus Zeitlinger
- Department of Clinical Pharmacology, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
12
|
Jomehzadeh N, Emrani SS. Assessment of biofilm formation, antibiotic resistance patterns, and the prevalence of adhesion-related genes in clinical Staphylococcus aureus isolates. Heliyon 2025; 11:e41537. [PMID: 39850422 PMCID: PMC11755045 DOI: 10.1016/j.heliyon.2024.e41537] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Revised: 12/13/2024] [Accepted: 12/26/2024] [Indexed: 01/25/2025] Open
Abstract
Background This study aimed to evaluate the biofilm formation abilities of clinical Staphylococcus aureus strains, assess their antibiotic susceptibility patterns, and identify the prevalence of adhesion-associated genes. Methodology In this study, a total of 60 S. aureus strains were collected from urine, pus, wounds, blood, body fluid, and sputum in health centers affiliated with Abadan University of Medical Sciences, Iran. Strains were identified via microbiological methods and polymerase chain reaction (PCR) to target the nuc gene. Antibiotic susceptibility testing (AST) was conducted via the disc diffusion method. Methicillin-resistant S. aureus (MRSA) strains were identified by cefoxitin disc diffusion and PCR targeting the mecA gene. Biofilm formation was assessed via a microtiter plate assay, and the prevalence of adhesion-encoding genes was evaluated via PCR. The data were analyzed in Excel and SPSS via statistical methods, with P-values <0.05 considered significant. Results Using AST, daptomycin and linezolid were the most effective antibiotics (100 % susceptibility rate). According to the results of the cefoxitin disc test, 48.3 % (n = 29/60) of the strains were MRSA. All the MRSA strains harbored the mecA gene. In total, 32 % of the strains were biofilm producers. Moreover, 56.2 %, 28.1 %, and 15.6 % of the strains produced weak, moderate, and strong biofilms, respectively. There were no significant differences between the MRSA and MSSA strains in terms of the association of biofilm formation with antibiotic resistance except for erythromycin (P-value = 0.0087), gentamicin (P-value = 0.0009), and penicillin (P-value = 0.0009). The most prevalent biofilm-encoding genes were icaA (76.7 %), followed by icaD (70 %), clfA (65.0 %), and fnbA (53.3 %). Conclusion This study identified MRSA strains with biofilm-forming abilities that possess adhesion-associated genes. The most prevalent biofilm-encoding gene was icaA. To prevent further spread of these strains, regional preventive measures are needed.
Collapse
Affiliation(s)
- Nabi Jomehzadeh
- Department of Basic Medical Sciences, Faculty of Medicine, Abadan University of Medical Sciences, Abadan, Iran
- Department of Microbiology, Faculty of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Sogol Seif Emrani
- Department of Basic Medical Sciences, Faculty of Medicine, Abadan University of Medical Sciences, Abadan, Iran
| |
Collapse
|
13
|
Lavrikova A, Janda M, Bujdáková H, Hensel K. Eradication of single- and mixed-species biofilms of P. aeruginosa and S. aureus by pulsed streamer corona discharge cold atmospheric plasma. THE SCIENCE OF THE TOTAL ENVIRONMENT 2025; 959:178184. [PMID: 39718071 DOI: 10.1016/j.scitotenv.2024.178184] [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: 08/27/2024] [Revised: 12/15/2024] [Accepted: 12/16/2024] [Indexed: 12/25/2024]
Abstract
Cold atmospheric plasma has recently gained much attention due to its antimicrobial effects. Among others, plasma has proven its potential to combat microbial biofilms. Yet, knowledge of complex network interactions between individual microbial species in natural infection environments of the biofilm as well as plasma-biofilm inactivation pathways is limited. This study reports the effects of a cold plasma generated by a pulsed streamer corona discharge in air on single- and mixed-species biofilms of P. aeruginosa and S. aureus. The plasma causes significant biofilm biomass reduction, bacteria inactivation, and alteration in intracellular metabolism. For single-species biofilms S. aureus is found more tolerant to plasma than P. aeruginosa, and mixed-species biofilms display higher tolerance of both bacteria to plasma than in single-species biofilms. A comparison between wet and dehydrated biofilms reveals reduced plasma efficacy in wet environments. Consequently, biofilm dehydration prior to the plasma treatment facilitates penetration of plasma reactive species leading to higher bacteria inactivation. The evaluation of plasma-generated gaseous reactive species reveals O3 and NO2 being dominant species contributing to the etching mechanism of the overall plasma anti-biofilm effect. Despite the strong anti-biofilm effect is obtained, the biofilm regrowth on the next day after plasma treatment implies on the inability of pulsed streamer corona discharge to permanently eradicate biofilms on a surface. The search for adequate plasma treatment conditions of biofilms remains crucial to avoid the appearance of more adaptive biofilms.
Collapse
Affiliation(s)
- Aleksandra Lavrikova
- Division of Environmental Physics, Faculty of Mathematics, Physics and Informatics, Comenius University, 842 48 Bratislava, Slovakia; Ecole Polytechnique Fédérale de Lausanne (EPFL), Swiss Plasma Center (SPC), CH-1015 Lausanne, Switzerland.
| | - Mário Janda
- Division of Environmental Physics, Faculty of Mathematics, Physics and Informatics, Comenius University, 842 48 Bratislava, Slovakia
| | - Helena Bujdáková
- Department of Microbiology and Virology, Faculty of Natural Sciences, Comenius University, 842 15 Bratislava, Slovakia
| | - Karol Hensel
- Division of Environmental Physics, Faculty of Mathematics, Physics and Informatics, Comenius University, 842 48 Bratislava, Slovakia
| |
Collapse
|
14
|
Fusar Poli S, Locatelli C, Monistero V, Freu G, Cremonesi P, Castiglioni B, Lecchi C, Longheu CM, Tola S, Guaraglia A, Allievi C, Villa L, Manfredi MT, Addis MF. Staphylococcus aureus and methicillin-resistant staphylococci and mammaliicocci in the bulk tank milk of dairy cows from a livestock-dense area in northern Italy. Res Vet Sci 2025; 182:105482. [PMID: 39612737 DOI: 10.1016/j.rvsc.2024.105482] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Revised: 11/18/2024] [Accepted: 11/24/2024] [Indexed: 12/01/2024]
Abstract
Staphylococcus aureus is the main etiologic agent of contagious dairy cow mastitis, while non-aureus staphylococci and mammaliicocci (NASM) are the bacteria most frequently isolated from milk. Beyond their impact on animal health, NASM can harbor antimicrobial resistance (AMR) genes with potential for bidirectional transfer with S. aureus, and methicillin-resistant (MR) staphylococci (MRS) can raise significant One Health concerns. In our study, we evaluated the prevalence and characteristics of MRS in the bulk tank milk (BTM) of 88 dairy farms in the livestock-dense province of Lodi, Lombardy, northern Italy. S. aureus was isolated from 32.95 % of BTM samples, with the Ribosomal Spacer PCR (RS-PCR) genotype B being the most prevalent, identified in 37.93 % of S. aureus positive farms. All isolates carried the ica genes (icaA, icaB, icaC, icaD) indicating the potential to produce biofilm. MRS were isolated in 56.81 % of farms. According to MALDI-TOF MS analysis, the most prevalent MR species included S. epidermidis (MRSE, 35.59 %) followed by S. aureus (MRSA, 18.64 %), M. sciuri (15.25 %), S. saprophyticus (11.86 %), S. borealis (6.78 %), S. haemolyticus (5.08 %), M. fleurettii, (3.39 %), S. cohnii, and S. pettenkoferi (1.70 % each). Most MR isolates carried the mecA gene, while none carried mecC. The staphylococcal cassette chromosome mec (SCCmec) was predominantly type V in MRSA (45.45 %) and type IV in MRSE (61.90 %). Given their relevance to One Health, monitoring AMR in all staphylococci and mammaliicocci isolated from milk is essential for understanding the prevalence, characteristics, and transmission dynamics of MR gene pools within dairy herds.
Collapse
Affiliation(s)
- Sara Fusar Poli
- Dipartimento di Medicina Veterinaria e Scienze Animali - DIVAS, Università degli Studi di Milano, 26900 Lodi, Italy; Laboratorio di Malattie Infettive degli Animali (MiLab), Università degli Studi di Milano, 26900 Lodi, Italy
| | - Clara Locatelli
- Dipartimento di Medicina Veterinaria e Scienze Animali - DIVAS, Università degli Studi di Milano, 26900 Lodi, Italy
| | - Valentina Monistero
- Dipartimento di Medicina Veterinaria e Scienze Animali - DIVAS, Università degli Studi di Milano, 26900 Lodi, Italy
| | - Gustavo Freu
- Department of Animal Nutrition and Production, School of Veterinary Medicine and Animal Science, University of São Paulo, Pirassununga 13635-900, São Paulo, Brazil
| | - Paola Cremonesi
- Consiglio Nazionale delle Ricerche, Istituto di Biologia e Biotecnologie Agrarie (CNR-IBBA), 20133 Milan, Italy
| | - Bianca Castiglioni
- Consiglio Nazionale delle Ricerche, Istituto di Biologia e Biotecnologie Agrarie (CNR-IBBA), 20133 Milan, Italy
| | - Cristina Lecchi
- Dipartimento di Medicina Veterinaria e Scienze Animali - DIVAS, Università degli Studi di Milano, 26900 Lodi, Italy
| | | | - Sebastiana Tola
- Istituto Zooprofilattico Sperimentale della Sardegna, 07100 Sassari, Italy
| | - Alessandro Guaraglia
- Dipartimento di Scienze Umanistiche e Sociali, Università degli Studi di Sassari, 07100 Sassari, Italy
| | - Carolina Allievi
- Dipartimento di Medicina Veterinaria e Scienze Animali - DIVAS, Università degli Studi di Milano, 26900 Lodi, Italy; Laboratorio di Malattie Parassitarie e Zoonosi (ParVetLab), Università degli Studi di Milano, 26900 Lodi, Italy
| | - Luca Villa
- Dipartimento di Medicina Veterinaria e Scienze Animali - DIVAS, Università degli Studi di Milano, 26900 Lodi, Italy; Laboratorio di Malattie Parassitarie e Zoonosi (ParVetLab), Università degli Studi di Milano, 26900 Lodi, Italy
| | - Maria Teresa Manfredi
- Dipartimento di Medicina Veterinaria e Scienze Animali - DIVAS, Università degli Studi di Milano, 26900 Lodi, Italy; Laboratorio di Malattie Parassitarie e Zoonosi (ParVetLab), Università degli Studi di Milano, 26900 Lodi, Italy
| | - Maria Filippa Addis
- Dipartimento di Medicina Veterinaria e Scienze Animali - DIVAS, Università degli Studi di Milano, 26900 Lodi, Italy; Laboratorio di Malattie Infettive degli Animali (MiLab), Università degli Studi di Milano, 26900 Lodi, Italy.
| |
Collapse
|
15
|
Lange A, Kutwin M, Zawadzka K, Ostrowska A, Strojny-Cieślak B, Nasiłowska B, Bombalska A, Jaworski S. Impaired Biofilm Development on Graphene Oxide-Metal Nanoparticle Composites. Nanotechnol Sci Appl 2024; 17:303-320. [PMID: 39734361 PMCID: PMC11681909 DOI: 10.2147/nsa.s485841] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2024] [Accepted: 12/10/2024] [Indexed: 12/31/2024] Open
Abstract
Purpose Biofilms are one of the main threats related to bacteria. Owing to their complex structure, in which bacteria are embedded in the extracellular matrix, they are extremely challenging to eradicate, especially since they can inhabit both biotic and abiotic surfaces. This study aimed to create an effective antibiofilm nanofilm based on graphene oxide-metal nanoparticles (GOM-NPs). Methods To create nanofilms, physicochemical analysis was performed, including zeta potential (Zp) (and the nanocomposites stability in time) and size distribution measurements, scanning transmission electron microscopy (STEM), energy dispersive X-ray analysis (EDX), and atomic force microscopy (AFM) of the nanofilm surfaces. During biological analysis, reactive oxygen species (ROS) and antioxidant capacity were measured in planktonic cells treated with the nanocomposites. Thereafter, biofilm formation was checked via crystal violet staining, biofilm thickness was assessed by confocal microscopy using double fluorescent staining, and biofilm structure was analyzed by scanning electron microscopy. Results The results showed that two of the three nanocomposites were effective in reducing biofilm formation (GOAg and GOZnO), although the nanofilms were characterized by the roughest surface, indicating that high surface roughness is unfavorable for biofilm formation by the tested bacterial species (Staphylococcus aureus (ATCC 25923), Salmonella enterica (ATCC 13076), Pseudomonas aeruginosa (ATCC 27853)). Conclusion The performed analysis indicated that graphene oxide may be a platform for metal nanoparticles that enhances their properties (eg colloidal stability, which is maintained over time). Nanocomposites based on graphene oxide with silver nanoparticles and other types of nanocomposites with zinc oxide were effective against biofilms, contributing to changes throughout the biofilm structure, causing a significant reduction in the thickness of the structure, and affecting cell distribution. A nanocomposite consisting of graphene oxide with copper nanoparticles inhibited the biofilm, but to a lesser extent.
Collapse
Affiliation(s)
- Agata Lange
- Department of Nanobiotechnology, Institute of Biology, Warsaw University of Life Sciences, Warsaw, Poland
| | - Marta Kutwin
- Department of Nanobiotechnology, Institute of Biology, Warsaw University of Life Sciences, Warsaw, Poland
| | - Katarzyna Zawadzka
- Department of Nanobiotechnology, Institute of Biology, Warsaw University of Life Sciences, Warsaw, Poland
| | - Agnieszka Ostrowska
- Department of Nanobiotechnology, Institute of Biology, Warsaw University of Life Sciences, Warsaw, Poland
| | - Barbara Strojny-Cieślak
- Department of Nanobiotechnology, Institute of Biology, Warsaw University of Life Sciences, Warsaw, Poland
| | - Barbara Nasiłowska
- Center for Biomedical Engineering, Institute of Optoelectronics, Military University of Technology, Warsaw, Poland
| | - Aneta Bombalska
- Department of Optoelectronic Technologies, Institute of Optoelectronics, Military University of Technology, Warsaw, Poland
| | - Sławomir Jaworski
- Department of Nanobiotechnology, Institute of Biology, Warsaw University of Life Sciences, Warsaw, Poland
| |
Collapse
|
16
|
Tartor YH, Enany ME, Ismail NI, El-Demerdash AS, Eidaroos NH, Algendy RM, Mahmmod Y, Elsohaby I. Vancomycin-resistant Staphylococcus aureus endangers Egyptian dairy herds. Sci Rep 2024; 14:30606. [PMID: 39715776 DOI: 10.1038/s41598-024-81516-6] [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: 08/20/2024] [Accepted: 11/27/2024] [Indexed: 12/25/2024] Open
Abstract
The emergence of pandrug-resistant (PDR) and extensive drug-resistant (XDR) methicillin-resistant and vancomycin-resistant Staphylococcus aureus (MRSA and VRSA) isolates from bovine milk samples along with biofilm formation ability and harboring various virulence genes complicates the treatment of bovine mastitis and highlights the serious threat to public health. This study investigated for the first time the frequency, antimicrobial resistance profiles, biofilm-forming ability, virulence factors, spa and staphylococcal cassette chromosome mec (SCCmec) types of MRSA and VRSA isolated from clinical and subclinical bovine mastitis in Egypt. A total of 808 milk samples were collected from each quarter of 202 dairy animals, including 31 buffaloes and 171 cattle. The frequency of mastitis in the collected milk samples was 48.4% (60/124) in buffaloes and 29.2% (200/684) in cattle. A total of 65 Staphylococcus species isolates were recovered, including 27 coagulase-positive S. aureus (CoPS) isolates and 38 coagulase-negative staphylococci (CoNS). The CoNS included 27 mammaliicocci (20 Mammaliicoccus lentus and 7 M. sciuri) and 11 Non-aureus staphylococci (S. lugdunensis) isolates. All the CoPS isolates were mecA positive and resistant to 20-33 tested antimicrobials with multiple antibiotic resistance index ranging from 0.61 to 1. Three isolates were PDR, four were XDR, and 20 were multidrug resistant isolates. VRSA was detected in 85.2% of CoPS isolates with minimal inhibitory concentration (MIC) ranging from 64 to 1024 µg/mL. The vanA gene was found in 60.8%, vanB in 73.9%, and both genes in 43.5% of VRSA isolates. All the CoPS isolates exhibited biofilm formation ability, with 55.6% being strong, and 44.4% moderate biofilm producers, and harbored icaA (74.1%) and icaD (74.1%) biofilm-forming genes. All S. aureus isolates harbored both beta-haemolysin (hlb) and leucotoxin (lukMF) genes, while 44.4% were positive for toxic shock syndrome toxin (tsst) gene. Enterotoxin genes sea, seb, sec, sed, and see were found in 59.3%, 40.7%, 18.5%, 33.3%, and 14.8% of isolates, respectively. Additionally, 70.4% of the isolates had spa X-region gene, and exhibited eight different MRSA spa types (t127, t267, t037, t011, t843, t1081, t2663, and t1575), with spa t127 being the most common. Three SCCmec types (I, II and III) were identified, with SCCmec I being predominant, and were further classified into subtypes 1.1.1, 1.1.2, 1.n.1, and 4.1.1. The ability of MRSA and VRSA isolates to produce biofilms and resist antimicrobials highlights the serious threat these pathogens pose to bovine milk safety, animal welfare, and public health. Therefore, strict hygiene practices and antimicrobial surveillance are crucial to reduce the risk of MRSA and VRSA colonization and dissemination.
Collapse
Affiliation(s)
- Yasmine H Tartor
- Department of Microbiology, Faculty of Veterinary Medicine, Zagazig University, Zagazig, 44511, Egypt.
| | - Mohamed E Enany
- Department of Bacteriology, Immunology, and Mycology, Faculty of Veterinary Medicine, Suez Canal University, Ismailia, 41522, Egypt
| | | | - Azza S El-Demerdash
- Agriculture Research Center (ARC), Animal Health Research Institute (AHRI), Zagazig, 44516, Egypt
| | - Nada H Eidaroos
- Department of Bacteriology, Immunology, and Mycology, Faculty of Veterinary Medicine, Suez Canal University, Ismailia, 41522, Egypt
| | - Reem M Algendy
- Department of Food Hygiene, Safety and Technology, Faculty of Veterinary Medicine, Zagazig University, Zagazig, 44511, Egypt
| | - Yasser Mahmmod
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine, Long Island University, 720 Northern Boulevard, Brookville, NY, 11548, USA
| | - Ibrahim Elsohaby
- Department of Infectious Diseases and Public Health, Jockey Club College of Veterinary Medicine and Life Sciences, City University of Hong Kong, Hong Kong, SAR, China
- Centre for Applied One Health Research and Policy Advice (OHRP), City University of Hong Kong, Hong Kong, Hong Kong SAR, China
- Department of Animal Medicine, Faculty of Veterinary Medicine, Zagazig University, Zagazig, 44511, Egypt
| |
Collapse
|
17
|
Omwenga EO, Awuor SO. The Bacterial Biofilms: Formation, Impacts, and Possible Management Targets in the Healthcare System. THE CANADIAN JOURNAL OF INFECTIOUS DISEASES & MEDICAL MICROBIOLOGY = JOURNAL CANADIEN DES MALADIES INFECTIEUSES ET DE LA MICROBIOLOGIE MEDICALE 2024; 2024:1542576. [PMID: 39717533 PMCID: PMC11666319 DOI: 10.1155/cjid/1542576] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Revised: 11/04/2024] [Accepted: 11/18/2024] [Indexed: 12/25/2024]
Abstract
Introduction: The persistent increase in multidrug-resistant pathogens has catalyzed the creation of novel strategies to address antivirulence and anti-infective elements. Such methodologies aim to diminish the selective pressure exerted on bacterial populations, decreasing the likelihood of resistance emergence. This review explores the role of biofilm formation as a significant virulence factor and its impact on the development of antimicrobial resistance (AMR). Case Presentation: The ability of bacteria to form a superstructure-biofilm-has made resistance cases in the microbial world a big concern to public health and other sectors as it is a crucial virulence factor that causes difficulties in the management of infections, hence enhancing chronic infection occurrence. Biofilm formation dates to about 3.4 billion years when prokaryotes were discovered to be forming them and since then due to evolution and growth in science, they are more understood. Management and Outcome: The unique microenvironments within bacterial biofilms diminish antibiotic effectiveness and help bacteria evade the host immune system. Biofilm production is a widespread capability among diverse bacterial species. Biofilm formation is enhanced by quorum sensing (QS), reduction of nutrients, or harsh environments for the bacteria. Conclusion: The rise of severe, treatment-resistant biofilm infections poses major challenges in medicine and agriculture, yet much about how these biofilms form remains unknown.
Collapse
Affiliation(s)
- Eric Omori Omwenga
- Department of Medical Microbiology & Parasitology, School of Health Sciences, Kisii University, Kisii, Kenya
| | - Silas Onyango Awuor
- Department of Applied Health Sciences, School of Health Sciences, Kisii University, Kisii, Kenya
- Department of Medical Microbiology, Jaramogi Oginga Odinga Teaching and Referral Hospital, Kisumu, Kenya
| |
Collapse
|
18
|
Deng J, Liu M, Gao S, Lei D, Su Z, Liang F, Tang S, Yang H, Huang YY, Xie W, Pan GY. Microneedles Constructed by Swellable Hydrogels Loaded with Celastrol for Efficient Treatment of Skin Infections Induced by Drug-Resistant Bacterial Strains. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2024; 40:26125-26136. [PMID: 39588917 DOI: 10.1021/acs.langmuir.4c03593] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/27/2024]
Abstract
The urgent need for new antimicrobial drugs arises from the limited efficacy of traditional antibiotics against emerging drug-resistant strains. Celastrol (CSL) demonstrates an exceptional antibacterial property that remains unaffected by bacterial resistance, but its poor water solubility limits its wide applications. This study uses the hydrophobic inner cavity of mono-(6-diethylenetriamine-6-deoxy)-β-cyclodextrin (mβ-CD) (a derivative of cyclodextrin) to encapsulate CSL, constructing an inclusion complex (CSL@mβ-CD) to enhance the water solubility of CSL. The obtained inclusion complex is further incorporated into a swellable hydrogel microneedle (MN) to obtain CSL@mβ-CD/MN. The fabricated CSL@mβ-CD/MN can enable the sustained release of CSL, achieving effective bacterial eradication at infected sites. In vivo experiments demonstrate that CSL@mβ-CD/MN has a remarkable efficacy in the treatment of methicillin-resistant Staphylococcus aureus-induced subcutaneous abscesses and wound infections. Specifically, CSL@mβ-CD/MN can effectively penetrate the stratum corneum of the skin to realize rapid elimination of the bacteria in wounds. Moreover, CSL@mβ-CD/MN can efficiently scavenge reactive oxygen species, promote M2 polarization of macrophages, and relieve local inflammation at the wound sites. These results reveal that CSL@mβ-CD/MN holds great promise in the clinical treatment of acute skin infections induced by drug-resistant bacteria.
Collapse
Affiliation(s)
- Jianbin Deng
- School of Pharmacy, Guilin Medical University, Guilin 541100, PR China
- Guangxi Key Laboratory of Drug Discovery and Optimization, Guilin 541100, PR China
| | - Mengqi Liu
- School of Pharmacy, Guilin Medical University, Guilin 541100, PR China
- Guangxi Key Laboratory of Drug Discovery and Optimization, Guilin 541100, PR China
| | - Shiqi Gao
- School of Pharmacy, Guilin Medical University, Guilin 541100, PR China
| | - Dongjie Lei
- School of Pharmacy, Guilin Medical University, Guilin 541100, PR China
- Guangxi Key Laboratory of Drug Discovery and Optimization, Guilin 541100, PR China
| | - Zhicheng Su
- School of Pharmacy, Guilin Medical University, Guilin 541100, PR China
| | - Fuqing Liang
- School of Pharmacy, Guilin Medical University, Guilin 541100, PR China
- Guangxi Key Laboratory of Drug Discovery and Optimization, Guilin 541100, PR China
| | - Songyun Tang
- School of Pharmacy, Guilin Medical University, Guilin 541100, PR China
- Guangxi Key Laboratory of Drug Discovery and Optimization, Guilin 541100, PR China
| | - Huiyuan Yang
- School of Intelligent Medicine and Biotechnology, Guilin Medical University, Guilin 541100, PR China
| | - Yuan-Yuan Huang
- School of Intelligent Medicine and Biotechnology, Guilin Medical University, Guilin 541100, PR China
| | - Weiquan Xie
- School of Pharmacy, Guilin Medical University, Guilin 541100, PR China
- Guangxi Key Laboratory of Drug Discovery and Optimization, Guilin 541100, PR China
| | - Guang-Yu Pan
- School of Pharmacy, Guilin Medical University, Guilin 541100, PR China
- Key Laboratory of Biochemistry and Molecular Biology, Guilin Medical University, Education Department of Guangxi Zhuang Autonomous Region, Guilin 541100, PR China
- School of Intelligent Medicine and Biotechnology, Guilin Medical University, Guilin 541100, PR China
| |
Collapse
|
19
|
Zhao H, Zhang L, Du D, Mai L, Liu Y, Morigen M, Fan L. The RIG-I-like receptor signaling pathway triggered by Staphylococcus aureus promotes breast cancer metastasis. Int Immunopharmacol 2024; 142:113195. [PMID: 39303544 DOI: 10.1016/j.intimp.2024.113195] [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: 06/30/2024] [Revised: 09/10/2024] [Accepted: 09/14/2024] [Indexed: 09/22/2024]
Abstract
Host microbes are increasingly recognized as key components in various types of cancer, although their exact impact remains unclear. This study investigated the functional significance of Staphylococcus aureus (S. aureus) in breast cancer tumorigenesis and progression. We found that S. aureus invasion resulted in a compromised DNA damage response process, as evidenced by the absence of G1-phase arrest and apoptosis in breast cells in the background of double strand breaks production and the activation of the ataxia-telangiectasia mutated (ATM)-p53 signaling pathway. The high-throughput mRNA sequencing, bioinformatics analysis and pharmacological studies revealed that S. aureus facilitates breast cell metastasis through the innate immune pathway, particularly in cancer cells. During metastasis, S. aureus initially induced the expression of RIG-I-like receptors (RIG-I in normal breast cells, RIG-I and MDA5 in breast cancer cells), which in turn activated NF-κB p65 expression. We further showed that NF-κB p65 activated the CCL5-CCR5 pathway, contributing to breast cell metastasis. Our study provides novel evidence that the innate immune system, triggered by bacterial infection, plays a role in bacterial-driven cancer metastasis.
Collapse
Affiliation(s)
- Haile Zhao
- Inner Mongolia Key Laboratory for Molecular Regulation of the Cell, State Key Laboratory of Reproductive Regulation & Breeding of Grassland livestock, School of Life Sciences, Inner Mongolia University, Hohhot, Inner Mongolia, PR China
| | - Linzhe Zhang
- Inner Mongolia Key Laboratory for Molecular Regulation of the Cell, State Key Laboratory of Reproductive Regulation & Breeding of Grassland livestock, School of Life Sciences, Inner Mongolia University, Hohhot, Inner Mongolia, PR China
| | - Dongdong Du
- Inner Mongolia Key Laboratory for Molecular Regulation of the Cell, State Key Laboratory of Reproductive Regulation & Breeding of Grassland livestock, School of Life Sciences, Inner Mongolia University, Hohhot, Inner Mongolia, PR China
| | - Lisu Mai
- Inner Mongolia Key Laboratory for Molecular Regulation of the Cell, State Key Laboratory of Reproductive Regulation & Breeding of Grassland livestock, School of Life Sciences, Inner Mongolia University, Hohhot, Inner Mongolia, PR China
| | - Yaping Liu
- Department of Gynecology and Obstetrics, The Affiliated Hospital of Inner Mongolia Medical University, Hohhot, Inner Mongolia, PR China
| | - Morigen Morigen
- Inner Mongolia Key Laboratory for Molecular Regulation of the Cell, State Key Laboratory of Reproductive Regulation & Breeding of Grassland livestock, School of Life Sciences, Inner Mongolia University, Hohhot, Inner Mongolia, PR China.
| | - Lifei Fan
- Inner Mongolia Key Laboratory for Molecular Regulation of the Cell, State Key Laboratory of Reproductive Regulation & Breeding of Grassland livestock, School of Life Sciences, Inner Mongolia University, Hohhot, Inner Mongolia, PR China.
| |
Collapse
|
20
|
Turner AB, Zermeño-Pérez D, Mysior MM, Giraldo-Osorno PM, García B, O'Gorman E, Oubihi S, Simpson JC, Lasa I, Ó Cróinín T, Trobos M. Biofilm morphology and antibiotic susceptibility of methicillin-resistant Staphylococcus aureus (MRSA) on poly-D,L-lactide- co-poly(ethylene glycol) (PDLLA-PEG) coated titanium. Biofilm 2024; 8:100228. [PMID: 39830519 PMCID: PMC11740804 DOI: 10.1016/j.bioflm.2024.100228] [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: 07/09/2024] [Revised: 09/27/2024] [Accepted: 10/03/2024] [Indexed: 01/22/2025] Open
Abstract
Biodegradable polymeric coatings are being explored as a preventive strategy for orthopaedic device-related infection. In this study, titanium surfaces (Ti) were coated with poly-D,L-lactide (PDLLA, (P)), polyethylene-glycol poly-D,L-lactide (PEGylated-PDLLA, (PP20)), or multi-layered PEGylated-PDLLA (M), with or without 1 % silver sulfadiazine. The aim was to evaluate their cytocompatibility, resistance to Staphylococcus aureus biofilm formation, and their potential to enhance the susceptibility of any biofilm formed to antibiotics. Using automated high-content screening confocal microscopy, biofilm formation of a clinical methicillin-resistant Staphylococcus aureus (MRSA) isolate expressing GFP was quantified, along with isogenic mutants that were unable to form polysaccharidic or proteinaceous biofilm matrices. The results showed that PEGylated-PDLLA coatings exhibited significant antibiofilm properties, with M showing the highest effect. This inhibitory effect was stronger in S. aureus biofilms with a matrix composed of proteins compared to those with an exopolysaccharide (PIA) biofilm matrix. Our data suggest that the antibiofilm effect may have been due to (i) inhibition of the initial attachment through microbial surface components recognising adhesive matrix molecules (MSCRAMMs), since PEG reduces protein surface adsorption via surface hydration layer and steric repulsion; and (ii) mechanical disaggregation and dispersal of microcolonies due to the bioresorbable/degradable nature of the polymers, which undergo hydration and hydrolysis over time. The disruption of biofilm morphology by the PDLLA-PEG co-polymers increased S. aureus susceptibility to antibiotics like rifampicin and fusidic acid. Adding 1 % AgSD provided additional early bactericidal effects on both biofilm and planktonic S. aureus. Additionally, the coatings were cytocompatible with immune cells, indicating their potential to enhance bacterial clearance and reduce bacterial colonisation of titanium-based orthopaedic biomaterials.
Collapse
Affiliation(s)
- Adam Benedict Turner
- Department of Biomaterials, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Centre for Antibiotic Resistance Research in Gothenburg (CARe), Gothenburg, Sweden
| | - David Zermeño-Pérez
- Ashland Specialties Ireland Ltd., Mullingar, Ireland
- School of Biomolecular and Biomedical Science, University College Dublin, Dublin, Ireland
| | - Margaritha M. Mysior
- Cell Screening Laboratory, UCD School of Biology & Environmental Science, University College Dublin, Dublin, Ireland
| | - Paula Milena Giraldo-Osorno
- Department of Biomaterials, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Centre for Antibiotic Resistance Research in Gothenburg (CARe), Gothenburg, Sweden
| | - Begoña García
- Microbial Pathogenesis Laboratory. Navarrabiomed-Complejo Hospitalario de Navarra (CHN)-Universidad Pública de Navarra (UPNA), IDISNA, Pamplona, Navarra, Spain
| | - Elizabeth O'Gorman
- School of Biomolecular and Biomedical Science, University College Dublin, Dublin, Ireland
| | - Shafik Oubihi
- School of Biomolecular and Biomedical Science, University College Dublin, Dublin, Ireland
| | - Jeremy C. Simpson
- Cell Screening Laboratory, UCD School of Biology & Environmental Science, University College Dublin, Dublin, Ireland
| | - Iñigo Lasa
- Microbial Pathogenesis Laboratory. Navarrabiomed-Complejo Hospitalario de Navarra (CHN)-Universidad Pública de Navarra (UPNA), IDISNA, Pamplona, Navarra, Spain
| | - Tadhg Ó Cróinín
- School of Biomolecular and Biomedical Science, University College Dublin, Dublin, Ireland
| | - Margarita Trobos
- Department of Biomaterials, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Centre for Antibiotic Resistance Research in Gothenburg (CARe), Gothenburg, Sweden
| |
Collapse
|
21
|
Cuellar-Gaviria TZ, Rincon-Benavides MA, Halipci Topsakal HN, Salazar-Puerta AI, Jaramillo-Garrido S, Kordowski M, Vasquez-Martinez CA, Nguyen KT, Rima XY, Rana PSJB, Combita-Heredia O, Deng B, Dathathreya K, McComb DW, Reategui E, Wozniak D, Higuita-Castro N, Gallego-Perez D. Tissue nano-transfection of antimicrobial genes drives bacterial biofilm killing in wounds and is potentially mediated by extracellular vesicles. J Control Release 2024; 376:1300-1315. [PMID: 39491627 PMCID: PMC11780627 DOI: 10.1016/j.jconrel.2024.10.071] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2024] [Revised: 10/06/2024] [Accepted: 10/30/2024] [Indexed: 11/05/2024]
Abstract
The emergence of bacteria that are resistant to antibiotics is on track to become a major global health crisis. Therefore, there is an urgent need for new treatment options. Here, we studied the implementation of tissue-nanotransfection (TNT) to treat Staphylococcus aureus-infected wounds by delivering gene cargos that boost the levels of naturally produced antimicrobial peptides. The Cathelicidin Antimicrobial Peptide gene (CAMP), which produces the antimicrobial peptide LL-37, was used as model gene cargo. In vitro evaluation showed successful transfection and an increase in the transcription and translation of CAMP-coding plasmid in mouse primary epithelial cells. Moreover, we found that the extracellular vesicles (EVs) derived from the transfected cells (in vitro and in vivo) carried significantly higher concentrations of CAMP transcripts and LL-37 peptide compared to control EVs, possibly mediating the trafficking of the antimicrobial contents to other neighboring cells. The TNT platform was then used in vivo on an excisional wound model in mice to nanotransfect the CAMP-coding plasmid on the edge of infected wounds. After 4 days of daily treatment, we observed a significant decrease in the bacterial load in the CAMP-treated group compared to the sham group. Moreover, histological analysis and bacterial load quantification also revealed that TNT of CAMP on S. aureus-infected wounds was effective in treating biofilm progression by reducing the bacterial load. Lastly, we observed a significant increase in macrophage recruitment to the infected tissue, a robust increase in vascularization, as well as and an increased expression of IL10 and Fli1. Our results demonstrate that TNT-based delivery of gene cargos coding for antimicrobial compounds to the wound is a promising approach for combating biofilm infections in wounds.
Collapse
Affiliation(s)
- Tatiana Z Cuellar-Gaviria
- Department of Biomedical Engineering, The Ohio State University, Columbus, OH 43210, USA; Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH 43210, USA; Gene Therapy Institute, The Ohio State University, Columbus, OH 43210, USA; Infectious Disease Institute, The Ohio State University, Columbus, OH 43210, USA
| | - Maria Angelica Rincon-Benavides
- Department of Biomedical Engineering, The Ohio State University, Columbus, OH 43210, USA; Biophysics Program, The Ohio State University, Columbus, OH 43210, USA
| | - Hatice Nur Halipci Topsakal
- Department of Biomedical Engineering, The Ohio State University, Columbus, OH 43210, USA; Istanbul Atlas University, Istanbul 34408, Turkiye
| | | | | | - Mia Kordowski
- Department of Biomedical Engineering, The Ohio State University, Columbus, OH 43210, USA; Biophysics Program, The Ohio State University, Columbus, OH 43210, USA
| | - Carlos A Vasquez-Martinez
- Department of Biomedical Engineering, The Ohio State University, Columbus, OH 43210, USA; CONACYT - Faculty of Medicine, Benito Juárez Autonomous University of Oaxaca, Oaxaca 68020, Mexico
| | - Kim Truc Nguyen
- William G. Lowrie Department of Chemical and Biomolecular Engineering, The Ohio State University, Columbus, OH 43210, USA
| | - Xilal Y Rima
- William G. Lowrie Department of Chemical and Biomolecular Engineering, The Ohio State University, Columbus, OH 43210, USA
| | - Pranav S J B Rana
- Department of Microbiology, The Ohio State University, Columbus, OH 43210, USA; Department of Microbial Infection and Immunity, The Ohio State University, Columbus, OH 43210, USA
| | | | - Binbin Deng
- Center for Electron Microscopy and Analysis (CEMAS), The Ohio State University, Columbus, OH 43210, USA
| | - Kavya Dathathreya
- Department of Biomedical Engineering, The Ohio State University, Columbus, OH 43210, USA
| | - David W McComb
- Center for Electron Microscopy and Analysis (CEMAS), The Ohio State University, Columbus, OH 43210, USA; Department of Materials Science and Engineering, The Ohio State University, Columbus, OH 43210, USA
| | - Eduardo Reategui
- William G. Lowrie Department of Chemical and Biomolecular Engineering, The Ohio State University, Columbus, OH 43210, USA; Comprehensive Cancer Center, The Ohio State University, Columbus, OH 43210, USA
| | - Daniel Wozniak
- Infectious Disease Institute, The Ohio State University, Columbus, OH 43210, USA; Department of Microbiology, The Ohio State University, Columbus, OH 43210, USA; Department of Microbial Infection and Immunity, The Ohio State University, Columbus, OH 43210, USA
| | - Natalia Higuita-Castro
- Department of Biomedical Engineering, The Ohio State University, Columbus, OH 43210, USA; Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH 43210, USA; Gene Therapy Institute, The Ohio State University, Columbus, OH 43210, USA; Infectious Disease Institute, The Ohio State University, Columbus, OH 43210, USA; Biophysics Program, The Ohio State University, Columbus, OH 43210, USA; Department of Neurological Surgery, The Ohio State University, Columbus, OH 43210, USA
| | - Daniel Gallego-Perez
- Department of Biomedical Engineering, The Ohio State University, Columbus, OH 43210, USA; Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH 43210, USA; Gene Therapy Institute, The Ohio State University, Columbus, OH 43210, USA; Infectious Disease Institute, The Ohio State University, Columbus, OH 43210, USA; Biophysics Program, The Ohio State University, Columbus, OH 43210, USA; Department of Surgery, The Ohio State University, Columbus, OH 43210, USA.
| |
Collapse
|
22
|
Ni L, Shen R, Luo H, Li X, Zhang X, Huang L, Deng Y, Liao X, Wu Y, Duan C, Xie X. GlmS plays a key role in the virulence factor expression and biofilm formation ability of Staphylococcus aureus promoted by advanced glycation end products. Virulence 2024; 15:2352476. [PMID: 38741276 PMCID: PMC11095574 DOI: 10.1080/21505594.2024.2352476] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Accepted: 05/03/2024] [Indexed: 05/16/2024] Open
Abstract
Staphylococcus aureus (S. aureus) is well known for its biofilm formation ability and is responsible for serious, chronic refractory infections worldwide. We previously demonstrated that advanced glycation end products (AGEs), a hallmark of chronic hyperglycaemia in diabetic tissues, enhanced biofilm formation by promoting eDNA release via sigB upregulation in S. aureus, contributing to the high morbidity and mortality of patients presenting a diabetic foot ulcer infection. However, the exact regulatory network has not been completely described. Here, we used pull-down assay and LC-MS/MS to identify the GlmS as a candidate regulator of sigB in S. aureus stimulated by AGEs. Dual-luciferase assays and electrophoretic mobility shift assays (EMSAs) revealed that GlmS directly upregulated the transcriptional activity of sigB. We constructed NCTC 8325 ∆glmS for further validation. qRT-PCR analysis revealed that AGEs promoted both glmS and sigB expression in the NCTC 8325 strain but had no effect on NCTC 8325 ∆glmS. NCTC 8325 ∆glmS showed a significant attenuation in biofilm formation and virulence factor expression, accompanied by a decrease in sigB expression, even under AGE stimulation. All of the changes, including pigment deficiency, decreased haemolysis ability, downregulation of hla and hld expression, and less and sparser biofilms, indicated that sigB and biofilm formation ability no longer responded to AGEs in NCTC 8325 ∆glmS. Our data extend the understanding of GlmS in the global regulatory network of S. aureus and demonstrate a new mechanism by which AGEs can upregulate GlmS, which directly regulates sigB and plays a significant role in mediating biofilm formation and virulence factor expression.
Collapse
Affiliation(s)
- Lijia Ni
- Department of Clinical Laboratory, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
- Institution of Antibiotic, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Rui Shen
- Department of Clinical Laboratory, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
- Institution of Antibiotic, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Hua Luo
- Department of Clinical Laboratory, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
- Institution of Antibiotic, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Xuexue Li
- Department of Clinical Laboratory, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Xiaofan Zhang
- Department of Clinical Laboratory, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Lisi Huang
- Department of Clinical Laboratory, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
- Institution of Antibiotic, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Yawen Deng
- Department of Clinical Laboratory, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
- Institution of Antibiotic, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Xiaoyan Liao
- Department of Clinical Laboratory, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
- Institution of Antibiotic, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Yonglin Wu
- Department of Clinical Laboratory, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Chaohui Duan
- Department of Clinical Laboratory, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Xiaoying Xie
- Department of Clinical Laboratory, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
- Institution of Antibiotic, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
23
|
Dewan D, Basu A, Dolai D, Pal S. Biological and Biophysical Methods for Evaluation of Inhibitors of Sortase A in Staphylococcus aureus: An Overview. Cell Biochem Funct 2024; 42:e70002. [PMID: 39470102 DOI: 10.1002/cbf.70002] [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: 06/15/2024] [Revised: 09/01/2024] [Accepted: 10/02/2024] [Indexed: 10/30/2024]
Abstract
Staphylococcus aureus, one of the most notorious pathogens, develops antibiotic resistance by the formation of a thick layer of exopolysaccharides known as biofilms. Sortase A, a transpeptidase responsible for biofilm formation and attachment to the host surface, has emerged as an important drug target for development of anti-virulence agents. A number of sortase A inhibitors, both peptide and non-peptides are reported which involved the use of several experiments which may provide insights regarding binding affinity, specificity, safety, and efficacy of ligands. In this review, we focus on the principles, pros and cons, and the type of information obtained from biophysical (FRET assay, Microscale Thermophoresis, Surface Plasmon Resonance, CD spectroscopy etc.) and biological (cell viability assay, biofilm formation assay, CLSM, western blot analysis, in vivo characterization on mice etc.) methods for estimation of probable sortase A inhibitors, which might be helpful to the researchers who might be interested to delve into the development of sortase A inhibitors as a drug, to address the burning question of antimicrobial resistance (AMR).
Collapse
|
24
|
Nandanwar N, Gu G, Gibson JE, Neely MN. Polymicrobial interactions influence Mycobacterium abscessus co-existence and biofilm forming capabilities. Front Microbiol 2024; 15:1484510. [PMID: 39654682 PMCID: PMC11627178 DOI: 10.3389/fmicb.2024.1484510] [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: 08/22/2024] [Accepted: 11/12/2024] [Indexed: 12/12/2024] Open
Abstract
The lungs of patients with cystic fibrosis (CF) are vulnerable to persistent polymicrobial colonization by bacterial pathogens including Pseudomonas aeruginosa, Staphylococcus aureus, and the non-tuberculous mycobacterium (NTM) Mycobacterium abscessus. The polymicrobial milieu within the CF lung impacts individual species fitness, influences biofilm-forming capabilities, pathogenicity, production of virulence factors and even antimicrobial responses, all potentially compromising therapeutic success. Interaction studies among these CF pathogens are very limited, especially studies on the influences of P. aeruginosa and S. aureus on M. abscessus co-existence and virulence. Based on the little known thus far about coinfection of these pathogens, we hypothesize that the co-existence of P. aeruginosa and S. aureus alters M. abscessus virulence and phenotypic characteristics. We evaluated the direct (co-culture) and indirect (using supernatant) effects of P. aeruginosa and S. aureus on M. abscessus growth rate, biofilm formation, macrophage internalization and glycopeptidolipids (GPL) expression. Our observations indicate that P. aeruginosa and S. aureus exert a competitive behavior toward M. abscessus during direct contact or indirect interaction in-vitro, probably as is the case of polymicrobial infections in the lungs of patients with CF. This is the first report that demonstrates S. aureus inhibitory effects on M. abscessus growth and biofilm forming capabilities. Collectively, co-culture studies enhance our understanding of polymicrobial interactions during coinfection and can guide to establish better management of coinfections and treatment strategies for M. abscessus.
Collapse
Affiliation(s)
- Nishant Nandanwar
- Division of Infectious Diseases, Department of Pediatrics, Children’s Hospital Los Angeles, Los Angeles, CA, United States
| | - Geoffery Gu
- Division of Infectious Diseases, Department of Pediatrics, Children’s Hospital Los Angeles, Los Angeles, CA, United States
- Department of Biological Sciences, University of Southern California, Los Angeles, CA, United States
| | - Joy E. Gibson
- Division of Infectious Diseases, Department of Pediatrics, Children’s Hospital Los Angeles, Los Angeles, CA, United States
- Department of Pediatrics, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Michael N. Neely
- Division of Infectious Diseases, Department of Pediatrics, Children’s Hospital Los Angeles, Los Angeles, CA, United States
- Department of Pediatrics, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| |
Collapse
|
25
|
Kilani AM, Alabi ED, Adeleke OE. Coexistence of the blaZ gene and selected virulence determinants in multidrug-resistant Staphylococcus aureus: insights from three Nigerian tertiary hospitals. BMC Infect Dis 2024; 24:1269. [PMID: 39528974 PMCID: PMC11552187 DOI: 10.1186/s12879-024-10171-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Accepted: 10/31/2024] [Indexed: 11/16/2024] Open
Abstract
BACKGROUND AND PURPOSE Infections caused by β-lactamase-producing strains of Staphylococcus aureus have become increasingly difficult to treat due to the expression of multiple virulence factors. This has heightened concerns about managing S. aureus-related infections. This study was conducted to characterize the blaZ gene and selected virulence determinants in β-lactam resistant S. aureus from human sources in three Nigerian tertiary hospitals. MATERIALS AND METHODS Three hundred and sixty samples were collected for the study. S. aureus was isolated and characterized following standard microbiological protocols and nuc gene amplification. Antibiotic susceptibility and minimum inhibitory concentration tests were performed using the disk diffusion method and E-tests, respectively. Biofilm formation and β-lactamase production were assessed using Congo red agar and nitrocefin kits, while the blaZ gene was examined using conventional PCR. Capsular polysaccharide genotyping, accessory gene regulator (agr) detection, Panton-valentine leucocidin (PVL), and PVL proteins were performed using PCR and Western blotting. RESULTS S. aureus was recovered from 145 samples, 50 (34.5%) of these isolates exhibited multidrug resistance, with MICs ranging from 0.125 to 1.00 µg/mL, and showed significant resistance to aminoglycosides, fluoroquinolones, and β-lactams. Of these, 31 strains produced β-lactamases, 30 of which carried the blaZ gene in combination with cap8 (80%) or cap5 (20%). Biofilm formation and PVL gene were observed in 85% of the 20 randomly selected blaZ-positive multidrug-resistant (MDR) strains. The agr2 allele was predominant, found in 70% of the selected MDR strains. No significant difference in the occurrence of the blaZ gene was found among the three clinical sources (p ≤ α0.05). CONCLUSION The co-occurrence of the blaZ gene with PVL, capsular polysaccharide genes, and agr alleles is associated with biofilm formation, indicating a high risk of β-lactam-resistant S. aureus infections. Our findings highlight the need for continuous molecular surveillance to enhance infection management, treatment options, and patient outcomes in the study locality. A limitation of this study is the random selection of MDR isolates, which may affect the comprehensiveness of the analyses.
Collapse
Affiliation(s)
- Adetunji Misbau Kilani
- Department of Microbiology, Federal University Dutsin-Ma, Dutsin-Ma, Katsina State, Nigeria
| | - Emmanuel Dayo Alabi
- Department of Microbiology, Federal University Dutsin-Ma, Dutsin-Ma, Katsina State, Nigeria.
| | | |
Collapse
|
26
|
Xiong Y, Wang R, Zheng J, Fang D, He P, Liu S, Lin Z, Chen X, Chen C, Shang Y, Yu Z, Liu X, Han S. Discovery of novel dihydropyrrolidone-thiadiazole compound crosstalk between the YycG/F two-component regulatory pathway and cell membrane homeostasis to combat methicillin-resistant Staphylococcus aureus. Eur J Med Chem 2024; 277:116770. [PMID: 39208742 DOI: 10.1016/j.ejmech.2024.116770] [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: 06/04/2024] [Revised: 07/30/2024] [Accepted: 08/11/2024] [Indexed: 09/04/2024]
Abstract
The rapid emergence and spread of multidrug-resistant (MDR) Gram-positive pathogens present a significant challenge to global healthcare. Methicillin-resistant Staphylococcus aureus (MRSA) is a particular concern because of its high resistance to most antibiotics. Based on our previously reported chemical structure of compound 62, a series of novel derivatives were synthesized and evaluated for their antibacterial activities. We found that some of these derivatives displayed effective antibacterial activity against Gram-positive pathogens, with minimal cytotoxicity (CC50>100 μM) and hemolytic activity (HC50>200 μM). Among these derivatives, the minimum inhibitory concentration (MIC) of 62-7c against Gram-positive bacterial isolates ranged from 6.25 to 25 μM. This derivative also exhibited significant synergistic antibacterial effects with daptomycin both in vitro and in vivo, with an ability to eradicate planktonic and persister cells of MRSA. Additionally, 62-7c inhibited biofilm formation and eradicated mature biofilms of MRSA. Mechanistic studies revealed that 62-7c inhibited the YycG kinase activity and disrupted the cell membrane by binding to cardiolipin (CL), leading to cell death. Importantly, no development of drug resistance was observed even after 20 serial passages. Furthermore, 62-7c exhibited high biosafety and potent effectiveness in combating infections in both mouse pneumonia and mouse wound models infected with MRSA. Thus, our study revealed that 62-7c has the potential to serve as a novel antibacterial agent for treating MRSA infections.
Collapse
Affiliation(s)
- Yanpeng Xiong
- College of Biotechnology and Pharmaceutical Engineering, Nanjing Tech University, Nanjing 211816, China; Department of Infectious Diseases, Shenzhen Nanshan People's Hospital, Shenzhen University Medical School, Shenzhen 518052, China
| | - Ruian Wang
- College of Biotechnology and Pharmaceutical Engineering, Nanjing Tech University, Nanjing 211816, China
| | - Jiaoyang Zheng
- College of Biotechnology and Pharmaceutical Engineering, Nanjing Tech University, Nanjing 211816, China
| | - Di Fang
- College of Biotechnology and Pharmaceutical Engineering, Nanjing Tech University, Nanjing 211816, China
| | - Peikun He
- Department of Infectious Diseases, Shenzhen Nanshan People's Hospital, Shenzhen University Medical School, Shenzhen 518052, China
| | - Shanghong Liu
- Department of Infectious Diseases, Shenzhen Nanshan People's Hospital, Shenzhen University Medical School, Shenzhen 518052, China
| | - Zhiwei Lin
- Laboratory of Respiratory Disease, People's Hospital of Yangjiang, Yangjiang 529500, China
| | - Xuecheng Chen
- College of Biotechnology and Pharmaceutical Engineering, Nanjing Tech University, Nanjing 211816, China
| | - Chengchun Chen
- Department of Infectious Diseases, Shenzhen Nanshan People's Hospital, Shenzhen University Medical School, Shenzhen 518052, China
| | - Yongpeng Shang
- Department of Infectious Diseases, Shenzhen Nanshan People's Hospital, Shenzhen University Medical School, Shenzhen 518052, China
| | - Zhijian Yu
- Department of Infectious Diseases, Shenzhen Nanshan People's Hospital, Shenzhen University Medical School, Shenzhen 518052, China.
| | - Xiaoju Liu
- Department of Infectious Diseases, Shenzhen Nanshan People's Hospital, Shenzhen University Medical School, Shenzhen 518052, China.
| | - Shiqing Han
- College of Biotechnology and Pharmaceutical Engineering, Nanjing Tech University, Nanjing 211816, China.
| |
Collapse
|
27
|
Brahma U, Singothu S, Suresh A, Vemula D, Munagalasetty S, Sharma P, Bhandari V. MMV 1804559 is a potential antistaphylococcal and antibiofilm agent targeting the clfA gene of Staphylococcus aureus. J Appl Microbiol 2024; 135:lxae276. [PMID: 39474886 DOI: 10.1093/jambio/lxae276] [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: 08/01/2024] [Revised: 09/27/2024] [Accepted: 10/28/2024] [Indexed: 11/28/2024]
Abstract
AIMS Staphylococcus aureus, a high-priority pathogen proclaimed to cause infections ranging from mild to life-threatening, presents significant challenges in treatment. New therapies can be developed quicker using open drug discovery platforms offering a distinct approach to expedite the development of innovative antibacterial and anti-biofilm therapeutics. This study set out to address these issues by finding new uses for current medications to find compounds that are effective against S. aureus. METHODS AND RESULTS In this study, we screened the global priority health box, launched by Medicines for Malaria Ventures containing 240 compounds, for their effectiveness against S. aureus. MMV1795508, MMV1542799, MMV027331, MMV1593278, and MMV1804559 showed potential antibacterial activity at 10 µM concentration. These compounds underwent further evaluation for their ability to clear intracellular bacteria, disrupt biofilm formation, and eradicate existing biofilms. MMV1804559 demonstrated strong efficacy across all tested parameters, achieving 94% inhibition of intracellular bacteria, 79.19% disruption of biofilm cells, and 66.18% inhibition of biofilm formation. Scanning electron microscopy revealed notable membrane perforations and blebbing in MMV1804559-treated cells, indicating its impact on bacterial membranes. Gene expression analysis of cells treated with MMV1804559 showed downregulation of clfA and clfB genes, critical for biofilm formation. Additionally, docking studies confirmed the binding affinity of MMV1804559 with clfA, supported by favorable docking scores, MM/GBSA binding energy, and increased hydrogen bond interactions in the binding pocket, suggesting clfA as a target for MMV1804559. CONCLUSIONS MMV1804559 could serve as a potential therapy for S. aureus by targeting biofilm development and cell adhesion processes.
Collapse
Affiliation(s)
- Umarani Brahma
- National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, Balanagar, Kukatpally Industrial Area, Hyderabad- 500037, Telangana, India
| | - Siva Singothu
- National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, Balanagar, Kukatpally Industrial Area, Hyderabad- 500037, Telangana, India
| | - Akash Suresh
- National Institute of Animal Biotechnology (NIAB)-DBT, Gowlidoddi, Hyderabad-500049, Telangana, India
- Manipal Academy of Higher Education, Manipal 576104, India
| | - Divya Vemula
- National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, Balanagar, Kukatpally Industrial Area, Hyderabad- 500037, Telangana, India
| | - Sharon Munagalasetty
- National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, Balanagar, Kukatpally Industrial Area, Hyderabad- 500037, Telangana, India
| | - Paresh Sharma
- National Institute of Animal Biotechnology (NIAB)-DBT, Gowlidoddi, Hyderabad-500049, Telangana, India
| | - Vasundhra Bhandari
- National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, Balanagar, Kukatpally Industrial Area, Hyderabad- 500037, Telangana, India
| |
Collapse
|
28
|
Baysal A, Saygin H, Soyocak A. A Comparative Study on the Interaction Between Protein and PET Micro/Nanoplastics: Structural and Surface Characteristics of Particles and Impacts on Lung Carcinoma Cells (A549) and Staphylococcus aureus. ENVIRONMENTAL TOXICOLOGY 2024; 39:4899-4926. [PMID: 38923375 DOI: 10.1002/tox.24366] [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: 02/24/2024] [Revised: 04/24/2024] [Accepted: 05/23/2024] [Indexed: 06/28/2024]
Abstract
The interaction between particles and proteins is a key factor determining the toxicity responses of particles. Therefore, this study aimed to examine the interaction between the emerging pollutant polyethylene terephthalate micro/nanoplastics from water bottles with bovine serum albumin. The physicochemical characteristics of micro/nanoplastics were investigated using nuclear magnetic resonance, x-ray diffraction, Fourier transform infrared, dynamic light scattering, and x-ray energy dispersive spectroscopy after exposure to various concentrations and durations of protein. Furthermore, the impact of protein-treated micro/nanoplastics on biological activities was examined using the mitochondrial activity and membrane integrity of A549 cells and the activity and biofilm production of Staphylococcus aureus. The structural characteristics of micro/nanoplastics revealed an interaction with protein. For instance, the assignment of protein-related new proton signals (e.g., CH2, methylene protons of CH2O), changes in available protons s (e.g., CH and CH3), crystallinity, functional groups, elemental ratios, zeta potentials (-11.3 ± 1.3 to -12.4 ± 1.7 to 25.5 ± 2.3 mV), and particle size (395 ± 76 to 496 ± 60 to 866 ± 82 nm) of micro/nanoplastics were significantly observed after protein treatment. In addition, the loading (0.012-0.027 mM) and releasing (0.008-0.013 mM) of protein also showed similar responses with structural characteristics. Moreover, the cell-based responses were changed regarding the structural and surface characteristics of micro/nanoplastics and the loading efficiencies of protein. For example, insignificant mitochondrial activity (2%-10%) and significant membrane integrity (12%-28%) of A549 cells increased compared with control, and reductions in bacterial activity (5%-40%) in many cases and biofilm production specifically at low dose of all treatment stages (13%-46% reduction) were observed.
Collapse
Affiliation(s)
- Asli Baysal
- Department of Chemistry, Faculty of Science and Letters, Istanbul Technical University, Istanbul, Turkey
| | - Hasan Saygin
- Application and Research Center for Advanced Studies, Istanbul Aydin University, Istanbul, Turkey
| | - Ahu Soyocak
- Department of Medical Biology, Faculty of Medicine, Istanbul Aydin University, Istanbul, Turkey
| |
Collapse
|
29
|
Ragupathi H, Pushparaj MM, Gopi SM, Govindarajan DK, Kandaswamy K. Biofilm matrix: a multifaceted layer of biomolecules and a defensive barrier against antimicrobials. Arch Microbiol 2024; 206:432. [PMID: 39402397 DOI: 10.1007/s00203-024-04157-3] [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: 08/09/2024] [Revised: 09/24/2024] [Accepted: 10/03/2024] [Indexed: 11/10/2024]
Abstract
Bacterial cells often exist in the form of sessile aggregates known as biofilms, which are polymicrobial in nature and can produce slimy Extracellular Polymeric Substances (EPS). EPS is often referred to as a biofilm matrix and is a heterogeneous mixture of various biomolecules such as polysaccharides, proteins, and extracellular DNA/RNA (eDNA/RNA). In addition, bacteriophage (phage) was also found to be an integral component of the matrix and can serve as a protective barrier. In recent years, the roles of proteins, polysaccharides, and phages in the virulence of biofilms have been well studied. However, a mechanistic understanding of the release of such biomolecules and their interactions with antimicrobials requires a thorough review. Therefore, this article critically reviews the various mechanisms of release of matrix polymers. In addition, this article also provides a contemporary understanding of interactions between various biomolecules to protect biofilms against antimicrobials. In summary, this article will provide a thorough understanding of the functions of various biofilm matrix molecules.
Collapse
Affiliation(s)
- Harini Ragupathi
- Research Center for Excellence in Microscopy, Department of Biotechnology, Kumaraguru College of Technology, Coimbatore, Tamil Nadu, 641049, India
| | - Mahamahima Muthuswamy Pushparaj
- Research Center for Excellence in Microscopy, Department of Biotechnology, Kumaraguru College of Technology, Coimbatore, Tamil Nadu, 641049, India
| | - Sarves Mani Gopi
- Research Center for Excellence in Microscopy, Department of Biotechnology, Kumaraguru College of Technology, Coimbatore, Tamil Nadu, 641049, India
| | - Deenadayalan Karaiyagowder Govindarajan
- Singapore Centre for Environmental Life Sciences Engineering, Nanyang Technological University, 60 Nanyang Drive, 637551, Singapore, Singapore
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, 21 Nanyang Drive, 637371, Singapore, Singapore
| | - Kumaravel Kandaswamy
- Research Center for Excellence in Microscopy, Department of Biotechnology, Kumaraguru College of Technology, Coimbatore, Tamil Nadu, 641049, India.
| |
Collapse
|
30
|
Shi L, Zhang Y, Zhan Y, Wang X, Xu J, Wang H, Zeng M, Lu Z. Genomic characteristics of antimicrobial resistance and virulence factors of carbapenem-resistant Stutzerimonas nitrititolerans isolated from the clinical specimen. BMC Microbiol 2024; 24:386. [PMID: 39358682 PMCID: PMC11448376 DOI: 10.1186/s12866-024-03546-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2023] [Accepted: 09/24/2024] [Indexed: 10/04/2024] Open
Abstract
BACKGROUND Stutzerimonas nitrititolerans (S. nitrititolerans) is a rare human pathogenic bacterium and has been inadequately explored at the genomic level. Here, we report the first case of carbapenem-resistant S. nitrititolerans isolated from the peritoneal dialysis fluid of a patient with chronic renal failure. This study analyzed the genomic features, antimicrobial resistance, and virulence factors of the isolated strain through whole genome sequencing (WGS). METHODS The bacterial isolate from the peritoneal dialysis fluid was named PDI170223, and preliminary identification was conducted through Matrix-assisted laser desorption ionization/time of flight mass spectrometry (MALDI-TOF MS). WGS of the strain PDI170223 was performed using the Illumina platform, and a phylogenetic tree was constructed based on the 16S rRNA gene sequences. Antimicrobial susceptibility test (AST) was conducted using the TDR-200B2 automatic bacteria identification/drug sensitivity tester. RESULTS S. nitrititolerans may emerge as a human pathogen due to its numerous virulence genes, including those encoding toxins, and those involved in flagellum and biofilm formation. The AST results revealed that the strain is multidrug- and carbapenem-resistant. The antimicrobial resistance genes of S. nitrititolerans are complex and diverse, including efflux pump genes and β⁃lactam resistance genes. CONCLUSION The analysis of virulence factors and antimicrobial resistance of S. nitrititolerans provides clinical insight into the pathogenicity and potential risks of this bacterium. It is crucial to explore the mechanisms through which S. nitrititolerans causes diseases and maintains its antimicrobial resistance, thereby contributing to development of effective treatment and prevention strategies.
Collapse
Affiliation(s)
- Lifeng Shi
- Department of Medical Laboratory, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yingmiao Zhang
- Department of Medical Laboratory, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yu Zhan
- Department of Medical Laboratory, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiuling Wang
- Department of Medical Laboratory, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jia Xu
- School of Laboratory Medicine, Hubei University of Chinese Medicine, Wuhan, China
| | - Hui Wang
- Department of Medical Laboratory, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ming Zeng
- Hubei Center for Clinical Laboratory, Wuhan, China.
| | - Zhongxin Lu
- Department of Medical Laboratory, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
- School of Laboratory Medicine, Hubei University of Chinese Medicine, Wuhan, China.
| |
Collapse
|
31
|
Konwar B, De S, Das G, Ramesh A. Napthalimide-based nuclease inhibitor: A multifunctional therapeutic material to bolster MRSA uptake by macrophage-like cells and mitigate pathogen adhesion on orthopaedic implant. Int J Biol Macromol 2024; 277:134023. [PMID: 39032881 DOI: 10.1016/j.ijbiomac.2024.134023] [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: 11/18/2023] [Revised: 07/14/2024] [Accepted: 07/17/2024] [Indexed: 07/23/2024]
Abstract
The healthcare burden rendered by methicillin-resistant Staphylococcus aureus (MRSA) warrants the development of therapeutics that offer a distinct benefit in the clinics as compared to conventional antibiotics. The present study describes the potential of napthalimide-based synthetic ligands (C1-C3) as inhibitors of the staphylococcal nuclease known as micrococcal nuclease (MNase), a key virulence factor of the pathogen. Amongst the ligands, the most potent MNase inhibitor C1 rendered non-competitive inhibition, reduced MNase turnover number (Kcat) and catalytic efficiency (Kcat/Km) with an IC50 value of ~950 nM. CD spectroscopy suggested distortion of MNase conformation in presence of C1. Flow cytometry and confocal microscopy indicated that C1 restored the ability of activated THP-1 cells to engulf DNA-entrapped MRSA cells. Interestingly, C1 could inhibit MRSA adhesion onto collagen. For potential application, C1-loaded pluronic F-127 micellar nanocarrier (C1-PMC) was generated, wherein the anti-adhesion activity of the pluronic carrier (PMC) and C1 was harnessed in tandem to deter MRSA cell adhesion onto collagen. MRSA biofilm formation was hindered on C1-PMC-coated titanium (Ti) wire, while eluates from C1-PMC-coated Ti wires were non-toxic to HEK 293, MG-63 and THP-1 cells. The multifunctional C1 provides a blueprint for designing therapeutic materials that hold translational potential for mitigation of MRSA infections.
Collapse
Affiliation(s)
- Barlina Konwar
- Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati 781039, Assam, India
| | - Sagnik De
- Department of Chemistry, Indian Institute of Technology Guwahati, Guwahati 781039, Assam, India
| | - Gopal Das
- Department of Chemistry, Indian Institute of Technology Guwahati, Guwahati 781039, Assam, India.
| | - Aiyagari Ramesh
- Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati 781039, Assam, India.
| |
Collapse
|
32
|
Emiliano JVDS, Fusieger A, Camargo AC, Rodrigues FFDC, Nero LA, Perrone ÍT, Carvalho AFD. Staphylococcus aureus in Dairy Industry: Enterotoxin Production, Biofilm Formation, and Use of Lactic Acid Bacteria for Its Biocontrol. Foodborne Pathog Dis 2024; 21:601-616. [PMID: 39021233 DOI: 10.1089/fpd.2023.0170] [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: 07/20/2024] Open
Abstract
Staphylococcus aureus is a well-known pathogen capable of producing enterotoxins during bacterial growth in contaminated food, and the ingestion of such preformed toxins is one of the major causes of food poisoning around the world. Nowadays 33 staphylococcal enterotoxins (SEs) and SE-like toxins have been described, but nearly 95% of confirmed foodborne outbreaks are attributed to classical enterotoxins SEA, SEB, SEC, SED, and SEE. The natural habitat of S. aureus includes the skin and mucous membranes of both humans and animals, allowing the contamination of milk, its derivatives, and the processing facilities. S. aureus is well known for the ability to form biofilms in food processing environments, which contributes to its persistence and cross-contamination in food. The biocontrol of S. aureus in foods by lactic acid bacteria (LAB) and their bacteriocins has been studied for many years. Recently, LAB and their metabolites have also been explored for controlling S. aureus biofilms. LAB are used in fermented foods since in ancient times and nowadays characterized strains (or their purified bacteriocin) can be intentionally added to prolong food shelf-life and to control the growth of potentially pathogenic bacteria. Regarding the use of these microorganism and their metabolites (such as organic acids and bacteriocins) to prevent biofilm development or for biofilm removal, it is possible to conclude that a complex network behind the antagonistic activity remains poorly understood at the molecular level. The use of approaches that allow the characterization of these interactions is necessary to enhance our understanding of the mechanisms that govern the inhibitory activity of LAB against S. aureus biofilms in food processing environments.
Collapse
Affiliation(s)
- Jean Victor Dos Santos Emiliano
- InovaLeite - Laboratório de Pesquisa em Leites e Derivados, Departamento de Tecnologia de Alimentos, Universidade Federal de Viçosa, Viçosa, Brazil
| | - Andressa Fusieger
- InovaLeite - Laboratório de Pesquisa em Leites e Derivados, Departamento de Tecnologia de Alimentos, Universidade Federal de Viçosa, Viçosa, Brazil
| | - Anderson Carlos Camargo
- InovaLeite - Laboratório de Pesquisa em Leites e Derivados, Departamento de Tecnologia de Alimentos, Universidade Federal de Viçosa, Viçosa, Brazil
- InsPOA - Laboratório de Inspeção de Produtos de Origem Animal, Departamento de Veterinária, Universidade Federal de Viçosa, Viçosa, Brazil
| | - Fabíola Faria da Cruz Rodrigues
- InovaLeite - Laboratório de Pesquisa em Leites e Derivados, Departamento de Tecnologia de Alimentos, Universidade Federal de Viçosa, Viçosa, Brazil
| | - Luís Augusto Nero
- InsPOA - Laboratório de Inspeção de Produtos de Origem Animal, Departamento de Veterinária, Universidade Federal de Viçosa, Viçosa, Brazil
| | - Ítalo Tuler Perrone
- Faculdade de Farmácia, Universidade Federal de Juiz de Fora, Juiz de Fora, Brazil
| | - Antônio Fernandes de Carvalho
- InovaLeite - Laboratório de Pesquisa em Leites e Derivados, Departamento de Tecnologia de Alimentos, Universidade Federal de Viçosa, Viçosa, Brazil
| |
Collapse
|
33
|
Salazar M, Shahbazi Nia S, German NA, Awosile B, Sabiu S, Calle A. Exploring diflunisal as a synergistic agent against Staphylococcus aureus biofilm formation. Front Microbiol 2024; 15:1399996. [PMID: 39386371 PMCID: PMC11461217 DOI: 10.3389/fmicb.2024.1399996] [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: 03/12/2024] [Accepted: 08/20/2024] [Indexed: 10/12/2024] Open
Abstract
Staphylococcus aureus is a bacterial pathogen of considerable significance in public health, capable of inducing a diverse range of infectious diseases. One of the most notorious mechanisms used by S. aureus to survive and colonize the site of infection is its ability to form biofilms. Diflunisal, a non-steroidal anti-inflammatory drug (NSAID), is a known inhibitor of the Agr system in S. aureus, which is key in regulating biofilm formation. This study evaluated the effect of broad-spectrum antibiotics in combination with diflunisal on S. aureus biofilm density. Eight antibiotics were tested independently at different concentrations and in combination with diflunisal to assess their effect on S. aureus biofilm formation. When using the antibiotics alone and with diflunisal, a significant control effect on biofilm formation was observed (p < 0.05), irrespective of diflunisal presence, but did not achieve a complete biofilm growth inhibition. Over time, diflunisal influenced biofilm formation; however, such an effect was correlated with antibiotic concentration and exposure time. With amikacin treatments, biofilm density increased with extended exposure time. In the case of imipenem, doripenem, levofloxacin, and ciprofloxacin, lower doses and absence of diflunisal showed higher control over biofilm growth with longer exposure. However, in all cases, diflunisal did not significantly affect the treatment effect on biofilm formation. In the absence of antibiotics, diflunisal significantly reduced biofilm formation by 53.12% (p < 0.05). This study suggests that diflunisal could be a potential treatment to control S. aureus biofilms, but it does not enhance biofilm inhibition when combined with antibiotics.
Collapse
Affiliation(s)
- Maria Salazar
- School of Veterinary Medicine, Texas Tech University, Amarillo, TX, United States
| | - Siavash Shahbazi Nia
- School of Pharmacy, Department of Pharmaceutical Sciences, Texas Tech University Health Sciences Center, Amarillo, TX, United States
| | - Nadezhda A. German
- School of Pharmacy, Department of Pharmaceutical Sciences, Texas Tech University Health Sciences Center, Amarillo, TX, United States
| | - Babafela Awosile
- School of Veterinary Medicine, Texas Tech University, Amarillo, TX, United States
| | - Saheed Sabiu
- Faculty of Applied Sciences, Department of Biotechnology and Food Science, Durban University of Technology, Durban, South Africa
| | - Alexandra Calle
- School of Veterinary Medicine, Texas Tech University, Amarillo, TX, United States
| |
Collapse
|
34
|
Hauserman MR, Sullivan LE, James KL, Ferraro MJ, Rice KC. Response of Staphylococcus aureus physiology and Agr quorum sensing to low-shear modeled microgravity. J Bacteriol 2024; 206:e0027224. [PMID: 39120147 PMCID: PMC11411946 DOI: 10.1128/jb.00272-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Accepted: 07/11/2024] [Indexed: 08/10/2024] Open
Abstract
Staphylococcus aureus is commonly isolated from astronauts returning from spaceflight. Previous analysis of omics data from S. aureus low Earth orbit cultures indicated significantly increased expression of the Agr quorum sensing system and its downstream targets in spaceflight samples compared to ground controls. In this current study, the rotary cell culture system (RCCS) was used to investigate the effect of low-shear modeled microgravity (LSMMG) on S. aureus physiology and Agr activity. When cultured in the same growth medium and temperature as the previous spaceflight experiment, S. aureus LSMMG cultures exhibited decreased agr expression and altered growth compared to normal gravity control cultures, which are typically oriented with oxygenation membrane on the bottom of the high aspect rotating vessel (HARV). When S. aureus was grown in an inverted gravity control orientation (oxygenation membrane on top of the HARV), reduced Agr activity was observed relative to both traditional control and LSMMG cultures, signifying that oxygen availability may affect the observed differences in Agr activity. Metabolite assays revealed increased lactate and decreased acetate excretion in both LSMMG and inverted control cultures. Secretomics analysis of LSMMG, control, and inverted control HARV culture supernatants corroborated these results, with inverted and LSMMG cultures exhibiting a decreased abundance of Agr-regulated virulence factors and an increased abundance of proteins expressed in low-oxygen conditions. Collectively, these studies suggest that the orientation of the HARV oxygenation membrane can affect S. aureus physiology and Agr quorum sensing in the RCCS, a variable that should be considered when interpreting data using this ground-based microgravity model.IMPORTANCES. aureus is commonly isolated from astronauts returning from spaceflight and from surfaces within human-inhabited closed environments such as spacecraft. Astronaut health and immune function are significantly altered in spaceflight. Therefore, elucidating the effects of microgravity on S. aureus physiology is critical for assessing its pathogenic potential during long-term human space habitation. These results also highlight the necessity of eliminating potential confounding factors when comparing simulated microgravity model data with actual spaceflight experiments.
Collapse
Affiliation(s)
- Matthew R. Hauserman
- Department of Microbiology and Cell Science, IFAS, University of Florida, Gainesville, Florida, USA
| | - Leia E. Sullivan
- Department of Microbiology and Cell Science, IFAS, University of Florida, Gainesville, Florida, USA
| | - Kimberly L. James
- Department of Biological Sciences, Florida Gulf Coast University, Fort Myers, Florida, USA
| | - Mariola J. Ferraro
- Department of Microbiology and Cell Science, IFAS, University of Florida, Gainesville, Florida, USA
| | - Kelly C. Rice
- Department of Microbiology and Cell Science, IFAS, University of Florida, Gainesville, Florida, USA
| |
Collapse
|
35
|
Hamza M, Sivaraman GK, Mothadaka MP. Genomic portraits of methicillin-resistant staphylococci (MRS) from food fish unveiled the genes associated with staphylococcal food poisoning (SFP), virulence and antimicrobial resistance. BMC Microbiol 2024; 24:334. [PMID: 39251908 PMCID: PMC11386389 DOI: 10.1186/s12866-024-03469-0] [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: 12/06/2023] [Accepted: 08/19/2024] [Indexed: 09/11/2024] Open
Abstract
BACKGROUND Characteristics of non-clinical strains of methicillin-resistant Staphylococcus aureus (MRSA) especially from fishery environment are poorly understood. This research, in addition to comprehensive characterisation, sought to delineate the genetic relatedness between the MRSA strains originating from clinical as well as non-clinical settings. Out of 39 methicillin-resistant staphylococcal isolates from 197 fish samples, 6 (Three each of methicillin-resistant S. haemolyticus (MRSH) and MRSA) with distinct resistance profiles were selected for whole-genome sequencing. Using respective bioinformatics tools, MRSA genomes were comprehensively characterized for resistome, virulomes, molecular epidemiology and phylogenetic analysis. Simultaneously, MRSH genomes were specifically examined to characterize antimicrobial resistance genes (ARGs), owing to the fact that MRSH is often recognized as a reservoir for resistance determinants. RESULTS Three MRSA clones identified in this study include ST672-IVd/t13599 (sequence type-SCCmec type/spa type), ST88-V/t2526, and ST672-IVa/t1309. Though, the isolates were phenotypically vancomycin-sensitive, five of the six genomes carried vancomycin resistance genes including the VanT (VanG cluster) or VanY (VanM cluster). Among the three MRSA, only one harbored the gene encoding Panton-Valentine Leukocidin (PVL) toxin, while staphylococcal enterotoxin (SEs) genes such as sea and seb, associated with staphylococcal food poisoning were identified in two other MRSA. Genomes of MRSH carried a composite of type V staphylococcal cassette chromosome mec (SCCmec) elements (5C2 & 5). This finding may be explained by the inversion and recombination events that may facilitate the integration of type V elements to the SCC elements of S. aureus with a methicillin-susceptible phenotype. Phylogenetically, MRSA from a non-clinical setting displayed a considerable relatedness to that from clinical settings. CONCLUSION This study highlights the genetic diversity and resistance profiles of MRSA and MRSH, with non-clinical MRSA showing notable relatedness to clinical strains. Future research should explore resistance gene transfer mechanisms and environmental reservoirs to better manage MRSA spread.
Collapse
Affiliation(s)
- Muneeb Hamza
- Faculty of Science, Cochin University of Science and Technology, Cochin-682022, India
- Microbiology Fermentation and Biotechnology (MFB) Division, ICAR- Central Institute of Fisheries Technology (ICAR-CIFT), Matsyapuri P.O., Willingdon, Cochin 682029, India
| | - Gopalan Krishnan Sivaraman
- Microbiology Fermentation and Biotechnology (MFB) Division, ICAR- Central Institute of Fisheries Technology (ICAR-CIFT), Matsyapuri P.O., Willingdon, Cochin 682029, India.
| | - Mukteswar Prasad Mothadaka
- Visakhapatnam Research Centre of ICAR-CIFT, Andhra University P.O., Ocean View Layout, Pandurangapuram, Visakhapatnam, Andhra Pradesh, 530 003, India.
| |
Collapse
|
36
|
Sarkar S, Roy A, Mitra R, Kundu S, Banerjee P, Acharya Chowdhury A, Ghosh S. Escaping the ESKAPE pathogens: A review on antibiofilm potential of nanoparticles. Microb Pathog 2024; 194:106842. [PMID: 39117012 DOI: 10.1016/j.micpath.2024.106842] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Revised: 07/31/2024] [Accepted: 08/05/2024] [Indexed: 08/10/2024]
Abstract
ESKAPE pathogens, a notorious consortium comprising Enterococcusfaecium, Staphylococcusaureus, Klebsiellapneumoniae, Acinetobacterbaumannii, Pseudomonasaeruginosa, and Enterobacter species, pose formidable challenges in healthcare settings due to their multidrug-resistant nature. The increasing global cases of antimicrobial-resistant ESKAPE pathogens are closely related to their remarkable ability to form biofilms. Thus, understanding the unique mechanisms of antimicrobial resistance of ESKAPE pathogens and the innate resilience of biofilms against traditional antimicrobial agents is important for developing innovative strategies to establish effective control methods against them. This review offers a thorough analysis of biofilm dynamics, with a focus on the general mechanisms of biofilm formation, the significant contribution of persister cells in the resistance mechanisms, and the recurrence of biofilms in comparison to planktonic cells. Additionally, this review highlights the potential strategies of nanoparticles for managing biofilms in the ESKAPE group of pathogens. Nanoparticles, with their unique physicochemical properties, provide promising opportunities for disrupting biofilm structures and improving antimicrobial effectiveness. The review has explored interactions between nanoparticles and biofilms, covering a range of nanoparticle types such as metal, metal-oxide, surface-modified, and functionalized nanoparticles, along with organic nanoparticles and nanomaterials. The additional focus of this review also encompasses green synthesis techniques of nanoparticles that involve plant extract and supernatants from bacterial and fungal cultures as reducing agents. Furthermore, the use of nanocomposites and nano emulsions in biofilm management of ESKAPE is also discussed. To conclude, the review addresses the current obstacles and future outlooks in nanoparticle-based biofilm management, stressing the necessity for further research and development to fully exploit the potential of nanoparticles in addressing biofilm-related challenges.
Collapse
Affiliation(s)
| | - Ankita Roy
- Department of Biosciences, JIS University, Kolkata, India
| | - Rangan Mitra
- Department of Biosciences, JIS University, Kolkata, India
| | - Sweta Kundu
- Department of Biosciences, JIS University, Kolkata, India
| | | | | | - Suparna Ghosh
- Department of Biosciences, JIS University, Kolkata, India.
| |
Collapse
|
37
|
Koo J, Hord J, Gilliam C, Rae ML, Staubach K, Nowacki K, Lyren A, Coffey M, Dandoy CE. Levofloxacin prophylaxis in pediatric oncology and hematopoietic stem cell transplantation: a literature review. Pediatr Hematol Oncol 2024; 41:432-448. [PMID: 38975680 PMCID: PMC11335452 DOI: 10.1080/08880018.2024.2353888] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Revised: 04/04/2023] [Accepted: 04/08/2023] [Indexed: 07/09/2024]
Abstract
Bloodstream infections (BSI) are one of the leading causes of morbidity and mortality in children and young adults receiving chemotherapy for malignancy or undergoing hematopoietic stem cell transplantation (HSCT). Antibiotic prophylaxis is commonly used to decrease the risk of BSI; however, antibiotics carry an inherent risk of complications. The aim of this manuscript is to review levofloxacin prophylaxis in pediatric oncology patients and HSCT recipients. We reviewed published literature on levofloxacin prophylaxis to prevent BSI in pediatric oncology patients and HSCT recipients. Nine manuscripts were identified. The use of levofloxacin is indicated in neutropenic children and young adults receiving intensive chemotherapy for leukemia or undergoing HSCT. These results support the efficacy of levofloxacin in pediatric patients with leukemia receiving intensive chemotherapy and should be considered in pediatric patients undergoing HSCT prior to engraftment.
Collapse
Affiliation(s)
- Jane Koo
- Division of Bone Marrow Transplantation and Immune Deficiency, Cincinnati Children’s Medical Center, Cincinnati, OH, USA
- University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Jeffrey Hord
- Showers Family Center for Childhood Cancer and Blood Disorders, Akron Children’s Hospital, Aakron, OH, USA
| | - Craig Gilliam
- Department of Infection Prevention and Control, St. Jude Children’s Research Hospital, Memphis, TN, USA
| | - Mary Lynn Rae
- Ann & Robert H. Lurie Children’s Hospital of Chicago, Chicago, IL, USA
| | - Katherine Staubach
- James M Anderson Center for Health Systems Excellence, Cincinnati Children’s Medical Center, Cincinnati, OH, USA
| | - Katherine Nowacki
- James M Anderson Center for Health Systems Excellence, Cincinnati Children’s Medical Center, Cincinnati, OH, USA
| | - Anne Lyren
- Case Western Reserve University Cleveland, University Hospital Rainbow Babies & Children’s Hospital Cleveland, Cleveland, OH, USA
| | | | - Christopher E. Dandoy
- Division of Bone Marrow Transplantation and Immune Deficiency, Cincinnati Children’s Medical Center, Cincinnati, OH, USA
- University of Cincinnati College of Medicine, Cincinnati, OH, USA
- James M Anderson Center for Health Systems Excellence, Cincinnati Children’s Medical Center, Cincinnati, OH, USA
| |
Collapse
|
38
|
Mondal SK, Alam SA, Roymahapatra G, Mandal SM. Anti-MRSA activity of chlorophenyl pyrrolo benzodiazepines compound. J Antibiot (Tokyo) 2024; 77:589-599. [PMID: 38890385 DOI: 10.1038/s41429-024-00747-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Revised: 04/25/2024] [Accepted: 05/15/2024] [Indexed: 06/20/2024]
Abstract
Antibiotic resistant is the major concern in public health to control the infectious diseases. MRSA (Methicillin-resistant Staphylococcus aureus) is a significant concern in healthcare settings due to its resistance to many antibiotics, including methicillin and other beta-lactams. MRSA infection difficult to treat and increases the risk of complications. Here, we have tested a series of highly condensed heterocyclic derivatives of pyrrolo[1,2-a][1,4]benzodiazepines. Compounds were tested against both, Gram-positive bacteria, Staphylococcus aureus and S. epidermidis, and Gram-negative bacteria, Escherichia coli and Pseudomonas aeruginosa, to assess the antimicrobial efficacy. Compared to Gram-negative bacteria, compounds showed much stronger antibacterial activity against Gram-positive bacteria. SM-5 [Ethyl2-(7-(4-chlorophenyl)-4-methoxy-6,7,8,13-tetrahydro-5H-benzo[e]benzo[5,6][1,4]diazepino[2,1-a]isoindol-15-yl)acetate] derivative was selected as best on the basis of higher therapeutic index among the tested compounds, showed MIC value of 7.81 µg. ml-1 against Staphylococcus strains. Molecular docking analysis between cell wall biosynthesis protein of S. aureus and SM-5 revealed that PBP2a showed the highest binding energy (-8.3 Kcal mol-1), followed by beta-lactam-inducible PBP4 (-7.7 Kcal mol-1), and lipoteichoic acid synthase (-7.5 Kcal mol-1) which is comparably higher than methicillin. Ground state energy calculations by DFT analysis revealed that compound SM-5 and SM-6, almost have equal electronegativity 0.11018 au which also satisfy the quality of the compound reactivity. Analysis of their biofilm inhibition in vitro and in silico toxicity analysis demonstrated their substantial potential to be a kind of future lead antibiotic.
Collapse
Affiliation(s)
- Suresh K Mondal
- Department of Bioscience and Biotechnology, Indian Institute of Technology Kharagpur, Kharagpur, 721302, WB, India
| | - Sk Aftabul Alam
- Department of Botany, Netaji Mahavidyalaya, Arambagh, Hooghly, WB, India
| | | | - Santi M Mandal
- Department of Bioscience and Biotechnology, Indian Institute of Technology Kharagpur, Kharagpur, 721302, WB, India.
| |
Collapse
|
39
|
Crossman L, Sims L, Dean R, Felgate H, Calvo TD, Hill C, McNamara I, Webber MA, Wain J. Sticking together: independent evolution of biofilm formation in different species of staphylococci has occurred multiple times via different pathways. BMC Genomics 2024; 25:812. [PMID: 39198733 PMCID: PMC11350952 DOI: 10.1186/s12864-024-10719-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Accepted: 08/16/2024] [Indexed: 09/01/2024] Open
Abstract
BACKGROUND Staphylococci cause a wide range of infections, including implant-associated infections which are difficult to treat due to the presence of biofilms. Whilst some proteins involved in biofilm formation are known, the differences in biofilm production between staphylococcal species remains understudied. Currently biofilm formation by Staphylococcus aureus is better understood than other members of the genus as more research has focused on this species. RESULTS We assembled a panel of 385 non-aureus Staphylococcus isolates of 19 species from a combination of clinical sources and reference strains. We used a high-throughput crystal violet assay to assess the biofilm forming ability of all strains and assign distinct biofilm formation categories. We compared the prevalence of Pfam domains between the categories and used machine learning to identify amino acid 20-mers linked to biofilm formation. This identified some domains within proteins already linked to biofilm formation and important domains not previously linked to biofilm formation in staphylococci. RT-qPCR confirmed the expression of selected genes predicted to encode important domains within biofilms in Staphylococcus epidermidis. The prevalence and distribution of biofilm associated domains showed a link to phylogeny, suggesting different Staphylococcus species have independently evolved different mechanisms of biofilm production. CONCLUSIONS This work has identified different routes to biofilm formation in diverse species of Staphylococcus and suggests independent evolution of biofilm has occurred multiple times across the genus. Understanding the mechanisms of biofilm formation in any given species is likely to require detailed study of relevant strains and the ability to generalise across the genus may be limited.
Collapse
Affiliation(s)
- Lisa Crossman
- Quadram Institute Bioscience, Norwich, UK
- School of Biological Sciences, University of East Anglia, Norwich, UK
- SequenceAnalysis.Co.Uk, Norwich, UK
| | | | | | | | - Teresa Diaz Calvo
- Quadram Institute Bioscience, Norwich, UK
- School of Medicine, University of East Anglia, Norwich, UK
| | | | | | - Mark A Webber
- Quadram Institute Bioscience, Norwich, UK.
- School of Medicine, University of East Anglia, Norwich, UK.
| | - John Wain
- Quadram Institute Bioscience, Norwich, UK
- School of Medicine, University of East Anglia, Norwich, UK
| |
Collapse
|
40
|
Firoozbahr M, Palombo EA, Kingshott P, Zaferanloo B. Antibacterial and Antibiofilm Properties of Native Australian Plant Endophytes against Wound-Infecting Bacteria. Microorganisms 2024; 12:1710. [PMID: 39203552 PMCID: PMC11357646 DOI: 10.3390/microorganisms12081710] [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: 07/15/2024] [Revised: 08/12/2024] [Accepted: 08/14/2024] [Indexed: 09/03/2024] Open
Abstract
The wound management field faces significant challenges due to antimicrobial resistance (AMR) and the complexity of chronic wound care. Effective wound treatment requires antimicrobial dressings to prevent bacterial infections. However, the rise of AMR necessitates new antimicrobial agents for wound dressings, particularly for addressing bacterial pathogens like methicillin-resistant Staphylococcus aureus (MRSA). Endophytic fungi, known for producing diverse bioactive compounds, represent a promising source of such new agents. This study tested thirty-two endophytic fungi from thirteen distinct Australian native plants for their antibacterial activity against S. aureus. Ethyl acetate (EtOAc) extracts from fungal culture filtrates exhibited inhibitory effects against both methicillin-sensitive S. aureus ATCC 25923 (MIC = 78.1 µg/mL) and MRSA M180920 (MIC = 78.1 µg/mL). DNA sequence analysis was employed for fungal identification. The most active sample, EL 19 (Chaetomium globosum), was selected for further analysis, revealing that its EtOAc extracts reduced S. aureus ATCC 25923 biofilm formation by 55% and cell viability by 57% to 68% at 12 × MIC. Furthermore, cytotoxicity studies using the brine shrimp lethality test demonstrated low cytotoxicity up to 6 × MIC (25% mortality rate) with an LC50 value of 639.1 µg/mL. Finally, the most active sample was incorporated into polycaprolactone (PCL) fiber mats via electrospinning, with resultant inhibition of S. aureus species. This research underscores the potential of endophytic fungi from Australian plants as sources of substances effective against common wound pathogens. Further exploration of the responsible compounds and their mechanisms could facilitate the development of wound dressings effective against MRSA and innovative biofilm-resistant electrospun fibers, contributing to the global efforts to combat AMR.
Collapse
Affiliation(s)
- Meysam Firoozbahr
- Department of Chemistry and Biotechnology, School of Science, Computing and Engineering Technologies, Swinburne University of Technology, Hawthorn, VIC 3122, Australia; (M.F.); (P.K.)
| | - Enzo A. Palombo
- Department of Chemistry and Biotechnology, School of Science, Computing and Engineering Technologies, Swinburne University of Technology, Hawthorn, VIC 3122, Australia; (M.F.); (P.K.)
- ARC Training Center for Biofilm Research and Innovation, School of Science, Computing and Engineering Technologies, Swinburne University of Technology, Hawthorn, VIC 3122, Australia
| | - Peter Kingshott
- Department of Chemistry and Biotechnology, School of Science, Computing and Engineering Technologies, Swinburne University of Technology, Hawthorn, VIC 3122, Australia; (M.F.); (P.K.)
- ARC Training Centre in Surface Engineering for Advanced Materials (SEAM), School of Engineering, Swinburne University of Technology, Hawthorn, VIC 3122, Australia
| | - Bita Zaferanloo
- Department of Chemistry and Biotechnology, School of Science, Computing and Engineering Technologies, Swinburne University of Technology, Hawthorn, VIC 3122, Australia; (M.F.); (P.K.)
| |
Collapse
|
41
|
Freiberg JA, Reyes Ruiz VM, Gimza BD, Murdoch CC, Green ER, Curry JM, Cassat JE, Skaar EP. Restriction of arginine induces antibiotic tolerance in Staphylococcus aureus. Nat Commun 2024; 15:6734. [PMID: 39112491 PMCID: PMC11306626 DOI: 10.1038/s41467-024-51144-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Accepted: 07/31/2024] [Indexed: 08/10/2024] Open
Abstract
Staphylococcus aureus is responsible for a substantial number of invasive infections globally each year. These infections are problematic because they are frequently recalcitrant to antibiotic treatment. Antibiotic tolerance, the ability of bacteria to persist despite normally lethal doses of antibiotics, contributes to antibiotic treatment failure in S. aureus infections. To understand how antibiotic tolerance is induced, S. aureus biofilms exposed to multiple anti-staphylococcal antibiotics are examined using both quantitative proteomics and transposon sequencing. These screens indicate that arginine metabolism is involved in antibiotic tolerance within a biofilm and support the hypothesis that depletion of arginine within S. aureus communities can induce antibiotic tolerance. Consistent with this hypothesis, inactivation of argH, the final gene in the arginine synthesis pathway, induces antibiotic tolerance. Arginine restriction induces antibiotic tolerance via inhibition of protein synthesis. In murine skin and bone infection models, an argH mutant has enhanced ability to survive antibiotic treatment with vancomycin, highlighting the relationship between arginine metabolism and antibiotic tolerance during S. aureus infection. Uncovering this link between arginine metabolism and antibiotic tolerance has the potential to open new therapeutic avenues targeting previously recalcitrant S. aureus infections.
Collapse
Affiliation(s)
- Jeffrey A Freiberg
- Division of Infectious Diseases, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA.
- Vanderbilt Institute for Infection, Immunology and Inflammation, Vanderbilt University Medical Center, Nashville, TN, USA.
| | - Valeria M Reyes Ruiz
- Vanderbilt Institute for Infection, Immunology and Inflammation, Vanderbilt University Medical Center, Nashville, TN, USA
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Brittney D Gimza
- Division of Pediatric Infectious Diseases, Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Caitlin C Murdoch
- Vanderbilt Institute for Infection, Immunology and Inflammation, Vanderbilt University Medical Center, Nashville, TN, USA
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Erin R Green
- Vanderbilt Institute for Infection, Immunology and Inflammation, Vanderbilt University Medical Center, Nashville, TN, USA
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA
- Department of Microbiology, University of Chicago, Chicago, IL, USA
| | - Jacob M Curry
- Division of Pediatric Infectious Diseases, Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN, USA
| | - James E Cassat
- Vanderbilt Institute for Infection, Immunology and Inflammation, Vanderbilt University Medical Center, Nashville, TN, USA
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA
- Division of Pediatric Infectious Diseases, Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN, USA
- Vanderbilt Center for Bone Biology, Vanderbilt University Medical Center, Nashville, TN, USA
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, USA
| | - Eric P Skaar
- Vanderbilt Institute for Infection, Immunology and Inflammation, Vanderbilt University Medical Center, Nashville, TN, USA.
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA.
| |
Collapse
|
42
|
Rimi SS, Ashraf MN, Sigma SH, Ahammed MT, Siddique MP, Zinnah MA, Rahman MT, Islam MS. Biofilm formation, agr typing and antibiotic resistance pattern in methicillin-resistant Staphylococcus aureus isolated from hospital environments. PLoS One 2024; 19:e0308282. [PMID: 39102390 PMCID: PMC11299820 DOI: 10.1371/journal.pone.0308282] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2024] [Accepted: 07/20/2024] [Indexed: 08/07/2024] Open
Abstract
Biofilm development significantly enhances the virulence of methicillin-resistant Staphylococcus aureus (MRSA), leading to severe infections and decreased susceptibility to antibiotics, especially in strains associated with hospital environments. This study examined the occurrence of MRSA, their ability to form biofilms, agr typing, and the antibiotic resistance profiles of biofilm-forming MRSA strains isolated from environmental surfaces at Mymensingh Medical College Hospital (MMCH). From 120 swab samples, 86 (71.67%) tested positive for S. aureus. MRSA was identified in 86 isolates using the disk diffusion technique, and by polymerase chain reaction (PCR), 56 (65.1%) isolates were confirmed to carry the mecA gene. The Crystal Violet Microtiter Plate (CVMP) test revealed that 80.35% (45 isolates) were biofilm-forming and 19.6% (11 isolates) were non-biofilm-forming. Out of 45 biofilm producer isolates 37.5% and 42.9% isolates exhibited strong and intermediate biofilm-forming characteristics, respectively. Molecular analysis revealed that 17.78% of MRSA isolates carried at least one gene related to biofilm formation, specifically icaA, icaB, and icaD genes were discovered in 13.33%, 8.89%, 6.67% of the MRSA isolates, respectively. In agr typing, the most prevalent group was agr I (71.11%), followed by group III (17.78%) and group II (11.11%). Group IV was not detected. The distribution of agr gene groups showed a significant difference among biofilm-forming isolates (p < 0.05). In agr group I, 18.75% of isolates carried the icaA gene, 12.5% carried the icaB gene, and 9.37% carried the icaD gene. Biofilm-forming genes were not detected in any of the isolates from agr groups II or III. There are no statistically significant differences between agr groups and the presence of these genes (p > 0.05). Antibiotic resistance varied significantly among agr groups, with agr group I displaying the highest resistance, agr group II, and agr group III exhibiting the least resistance (p < 0.05). Seventy-three (73.3%) of the isolates were multi-drug resistant, with agr group I displaying nineteen MDR patterns. The occurrence of MRSA in hospital environments and their capacity to form biofilm raises concerns for public health. These findings support the importance of further research focused on agr quorum sensing systems as a basis for developing novel antibacterial agents.
Collapse
Affiliation(s)
- Sabrina Sultana Rimi
- Department of Microbiology and Hygiene, Faculty of Veterinary Science, Bangladesh Agricultural University, Mymensingh, Bangladesh
| | - Md. Nahid Ashraf
- Department of Microbiology and Hygiene, Faculty of Veterinary Science, Bangladesh Agricultural University, Mymensingh, Bangladesh
| | - Sanzila Hossain Sigma
- Department of Microbiology and Hygiene, Faculty of Veterinary Science, Bangladesh Agricultural University, Mymensingh, Bangladesh
| | - Md. Tanjir Ahammed
- Department of Microbiology and Hygiene, Faculty of Veterinary Science, Bangladesh Agricultural University, Mymensingh, Bangladesh
| | - Mahbubul Pratik Siddique
- Department of Microbiology and Hygiene, Faculty of Veterinary Science, Bangladesh Agricultural University, Mymensingh, Bangladesh
| | - Mohammad Ali Zinnah
- Department of Microbiology and Public Health, Faculty of Veterinary Medicine and Animal Science, Bangabandhu Sheikh Mujibur Rahman Agricultural University, Gazipur, Bangladesh
| | - Md. Tanvir Rahman
- Department of Microbiology and Hygiene, Faculty of Veterinary Science, Bangladesh Agricultural University, Mymensingh, Bangladesh
| | - Md. Shafiqul Islam
- Department of Microbiology and Hygiene, Faculty of Veterinary Science, Bangladesh Agricultural University, Mymensingh, Bangladesh
| |
Collapse
|
43
|
Yousuf B, Pasha R, Pineault N, Ramirez-Arcos S. Modulation of Staphylococcus aureus gene expression during proliferation in platelet concentrates with focus on virulence and platelet functionality. PLoS One 2024; 19:e0307920. [PMID: 39052660 PMCID: PMC11271859 DOI: 10.1371/journal.pone.0307920] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Accepted: 07/09/2024] [Indexed: 07/27/2024] Open
Abstract
Staphylococcus aureus is a well-documented bacterial contaminant in platelet concentrates (PCs), a blood component used to treat patients with platelet deficiencies. This bacterium can evade routine PC culture screening and cause septic transfusion reactions. Here, we investigated the gene expression modulation within the PC niche versus trypticase soy media (TSB) of S. aureus CBS2016-05, a strain isolated from a septic reaction, in comparison to PS/BAC/317/16/W, a strain identified during PC screening. RNA-seq analysis revealed upregulation of the capsule biosynthesis operon (capA-H), surface adhesion factors (sasADF), clumping factor A (clfA), protein A (spa), and anaerobic metabolism genes (pflAB, nrdDG) in CBS2016-05 when grown in PCs versus TSB, implying its enhanced pathogenicity in this milieu, in contrast to the PS/BAC/317/16/W strain. Furthermore, we investigated the impact of S. aureus CBS2016-05 on platelet functionality in spiked PCs versus non-spiked PC units. Flow cytometry analyses revealed a significant decrease in glycoprotein (GP) IIb (CD41) and GPIbα (CD42b) expression, alongside increased P-selectin (CD62P) and phosphatidylserine (annexin V) expression in spiked PCs compared to non-spiked PCs (p = 0.01). Moreover, spiked PCs exhibited a drastic reduction in MitoTrack Red FM and Calcein AM positive platelets (87.3% vs. 29.4%, p = 0.0001 and 95.4% vs. 24.7%, p = 0.0001) in a bacterial cell density manner. These results indicated that S. aureus CBS2016-05 triggers platelet activation and apoptosis, and compromises mitochondrial functionality and platelet viability, in contaminated PCs. Furthermore, this study enhanced our understanding of the effects of platelet-bacteria interactions in the unique PC niche, highlighting S. aureus increased pathogenicity and deleterious effect on platelet functionality in a strain specific manner. Our novel insights serve as a platform to improve PC transfusion safety.
Collapse
Affiliation(s)
- Basit Yousuf
- Medical Affairs and Innovation, Canadian Blood Services, Ottawa, Canada
- Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, Canada
| | - Roya Pasha
- Medical Affairs and Innovation, Canadian Blood Services, Ottawa, Canada
| | - Nicolas Pineault
- Medical Affairs and Innovation, Canadian Blood Services, Ottawa, Canada
- Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, Canada
| | - Sandra Ramirez-Arcos
- Medical Affairs and Innovation, Canadian Blood Services, Ottawa, Canada
- Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, Canada
| |
Collapse
|
44
|
Yu J, Han W, Xu Y, Shen L, Zhao H, Zhang J, Xiao Y, Guo Y, Yu F. Biofilm-producing ability of methicillin-resistant Staphylococcus aureus clinically isolated in China. BMC Microbiol 2024; 24:241. [PMID: 38961344 PMCID: PMC11223284 DOI: 10.1186/s12866-024-03380-8] [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: 01/11/2024] [Accepted: 06/18/2024] [Indexed: 07/05/2024] Open
Abstract
BACKGROUND Staphylococcus aureus, a commensal bacterium, colonizes the skin and mucous membranes of approximately 30% of the human population. Apart from conventional resistance mechanisms, one of the pathogenic features of S. aureus is its ability to survive in a biofilm state on both biotic and abiotic surfaces. Due to this characteristic, S. aureus is a major cause of human infections, with Methicillin-Resistant Staphylococcus aureus (MRSA) being a significant contributor to both community-acquired and hospital-acquired infections. RESULTS Analyzing non-repetitive clinical isolates of MRSA collected from seven provinces and cities in China between 2014 and 2020, it was observed that 53.2% of the MRSA isolates exhibited varying degrees of ability to produce biofilm. The biofilm positivity rate was notably high in MRSA isolates from Guangdong, Jiangxi, and Hubei. The predominant MRSA strains collected in this study were of sequence types ST59, ST5, and ST239, with the biofilm-producing capability mainly distributed among moderate and weak biofilm producers within these ST types. Notably, certain sequence types, such as ST88, exhibited a high prevalence of strong biofilm-producing strains. The study found that SCCmec IV was the predominant type among biofilm-positive MRSA, followed by SCCmec II. Comparing strains with weak and strong biofilm production capabilities, the positive rates of the sdrD and sdrE were higher in strong biofilm producers. The genetic determinants ebp, icaA, icaB, icaC, icaD, icaR, and sdrE were associated with strong biofilm production in MRSA. Additionally, biofilm-negative MRSA isolates showed higher sensitivity rates to cefalotin (94.8%), daptomycin (94.5%), mupirocin (86.5%), teicoplanin (94.5%), fusidic acid (81.0%), and dalbavancin (94.5%) compared to biofilm-positive MRSA isolates. The biofilm positivity rate was consistently above 50% in all collected specimen types. CONCLUSIONS MRSA strains with biofilm production capability warrant increased vigilance.
Collapse
Affiliation(s)
- Jingyi Yu
- Department of Clinical Laboratory, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Weihua Han
- 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
| | - Li Shen
- 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
| | - Jiao Zhang
- Department of Clinical Laboratory, Key Laboratory of Clinical Laboratory Diagnosis and Translational Research of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Yanghua Xiao
- School of Public Health, Nanchang University, Nanchang, Jiangxi, China
| | - Yinjuan Guo
- 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.
| |
Collapse
|
45
|
Narayana Iyengar S, Dowden B, Ragheb K, Patsekin V, Rajwa B, Bae E, Robinson JP. Identifying antibiotic-resistant strains via cell sorting and elastic-light-scatter phenotyping. Appl Microbiol Biotechnol 2024; 108:406. [PMID: 38958764 PMCID: PMC11222266 DOI: 10.1007/s00253-024-13232-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Revised: 03/04/2024] [Accepted: 03/19/2024] [Indexed: 07/04/2024]
Abstract
The proliferation and dissemination of antimicrobial-resistant bacteria is an increasingly global challenge and is attributed mainly to the excessive or improper use of antibiotics. Currently, the gold-standard phenotypic methodology for detecting resistant strains is agar plating, which is a time-consuming process that involves multiple subculturing steps. Genotypic analysis techniques are fast, but they require pure starting samples and cannot differentiate between viable and non-viable organisms. Thus, there is a need to develop a better method to identify and prevent the spread of antimicrobial resistance. This work presents a novel method for detecting and identifying antibiotic-resistant strains by combining a cell sorter for bacterial detection and an elastic-light-scattering method for bacterial classification. The cell sorter was equipped with safety mechanisms for handling pathogenic organisms and enabled precise placement of individual bacteria onto an agar plate. The patterning was performed on an antibiotic-gradient plate, where the growth of colonies in sections with high antibiotic concentrations confirmed the presence of a resistant strain. The antibiotic-gradient plate was also tested with an elastic-light-scattering device where each colony's unique colony scatter pattern was recorded and classified using machine learning for rapid identification of bacteria. Sorting and patterning bacteria on an antibiotic-gradient plate using a cell sorter reduced the number of subculturing steps and allowed direct qualitative binary detection of resistant strains. Elastic-light-scattering technology is a rapid, label-free, and non-destructive method that permits instantaneous classification of pathogenic strains based on the unique bacterial colony scatter pattern. KEY POINTS: • Individual bacteria cells are placed on gradient agar plates by a cell sorter • Laser-light scatter patterns are used to recognize antibiotic-resistant organisms • Scatter patterns formed by colonies correspond to AMR-associated phenotypes.
Collapse
Affiliation(s)
| | - Brianna Dowden
- Department of Basic Medical Sciences, Purdue University, West Lafayette, IN, 47907, USA
| | - Kathy Ragheb
- Department of Basic Medical Sciences, Purdue University, West Lafayette, IN, 47907, USA
| | - Valery Patsekin
- Department of Basic Medical Sciences, Purdue University, West Lafayette, IN, 47907, USA
| | - Bartek Rajwa
- Bindley Bioscience Center, Purdue University, West Lafayette, IN, 47907, USA
| | - Euiwon Bae
- School of Mechanical Engineering, Purdue University, West Lafayette, IN, 47907, USA
| | - J Paul Robinson
- Department of Basic Medical Sciences, Purdue University, West Lafayette, IN, 47907, USA.
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN, 47907, USA.
| |
Collapse
|
46
|
Aalilou Y, Moussa H, Lee LH, Bouyahya A, Zengin G, Faouzi MEA. What hidden treasure resides beneath the waves?: Phytochemistry, pharmacological properties and uses of Halopteris scoparia (Linnaeus) Sauvageau 1904: An overview. Fitoterapia 2024; 176:106016. [PMID: 38740345 DOI: 10.1016/j.fitote.2024.106016] [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: 01/03/2024] [Revised: 05/09/2024] [Accepted: 05/11/2024] [Indexed: 05/16/2024]
Abstract
Over the years, the biological activities of seaweeds could have piqued research interest due to their specific functional phytochemistry, which may not be available in terrestrial plants. Seaweeds produce these compounds to overcome and control stressful biotic and abiotic conditions. Additionally, they are potentially excellent sources of highly useful leads in the development of new drugs. Our study aims to unveil, for the first time, an overview of Halopteris scoparia, a species belonging to the Phaeophyceae class and the Stypocaulacea family, by summarizing all available literature data. In this work, we attempt to shed light on its phytochemistry, nutritional values, pharmacological activities, and industrial uses and applications. To gather information related to H. scoparia, relevant keywords were used to search internet databases including Google Scholar, PubMed, ResearchGate, Web of Science, Algae Database, WoRMS database, and DORIS database. The chemical structures were drawn using Chemdraw and verified using the PubChem database. Chemically, this species contains a wide variety of secondary metabolites, such as terpenoids and phenolic compounds. Additionally, other chemical components with nutraceutical value have been identified, such as carbohydrates, proteins, lipids, pigments, minerals and mycosporine like amino acids. Then, holding several reported pharmacological properties, including antioxidant, anti-inflammatory, cytotoxic, dermoprotective, antidepressive, antibacterial, antibiofilm, antifungal, anti-parasitic activities and acute toxicity. In addition to other their applications such as bioconversion and antifouling activities. To confirm the previous pharmacological properties, more comprehensive and systematic in vivo, preclinical, and clinical studies are needed. Furthermore, research is required to uncover the mechanisms of its active compounds and their potential therapeutic effects in treating other diseases such as atherosclerosis, neurodegenerative diseases, and viral infections.
Collapse
Affiliation(s)
- Youssra Aalilou
- Laboratories of Pharmacology and Toxicology, Pharmaceutical and Toxicological Analysis Research Team, Faculty of Medicine and Pharmacy, Mohammed V University in Rabat, Rabat, Morocco
| | - Hanaa Moussa
- Applied Phycology-Mycology Group, Applied Botany Laboratory, Department of Biology, Faculty of Sciences, Abdelmalek Essaâdi University, M'Hannech II, 93030 Tetouan, Morocco
| | - Learn Han Lee
- Microbiome Research Group, Research Centre for Life Science and Healthcare, Nottingham Ningbo China Beacons of Excellence Research and Innovation Institute (CBI), University of Nottingham Ningbo China, Ningbo, China 315000, China; Novel Bacteria and Drug Discovery Research Group (NBDD), Microbiome and Bioresource Research Strength (MBRS), Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Subang Jaya, Selangor 47500, Malaysia.
| | - Abdelhakim Bouyahya
- Laboratory of Human Pathologies Biology, Faculty of Sciences, Mohammed V University in Rabat, Rabat 10106, Morocco
| | - Gokhan Zengin
- Physiology and Biochemistry Laboratory, Department of Biology, Science Faculty, Selcuk University, Konya 42130, Turkey.
| | - My El Abbes Faouzi
- Laboratories of Pharmacology and Toxicology, Pharmaceutical and Toxicological Analysis Research Team, Faculty of Medicine and Pharmacy, Mohammed V University in Rabat, Rabat, Morocco
| |
Collapse
|
47
|
Schaffler BC, Longwell M, Byers B, Kreft R, Ramot R, Ramot Y, Schwarzkopf R. Nanoparticle ultrasonication outperforms conventional irrigation solutions in eradicating Staphylococcus aureus biofilm from titanium surfaces: an in vitro study. EUROPEAN JOURNAL OF ORTHOPAEDIC SURGERY & TRAUMATOLOGY : ORTHOPEDIE TRAUMATOLOGIE 2024; 34:2729-2734. [PMID: 38761198 DOI: 10.1007/s00590-024-03982-y] [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: 03/15/2024] [Accepted: 04/29/2024] [Indexed: 05/20/2024]
Abstract
PURPOSE Bacterial biofilms create a challenge in the treatment of prosthetic joint infection (PJI), and failure to eradicate biofilms is often implicated in the high rates of recurrence. In this study, we aimed to compare the effectiveness of a novel nanoparticle ultrasonication technology on Staphylococcus aureus biofilm eradication compared to commonly used orthopedic irrigation solutions. METHODS Twenty-four sterile, titanium alloy discs were inoculated with a standardized concentration of methicillin-resistant S. aureus and cultured for seven days to allow for biofilm formation. Discs were then treated with either ultrasonicated nanoparticle therapy or irrigation with chlorhexidine gluconate, povidone-iodine or normal saline. The remaining bacteria on each surface was subsequently plated for colony-forming units of S. aureus. Bacterial eradication was reported as a decrease in CFUs relative to the control group. Mann-Whitney U tests were used to compare between groups. RESULTS Treatment with ultrasonicated nanoparticles resulted in a significant mean decrease in CFUs of 99.3% compared to controls (p < 0.0001). Irrigation with povidone-iodine also resulted in a significant 77.5% reduction in CFUs compared to controls (p < 0.0001). Comparisons between ultrasonicated nanoparticles and povidone-iodine demonstrated a significantly higher reduction in bacterial CFUs in the nanoparticle group (p < 0.0001). CONCLUSION Ultrasonicated nanoparticle were superior to commonly used bactericidal irrigation solutions in the eradication of S. aureus from a titanium surface. Future clinical studies are warranted to evaluate this ultrsonication technology in the treatment of PJI.
Collapse
Affiliation(s)
- Benjamin C Schaffler
- Department of Orthopedic Surgery, NYU Langone Orthopedic Hospital, 301 East 17Th Street, New York, NY, 10003, USA
| | - Mark Longwell
- Center for Excellence in Biofilm Research, Allegheny Health Network Research Institute, Pittsburgh, PA, USA
| | - Barbara Byers
- Center for Excellence in Biofilm Research, Allegheny Health Network Research Institute, Pittsburgh, PA, USA
| | - Rachel Kreft
- Center for Excellence in Biofilm Research, Allegheny Health Network Research Institute, Pittsburgh, PA, USA
| | - Roi Ramot
- Center for Excellence in Biofilm Research, Allegheny Health Network Research Institute, Pittsburgh, PA, USA
| | - Yair Ramot
- Center for Excellence in Biofilm Research, Allegheny Health Network Research Institute, Pittsburgh, PA, USA
| | - Ran Schwarzkopf
- Department of Orthopedic Surgery, NYU Langone Orthopedic Hospital, 301 East 17Th Street, New York, NY, 10003, USA.
| |
Collapse
|
48
|
Santajit S, Tunyong W, Horpet D, Binmut A, Kong-Ngoen T, Wisessaowapak C, Thavorasak T, Pumirat P, Indrawattana N. Unveiling the Antimicrobial, Anti-Biofilm, and Anti-Quorum-Sensing Potential of Paederia foetida Linn. Leaf Extract against Staphylococcus aureus: An Integrated In Vitro-In Silico Investigation. Antibiotics (Basel) 2024; 13:613. [PMID: 39061295 PMCID: PMC11273848 DOI: 10.3390/antibiotics13070613] [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: 05/21/2024] [Revised: 06/18/2024] [Accepted: 06/19/2024] [Indexed: 07/28/2024] Open
Abstract
Antimicrobial resistance poses a global health threat, with Staphylococcus aureus emerging as a notorious pathogen capable of forming stubborn biofilms and regulating virulence through quorum sensing (QS). In the quest for novel therapeutic strategies, this groundbreaking study unveils the therapeutic potential of Paederia foetida Linn., an Asian medicinal plant containing various bioactive compounds, contributing to its antimicrobial activities, in the battle against S. aureus. Through a comprehensive approach, we investigated the effect of ethanolic P. foetida leaf extract on S. aureus biofilms, QS, and antimicrobial activity. The extract exhibited promising inhibitory effects against S. aureus including the biofilm-forming strain and MRSA. Real-time PCR analysis revealed significant downregulation of key virulence and biofilm genes, suggesting interference with QS. Biofilm assays quantified the extract's ability to disrupt and prevent biofilm formation. LC-MS/MS analysis identified quercetin and kaempferol glycosides as potential bioactive constituents, while molecular docking studies explored their binding to the QS transcriptional regulator SarA. Computational ADMET predictions highlighted favorable intestinal absorption but potential P-glycoprotein interactions limiting oral bioavailability. While promising anti-virulence effects were demonstrated, the high molecular weights and excessive hydrogen bond donors/acceptors of the flavonoid glycosides raise concerns regarding drug-likeness and permeability. This integrated study offers valuable insights for developing novel anti-virulence strategies to combat antimicrobial resistance.
Collapse
Affiliation(s)
- Sirijan Santajit
- Department of Medical Technology, School of Allied Health Sciences, Walailak University, Tha Sala 80160, Thailand; (S.S.); (D.H.); (A.B.)
- Research Center in Tropical Pathobiology, Walailak University, Tha Sala 80160, Thailand
| | - Witawat Tunyong
- Department of Microbiology and Immunology, Faculty of Tropical Medicine, Mahidol University, Bangkok 10400, Thailand; (W.T.); (T.K.-N.); (P.P.)
| | - Dararat Horpet
- Department of Medical Technology, School of Allied Health Sciences, Walailak University, Tha Sala 80160, Thailand; (S.S.); (D.H.); (A.B.)
| | - Asma Binmut
- Department of Medical Technology, School of Allied Health Sciences, Walailak University, Tha Sala 80160, Thailand; (S.S.); (D.H.); (A.B.)
| | - Thida Kong-Ngoen
- Department of Microbiology and Immunology, Faculty of Tropical Medicine, Mahidol University, Bangkok 10400, Thailand; (W.T.); (T.K.-N.); (P.P.)
| | | | - Techit Thavorasak
- Center of Research Excellence in Therapeutic Proteins and Antibody Engineering, Department of Parasitology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand;
| | - Pornpan Pumirat
- Department of Microbiology and Immunology, Faculty of Tropical Medicine, Mahidol University, Bangkok 10400, Thailand; (W.T.); (T.K.-N.); (P.P.)
| | - Nitaya Indrawattana
- Department of Microbiology and Immunology, Faculty of Tropical Medicine, Mahidol University, Bangkok 10400, Thailand; (W.T.); (T.K.-N.); (P.P.)
- Siriraj Center of Research Excellence in Allergy and Immunology, Department of Research, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand
| |
Collapse
|
49
|
Las Heras K, Garcia-Orue I, Rancan F, Igartua M, Santos-Vizcaino E, Hernandez RM. Modulating the immune system towards a functional chronic wound healing: A biomaterials and Nanomedicine perspective. Adv Drug Deliv Rev 2024; 210:115342. [PMID: 38797316 DOI: 10.1016/j.addr.2024.115342] [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: 01/26/2024] [Revised: 05/16/2024] [Accepted: 05/18/2024] [Indexed: 05/29/2024]
Abstract
Chronic non-healing wounds persist as a substantial burden for healthcare systems, influenced by factors such as aging, diabetes, and obesity. In contrast to the traditionally pro-regenerative emphasis of therapies, the recognition of the immune system integral role in wound healing has significantly grown, instigating an approach shift towards immunological processes. Thus, this review explores the wound healing process, highlighting the engagement of the immune system, and delving into the behaviors of innate and adaptive immune cells in chronic wound scenarios. Moreover, the article investigates biomaterial-based strategies for the modulation of the immune system, elucidating how the adjustment of their physicochemical properties or their synergistic combination with other agents such as drugs, proteins or mesenchymal stromal cells can effectively modulate the behaviors of different immune cells. Finally this review explores various strategies based on synthetic and biological nanostructures, including extracellular vesicles, to finely tune the immune system as natural immunomodulators or therapeutic nanocarriers with promising biophysical properties.
Collapse
Affiliation(s)
- Kevin Las Heras
- NanoBioCel Research Group, Laboratory of Pharmaceutics, School of Pharmacy, University of the Basque Country (UPV-EHU), Vitoria-Gasteiz, Spain; Bioaraba, NanoBioCel Research Group, Vitoria-Gasteiz, Spain
| | - Itxaso Garcia-Orue
- NanoBioCel Research Group, Laboratory of Pharmaceutics, School of Pharmacy, University of the Basque Country (UPV-EHU), Vitoria-Gasteiz, Spain; Bioaraba, NanoBioCel Research Group, Vitoria-Gasteiz, Spain; Biomedical Research Networking Centre in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN). Institute of Health Carlos III, Madrid, Spain
| | - Fiorenza Rancan
- Department of Dermatology, Venereology und Allergology,Clinical Research Center for Hair and Skin Science, Charité - Universitätsmedizin Berlin
| | - Manoli Igartua
- NanoBioCel Research Group, Laboratory of Pharmaceutics, School of Pharmacy, University of the Basque Country (UPV-EHU), Vitoria-Gasteiz, Spain; Bioaraba, NanoBioCel Research Group, Vitoria-Gasteiz, Spain; Biomedical Research Networking Centre in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN). Institute of Health Carlos III, Madrid, Spain
| | - Edorta Santos-Vizcaino
- NanoBioCel Research Group, Laboratory of Pharmaceutics, School of Pharmacy, University of the Basque Country (UPV-EHU), Vitoria-Gasteiz, Spain; Bioaraba, NanoBioCel Research Group, Vitoria-Gasteiz, Spain; Biomedical Research Networking Centre in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN). Institute of Health Carlos III, Madrid, Spain.
| | - Rosa Maria Hernandez
- NanoBioCel Research Group, Laboratory of Pharmaceutics, School of Pharmacy, University of the Basque Country (UPV-EHU), Vitoria-Gasteiz, Spain; Bioaraba, NanoBioCel Research Group, Vitoria-Gasteiz, Spain; Biomedical Research Networking Centre in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN). Institute of Health Carlos III, Madrid, Spain.
| |
Collapse
|
50
|
Wei PW, Wang X, Wang C, Chen M, Liu MZ, Liu WX, He YL, Xu GB, Zheng XH, Zhang H, Liu HM, Wang B. Ginkgo biloba L. exocarp petroleum ether extract inhibits methicillin-resistant Staphylococcus aureus by modulating ion transport, virulence, and biofilm formation in vitro and in vivo. JOURNAL OF ETHNOPHARMACOLOGY 2024; 328:117957. [PMID: 38493904 DOI: 10.1016/j.jep.2024.117957] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Revised: 02/10/2024] [Accepted: 02/19/2024] [Indexed: 03/19/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE As reported in the Ancient Chinese Medicinal Books, Ginkgo biloba L. fruit has been used as a traditional Chinese medicine for the treatment asthma and cough or as a disinfectant. Our previous study demonstrated that G. biloba exocarp extract (GBEE), an extract of a traditional Chinese herb, inhibits the formation of methicillin-resistant Staphylococcus aureus (MRSA) biofilms. However, GBEE is a crude extract that contains many components, and the underlying mechanisms of purified GBEE fractions extracted with solvents of different polarities are unknown. AIM OF THE STUDY This study aimed to investigate the different components in GBEE fractions extracted with solvents of different polarities and their antibacterial effects and mechanisms against MRSA and Staphylococcus haemolyticus biofilms both in vitro and in vivo. METHODS The components in different fractions were detected by high-performance liquid chromatography-high resolution mass spectrometry (HPLC-HRMS). Microbroth dilution assays and time growth curves were used to determine the antibacterial effects of the fractions on 15 clinical bacterial isolates. Crystal violet staining, scanning electron microscopy (SEM) and transmission electron microscopy (TEM) were utilized to identify the fractions that affected bacterial biofilm formation. The potential MRSA targets of the GBEE fraction obtained with petroleum ether (PE), denoted GBEE-PE, were screened by transcriptome sequencing, and the gene expression profile was verified by quantitative polymerase chain reaction (qPCR). RESULTS HPLC-HRMS analysis revealed that the four GBEE fractions (extracted with petroleum ether, ethyl acetate, n-butanol, and water) contained different ginkgo components, and the antibacterial effects decreased as the polarity of the extraction solvent increased. The antibacterial activity of GBEE-PE was greater than that of the GBEE fraction extracted with ethyl acetate (EA). GBEE-PE improved H. illucens survival and reduced MRSA colonization in model mouse organs. Crystal violet staining and SEM and TEM analyses revealed that GBEE-PE inhibited MRSA and S. haemolyticus biofilm formation. Transcriptional analysis revealed that GBEE-PE inhibits MRSA biofilms by altering ion transport, cell wall metabolism and virulence-related gene expression. In addition, the LO2 cell viability and H. illucens toxicity assay data showed that GBEE-PE at 20 mg/kg was nontoxic. CONCLUSION The GBEE fractions contained different components, and their antibacterial effects decreased with increases in the polarity of the extraction solvent. GBEE-PE limited MRSA growth and biofilm formation by affecting ion transport, cell wall synthesis, and virulence-related pathways. This research provides a more detailed overview of the mechanism by which GBEE-PE inhibits MRSA both in vitro and in vivo and suggests that GBEE-PE is a new prospective antimicrobial with the potential to be used in MRSA therapeutics in the future.
Collapse
Affiliation(s)
- Peng-Wei Wei
- Engineering Research Center of Health Medicine Biotechnology of Guizhou Province, Key Laboratory of Biology and Medical Engineering, School of Biology and Engineering (Modern Industry College of Health Medicine), Guizhou Medical University, Guiyang, 561113, Guizhou, China
| | - Xu Wang
- Engineering Research Center of Health Medicine Biotechnology of Guizhou Province, Key Laboratory of Biology and Medical Engineering, School of Biology and Engineering (Modern Industry College of Health Medicine), Guizhou Medical University, Guiyang, 561113, Guizhou, China
| | - Cong Wang
- The First Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Formulation (R&D) Department, Guiyang, 550001, China
| | - Ming Chen
- Engineering Research Center of Health Medicine Biotechnology of Guizhou Province, Key Laboratory of Biology and Medical Engineering, School of Biology and Engineering (Modern Industry College of Health Medicine), Guizhou Medical University, Guiyang, 561113, Guizhou, China; Key Laboratory of Environmental Pollution Monitoring and Disease Control, China Ministry of Education (School of Public Health, Guizhou Medical University), Guiyang, 561113, Guizhou, China
| | - Meng-Zhu Liu
- Engineering Research Center of Health Medicine Biotechnology of Guizhou Province, Key Laboratory of Biology and Medical Engineering, School of Biology and Engineering (Modern Industry College of Health Medicine), Guizhou Medical University, Guiyang, 561113, Guizhou, China; Key Laboratory of Environmental Pollution Monitoring and Disease Control, China Ministry of Education (School of Public Health, Guizhou Medical University), Guiyang, 561113, Guizhou, China
| | - Wen-Xia Liu
- Engineering Research Center of Health Medicine Biotechnology of Guizhou Province, Key Laboratory of Biology and Medical Engineering, School of Biology and Engineering (Modern Industry College of Health Medicine), Guizhou Medical University, Guiyang, 561113, Guizhou, China; Key Laboratory of Microbiology and Parasitology of Education Department of Guizhou, School of Basic Medical Science, Guizhou Medical University, Guiyang, 561113, Guizhou, China
| | - Yan-Ling He
- Zhejiang Hisun Pharmaceutical Co., Ltd., Taizhou, 318000, Zhejiang, China
| | - Guo-Bo Xu
- State Key Laboratory of Functions and Applications of Medicinal Plants & School of Pharmacy, Guizhou Medical University, Guian New Area, 561113, Guizhou, China.
| | - Xiao-He Zheng
- Zhejiang Hisun Pharmaceutical Co., Ltd., Taizhou, 318000, Zhejiang, China
| | - Hua Zhang
- Department of Laboratory Medicine, Guizhou Provincial People's Hospital, Affiliated Hospital of Guizhou University, Guiyang, 550002, Guizhou, China.
| | - Hong-Mei Liu
- Engineering Research Center of Health Medicine Biotechnology of Guizhou Province, Key Laboratory of Biology and Medical Engineering, School of Biology and Engineering (Modern Industry College of Health Medicine), Guizhou Medical University, Guiyang, 561113, Guizhou, China.
| | - Bing Wang
- Engineering Research Center of Health Medicine Biotechnology of Guizhou Province, Key Laboratory of Biology and Medical Engineering, School of Biology and Engineering (Modern Industry College of Health Medicine), Guizhou Medical University, Guiyang, 561113, Guizhou, China; Key Laboratory of Environmental Pollution Monitoring and Disease Control, China Ministry of Education (School of Public Health, Guizhou Medical University), Guiyang, 561113, Guizhou, China; Key Laboratory of Microbiology and Parasitology of Education Department of Guizhou, School of Basic Medical Science, Guizhou Medical University, Guiyang, 561113, Guizhou, China.
| |
Collapse
|