1
|
Tan L, Yang J, He Z, Wan Y, Li Z, Song J, Zhang W, Yang X. Inhibitory effects of extracts from Prunella vulgaris on biofilm formation of Staphylococcus aureus. Microb Pathog 2025; 205:107694. [PMID: 40355056 DOI: 10.1016/j.micpath.2025.107694] [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: 05/29/2024] [Revised: 05/07/2025] [Accepted: 05/09/2025] [Indexed: 05/14/2025]
Abstract
Staphylococcus aureus (S. aureus) is a highly prevalent pathogen capable of strongly adhering to food processing equipment and the contact surfaces, where it forms resilient biofilms that are difficult to eliminate. Prunella vulgaris (P. vulgaris), a traditional Chinese herbal medicine, has demonstrated strong potential in inhibiting S. aureus biofilm formation. This study investigated the inhibitory mechanisms of P. vulgaris extracts against S. aureus growth and biofilm formation, evaluating the biofilm inhibitory concentration, bactericidal concentration and their effects on ica operon gene expression. The P. vulgaris extracts exhibited a minimum inhibitory concentration (MIC) and minimum bactericidal concentration (MBC) of 1.25 mg/mL. At the MIC level, the extracts not only suppressed S. aureus growth and metabolic viability but also inhibited polysaccharide intercellular adhesion (PIA), prevented biofilm formation and disrupted mature biofilms. Furthermore, P. vulgaris extracts demonstrated concentration-dependent effects on extracellular polymeric substances (EPS) production: while 1/2 MIC concentrations stimulated EPS synthesis, double-MIC concentrations markedly suppressed it. Notably, the extracts consistently downregulated icaA and icaD expression at both MIC and 2 × MIC concentrations. Therefore, P. vulgaris exhibits significant potential against S. aureus-induced foodborne diseases, demonstrating promise as a novel antibacterial agent for future applications in both pharmaceutical development and food safety enhancement.
Collapse
Affiliation(s)
- Luyi Tan
- Food Safety and Health Research Center, NMPA Key Laboratory for Safety Evaluation of Cosmetics, Guangdong-Hongkong-Macao Joint Laboratory for Contaminants Exposure and Health, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, 510515, PR China; Department of Public Health and Preventive Medicine, School of Medicine, Jinan University, Guangzhou, Guangdong, 510632, PR China
| | - Jiani Yang
- Food Safety and Health Research Center, NMPA Key Laboratory for Safety Evaluation of Cosmetics, Guangdong-Hongkong-Macao Joint Laboratory for Contaminants Exposure and Health, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, 510515, PR China; Department of Public Health and Preventive Medicine, School of Medicine, Jinan University, Guangzhou, Guangdong, 510632, PR China
| | - Zhini He
- Food Safety and Health Research Center, NMPA Key Laboratory for Safety Evaluation of Cosmetics, Guangdong-Hongkong-Macao Joint Laboratory for Contaminants Exposure and Health, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, 510515, PR China
| | - Yu Wan
- Food Safety and Health Research Center, NMPA Key Laboratory for Safety Evaluation of Cosmetics, Guangdong-Hongkong-Macao Joint Laboratory for Contaminants Exposure and Health, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, 510515, PR China
| | - Ziyin Li
- Food Safety and Health Research Center, NMPA Key Laboratory for Safety Evaluation of Cosmetics, Guangdong-Hongkong-Macao Joint Laboratory for Contaminants Exposure and Health, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, 510515, PR China
| | - Jia Song
- Food Safety and Health Research Center, NMPA Key Laboratory for Safety Evaluation of Cosmetics, Guangdong-Hongkong-Macao Joint Laboratory for Contaminants Exposure and Health, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, 510515, PR China
| | - Wenjuan Zhang
- Department of Public Health and Preventive Medicine, School of Medicine, Jinan University, Guangzhou, Guangdong, 510632, PR China.
| | - Xingfen Yang
- Food Safety and Health Research Center, NMPA Key Laboratory for Safety Evaluation of Cosmetics, Guangdong-Hongkong-Macao Joint Laboratory for Contaminants Exposure and Health, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, 510515, PR China.
| |
Collapse
|
2
|
Vatansever C, Aksoy N, Adaklı Aksoy B, Fışgın T. Evaluating the Antibiofilm Effects of Antibiotics on Staphylococcus Species from Pediatric Hematology-Oncology Patients. Microb Drug Resist 2025. [PMID: 40340597 DOI: 10.1089/mdr.2025.0006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/10/2025] Open
Abstract
Biofilms are microbial communities and occur on different medical devices such as catheters. The formation of bacterial biofilms on medical devices leads to indwelling medical device-related infections. Since biofilm bacteria are more resistant to antibiotics than planktonic bacteria, using these antibiotics in indwelling medical device-related infections causes recurrence of infections, treatment failure, and death. Minimum inhibitory concentration (MIC) is an important reference in treating acute infections caused by planktonic bacteria. However, MIC is ineffective in indwelling medical device-related infections caused by biofilm bacteria. The study aims to demonstrate the necessity and development of effective and standard methods such as minimum biofilm prevention concentration, minimum biofilm inhibitory concentration, and minimum biofilm eradication concentration in the case of indwelling medical device-related infection. The study was conducted with 10 isolates of Staphylococcus species from patients who developed infections in the Pediatric Hematology-Oncology Department at Medical Park Bahcelievler Hospital. According to the study results, even if planktonic bacteria are sensitive to antibiotics, they can become resistant to this antibiotic when they are in a biofilm (p < 0.05, Crosstab). Also, inhibiting the growth of planktonic bacteria does not prevent biofilm formation. The study additionally revealed that inhibiting and eradicating biofilm is more difficult than preventing biofilm formation (p < 0.05).
Collapse
Affiliation(s)
- Cansu Vatansever
- Department of Pharmaceutical Microbiology, School of Pharmacy, Altınbaş University, Istanbul, Turkey
| | - Nilay Aksoy
- Department of Clinical Pharmacy, School of Pharmacy, Altınbaş University, Istanbul, Turkey
| | - Başak Adaklı Aksoy
- Department of Pediatric Hematology-Oncology and Pediatric Bone Marrow Transplantation, Medical Park Bahcelievler Hospital, Altınbaş University, Istanbul, Turkey
- Department of Child Health and Diseases, School of Medicine, Altınbaş University, Istanbul, Turkey
| | - Tunç Fışgın
- Department of Pediatric Hematology-Oncology and Pediatric Bone Marrow Transplantation, Medical Park Bahcelievler Hospital, Altınbaş University, Istanbul, Turkey
- Department of Child Health and Diseases, School of Medicine, Altınbaş University, Istanbul, Turkey
| |
Collapse
|
3
|
Park S, Lee G, Yoon KJ, Yoo K. Elucidating airborne bacterial communities and their potential pathogenic risks in urban metro environments. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2025; 292:117936. [PMID: 39987686 DOI: 10.1016/j.ecoenv.2025.117936] [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: 11/24/2024] [Revised: 02/10/2025] [Accepted: 02/17/2025] [Indexed: 02/25/2025]
Abstract
Metros are the predominant mode of transportation for urban residents. Because of high passenger volume and pollutant concentrations, concern is growing regarding the potential health hazards of exposure to potential pathogenic airborne bacteria in metros. However, the risks of airborne bacterial communities in metros have not been assessed. Therefore, this study was conducted to explore the airborne bacterial communities and potential pathogenic risk of bacteria in the inner metro train (IM) and metro stations (MS) in Busan, South Korea. The concentrations of culturable total airborne bacteria (CABs) and culturable total airborne Staphylococcus (CAS) were higher in the MS samples than in the IM samples. Bacterial community analysis revealed that although the overall metro environment was dominated by human-associated bacteria, such as Corynebacterium and Staphylococcus genera, the IM and MS samples exhibited significantly distinct core bacterial taxa despite their similar bacterial communities; this is a result of human activity rather than the presence of passengers. Through multilocus sequence typing (MLST), the isolated S. epidermidis from both the IM and MS samples was identified as a human pathogen with four sequence types (ST190, ST54, ST992, and ST817). Furthermore, the MLST results were significantly positively correlated with the CABs and CASs in both the IM and MS samples. The S. aureus infection pathway was predicted in all samples using PICRUSt2 and was significantly higher in the IM samples than in the MS samples. The findings of this study can serve as a reference for developing microbial public health provisions for metro systems.
Collapse
Affiliation(s)
- Sena Park
- Department of Environmental Engineering, National Korea Maritime and Ocean University, Busan 49112, Republic of Korea; Interdisciplinary Major of Ocean Renewable Energy Engineering, National Korea Maritime and Ocean University, Busan 49112, Republic of Korea
| | - Gihan Lee
- Department of Environmental Engineering, National Korea Maritime and Ocean University, Busan 49112, Republic of Korea; Interdisciplinary Major of Ocean Renewable Energy Engineering, National Korea Maritime and Ocean University, Busan 49112, Republic of Korea
| | - Keum Ju Yoon
- Department of Environmental Engineering, National Korea Maritime and Ocean University, Busan 49112, Republic of Korea
| | - Keunje Yoo
- Department of Environmental Engineering, National Korea Maritime and Ocean University, Busan 49112, Republic of Korea; Interdisciplinary Major of Ocean Renewable Energy Engineering, National Korea Maritime and Ocean University, Busan 49112, Republic of Korea.
| |
Collapse
|
4
|
Nugrahani NA, Nurilyana MM, Faizal IA, Kholifa M, Hafizi I. Efficacy of avocado seed extract in preventing, inhibiting, and eliminating Prevotella intermedia biofilms: An in vitro study. Vet World 2025; 18:408-418. [PMID: 40182820 PMCID: PMC11963592 DOI: 10.14202/vetworld.2025.408-418] [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: 11/19/2024] [Accepted: 01/16/2025] [Indexed: 04/05/2025] Open
Abstract
Background and Aim Prevotella intermedia is a significant contributor to periodontitis, capable of forming biofilms that resist antibiotics and complicate treatment. Avocado seeds (Persea americana Mill.) are rich in bioactive compounds, including flavonoids, tannins, saponins, and alkaloids, which exhibit potential antibiofilm activity. This study aims to evaluate the efficacy of avocado seed ethanol extract in preventing biofilm attachment, inhibiting biofilm formation, and eradicating established biofilms of P. intermedia in vitro. Materials and Methods A post-test-only control group design was employed using P. intermedia (ATCC 25611). Ten groups were included: Bacterial and negative controls, a positive control (chlorhexidine), and experimental groups with ethanol extract concentrations (3.25%-9.25%). Biofilm activity was assessed using 96-well microtiter plates, crystal violet staining, and optical density measurements at 595 nm to determine the minimum biofilm prevention (MBPC), inhibition (MBIC), and eradication concentrations (MBEC). Statistical analysis was conducted using one-way ANOVA and Bonferroni post hoc tests. Results Biofilm assays showed a dose-dependent increase in antibiofilm efficacy. The highest attachment prevention (82.67%), biofilm formation inhibition (84.26%), and biofilm eradication (86.04%) were observed at 9.25%. Significant differences (p < 0.05) were found between the extract and negative control groups, with no significant differences (p > 0.05) between the 8.25%-9.25% extracts and chlorhexidine. The MBPC50, MBIC50, and MBEC50 were identified at a concentration of 6.25%, achieving >50% efficacy in biofilm prevention, inhibition, and eradication. Conclusion Avocado seed ethanol extract demonstrated significant antibiofilm properties against P. intermedia, comparable to chlorhexidine at higher concentrations. The bioactive compounds - flavonoids, tannins, saponins, and alkaloids - likely contributed to these effects through mechanisms such as quorum sensing inhibition, disruption of bacterial adhesion, and destabilization of biofilm structures. These findings highlight avocado seed extract as a promising natural alternative for managing periodontitis-related biofilm infections.
Collapse
Affiliation(s)
- Nur Ariska Nugrahani
- Department of Oral Biology, Faculty of Dentistry, Muhammadiyah University of Surakarta, 57141, Surakarta, Indonesia
| | - Maulita Misi Nurilyana
- Department of Oral Biology, Faculty of Dentistry, Muhammadiyah University of Surakarta, 57141, Surakarta, Indonesia
| | - Imam Agus Faizal
- Department of Applied Bachelor's Degree of Medical Laboratory Technology, Faculty of Pharmacy, Science, and Technology, Al-Irsyad University of Cilacap, 53223, Cilacap, Indonesia
| | - Mahmud Kholifa
- Department of Oral Biology, Faculty of Dentistry, Muhammadiyah University of Surakarta, 57141, Surakarta, Indonesia
| | - Ikmal Hafizi
- Department of Orthodontics Dentistry, Faculty of Dentistry, Muhammadiyah University of Surakarta, 57141, Surakarta, Indonesia
| |
Collapse
|
5
|
Roque‐Borda CA, Primo LMDG, Medina‐Alarcón KP, Campos IC, Nascimento CDF, Saraiva MMS, Berchieri Junior A, Fusco‐Almeida AM, Mendes‐Giannini MJS, Perdigão J, Pavan FR, Albericio F. Antimicrobial Peptides: A Promising Alternative to Conventional Antimicrobials for Combating Polymicrobial Biofilms. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2410893. [PMID: 39530703 PMCID: PMC11714181 DOI: 10.1002/advs.202410893] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/06/2024] [Revised: 10/23/2024] [Indexed: 11/16/2024]
Abstract
Polymicrobial biofilms adhere to surfaces and enhance pathogen resistance to conventional treatments, significantly contributing to chronic infections in the respiratory tract, oral cavity, chronic wounds, and on medical devices. This review examines antimicrobial peptides (AMPs) as a promising alternative to traditional antibiotics for treating biofilm-associated infections. AMPs, which can be produced as part of the innate immune response or synthesized therapeutically, have broad-spectrum antimicrobial activity, often disrupting microbial cell membranes and causing cell death. Many specifically target negatively charged bacterial membranes, unlike host cell membranes. Research shows AMPs effectively inhibit and disrupt polymicrobial biofilms and can enhance conventional antibiotics' efficacy. Preclinical and clinical research is advancing, with animal studies and clinical trials showing promise against multidrug-resistant bacteria and fungi. Numerous patents indicate increasing interest in AMPs. However, challenges such as peptide stability, potential cytotoxicity, and high production costs must be addressed. Ongoing research focuses on optimizing AMP structures, enhancing stability, and developing cost-effective production methods. In summary, AMPs offer a novel approach to combating biofilm-associated infections, with their unique mechanisms and synergistic potential with existing antibiotics positioning them as promising candidates for future treatments.
Collapse
Affiliation(s)
- Cesar Augusto Roque‐Borda
- Department of Biological SciencesSchool of Pharmaceutical SciencesUniversidade Estadual Paulista (UNESP)AraraquaraSao Paulo14800‐903Brazil
- iMed.ULisboa–Institute for Medicines ResearchFaculty of PharmacyUniversity of LisbonLisbon1649004Portugal
- Vicerrectorado de InvestigaciónUniversidad Católica de Santa MaríaArequipa04000Peru
| | - Laura Maria Duran Gleriani Primo
- Department of Biological SciencesSchool of Pharmaceutical SciencesUniversidade Estadual Paulista (UNESP)AraraquaraSao Paulo14800‐903Brazil
| | - Kaila Petronila Medina‐Alarcón
- Department of Clinical AnalysisSchool of Pharmaceutical SciencesUniversidade Estadual Paulista (UNESP)AraraquaraSao Paulo14800‐903Brazil
| | - Isabella C. Campos
- São Paulo State University (UNESP)School of Agricultural and Veterinarian SciencesJaboticabalSao Paulo14884‐900Brazil
| | - Camila de Fátima Nascimento
- São Paulo State University (UNESP)School of Agricultural and Veterinarian SciencesJaboticabalSao Paulo14884‐900Brazil
| | - Mauro M. S. Saraiva
- São Paulo State University (UNESP)School of Agricultural and Veterinarian SciencesJaboticabalSao Paulo14884‐900Brazil
| | - Angelo Berchieri Junior
- São Paulo State University (UNESP)School of Agricultural and Veterinarian SciencesJaboticabalSao Paulo14884‐900Brazil
| | - Ana Marisa Fusco‐Almeida
- Department of Clinical AnalysisSchool of Pharmaceutical SciencesUniversidade Estadual Paulista (UNESP)AraraquaraSao Paulo14800‐903Brazil
| | - Maria José Soares Mendes‐Giannini
- Department of Clinical AnalysisSchool of Pharmaceutical SciencesUniversidade Estadual Paulista (UNESP)AraraquaraSao Paulo14800‐903Brazil
| | - João Perdigão
- iMed.ULisboa–Institute for Medicines ResearchFaculty of PharmacyUniversity of LisbonLisbon1649004Portugal
| | - Fernando Rogério Pavan
- Department of Biological SciencesSchool of Pharmaceutical SciencesUniversidade Estadual Paulista (UNESP)AraraquaraSao Paulo14800‐903Brazil
| | - Fernando Albericio
- Peptide Science LaboratorySchool of Chemistry and PhysicsUniversity of KwaZulu‐NatalDurban4001South Africa
- CIBER‐BBNNetworking Centre on BioengineeringBiomaterials and Nanomedicineand Department of Organic ChemistryUniversity of BarcelonaBarcelona08028Spain
| |
Collapse
|
6
|
Lee SM, Song JH, Lee KS, Yoo KH. Pathogenic Bacterial Detection Using Vertical-Capacitance Sensor Array Immobilized with the Antimicrobial Peptide Melittin. SENSORS (BASEL, SWITZERLAND) 2024; 25:12. [PMID: 39796801 PMCID: PMC11722802 DOI: 10.3390/s25010012] [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: 10/21/2024] [Revised: 12/18/2024] [Accepted: 12/20/2024] [Indexed: 01/13/2025]
Abstract
The rapid and reliable detection of pathogenic bacteria remains a significant challenge in clinical microbiology. Consequently, the demand for simple and rapid techniques, such as antimicrobial peptide (AMP)-based sensors, has recently increased as an alternative to traditional methods. Melittin, a broad-spectrum AMP, rapidly associates with the cell membranes of various gram-positive and gram-negative bacteria. It also inhibits bacterial biofilm formation in blood culture media. In our study, bacterial growth was measured using electrical vertical-capacitance sensors with interdigitated electrodes functionalized with melittin, a widely studied AMP. The melittin-immobilized vertical-capacitance sensors demonstrated real-time detection of both standard and clinically isolated bacteria in media. Furthermore, these sensors successfully detected clinically isolated bacteria in blood culture media while inhibiting bacterial biofilm formation. Melittin-immobilized vertical-capacitance sensors provide a rapid and sensitive pathogen detection platform, with significant potential for improving patient care.
Collapse
Affiliation(s)
- Sun-Mi Lee
- Department of Physics, Yonsei University, Seoul 03722, Republic of Korea; (J.-H.S.)
- Nanomedical Graduate Program, Yonsei University, Seoul 03722, Republic of Korea
- Division of Rheumatology, Department of Internal Medicine, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
| | - Jun-Ho Song
- Department of Physics, Yonsei University, Seoul 03722, Republic of Korea; (J.-H.S.)
| | - Kyo-Seok Lee
- Department of Physics, Yonsei University, Seoul 03722, Republic of Korea; (J.-H.S.)
| | - Kyung-Hwa Yoo
- Department of Physics, Yonsei University, Seoul 03722, Republic of Korea; (J.-H.S.)
- Nanomedical Graduate Program, Yonsei University, Seoul 03722, Republic of Korea
| |
Collapse
|
7
|
Nazari M, Taheri M, Nouri F, Bahmanzadeh M, Alikhani MY. The antimicrobial and antibiofilm effects of gentamicin, imipenem, and fucoidan combinations against dual-species biofilms of Staphylococcus aureus and Acinetobacter baumannii isolated from diabetic foot ulcers. Ann Clin Microbiol Antimicrob 2024; 23:101. [PMID: 39548455 PMCID: PMC11568526 DOI: 10.1186/s12941-024-00760-w] [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: 08/19/2024] [Accepted: 11/05/2024] [Indexed: 11/18/2024] Open
Abstract
INTRODUCTION Diabetes mellitus is a chronic metabolic disorder characterized by persistent hyperglycemia due to impaired insulin production or utilization, leading to severe health complications. Diabetic foot ulcers (DFUs) represent a major complication, often exacerbated by polymicrobial infections involving Staphylococcus aureus and Acinetobacter baumannii. These pathogens, notorious for their resistance to antibiotics, complicate treatment efforts, especially due to biofilm formation, which enhances bacterial survival and resistance. This study explores the synergistic effects of combining gentamicin, imipenem, and fucoidan, a sulfated polysaccharide with antimicrobial properties, against both planktonic and biofilm forms of S. aureus and A. baumannii. METHODS Isolates of S. aureus and A. baumannii were collected from DFUs and genetically confirmed. Methicillin resistance in S. aureus was identified through disk diffusion and PCR. Biofilm formation, including dual-species biofilms, was analyzed using the microtiter plate method. The antimicrobial efficacy of gentamicin, imipenem, and fucoidan was assessed by determining the minimum inhibitory concentration (MIC), minimum bactericidal concentration (MBC), minimum biofilm inhibitory concentration (MBIC), and minimum biofilm eradication concentration (MBEC). Synergistic interactions were evaluated using the fractional inhibitory concentration index (FICi) and fractional bactericidal concentration index (FBCi). The expression of biofilm-associated genes (icaA in S. aureus and bap in A. baumannii) was analyzed, and the cytotoxicity of fucoidan was assessed. RESULTS The study revealed that 77.4% of S. aureus and all A. baumannii isolates showed multidrug resistance. Among 837 tested conditions for dual-species biofilm formation, 72 resulted in strong biofilm formation and 67 in moderate biofilm formation. The geometric mean MIC values for gentamicin were 12.2 µg/mL for S. aureus, 22.62 µg/mL for A. baumannii, and 5.87 µg/mL for their co-culture; for imipenem, they were 19.84, 9.18, and 3.70 µg/mL, respectively, and for fucoidan, 48.50, 31.20, and 19.65 µg/mL, respectively. The MBC values for gentamicin were 119.42, 128, and 11.75 µg/mL; for imipenem, they were 48.50, 14.92, and 8 µg/mL; and for fucoidan, they were 88.37, 62.62, and 42.48 µg/mL. The MBIC values were 55.71, 119.42, and 18.66 µg/mL for gentamicin; 68.59, 48.50, and 25.39 µg/mL for imipenem; and 153.89, 101.49, and 53.53 µg/mL for fucoidan. The MBEC values were 315.17, 362.03, and 59.25 µg/mL for gentamicin; 207.93, 157.58, and 74.65 µg/mL for imipenem; and 353.55, 189.46, and 99.19 µg/mL for fucoidan. When cultured in planktonic form, the geometric mean FICi and FBCi values indicated additive effects, while co-culture showed FICi values of ≤ 0.5, suggesting a synergistic interaction. Treatment with gentamicin and fucoidan led to significant downregulation of the icaA and bap genes in both single-species and dual-species biofilms of S. aureus and A. baumannii. The reductions in gene expression were more pronounced in dual-species biofilms compared to single-species biofilms. Additionally, treatment with imipenem and fucoidan also resulted in significant downregulation of these genes in both biofilm types. Cytotoxicity assessments indicated that higher concentrations of fucoidan were toxic, yet no harmful effects were noted at the optimal synergistic concentrations used with antibiotics. CONCLUSION In our investigation, we found that combining gentamicin, imipenem, and fucoidan had a synergistic effect on dual-species biofilms of S. aureus and A. baumannii, suggesting potential benefits for treating such infections effectively. This underscores the importance of understanding microbial interactions, antibiotic susceptibility, and biofilm formation in DFUs.
Collapse
Affiliation(s)
- Mohsen Nazari
- Department of Microbiology, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Mohammad Taheri
- Department of Microbiology, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Fatemeh Nouri
- Department of Pharmaceutical Biotechnology, School of Pharmacy, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Maryam Bahmanzadeh
- Department of Anatomical Sciences, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
- Fertility and Infertility Research Center, Avicenna Institute of Clinical Sciences, Avicenna Health Research Institute , Hamadan University of Medical Sciences, Hamadan, Iran
| | - Mohammad Yousef Alikhani
- Department of Microbiology, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran.
- Infectious Disease Research Center, Avicenna Institute of Clinical Sciences, Avicenna Health Research Institute , Hamadan University of Medical Sciences, P.O. Box: 6517838678, Hamadan, Iran.
| |
Collapse
|
8
|
Chegini Z, Shariati A, Alikhani MY, Safaiee M, Rajaeih S, Arabestani M, Azizi M. Antibacterial and antibiofilm activity of silver nanoparticles stabilized with C-phycocyanin against drug-resistant Pseudomonas aeruginosa and Staphylococcus aureus. Front Bioeng Biotechnol 2024; 12:1455385. [PMID: 39524122 PMCID: PMC11544008 DOI: 10.3389/fbioe.2024.1455385] [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: 06/26/2024] [Accepted: 10/07/2024] [Indexed: 11/16/2024] Open
Abstract
Background Biofilms are bacterial communities that can protect them against external factors, including antibiotics. In this study, silver nanoparticles (AgNPs) were formed by modifying AgNPs with C-phycocyanin (Ag-Pc) to inhibit the growth of carbapenem-resistant Pseudomonas aeruginosa (CR P. aeruginosa) and methicillin-resistant Staphylococcus aureus (MRSA) and destroy biofilm of these bacteria. Methods The AgNPs were prepared with the green synthesis method, and Pc was used to stabilize the AgNPs. The Ag-Pc's antibacterial and antibiofilm effects were evaluated using the Microbroth dilution method and microtiter plate assay. The inhibitory effect of Ag-Pc on the expression of biofilm-related genes was evaluated by real-time PCR. Moreover, the MTT assay was used to assess the Ag-Pc toxicity. Results The Ag-Pc minimum inhibitory concentration (MIC) was 7.4 μg/mL for CR P. aeruginosa and MRSA. Pc did not show antibacterial effects against any of the strains. Ag-Pc suppressed biofilm formation and destroyed matured biofilm in both bacteria more efficiently than the AgNPs (P< 0.05). The expression of all genes was not significantly reduced in the presence of synthesized nanoparticles. Finally, the MTT assay results did not show toxicity against a murine fibroblast cell line (L929) at MIC concentration. Conclusion The present study showed the promising potential of Pc for improving the antibacterial and antibiofilm function of AgNPs and inhibiting drug-resistant bacteria. Therefore, Ag-Pc nanoparticles can be considered a promising therapeutic approach for the managing of the bacterial biofilm.
Collapse
Affiliation(s)
- Zahra Chegini
- Department of Microbiology, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Aref Shariati
- Infectious Diseases Research Center (IDRC), Arak University of medical sciences, Arak, Iran
| | - Mohammad Yousef Alikhani
- Department of Microbiology, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Maliheh Safaiee
- Department of Organic Chemistry, Faculty of Chemistry and Petroleum Sciences, Bu-Ali Sina University, Hamedan, Iran
| | - Shahin Rajaeih
- ENT and Head and Neck Research Center and Department, The Five Senses Health Institute, Iran University of Medical Sciences, Tehran, Iran
| | - Mohammadreza Arabestani
- Department of Microbiology, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
- Infectious Diseases Research Centre, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Mehdi Azizi
- Department of Tissue Engineering and Regenerative Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| |
Collapse
|
9
|
Chan YL, Tang SN, Osman CP, Chee CF, Tay ST. Exploring naphthoquinone and anthraquinone derivatives as antibiotic adjuvants against Staphylococcus aureus biofilms: Synergistic effects of menadione. Microb Pathog 2024; 195:106886. [PMID: 39182855 DOI: 10.1016/j.micpath.2024.106886] [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: 06/26/2024] [Revised: 08/11/2024] [Accepted: 08/23/2024] [Indexed: 08/27/2024]
Abstract
Given the ability of Staphylococcus aureus to form biofilms and produce persister cells, making infections difficult to treat with antibiotics alone, there is a pressing need for an effective antibiotic adjuvant to address this public health threat. In this study, a series of quinone derivatives were evaluated for their antimicrobial and antibiofilm activities against methicillin-susceptible and methicillin-resistant S. aureus reference strains. Following analyses using broth microdilution, growth curve analysis, checkerboard assay, time-kill experiments, and confocal laser scanning microscopy, menadione was identified as a hit compound. Menadione exhibited a notable antibacterial profile (minimum inhibitory concentration, MIC = 4-16 μg/ml; minimum bactericidal concentration, MBC = 256 μg/ml) against planktonic S. aureus and its biofilms (minimum biofilm inhibitory concentration, MBIC50 = 0.0625-0.25 μg/ml). When combined with oxacillin, erythromycin, and vancomycin, menadione exhibited a synergistic or additive effect against planktonic cells and biofilms of two S. aureus reference strains and six clinical isolates, highlighting its potential as a suitable adjuvant for further development against S. aureus biofilm-associated infections.
Collapse
Affiliation(s)
- Yun Li Chan
- Department of Medical Microbiology, Faculty of Medicine, Universiti Malaya, 50603, Kuala Lumpur, Malaysia
| | - Soo Nee Tang
- Department of Medical Microbiology, Faculty of Medicine, Universiti Malaya, 50603, Kuala Lumpur, Malaysia
| | - Che Puteh Osman
- School of Chemistry and Environment, Faculty of Applied Sciences, Universiti Teknologi MARA, 40450, Shah Alam, Selangor Darul Ehsan, Malaysia
| | - Chin Fei Chee
- Nanotechnology and Catalysis Research Centre, Universiti Malaya, 50603, Kuala Lumpur, Malaysia.
| | - Sun Tee Tay
- Department of Medical Microbiology, Faculty of Medicine, Universiti Malaya, 50603, Kuala Lumpur, Malaysia.
| |
Collapse
|
10
|
El-Didamony SE, Kalaba MH, Sharaf MH, El-Fakharany EM, Osman A, Sitohy M, Sitohy B. Melittin alcalase-hydrolysate: a novel chemically characterized multifunctional bioagent; antibacterial, anti-biofilm and anticancer. Front Microbiol 2024; 15:1419917. [PMID: 39091304 PMCID: PMC11293514 DOI: 10.3389/fmicb.2024.1419917] [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: 04/19/2024] [Accepted: 06/27/2024] [Indexed: 08/04/2024] Open
Abstract
The prevalent life-threatening microbial and cancer diseases and lack of effective pharmaceutical therapies created the need for new molecules with antimicrobial and anticancer potential. Bee venom (BV) was collected from honeybee workers, and melittin (NM) was extracted from BV and analyzed by urea-polyacrylamide gel electrophoresis (urea-PAGE). The isolated melittin was hydrolyzed with alcalase into new bioactive peptides and evaluated for their antimicrobial and anticancer activity. Gel filtration chromatography fractionated melittin hydrolysate (HM) into three significant fractions (F1, F2, and F3), that were characterized by electrospray ionization mass spectrometry (ESI-MS) and evaluated for their antimicrobial, anti-biofilm, antitumor, and anti-migration activities. All the tested peptides showed antimicrobial and anti-biofilm activities against Gram-positive and Gram-negative bacteria. Melittin and its fractions significantly inhibited the proliferation of two types of cancer cells (Huh-7 and HCT 116). Yet, melittin and its fractions did not affect the viability of normal human lung Wi-38 cells. The IC50 and selectivity index data evidenced the superiority of melittin peptide fractions over intact melittin. Melittin enzymatic hydrolysate is a promising novel product with high potential as an antibacterial and anticancer agent.
Collapse
Affiliation(s)
- Samia E. El-Didamony
- Department of Zoology and Entomology, Faculty of Science, Al-Azhar University (Girls), Nasr City, Egypt
| | - Mohamed H. Kalaba
- Department of Botany and Microbiology, Faculty of Science, Al-Azhar University (Boys), Cairo, Egypt
| | - Mohamed H. Sharaf
- Department of Botany and Microbiology, Faculty of Science, Al-Azhar University (Boys), Cairo, Egypt
| | - Esmail M. El-Fakharany
- Protein Research Department, Genetic Engineering and Biotechnology Research Institute (GEBRI), City of Scientific Research and Technological Applications (SRTA-City), New Borg Al-Arab City, Alexandria, Egypt
- Pharmaceutical and Fermentation Industries Development Center, City of Scientific Research and Technological Applications (SRTA-City), New Borg Al-Arab City, Alexandria, Egypt
- Pharos University in Alexandria, Alexandria, Egypt
| | - Ali Osman
- Department of Biochemistry, Faculty of Agriculture, Zagazig University, Zagazig, Egypt
- Department of Clinical Microbiology, Infection, and Immunology, Umeå University, Umeå, Sweden
- Department of Diagnostics and Intervention, Oncology, Umeå University, Umeå, Sweden
| | - Mahmoud Sitohy
- Department of Biochemistry, Faculty of Agriculture, Zagazig University, Zagazig, Egypt
| | - Basel Sitohy
- Department of Clinical Microbiology, Infection, and Immunology, Umeå University, Umeå, Sweden
- Department of Diagnostics and Intervention, Oncology, Umeå University, Umeå, Sweden
| |
Collapse
|
11
|
Hemmati J, Chiani M, Asghari B, Roshanaei G, Soleimani Asl S, Shafiei M, Arabestani MR. Antibacterial and antibiofilm potentials of vancomycin-loaded niosomal drug delivery system against methicillin-resistant Staphylococcus aureus (MRSA) infections. BMC Biotechnol 2024; 24:47. [PMID: 38978013 PMCID: PMC11229259 DOI: 10.1186/s12896-024-00874-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2024] [Accepted: 07/01/2024] [Indexed: 07/10/2024] Open
Abstract
The threat of methicillin-resistant Staphylococcus aureus (MRSA) is increasing worldwide, making it significantly necessary to discover a novel way of dealing with related infections. The quick spread of MRSA isolates among infected individuals has heightened public health concerns and significantly limited treatment options. Vancomycin (VAN) can be applied to treat severe MRSA infections, and the indiscriminate administration of this antimicrobial agent has caused several concerns in medical settings. Owing to several advantageous characteristics, a niosomal drug delivery system may increase the potential of loaded antimicrobial agents. This work aims to examine the antibacterial and anti-biofilm properties of VAN-niosome against MRSA clinical isolates with emphasis on cytotoxicity and stability studies. Furthermore, we aim to suggest an effective approach against MRSA infections by investigating the inhibitory effect of formulated niosome on the expression of the biofilm-associated gene (icaR). The thin-film hydration approach was used to prepare the niosome (Tween 60, Span 60, and cholesterol), and field emission scanning electron microscopy (FE-SEM), an in vitro drug release, dynamic light scattering (DLS), and entrapment efficiency (EE%) were used to investigate the physicochemical properties. The physical stability of VAN-niosome, including hydrodynamic size, polydispersity index (PDI), and EE%, was analyzed for a 30-day storage time at 4 °C and 25 °C. In addition, the human foreskin fibroblast (HFF) cell line was used to evaluate the cytotoxic effect of synthesized niosome. Moreover, minimum inhibitory and bactericidal concentrations (MICs/MBCs) were applied to assess the antibacterial properties of niosomal VAN formulation. Also, the antibiofilm potential of VAN-niosome was investigated by microtiter plate (MTP) and real-time PCR methods. The FE-SEM result revealed that synthesized VAN-niosome had a spherical morphology. The hydrodynamic size and PDI of VAN-niosome reported by the DLS method were 201.2 nm and 0.301, respectively. Also, the surface zeta charge of the prepared niosome was - 35.4 mV, and the EE% ranged between 58.9 and 62.5%. Moreover, in vitro release study revealed a sustained-release profile for synthesized niosomal formulation. Our study showed that VAN-niosome had acceptable stability during a 30-day storage time. Additionally, the VAN-niosome had stronger antibacterial and anti-biofilm properties against MRSA clinical isolates compared with free VAN. In conclusion, the result of our study demonstrated that niosomal VAN could be promising as a successful drug delivery system due to sustained drug release, negligible toxicity, and high encapsulation capacity. Also, the antibacterial and anti-biofilm studies showed the high capacity of VAN-niosome against MRSA clinical isolates. Furthermore, the results of real-time PCR exhibited that VAN-niosome could be proposed as a powerful strategy against MRSA biofilm via down-regulation of icaR gene expression.
Collapse
Affiliation(s)
- Jaber Hemmati
- Department of Microbiology, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
- Department of Bacteriology, Pasteur Institute of Iran, Tehran, Iran
| | - Mohsen Chiani
- Department of NanoBiotechnology, Pasteur Institute of Iran, Tehran, Iran
| | - Babak Asghari
- Department of Microbiology, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Ghodratollah Roshanaei
- Department of Biostatistics, School of Public Health, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Sara Soleimani Asl
- Department of Anatomy, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Morvarid Shafiei
- Department of Bacteriology, Pasteur Institute of Iran, Tehran, Iran.
| | - Mohammad Reza Arabestani
- Department of Microbiology, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran.
- Infectious Disease Research Center, Hamadan University of Medical Sciences, Hamadan, Iran.
| |
Collapse
|
12
|
Govindan R, Gnanasekaran C, Govindan R, Muthuchamy M, Quero F, Jothi A, Chelliah CK, Arunachalam A, Viswanathan MR, Natesan M, Kadaikunnan S, Li WJ. Anti-quorum Sensing and Anti-biofilm Effect of Nocardiopsis synnemataformans RMN 4 (MN061002) Compound 2,6-Di-tert-butyl, 1,4-Benzoquinone Against Biofilm-Producing Bacteria. Appl Biochem Biotechnol 2024; 196:3914-3948. [PMID: 37792174 DOI: 10.1007/s12010-023-04738-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/15/2023] [Indexed: 10/05/2023]
Abstract
In this study, the anti-biofilm compound of 2,6-Di-tert-butyl, 1,4-benzoquinone was purified from Nocardiopsis synnemataformans (N. synnemataformans) RMN 4 (MN061002). To confirm the compound, various spectroscopy analyses were done including ultraviolet (UV) spectrometer, Fourier transform infrared spectroscopy (FTIR), analytical high-performance liquid chromatography (HPLC), preparative HPLC, gas chromatography-mass spectroscopy (GC-MS), liquid chromatography-mass spectroscopy (LC-MS), and 2D nuclear magnetic resonance (NMR). Furthermore, the purified compound was shown 94% inhibition against biofilm-producing Proteus mirabilis (P. mirabilis) (MN396686) at 70 µg/mL concentrations. Furthermore, the metabolic activity, exopolysaccharide damage, and hydrophobicity degradation results of identified compound exhibited excellent inhibition at 100 µg/mL concentration. Furthermore, the confocal laser scanning electron microscope (CLSM) and scanning electron microscope (SEM) results were shown with intracellular damages and architectural changes in bacteria. Consecutively, the in vivo toxicity effect of the compound against Artemia franciscana (A. franciscana) was shown to have a low mortality rate at 100 µg/mL. Finally, the molecular docking interaction between the quorum sensing (QS) genes and identified compound clearly suggested that the identified compound 2,6-Di-tert-butyl, 1,4-benzoquinone has anti-quorum sensing and anti-biofilm activities against P. mirabilis (MN396686).
Collapse
Affiliation(s)
- Rajivgandhi Govindan
- State Key Laboratory of Biocontrol, Guangdong Provincial Key Laboratory of Plant Resources and Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), School of Life Sciences, Sun Yat-Sen University, Guangzhou, 510275, People's Republic of China
- Marine Pharmacology & Toxicology Lab, Department of Marine Science, Bharathidasan University, Tiruchirappalli, 620024, Tamil Nadu, India
- Laboratorio de Nanocelulosa y Biomateriales, Departamento de Ingeniería Química, Facultad de CienciasFísicas y Matemáticas, Universidad de Chile, Biotecnología y MaterialesAvenida Beauchef 851, 8370456, Santiago, Chile
| | - Chackaravarthi Gnanasekaran
- Marine Pharmacology & Toxicology Lab, Department of Marine Science, Bharathidasan University, Tiruchirappalli, 620024, Tamil Nadu, India
| | - Ramachandran Govindan
- Marine Pharmacology & Toxicology Lab, Department of Marine Science, Bharathidasan University, Tiruchirappalli, 620024, Tamil Nadu, India.
| | - Maruthupandy Muthuchamy
- Laboratorio de Nanocelulosa y Biomateriales, Departamento de Ingeniería Química, Facultad de CienciasFísicas y Matemáticas, Universidad de Chile, Biotecnología y MaterialesAvenida Beauchef 851, 8370456, Santiago, Chile
- Lab of Toxicology, Department of Health Sciences, The Graduate School of Dong-A University, 37, Nakdong-Dearo 550 Beon-Gil, Saha-Gu, Busan, 49315, South Korea
| | - Franck Quero
- Laboratorio de Nanocelulosa y Biomateriales, Departamento de Ingeniería Química, Facultad de CienciasFísicas y Matemáticas, Universidad de Chile, Biotecnología y MaterialesAvenida Beauchef 851, 8370456, Santiago, Chile
| | - Arunachalam Jothi
- School of Chemical & Biotechnology, SASTRA Deemed University, Tamil Nadu, Tanjore, India, 401
| | - Chenthis Knaisha Chelliah
- Department of Nanotechnology, Noorul Islam Centre for Higher Education, Tamil Nadu, Kumaracoil, Kanyakumari, 629180, India
| | - Arulraj Arunachalam
- Departamento de Electricidad, Facultad de Ingeniería, Universidad Tecnológica Metropolitana (UTEM), Macul, Santiago, Chile
| | - Mangalaraja Ramalinga Viswanathan
- Faculty of Engineering and Sciences, Universidad Adolfo Ibáñez, Región Metropolitana, Diag. Las Torres 2640, 7941169, Peñalolén, Santiago, Chile
| | - Manoharan Natesan
- Marine Pharmacology & Toxicology Lab, Department of Marine Science, Bharathidasan University, Tiruchirappalli, 620024, Tamil Nadu, India
| | - Shine Kadaikunnan
- Department of Botany and Microbiology, College of Science, King Saud University, P. O. Box 2455, 11451, Riyadh, Saudi Arabia
| | - Wen-Jun Li
- State Key Laboratory of Biocontrol, Guangdong Provincial Key Laboratory of Plant Resources and Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), School of Life Sciences, Sun Yat-Sen University, Guangzhou, 510275, People's Republic of China.
- State Key Laboratory of Desert and Oasis Ecology, Xinjiang Institute of Ecology and Geography, Chinese Academy of Sciences, Urumqi, 830011, People's Republic of China.
| |
Collapse
|
13
|
Hemmati J, Chiani M, Chegini Z, Seifalian A, Arabestani MR. Surface modified niosomal quercetin with cationic lipid: an appropriate drug delivery system against Pseudomonas aeruginosa Infections. Sci Rep 2024; 14:13362. [PMID: 38862754 PMCID: PMC11167023 DOI: 10.1038/s41598-024-64416-7] [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: 03/25/2024] [Accepted: 06/08/2024] [Indexed: 06/13/2024] Open
Abstract
The Increase in infections caused by resistant strains of Pseudomonas aeruginosa poses a formidable challenge to global healthcare systems. P. aeruginosa is capable of causing severe human infections across diverse anatomical sites, presenting considerable therapeutic obstacles due to its heightened drug resistance. Niosomal drug delivery systems offer enhanced pharmaceutical potential for loaded contents due to their desirable properties, mainly providing a controlled-release profile. This study aimed to formulate an optimized niosomal drug delivery system incorporating stearylamine (SA) to augment the anti-bacterial and anti-biofilm activities of quercetin (QCT) against both standard and clinical strains of P. aeruginosa. QCT-loaded niosome (QCT-niosome) and QCT-loaded SA- niosome (QCT-SA- niosome) were synthesized by the thin-film hydration technique, and their physicochemical characteristics were evaluated by field emission scanning electron microscopy (FE-SEM), zeta potential measurement, entrapment efficacy (EE%), and in vitro release profile. The anti-P. aeruginosa activity of synthesized niosomes was assessed using minimum inhibitory and bactericidal concentrations (MICs/MBCs) and compared with free QCT. Additionally, the minimum biofilm inhibitory and eradication concentrations (MBICs/MBECs) were carried out to analyze the ability of QCT-niosome and QCT-SA-niosome against P. aeruginosa biofilms. Furthermore, the cytotoxicity assay was conducted on the L929 mouse fibroblasts cell line to evaluate the biocompatibility of the formulated niosomes. FE-SEM analysis revealed that both synthesized niosomal formulations exhibited spherical morphology with different sizes (57.4 nm for QCT-niosome and 178.9 nm for QCT-SA-niosome). The EE% for cationic and standard niosomal formulations was reported at 75.9% and 59.6%, respectively. Both formulations showed an in vitro sustained-release profile, and QCT-SA-niosome exhibited greater stability during a 4-month storage time compared to QCT-niosome. Microbial experiments indicated that both prepared formulations had higher anti-bacterial and anti-biofilm activities than free QCT. Also, the QCT-SA-niosome exhibited greater reductions in MIC, MBC, MBIC, and MBEC values compared to the QCT-niosome at equivalent concentrations. This study supports the potential of QCT-niosome and QCT-SA-niosome as effective agents against P. aeruginosa infections, manifesting significant anti-bacterial and anti-biofilm efficacy alongside biocompatibility with L929 cell lines. Furthermore, our results suggest that optimized QCT-niosome with cationic lipids could efficiently target P. aeruginosa cells with negligible cytotoxic effect.
Collapse
Affiliation(s)
- Jaber Hemmati
- Department of Microbiology, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
- Student Research Committee, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Mohsen Chiani
- Department of NanoBiotechnology, Pasteur Institute of Iran, Tehran, Iran
| | - Zahra Chegini
- Department of Microbiology, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Alexander Seifalian
- Nanotechnology & Regenerative Medicine Commercialization Centre (NanoRegMed Ltd, Nanoloom Ltd & Liberum Health Ltd), LBIC, University of London, London, UK
| | - Mohammad Reza Arabestani
- Department of Microbiology, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran.
- Infectious Disease Research Center, Hamadan University of Medical Sciences, Hamadan, Iran.
| |
Collapse
|
14
|
Mahdizade Ari M, Mirkalantari S, Darban-Sarokhalil D, Darbandi A, Razavi S, Talebi M. Investigating the antimicrobial and anti-inflammatory effects of Lactobacillus and Bifidobacterium spp. on cariogenic and periodontitis pathogens. Front Microbiol 2024; 15:1383959. [PMID: 38881669 PMCID: PMC11177620 DOI: 10.3389/fmicb.2024.1383959] [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: 02/08/2024] [Accepted: 05/03/2024] [Indexed: 06/18/2024] Open
Abstract
Background The use of probiotics is emerging as an innovative approach to managing oral health issues and mediating the immune system. The current study assessed the in vitro impacts of non-orally isolated probiotics on periodontitis and tooth decay pathogens. Methods Briefly, the persistence of probiotics in exposure to oral cavity enzymes, hydrogen peroxide, and saliva samples was examined. It was also investigated the biofilm formation and aggregation ability of probiotics, the adherence of probiotics in human gingival fibroblast cell (HGFC) lines and molar teeth samples, and the potential of probiotics to co-aggregate with oral pathogens. Additionally, the current study evaluated the effects of live probiotics on virulence gene expression, biofilm production of main oral pathogens, and changes in inflammation markers. Results The probiotics remained alive when exposed to enzymes in the oral cavity, hydrogen peroxide, and saliva at baseline, 1, 3, and 5 h after incubation at 37°C (p-value <0.05). Probiotics demonstrated to produce biofilm and aggregation, as well as adherence to HGFCs and maxillary molars (p-value >0.05). They showed significant co-aggregation with oral pathogens, which were recorded as 65.57% for B. bifidum 1001 with S. mutans, 50.06% for B. bifidum 1005 with P. gingivalis, 35.6% for L. plantarum 156 with F. nucleatum, and 18.7% for B. longum 1044 with A. actinomycetemcomitans after 8 h of incubation. A balance between pro-inflammatory and anti-inflammatory cytokines, along with inhibition of biofilm formation and changes in virulence gene transcripts, were observed. However, most of these changes were not statistically significant (p-value >0.05). Conclusion This study demonstrated the direct link between adhesiveness, aggregation, and biofilm formation with probiotic antibacterial activity. In addition to the careful selection of suitable probiotic strains, the concentration and origin of probiotic isolates should be considered.
Collapse
Affiliation(s)
- Marzie Mahdizade Ari
- Department of Microbiology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Shiva Mirkalantari
- Department of Microbiology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Davood Darban-Sarokhalil
- Department of Microbiology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Atieh Darbandi
- Department of Microbiology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Shabnam Razavi
- Department of Microbiology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
- Microbial Biotechnology Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Malihe Talebi
- Department of Microbiology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
15
|
Chen C, Shi J, Wang D, Kong P, Wang Z, Liu Y. Antimicrobial peptides as promising antibiotic adjuvants to combat drug-resistant pathogens. Crit Rev Microbiol 2024; 50:267-284. [PMID: 36890767 DOI: 10.1080/1040841x.2023.2186215] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Revised: 07/19/2022] [Accepted: 10/26/2022] [Indexed: 03/10/2023]
Abstract
The widespread antimicrobial resistance (AMR) calls for the development of new antimicrobial strategies. Antibiotic adjuvant rescues antibiotic activity and increases the life span of the antibiotics, representing a more productive, timely, and cost-effective strategy in fighting drug-resistant pathogens. Antimicrobial peptides (AMPs) from synthetic and natural sources are considered new-generation antibacterial agents. Besides their direct antimicrobial activity, growing evidence shows that some AMPs effectively enhance the activity of conventional antibiotics. The combinations of AMPs and antibiotics display an improved therapeutic effect on antibiotic-resistant bacterial infections and minimize the emergence of resistance. In this review, we discuss the value of AMPs in the age of resistance, including modes of action, limiting evolutionary resistance, and their designing strategies. We summarise the recent advances in combining AMPs and antibiotics against antibiotic-resistant pathogens, as well as their synergistic mechanisms. Lastly, we highlight the challenges and opportunities associated with the use of AMPs as potential antibiotic adjuvants. This will shed new light on the deployment of synergistic combinations to address the AMR crisis.
Collapse
Affiliation(s)
- Chen Chen
- College of Veterinary Medicine, Yangzhou University, Yangzhou, China
| | - Jingru Shi
- College of Veterinary Medicine, Yangzhou University, Yangzhou, China
| | - Dejuan Wang
- College of Veterinary Medicine, Yangzhou University, Yangzhou, China
| | - Pan Kong
- College of Veterinary Medicine, Yangzhou University, Yangzhou, China
| | - Zhiqiang Wang
- College of Veterinary Medicine, Yangzhou University, Yangzhou, China
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou, China
- Joint International Research Laboratory of Agriculture and Agri-Product Safety, Ministry of Education of China, Yangzhou University, Yangzhou, China
| | - Yuan Liu
- College of Veterinary Medicine, Yangzhou University, Yangzhou, China
- Institute of Comparative Medicine, Yangzhou University, Yangzhou, China
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou, China
- Joint International Research Laboratory of Agriculture and Agri-Product Safety, Ministry of Education of China, Yangzhou University, Yangzhou, China
| |
Collapse
|
16
|
Icriverzi M, Florian PE, Bonciu A, Dumitrescu LN, Moldovan A, Pelinescu D, Ionescu R, Avram I, Munteanu CVA, Sima LE, Dinca V, Rusen L, Roseanu A. Hybrid bio-nanoporous peptide loaded-polymer platforms with anticancer and antibacterial activities. NANOSCALE ADVANCES 2024; 6:2038-2058. [PMID: 38633049 PMCID: PMC11019497 DOI: 10.1039/d3na00947e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Accepted: 02/26/2024] [Indexed: 04/19/2024]
Abstract
In this study, hybrid bio-nanoporous peptides loaded onto poly(N-isopropylacrylamide-co-butylacrylate) (pNIPAM-co-BA) coatings were designed and obtained via matrix-assisted pulsed laser evaporation (MAPLE) technique. The incorporation of cationic peptides magainin (MG) and melittin (Mel) and their combination was tailored to target synergistic anticancer and antibacterial activities with low toxicity on normal mammalian cells. Atomic force microscopy, scanning electron microscopy, X-ray photoelectron spectroscopy, Fourier transform infrared spectroscopy as well as contact angle and surface energy measurements revealed the successful and functional incorporation of both the peptides within porous polymeric nanolayers as well as surface modifications (i.e. variation in the pore size diameter, surface roughness, and wettability) after Mel, MG or Mel-MG incorporation compared to pNIPAM-co-BA. In vitro testing revealed the impairment of biofilm formation on all the hybrid coatings while testing with S. aureus, E. coli and P. aeruginosa. Moreover, MG was shown to modulate the effect of Mel in the combined Mel-MG extract formulation released via pNIPAM-platforms, thus significantly reducing cancer cell proliferation through apoptosis/necrosis as revealed by flow cytometry analysis performed in vitro on HEK293T, A375, B16F1 and B16F10 cells. To the best of our knowledge, Mel-MG combination entrapped in the pNIPAM-co-BA copolymer has not yet been reported as a new promising candidate with anticancer and antibacterial properties for improved utility in the biomedical field. Mel-MG incorporation compared to pNIPAM-co-BA in in vitro testing revealed the impairment of biofilm formation in all the hybrid formulations.
Collapse
Affiliation(s)
- Madalina Icriverzi
- Institute of Biochemistry of the Romanian Academy 060031 Bucharest Romania
| | | | - Anca Bonciu
- National Institute for Lasers, Plasma, and Radiation Physics 409 Atomistilor Street 077125 Magurele Romania
| | | | - Antoniu Moldovan
- National Institute for Lasers, Plasma, and Radiation Physics 409 Atomistilor Street 077125 Magurele Romania
| | - Diana Pelinescu
- Faculty of Biology, University of Bucharest, Department of Genetics Intrarea Portocalelor no. 1-3, Sector 6 Bucharest Romania
| | - Robertina Ionescu
- Faculty of Biology, University of Bucharest, Department of Genetics Intrarea Portocalelor no. 1-3, Sector 6 Bucharest Romania
| | - Ionela Avram
- Faculty of Biology, University of Bucharest, Department of Genetics Intrarea Portocalelor no. 1-3, Sector 6 Bucharest Romania
| | | | - Livia Elena Sima
- Institute of Biochemistry of the Romanian Academy 060031 Bucharest Romania
| | - Valentina Dinca
- National Institute for Lasers, Plasma, and Radiation Physics 409 Atomistilor Street 077125 Magurele Romania
| | - Laurentiu Rusen
- National Institute for Lasers, Plasma, and Radiation Physics 409 Atomistilor Street 077125 Magurele Romania
| | - Anca Roseanu
- Institute of Biochemistry of the Romanian Academy 060031 Bucharest Romania
| |
Collapse
|
17
|
Jalalifar S, Razavi S, Mirzaei R, Irajian G, Pooshang Bagheri K. A hope for ineffective antibiotics to return to treatment: investigating the anti-biofilm potential of melittin alone and in combination with penicillin and oxacillin against multidrug resistant-MRSA and -VRSA. Front Microbiol 2024; 14:1269392. [PMID: 38370578 PMCID: PMC10870424 DOI: 10.3389/fmicb.2023.1269392] [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: 07/29/2023] [Accepted: 12/27/2023] [Indexed: 02/20/2024] Open
Abstract
BACKGROUND The emergence and rapid spread of multi-drug resistant (MDR) bacterial strains, such as methicillin-resistant Staphylococcus aureus (MRSA) and vancomycin-resistant S. aureus (VRSA), have posed a significant challenge to the medical community due to their ability to form biofilm and develop resistance to common antibiotics. Traditional antibiotics that were once effective in treating bacterial infections are now becoming increasingly ineffective, leading to severe consequences for patient outcomes. This concerning situation has called for urgent research to explore alternative treatment strategies. Recent studies have shown that antimicrobial peptides (AMPs) hold promise as effective agents against biofilm-associated drug-resistant infections as well as to enhance the efficacy of conventional antibiotics. Accordingly, we aimed to investigate the antimicrobial and antibiofilm effects of melittin AMP, both alone and in combination with penicillin and oxacillin, against biofilm-forming MDR-MRSA and -VRSA. METHODS In this study, we investigated the kinetics of biofilm formation and assessed various parameters related to the antimicrobial and antibiofilm efficacy of melittin and antibiotics, both alone and in combination, against MDR-MRSA and -VRSA. The antimicrobial parameters included the Minimum Inhibitory Concentration (MIC), Minimum Bactericidal Concentration (MBC), Fractional Inhibitory Concentration Index (FICi), Fractional Bactericidal Concentration Index (FBCi), and the antibiofilm activity of melittin and antibiotics indicated by the Minimum Biofilm Inhibitory Concentration (MBIC), Minimal Biofilm Eradication Concentration (MBEC), Fractional Biofilm Inhibitory Concentration Index (FBICi), and Fractional Biofilm Eradication Concentration Index (FBECi). RESULTS The MIC results showed that all S. aureus isolates were resistant to penicillin (≥0.25 μg/mL), and 66% of isolates were resistant to oxacillin. The geometric means of the MIC values for penicillin, oxacillin, and melittin were 19.02, 16, and 1.62 μg/ml, respectively, and the geometric means of the MBC values for penicillin, oxacillin, and melittin were 107.63, 49.35, and 5.45 μg/ml, respectively. The study revealed that the combination indexes of melittin-penicillin and melittin-oxacillin, as determined by FIC values against all isolates, were 0.37 and 0.03, respectively. Additionally, melittin-penicillin and melittin-oxacillin exhibited combination indexes based on FBC values against all isolates at 1.145 and 0.711, respectively. Besides, melittin inhibited the biofilm formation of all S. aureus isolates, with MBIC values ranging from 10 to 1.25 μg/mL, and MBEC values ranging from 40 to 10 μg/mL. Generally, the combination indexes of melittin-penicillin and melittin-oxacillin, determined using FBIC values against all isolates, were 0.23 and 0.177, respectively. Moreover, melittin-penicillin and melittin-oxacillin typically had combination indexes based on FBEC values against all isolates at 5 and 2.97, respectively. CONCLUSION In conclusion, our study provides evidence that melittin is effective against both planktonik and biofilm forms of MRSA and VRSA and exhibits significant synergistic effects when combined with antibiotics. These results suggest that melittin and antibiotics could be a potential candidate for further investigation for in vivo infections caused by MDR S. aureus. Furthermore, melittin has the potential to restore the efficacy of penicillin and oxacillin antibiotics in the treatment of MDR infections. Applying AMPs, like melittin, to revive beta-lactam antibiotics against MRSA and VRSA is an innovative approach against antibiotic-resistant bacteria. Further research is needed to optimize dosage and understand melittin mechanism and interactions with beta-lactam antibiotics for successful clinical applications.
Collapse
Affiliation(s)
- Saba Jalalifar
- Microbial Biotechnology Research Center, Iran University of Medical Sciences, Tehran, Iran
- Department of Microbiology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Shabnam Razavi
- Microbial Biotechnology Research Center, Iran University of Medical Sciences, Tehran, Iran
- Department of Microbiology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Rasoul Mirzaei
- Venom and Biotherapeutics Molecules Lab., Department of Medical Biotechnology, Biotechnology Research Center, Pasteur Institute of Iran, Tehran, Iran
| | - Gholamreza Irajian
- Microbial Biotechnology Research Center, Iran University of Medical Sciences, Tehran, Iran
- Department of Microbiology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Kamran Pooshang Bagheri
- Venom and Biotherapeutics Molecules Lab., Department of Medical Biotechnology, Biotechnology Research Center, Pasteur Institute of Iran, Tehran, Iran
| |
Collapse
|
18
|
Yang H, Ma R, Chen J, Xie Q, Luo W, Sun P, Liu Z, Guo J. Discovery of Melittin as Triple-Action Agent: Broad-Spectrum Antibacterial, Anti-Biofilm, and Potential Anti-Quorum Sensing Activities. Molecules 2024; 29:558. [PMID: 38338303 PMCID: PMC10856726 DOI: 10.3390/molecules29030558] [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: 11/10/2023] [Revised: 01/13/2024] [Accepted: 01/17/2024] [Indexed: 02/12/2024] Open
Abstract
The development of antibiotic-resistant microorganisms is a major global health concern. Recently, there has been an increasing interest in antimicrobial peptides as a therapeutic option. This study aimed to evaluate the triple-action (broad-spectrum antibacterial, anti-biofilm, and anti-quorum sensing activities) of melittin, a membrane-active peptide present in bee venom. The minimum inhibitory concentration and minimum bactericidal concentration of the melittin were determined using the microdilution method and agar plate counting. Growth curve analysis revealed that melittin showed a concentration-dependent antibacterial activity. Scanning electron microscope analysis revealed that melittin treatment altered the morphology. Confocal laser scanning microscope revealed that melittin increased the membrane permeability and intracellular ROS generation in bacteria, all of which contribute to bacterial cell death. In addition, the crystal violet (CV) assay was used to test the anti-biofilm activity. The CV assay demonstrated that melittin inhibited biofilm formation and eradicated mature biofilms. Biofilm formation mediated by quorum sensing (QS) plays a major role in this regard, so molecular docking and molecular dynamics analysis confirmed that melittin interacts with LasR receptors through hydrogen bonds, and further evaluates the anti-QS activity of melittin through the production of virulence factors (pyocyanin, elastase, and rhamnolipid), exopolysaccharides secretion, and bacterial motility, that may be the key to inhibiting the biofilm formation mechanism. The present findings highlight the promising role of melittin as a broad-spectrum antibacterial, anti-biofilm agent, and potential QS inhibitor, providing a new perspective and theoretical basis for the development of alternative antibiotics.
Collapse
Affiliation(s)
- Hongyan Yang
- School of Medicine, Foshan University, Foshan 528000, China (J.C.); (Q.X.)
- College of Pharmacy, Jinan University, Guangzhou 510632, China;
| | - Rong Ma
- School of Medicine, Foshan University, Foshan 528000, China (J.C.); (Q.X.)
| | - Jiarou Chen
- School of Medicine, Foshan University, Foshan 528000, China (J.C.); (Q.X.)
| | - Qian Xie
- School of Medicine, Foshan University, Foshan 528000, China (J.C.); (Q.X.)
| | - Wenhui Luo
- Guangdong Yifang Pharmaceutical Co., Ltd., Foshan 528244, China;
| | - Pinghua Sun
- College of Pharmacy, Jinan University, Guangzhou 510632, China;
| | - Zheng Liu
- School of Medicine, Foshan University, Foshan 528000, China (J.C.); (Q.X.)
| | - Jialiang Guo
- School of Medicine, Foshan University, Foshan 528000, China (J.C.); (Q.X.)
- College of Pharmacy, Jinan University, Guangzhou 510632, China;
| |
Collapse
|
19
|
Sharafi T, Ghaemi EA, Rafiee M, Ardebili A. Combination antimicrobial therapy: in vitro synergistic effect of anti-staphylococcal drug oxacillin with antimicrobial peptide nisin against Staphylococcus epidermidis clinical isolates and Staphylococcus aureus biofilms. Ann Clin Microbiol Antimicrob 2024; 23:7. [PMID: 38245727 PMCID: PMC10800071 DOI: 10.1186/s12941-024-00667-6] [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: 09/27/2023] [Accepted: 01/04/2024] [Indexed: 01/22/2024] Open
Abstract
The ability of Staphylococcus epidermidis and S. aureus to form strong biofilm on plastic devices makes them the major pathogens associated with device-related infections (DRIs). Biofilm-embedded bacteria are more resistant to antibiotics, making biofilm infections very difficult to effectively treat. Here, we evaluate the in vitro activities of anti-staphylococcal drug oxacillin and antimicrobial peptide nisin, alone and in combination, against methicillin-resistant S. epidermidis (MRSE) clinical isolates and the methicillin-resistant S. aureus ATCC 43,300. The minimum inhibitory concentrations (MIC) and minimum biofilm eradication concentrations (MBEC) of oxacillin and nisin were determined using the microbroth dilution method. The anti-biofilm activities of oxacillin and nisin, alone or in combination, were evaluated. In addition, the effects of antimicrobial agents on the expression of icaA gene were examined by quantitative real-time PCR. MIC values for oxacillin and nisin ranged 4-8 µg/mL and 64-128 µg/mL, respectively. Oxacillin and nisin reduced biofilm biomass in all bacteria in a dose-dependent manner and this inhibitory effect was enhanced with combinatorial treatment. MBEC ranges for oxacillin and nisin were 2048-8192 µg/mL and 2048-4096 µg/mL, respectively. The addition of nisin significantly decreased the oxacillin MBECs from 8- to 32-fold in all bacteria. At the 1× MIC and 1/2× MIC, both oxacillin and nisin decreased significantly the expression of icaA gene in comparison with untreated control. When two antimicrobial agents were combined at 1/2× MIC concentration, the expression of icaA were significantly lower than when were used alone. Nisin/conventional oxacillin combination showed considerable anti-biofilm effects, including inhibition of biofilm formation, eradication of mature biofilm, and down-regulation of biofilm-related genes, proposing its applications for treating or preventing staphylococcal biofilm-associated infections, including device-related infections.
Collapse
Affiliation(s)
- Toktam Sharafi
- Infectious Disease Research Center, Faculty of Medicine, Golestan University of Medical Sciences, Gorgan, Iran
- Department of Microbiology, Faculty of Medicine, Golestan University of Medical Sciences, Gorgan, Iran
| | - Ezzat Allah Ghaemi
- Infectious Disease Research Center, Faculty of Medicine, Golestan University of Medical Sciences, Gorgan, Iran
- Department of Microbiology, Faculty of Medicine, Golestan University of Medical Sciences, Gorgan, Iran
| | - Maryam Rafiee
- Infectious Disease Research Center, Faculty of Medicine, Golestan University of Medical Sciences, Gorgan, Iran
- Department of Microbiology, Faculty of Medicine, Golestan University of Medical Sciences, Gorgan, Iran
| | - Abdollah Ardebili
- Infectious Disease Research Center, Faculty of Medicine, Golestan University of Medical Sciences, Gorgan, Iran.
- Department of Microbiology, Faculty of Medicine, Golestan University of Medical Sciences, Gorgan, Iran.
| |
Collapse
|
20
|
Naman A, Tahseen H, Nawaz H, Majeed MI, Ali A, Haque A, Akbar MU, Mehmood N, Nosheen R, Nadeem S, Shahzadi A, Imran M. Surface-enhanced Raman spectroscopy for characterization of supernatant samples of biofilm forming bacterial strains. SPECTROCHIMICA ACTA. PART A, MOLECULAR AND BIOMOLECULAR SPECTROSCOPY 2024; 305:123414. [PMID: 37852119 DOI: 10.1016/j.saa.2023.123414] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/23/2023] [Revised: 09/07/2023] [Accepted: 09/13/2023] [Indexed: 10/20/2023]
Abstract
Staphylococcus epidermidis is considered major cause of nosocomial infections. Its pathogenicity is mainly due to the ability to form biofilms on different surfaces, particularly indwelling medical devices. This bacterium consists of different strains consisting of non, medium and strong biofilm forming ones. Surface-enhanced Raman spectroscopy (SERS) is a powerful analytical technique that can be used to detect and analyze biochemical composition of the supernatant samples of different strains of bacteria including non, medium and strong biofilm forming bacterial strains. SERS is a powerful technique for the robust, reliable, rapid detection and discrimination of bacteria in the form of characteristic SERS spectral features which can be used for detection and classification. SERS is used to differentiate three classes of bacteria with respect to their biofilm forming ability. Silver nanoparticles (Ag NPs) are used as SERS substrate and synthesized with chemical reduction method. Principal component analysis (PCA) and partial least square discriminant analysis (PLS-DA) are used to discriminate SERS spectral data sets of non, medium and strong biofilm forming bacteria. PLS-DA analysis is a multivariate statistical technique that can be used to analyze data from bacterial sets.
Collapse
Affiliation(s)
- Abdul Naman
- Department of Chemistry, University of Agriculture Faisalabad, Faisalabad 38000, Pakistan
| | - Hira Tahseen
- Department of Chemistry, University of Agriculture Faisalabad, Faisalabad 38000, Pakistan
| | - Haq Nawaz
- Department of Chemistry, University of Agriculture Faisalabad, Faisalabad 38000, Pakistan.
| | - Muhammad Irfan Majeed
- Department of Chemistry, University of Agriculture Faisalabad, Faisalabad 38000, Pakistan.
| | - Aamir Ali
- National Institute for Biotechnology and Genetic Engineering College, Pakistan Institute of Engineering and Applied Sciences (NIBGE-C, PIEAS), Jhang Road, Faisalabad 38000, Pakistan
| | - Asma Haque
- Department of Bioinformatics and Biotechnology, Government College University Faisalabad (GCUF), Faisalabad 38000, Pakistan
| | - Muhammad Umair Akbar
- Department of Bioinformatics and Biotechnology, Government College University Faisalabad (GCUF), Faisalabad 38000, Pakistan
| | - Nasir Mehmood
- Department of Chemistry, University of Agriculture Faisalabad, Faisalabad 38000, Pakistan
| | - Rashid Nosheen
- Department of Chemistry, University of Education, Faisalabad Campus, Faisalabad 38000, Pakistan.
| | - Sana Nadeem
- Department of Chemistry, University of Agriculture Faisalabad, Faisalabad 38000, Pakistan
| | - Aqsa Shahzadi
- Department of Chemistry, University of Agriculture Faisalabad, Faisalabad 38000, Pakistan
| | - Muhammad Imran
- Department of Chemistry, Faculty of Science, King Khalid University, P.O. Box 9004, Abha 61413, Saudi Arabia
| |
Collapse
|
21
|
Pereira AFM, Sani AA, Zapata TB, de Sousa DSM, Rossini BC, dos Santos LD, Rall VLM, Riccardi CDS, Fernandes Júnior A. Synergistic Antibacterial Efficacy of Melittin in Combination with Oxacillin against Methicillin-Resistant Staphylococcus aureus (MRSA). Microorganisms 2023; 11:2868. [PMID: 38138012 PMCID: PMC10745785 DOI: 10.3390/microorganisms11122868] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2023] [Revised: 11/17/2023] [Accepted: 11/20/2023] [Indexed: 12/24/2023] Open
Abstract
Methicillin-resistant Staphylococcus aureus (MRSA) often cause infections with high mortality rates. Antimicrobial peptides are a source of molecules for developing antimicrobials; one such peptide is melittin, a fraction from the venom of the Apis mellifera bee. This study aimed to evaluate the antibacterial and antibiofilm activities of melittin and its association with oxacillin (mel+oxa) against MRSA isolates, and to investigate the mechanisms of action of the treatments on MRSA. Minimum inhibitory concentrations (MICs) were determined, and synergistic effects of melittin with oxacillin and cephalothin were assessed. Antibiofilm and cytotoxic activities, as well as their impact on the cell membrane, were evaluated for melittin, oxacillin, and mel+oxa. Proteomics evaluated the effects of the treatments on MRSA. Melittin mean MICs for MRSA was 4.7 μg/mL and 12 μg/mL for oxacillin. Mel+oxa exhibited synergistic effects, reducing biofilm formation, and causing leakage of proteins, nucleic acids, potassium, and phosphate ions, indicating action on cell membrane. Melittin and mel+oxa, at MIC values, did not induce hemolysis and apoptosis in HaCaT cells. The treatments resulted in differential expression of proteins associated with protein synthesis and energy metabolism. Mel+oxa demonstrated antibacterial activity against MRSA, suggesting a potential as a candidate for the development of new antibacterial agents against MRSA.
Collapse
Affiliation(s)
- Ana Flávia Marques Pereira
- The Center for the Study of Venoms and Venomous Animals of UNESP (CEVAP), São Paulo State University (UNESP), Botucatu 18619-002, São Paulo, Brazil;
| | - Alessandra Aguirra Sani
- Department of Chemical and Biological Sciences, Microbiology and Immunology Sector, Institute of Biosciences of Botucatu (IBB), São Paulo State University (UNESP), Botucatu 18618-689, São Paulo, Brazil; (A.A.S.); (T.B.Z.); (D.S.M.d.S.); (V.L.M.R.)
| | - Tatiane Baptista Zapata
- Department of Chemical and Biological Sciences, Microbiology and Immunology Sector, Institute of Biosciences of Botucatu (IBB), São Paulo State University (UNESP), Botucatu 18618-689, São Paulo, Brazil; (A.A.S.); (T.B.Z.); (D.S.M.d.S.); (V.L.M.R.)
| | - Débora Silva Marques de Sousa
- Department of Chemical and Biological Sciences, Microbiology and Immunology Sector, Institute of Biosciences of Botucatu (IBB), São Paulo State University (UNESP), Botucatu 18618-689, São Paulo, Brazil; (A.A.S.); (T.B.Z.); (D.S.M.d.S.); (V.L.M.R.)
| | - Bruno César Rossini
- Institute of Biotechnology (IBTEC), São Paulo State University (UNESP), Botucatu 18607-440, São Paulo, Brazil; (B.C.R.); (L.D.d.S.)
| | - Lucilene Delazari dos Santos
- Institute of Biotechnology (IBTEC), São Paulo State University (UNESP), Botucatu 18607-440, São Paulo, Brazil; (B.C.R.); (L.D.d.S.)
- Graduate Program in Tropical Diseases and Graduate Program in Research and Development (Medical Biotechnology), Botucatu Medical School (FMB), São Paulo State University (UNESP), Botucatu 18618-687, São Paulo, Brazil
| | - Vera Lúcia Mores Rall
- Department of Chemical and Biological Sciences, Microbiology and Immunology Sector, Institute of Biosciences of Botucatu (IBB), São Paulo State University (UNESP), Botucatu 18618-689, São Paulo, Brazil; (A.A.S.); (T.B.Z.); (D.S.M.d.S.); (V.L.M.R.)
| | - Carla dos Santos Riccardi
- Department of Bioprocesses and Biotechnology, Faculty of Agricultural Sciences (FCA), São Paulo State University (UNESP), Botucatu 18610-034, São Paulo, Brazil;
| | - Ary Fernandes Júnior
- Department of Chemical and Biological Sciences, Microbiology and Immunology Sector, Institute of Biosciences of Botucatu (IBB), São Paulo State University (UNESP), Botucatu 18618-689, São Paulo, Brazil; (A.A.S.); (T.B.Z.); (D.S.M.d.S.); (V.L.M.R.)
| |
Collapse
|
22
|
Xiao G, Li J, Sun Z. The Combination of Antibiotic and Non-Antibiotic Compounds Improves Antibiotic Efficacy against Multidrug-Resistant Bacteria. Int J Mol Sci 2023; 24:15493. [PMID: 37895172 PMCID: PMC10607837 DOI: 10.3390/ijms242015493] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Revised: 10/19/2023] [Accepted: 10/21/2023] [Indexed: 10/29/2023] Open
Abstract
Bacterial antibiotic resistance, especially the emergence of multidrug-resistant (MDR) strains, urgently requires the development of effective treatment strategies. It is always of interest to delve into the mechanisms of resistance to current antibiotics and target them to promote the efficacy of existing antibiotics. In recent years, non-antibiotic compounds have played an important auxiliary role in improving the efficacy of antibiotics and promoting the treatment of drug-resistant bacteria. The combination of non-antibiotic compounds with antibiotics is considered a promising strategy against MDR bacteria. In this review, we first briefly summarize the main resistance mechanisms of current antibiotics. In addition, we propose several strategies to enhance antibiotic action based on resistance mechanisms. Then, the research progress of non-antibiotic compounds that can promote antibiotic-resistant bacteria through different mechanisms in recent years is also summarized. Finally, the development prospects and challenges of these non-antibiotic compounds in combination with antibiotics are discussed.
Collapse
Affiliation(s)
| | | | - Zhiliang Sun
- College of Veterinary Medicine, Hunan Agricultural University, Changsha 410128, China; (G.X.); (J.L.)
| |
Collapse
|
23
|
Zhang LM, Yang M, Zhou SW, Zhang H, Feng Y, Shi L, Li DS, Lu QM, Zhang ZH, Zhao M. Blapstin, a Diapause-Specific Peptide-Like Peptide from the Chinese Medicinal Beetle Blaps rhynchopetera, Has Antifungal Function. Microbiol Spectr 2023; 11:e0308922. [PMID: 37140456 PMCID: PMC10269622 DOI: 10.1128/spectrum.03089-22] [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: 08/07/2022] [Accepted: 04/10/2023] [Indexed: 05/05/2023] Open
Abstract
Drug resistance against bacteria and fungi has become common in recent years, and it is urgent to discover novel antimicrobial peptides to manage this problem. Many antimicrobial peptides from insects have been reported to have antifungal activity and are candidate molecules in the treatment of human diseases. In the present study, we characterized an antifungal peptide named blapstin that was isolated from the Chinese medicinal beetle Blaps rhynchopetera used in folk medicine. The complete coding sequence was cloned from the cDNA library prepared from the midgut of B. rhynchopetera. It is a 41-amino-acid diapause-specific peptide (DSP)-like peptide stabilized by three disulfide bridges and shows antifungal activity against Candida albicans and Trichophyton rubrum with MICs of 7 μM and 5.3 μM, respectively. The C. albicans and T. rubrum treated with blapstin showed irregular and shrunken cell membranes. In addition, blapstin inhibited the activity of C. albicans biofilm and showed little hemolytic or toxic activity on human cells and it is highly expressed in the fat body, followed by the hemolymph, midgut, muscle, and defensive glands. These results indicate that blapstin may help insects fight against fungi and showed a potential application in the development of antifungal reagents. IMPORTANCE Candida albicans is one of the conditional pathogenic fungi causing severe nosocomial infections. Trichophyton rubrum and other skin fungi are the main pathogens of superficial cutaneous fungal diseases, especially in children and the elderly. At present, antibiotics such as amphotericin B, ketoconazole, and fluconazole are the main drugs for the clinical treatment of C. albicans and T. rubrum infections. However, these drugs have certain acute toxicity. Long-term use can increase kidney damage and other side effects. Therefore, obtaining broad-spectrum antifungal drugs with high efficiency and low toxicity for the treatment of C. albicans and T. rubrum infections is a top priority. Blapstin is an antifungal peptide which shows activity against C. albicans and T. rubrum. The discovery of blapstin provides a novel clue for our understanding of the innate immunity of Blaps rhynchopetera and provides a template for designing antifungal drugs.
Collapse
Affiliation(s)
- La-Mei Zhang
- Institute of Highland Forest Science, Chinese Academy of Forestry, Kunming, China
- College of Forestry, Nanjing Forestry University, Nanjing, Jiangsu, China
- Key Laboratory of Breeding and Utilization of Resource Insects, National Forestry and Grassland Administration, Kunming, China
| | - Min Yang
- Key Laboratory of Bioactive Peptides of Yunnan Province, Kunming Institute of Zoology, Kunming, Yunnan, China
- Kunming College of Life Science, University of Chinese Academy of Sciences, Beijing, China
| | - Sheng-Wen Zhou
- Key Laboratory of Bioactive Peptides of Yunnan Province, Kunming Institute of Zoology, Kunming, Yunnan, China
- Kunming College of Life Science, University of Chinese Academy of Sciences, Beijing, China
| | - Hao Zhang
- Key Laboratory of Bioactive Peptides of Yunnan Province, Kunming Institute of Zoology, Kunming, Yunnan, China
- Kunming College of Life Science, University of Chinese Academy of Sciences, Beijing, China
| | - Ying Feng
- Institute of Highland Forest Science, Chinese Academy of Forestry, Kunming, China
- Key Laboratory of Breeding and Utilization of Resource Insects, National Forestry and Grassland Administration, Kunming, China
| | - Lei Shi
- Institute of Highland Forest Science, Chinese Academy of Forestry, Kunming, China
- Key Laboratory of Breeding and Utilization of Resource Insects, National Forestry and Grassland Administration, Kunming, China
| | - Dong-Sheng Li
- Key Laboratory of Bioactive Peptides of Yunnan Province, Kunming Institute of Zoology, Kunming, Yunnan, China
- Sino-African Joint Research Center, Chinese Academy of Science, Wuhan, Hubei, China
| | - Qiu-Min Lu
- Key Laboratory of Bioactive Peptides of Yunnan Province, Kunming Institute of Zoology, Kunming, Yunnan, China
- Sino-African Joint Research Center, Chinese Academy of Science, Wuhan, Hubei, China
| | - Zhong-He Zhang
- Institute of Highland Forest Science, Chinese Academy of Forestry, Kunming, China
- Key Laboratory of Breeding and Utilization of Resource Insects, National Forestry and Grassland Administration, Kunming, China
| | - Min Zhao
- Institute of Highland Forest Science, Chinese Academy of Forestry, Kunming, China
- Key Laboratory of Breeding and Utilization of Resource Insects, National Forestry and Grassland Administration, Kunming, China
| |
Collapse
|
24
|
Liu T, Sun Z, Yang Z, Qiao X. Microbiota-derived short-chain fatty acids and modulation of host-derived peptides formation: Focused on host defense peptides. Biomed Pharmacother 2023; 162:114586. [PMID: 36989711 DOI: 10.1016/j.biopha.2023.114586] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Revised: 03/12/2023] [Accepted: 03/21/2023] [Indexed: 03/29/2023] Open
Abstract
The byproducts of bacterial fermentation known as short-chain fatty acids (SCFAs) are chemically comprised of a carboxylic acid component and a short hydrocarbon chain. Recent investigations have demonstrated that SCFAs can affect intestinal immunity by inducing endogenous host defense peptides (HDPs) and their beneficial effects on barrier integrity, gut health, energy supply, and inflammation. HDPs, which include defensins, cathelicidins, and C-type lectins, perform a significant function in innate immunity in gastrointestinal mucosal membranes. SCFAs have been demonstrated to stimulate HDP synthesis by intestinal epithelial cells via interactions with G protein-coupled receptor 43 (GPR43), activation of the Jun N-terminal kinase (JNK) and Mitogen-activated protein kinase kinase (MEK)/extracellular signal-regulated kinase (ERK) pathways, and the cell growth pathways. Furthermore, SCFA butyrate has been demonstrated to enhance the number of HDPs released from macrophages. SCFAs promote monocyte-to-macrophage development and stimulate HDP synthesis in macrophages by inhibiting histone deacetylase (HDAC). Understanding the etiology of many common disorders might be facilitated by studies into the function of microbial metabolites, such as SCFAs, in the molecular regulatory processes of immune responses (e.g., HDP production). This review will focus on the current knowledge of the role and mechanism of microbiota-derived SCFAs in influencing the synthesis of host-derived peptides, particularly HDPs.
Collapse
|
25
|
Daffinee KE, O'Neill ET, Bleick CR, Williams G, Antoci V, Garcia D, LaPlante KL. Staphylococcal Biofilm: Penetration and bioavailability of vancomycin with or without rifampin. Diagn Microbiol Infect Dis 2023; 106:115947. [PMID: 37116243 DOI: 10.1016/j.diagmicrobio.2023.115947] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Revised: 03/14/2023] [Accepted: 03/25/2023] [Indexed: 03/31/2023]
Abstract
We measured antibiotic penetration and bioavailability in staphylococcus biofilms using simulated humanized concentrations of fluorescent vancomycin plus or minus rifampin. Vancomycin percent recovery across biofilm layers was:upper = 46%, middle = 40%, and lower = 33%. Vancomycin plus rifampin was not significantly different (P = 0.65). Addition of rifampin did not improve vancomycin penetration across biofilm layers.
Collapse
Affiliation(s)
- Kathryn E Daffinee
- Infectious Diseases Research Program, Providence Veterans Affairs Medical Center, Providence, RI, USA
| | - Emily T O'Neill
- Infectious Diseases Research Program, Providence Veterans Affairs Medical Center, Providence, RI, USA; College of Pharmacy, University of Rhode Island, Kingston, RI, USA
| | - Callan R Bleick
- College of Pharmacy, University of Rhode Island, Kingston, RI, USA
| | - Geoff Williams
- Leduc Bioimaging Facility, Brown University, Division of Biology and Medicine, Providence, RI, USA
| | - Valentin Antoci
- Department of Orthopaedics, The Warren Alpert School of Medicine, Brown University, Providence, RI, USA; The Diane N. Weiss Center for Orthopaedic Trauma Research, Rhode Island Hospital, Providence, RI, USA
| | - Dioscaris Garcia
- Department of Orthopaedics, The Warren Alpert School of Medicine, Brown University, Providence, RI, USA; The Diane N. Weiss Center for Orthopaedic Trauma Research, Rhode Island Hospital, Providence, RI, USA
| | - Kerry L LaPlante
- Infectious Diseases Research Program, Providence Veterans Affairs Medical Center, Providence, RI, USA; College of Pharmacy, University of Rhode Island, Kingston, RI, USA; Warren Alpert Medical School of Brown University, Division of Infectious Diseases, Providence, RI, USA.
| |
Collapse
|
26
|
Mirzaei R, Esmaeili Gouvarchin Ghaleh H, Ranjbar R. Antibiofilm effect of melittin alone and in combination with conventional antibiotics toward strong biofilm of MDR-MRSA and - Pseudomonas aeruginosa. Front Microbiol 2023; 14:1030401. [PMID: 36910230 PMCID: PMC9994733 DOI: 10.3389/fmicb.2023.1030401] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2022] [Accepted: 02/01/2023] [Indexed: 02/22/2023] Open
Abstract
INTRODUCTION Multidrug-resistant (MDR) pathogens are being recognized as a critical threat to human health if they can form biofilm and, in this sense, biofilm-forming MDR-methicillin resistant Staphylococcus aureus (MRSA) and -Pseudomonas aeruginosa strains are a worse concern. Hence, a growing body of documents has introduced antimicrobial peptides (AMPs) as a substitute candidate for conventional antimicrobial agents against drug-resistant and biofilm-associated infections. We evaluated melittin's antibacterial and antibiofilm activity alone and/or in combination with gentamicin, ciprofloxacin, rifampin, and vancomycin on biofilm-forming MDR-P. aeruginosa and MDR-MRSA strains. METHODS Antibacterial tests [antibiogram, minimum inhibitory concentration (MIC), and minimum bactericidal concentration (MBC)], anti-biofilm tests [minimum biofilm inhibition concentration (MBIC), and minimum biofilm eradication concentration (MBEC)], as well as synergistic antibiofilm activity of melittin and antibiotics, were performed. Besides, the influence of melittin alone on the biofilm encoding genes and the cytotoxicity and hemolytic effects of melittin were examined. RESULTS MIC, MBC, MBIC, and MBEC indices for melittin were in the range of 0.625-5, 1.25-10, 2.5-20, and 10-40 μg/ml, respectively. The findings found that the combination of melittin AMP with antibiotics was synergistic and fractional biofilm inhibitory concentration index (FBICi) for most tested concentrations was <0.5, resulting in a significant reduction in melittin, gentamicin, ciprofloxacin, vancomycin, and rifampin concentrations by 2-256.4, 2-128, 2-16, 4-64 and 4-8 folds, respectively. This phenomenon reduced the toxicity of melittin, whereby its synergist concentration required for biofilm inhibition did not show cytotoxicity and hemolytic activity. Our findings found that melittin decreased the expression of icaA in S. aureus and LasR in P. aeruginosa genes from 0.1 to 4.11 fold for icaA, and 0.11 to 3.7 fold for LasR, respectively. CONCLUSION Overall, the results obtained from our study show that melittin alone is effective against the strong biofilm of MDR pathogens and also offers sound synergistic effects with antibiotics without toxicity. Hence, combining melittin and antibiotics can be a potential candidate for further evaluation of in vivo infections by MDR pathogens.
Collapse
Affiliation(s)
- Rasoul Mirzaei
- Department of Microbiology, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | | | - Reza Ranjbar
- Molecular Biology Research Center, Systems Biology and Poisonings Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| |
Collapse
|
27
|
Alimohamadi M, Khataee A, Arefi-Oskoui S, Vahid B, Orooji Y, Yoon Y. Catalytic activation of hydrogen peroxide by Cr 2AlC MAX phase under ultrasound waves for a treatment of water contaminated with organic pollutants. ULTRASONICS SONOCHEMISTRY 2023; 93:106294. [PMID: 36640461 PMCID: PMC9852641 DOI: 10.1016/j.ultsonch.2023.106294] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/05/2022] [Revised: 12/28/2022] [Accepted: 01/06/2023] [Indexed: 06/17/2023]
Abstract
This study aims to investigate the sonocatalytic activation of hydrogen peroxide (H2O2) using Cr2AlC MAX phase prepared by the reactive sintering process. The hexagonal structure of the crystalline MAX phase was confirmed by X-ray diffraction. Moreover, the compacted layered structure of the MAX phase was observed via scanning electron microscopy and high-resolution transmission electron microscopy. Under the desired operating conditions, Cr2AlC MAX phase (0.75 g/L) showed suitable potential to activate H2O2 (1 mmol/L) under sonication, thereby allowing a considerable removal efficiency for various organic pollutants, including dimethyl phthalate (69.1%), rifampin (94.5%), hydroxychloroquine (100%), and acid blue 7 (91.5%) with initial concentration of 15 mg/L within 120 min of treatment. Kinetic analysis proved that the degradation reaction followed pseudo-first-order kinetics. Scavenging tests demonstrated that hydroxyl radicals and singlet oxygen were effective species during degradation. Furthermore, a probable mechanism for dimethyl phthalate degradation was suggested according to gas chromatography-mass spectroscopy and nuclear magnetic resonance analyses. The obtained results confirmed the capability of the triple Cr2AlC/H2O2/US process as a promising method for treating contaminated water.
Collapse
Affiliation(s)
- Monireh Alimohamadi
- Research Laboratory of Advanced Water and Wastewater Treatment Processes, Department of Applied Chemistry, Faculty of Chemistry, University of Tabriz, 51666-16471 Tabriz, Iran
| | - Alireza Khataee
- Research Laboratory of Advanced Water and Wastewater Treatment Processes, Department of Applied Chemistry, Faculty of Chemistry, University of Tabriz, 51666-16471 Tabriz, Iran
- Рeoples’ Friendship University of Russia (RUDN University), 6 Miklukho-Maklaya Street, Moscow, 117198, Russian Federation
| | - Samira Arefi-Oskoui
- Research Laboratory of Advanced Water and Wastewater Treatment Processes, Department of Applied Chemistry, Faculty of Chemistry, University of Tabriz, 51666-16471 Tabriz, Iran
- Department of Chemical Industry, Technical and Vocational University (TVU), Tehran, Iran
| | - Behrouz Vahid
- Department of Chemical Engineering, Tabriz Branch, Islamic Azad University, Tabriz, Iran
| | - Yasin Orooji
- College of Geography and Environmental Sciences, Zhejiang Normal University, 321004 Jinhua, China
| | - Yeojoon Yoon
- Department of Environmental and Energy Engineering, Yonsei University, Wonju, Republic of Korea
| |
Collapse
|
28
|
Jalalifar S, Mirzaei R, Motallebirad T, Razavi S, Talebi M. The Emerging Role of Probiotics and their Derivatives against Biofilm-Producing MRSA: A Scoping Review. BIOMED RESEARCH INTERNATIONAL 2022; 2022:4959487. [PMID: 36605101 PMCID: PMC9810406 DOI: 10.1155/2022/4959487] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Revised: 11/20/2022] [Accepted: 12/01/2022] [Indexed: 12/29/2022]
Abstract
BACKGROUND Methicillin-resistant Staphylococcus aureus (MRSA) is one of the main bacterial pathogens causing chronic infections, mainly because of its capacity to produce biofilm. Biofilm production is one of the underlying strategies for antibacterial drug resistance. Accordingly, preventing and attenuating biofilm production has become an emerging approach to controlling persistent infections. Therefore, this scoping review is aimed at surveying the published literature describing the usage of probiotics and their derivatives against biofilm-producing MRSA. METHODS Updated literature searches were conducted across seven electronic databases including Web of Science, PubMed, Scopus, Cochrane Library, ProQuest, Embase, and Google Scholar to identify all original published articles about probiotics against MRSA. In this regard, studies were summarized and analyzed in the present review. RESULTS In the reviewed studies, various microorganisms and compounds were used as probiotics as follows: Lactobacillus species (8 studies), Enterococcus species (4 studies), Bacillus species (2 studies), Streptomyces species (2 studies), Saccharomyces cerevisiae (1 study), Corynebacterium accolens (1 study), and Lactococcus lactis derived Nisin (3 studies). Based on our comprehensive search, 21 studies with eligibility criteria were included in the present review including 12 studies on clinical strains, 6 studies on ATCC, 2 studies simultaneously on clinical and standard strains, and finally 1 study on food sample. CONCLUSIONS Our study showed that there was an increasing trend in the number of publications reporting probiotics against biofilm-producing MRSA. The results of this scoping review could use to guide the undertaking of the subsequent systematic reviews. In summary, probiotics with antimicrobial and antibiofilm properties can use as an embedded agent in food products or as a biopharmaceutical in the prevention and treatment of MRSA infections.
Collapse
Affiliation(s)
- Saba Jalalifar
- Microbial Biotechnology Research Center, Iran University of Medical Sciences, Tehran, Iran
- Department of Microbiology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Rasoul Mirzaei
- Venom and Biotherapeutics Molecules Lab, Medical Biotechnology Department, Biotechnology Research Center, Pasteur Institute of Iran, Tehran, Iran
| | - Tahereh Motallebirad
- Department of Microbiology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Shabnam Razavi
- Microbial Biotechnology Research Center, Iran University of Medical Sciences, Tehran, Iran
- Department of Microbiology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Malihe Talebi
- Microbial Biotechnology Research Center, Iran University of Medical Sciences, Tehran, Iran
- Department of Microbiology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
29
|
Mirzaei R, Yousefimashouf R, Arabestani MR, Sedighi I, Alikhani MY. The issue beyond resistance: Methicillin-resistant Staphylococcus epidermidis biofilm formation is induced by subinhibitory concentrations of cloxacillin, cefazolin, and clindamycin. PLoS One 2022; 17:e0277287. [PMID: 36350834 PMCID: PMC9645612 DOI: 10.1371/journal.pone.0277287] [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: 02/16/2022] [Accepted: 10/25/2022] [Indexed: 11/11/2022] Open
Abstract
Staphylococcus epidermis is one of the most frequent causes of device-associated infections due to biofilm formation. Current reports noted that subinhibitory concentrations of antibiotics induce biofilm production in some bacteria. Accordingly, we evaluated the effect of exposure of different subinhibitory concentrations of cloxacillin, cefazolin, clindamycin, and vancomycin on the biofilm formation of methicillin-resistant S. epidermidis (MRSE). Antimicrobial susceptibility testing and minimum inhibitory/bactericidal concentration of antimicrobial agents were determined. MRSE isolates were selected, and their biofilm formation ability was evaluated. The effect of subinhibitory concentrations of cloxacillin, cefazolin, clindamycin, and vancomycin, antibiotics selected among common choices in the clinic, on MRSE biofilm formation was determined by the microtitre method. Besides, the effect of subinhibitory concentrations of cloxacillin, cefazolin, clindamycin, and vancomycin on the expression of the biofilm-associated genes icaA and atlE was evaluated by Reverse-transcription quantitative real-time polymerase chain reaction (RT-qPCR). Antimicrobial susceptibility patterns of MRSE strains showed a high level of resistance as follows: 80%, 53.3%, 33.3%, 33.3%, and 26.6%, for erythromycin, trimethoprim-sulfamethoxazole, tetracycline, clindamycin, and gentamicin, respectively. Besides, 73.3% of S. epidermidis strains were Multidrug-resistant (MDR). Minimum inhibitory concentration (MIC) and minimum bactericidal concentration (MBC) values were in the range of 0.5 to512 μg/mL and 1 to1024 μg/mL for cloxacillin, 0.125 to256 μg/mL and 1 to512 μg/mL for cefazolin, 0.125 to64 μg/mL and 4 to>1024 μg/mL for clindamycin, and 2 to32 μg/mL and 4 to32 μg/mL for vancomycin, respectively. The findings showed that subinhibitory concentrations of cloxacillin, cefazolin, and clindamycin induce biofilm production in MRSE strains. In particular, the OD values of strains were in the range of 0.09-0.95, 0.05-0.86, and 0.06-1 toward cloxacillin, cefazolin, and clindamycin, respectively. On the other hand, exposure to subinhibitory vancomycin concentrations did not increase the biofilm formation in MRSE strains. The findings also demonstrated that sub-MIC of antibiotics up-regulated biofilm-associated genes. In particular, atlE and icaA were up-regulated 0.062 to 1.16 and 0.078 to 1.48 folds, respectively, for cloxacillin, 0.11 to 0.8, and 0.1 to 1.3 folds for cefazolin, 0.18 to 0.98, and 0.19 to 1.4 folds, respectively, for clindamycin. In contrast, the results showed that sub-MIC of vancomycin did not increase the biofilm-associated genes. These findings overall show that exposure to sub-MIC of traditional antibiotics can cause biofilm induction in MRSE, thereby increasing the survival and persistence on various surfaces that worsen the condition of comorbid infections.
Collapse
Affiliation(s)
- Rasoul Mirzaei
- Department of Microbiology, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Rasoul Yousefimashouf
- Department of Microbiology, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Mohammad Reza Arabestani
- Department of Microbiology, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Iraj Sedighi
- Department of Pediatrics, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Mohammad Yousef Alikhani
- Department of Microbiology, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
- Brucellosis Research Center, Hamadan University of Medical Sciences, Hamadan, Iran
| |
Collapse
|
30
|
Mirzaei R, Alikhani MY, Arciola CR, Sedighi I, Irajian G, Jamasbi E, Yousefimashouf R, Bagheri KP. Highly Synergistic Effects of Melittin With Vancomycin and Rifampin Against Vancomycin and Rifampin Resistant Staphylococcus epidermidis. Front Microbiol 2022; 13:869650. [PMID: 35814659 PMCID: PMC9260053 DOI: 10.3389/fmicb.2022.869650] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Accepted: 05/11/2022] [Indexed: 12/27/2022] Open
Abstract
Methicillin-resistant Staphylococcus epidermidis (MRSE) strains are increasingly emerging as serious pathogens because they can be resistant to many antibiotics called multidrug resistance (MDR) that limit the therapeutic options. In the case of vancomycin- and rifampin-resistant MDR-MRSE, the physicians are not allowed to increase the doses of antibiotics because of severe toxicity. Accordingly, we investigated the synergistic activity of melittin antimicrobial peptide with vancomycin and rifampin against vancomycin-resistant, and rifampin-resistant MDR-MRSE isolates. Minimum inhibitory concentration (MIC), minimum bactericidal concentration (MBC), fractional inhibitory concentration index (FICi), and fractional bactericidal concentration index (FBCi) of antimicrobial agents against isolates were determined. Coagulate activities and serum and salt stability as well as melittin cytotoxicity on the human embryonic kidney (HEK) 293 cells and human red blood cells (RBCs) at their synergistic concentrations. MIC and MBC values for melittin were in the range of 0.312-2.5 and 0.312-5, respectively. Results also showed that the interaction of melittin with drugs was highly synergistic in which the geometric means of FICi and FBCi were < 0.5. Induced synergism led to a decrease in melittin, rifampin, and vancomycin concentrations by 8-1,020, 2-16, and 4-16-folds, respectively. This phenomenon caused a reduction in melittin toxicity by which the synergistic concentration of melittin needed to kill bacteria did not show cytotoxicity and hemolytic activity. Besides, no coagulation activity was found for the synergistic and alone concentrations of melittin in both Prothrombin Time (PT) and Partial Thromboplastin Time (PTT). Interestingly, the antibacterial activity of melittin in Mueller Hinton Broth (MHB) containing human serum did no significant differences between MIC and MBC values of melittin in MHB and MHB containing 10% human serum. The present findings showed that the therapeutic index of melittin was improved by 32.08- and 12.82-folds when combined with vancomycin and rifampin, respectively. Taken together, the obtained data show that melittin alone was effective against MDR-MRSE isolates and this antimicrobial peptide showed highly synergistic effects with vancomycin and rifampin without causing toxicity. Therefore, the combination of melittin and traditional antibiotics could be a promising strategy for the treatment of infections caused by MDR-MRSE.
Collapse
Affiliation(s)
- Rasoul Mirzaei
- Department of Microbiology, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
- Venom and Biotherapeutics Molecules Laboratory, Medical Biotechnology Department, Biotechnology Research Center, Pasteur Institute of Iran, Tehran, Iran
| | - Mohammad Yousef Alikhani
- Department of Microbiology, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Carla Renata Arciola
- Laboratorio di Patologia delle Infezioni Associate all’Impianto, IRCCS Istituto Ortopedico Rizzoli, Bologna, Italy
- Laboratorio di Immunoreumatologia e Rigenerazione Tissutale, IRCCS Istituto Ortopedico Rizzoli, Bologn, Italy
- Department of Experimental, Diagnostic and Specialty Medicine, University of Bologna, Bologna, Italy
| | - Iraj Sedighi
- Department of Pediatrics, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - GholamReza Irajian
- Microbial Biotechnology Research Center, Iran University of Medical Sciences, Tehran, Iran
- Department of Microbiology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Elaheh Jamasbi
- Research Center of Oils and Fats, Kermanshah University of Medical Science, Kermanshah, Iran
| | - Rasoul Yousefimashouf
- Department of Microbiology, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Kamran Pooshang Bagheri
- Venom and Biotherapeutics Molecules Laboratory, Medical Biotechnology Department, Biotechnology Research Center, Pasteur Institute of Iran, Tehran, Iran
| |
Collapse
|
31
|
Akbarzadeh-Khiavi M, Torabi M, Olfati AH, Rahbarnia L, Safary A. Bio-nano scale modifications of melittin for improving therapeutic efficacy. Expert Opin Biol Ther 2022; 22:895-909. [PMID: 35687355 DOI: 10.1080/14712598.2022.2088277] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
INTRODUCTION Melittin (MLT), a natural membrane-active component, is the most prominent cytolytic peptide from bee venom. Remarkable biological properties of MLT, including anti-inflammatory, antimicrobial, anticancer, anti-protozoan, and antiarthritic activities, make it an up-and-coming therapeutic candidate for a wide variety of human diseases. Therapeutic applications of MLT may be hindered due to low stability, high toxicity, and weak tissue penetration. Different bio-nano scale modifications hold promise for improving its functionality and therapeutic efficacy. AREAS COVERED In the current review, we aimed to provide a comprehensive insight into strategies used for MLT conjugations and modifications, cellular delivery of modified forms, and their clinical perspectives by reviewing the published literature on PubMed, Scopus, and Google Scholar databases. We also emphasized the MLT structure modifications, mechanism of action, and cellular toxicity. EXPERT OPINION Developing new analogs and conjugates of MLT as a natural drug with improved functions and fewer side effects is crucial for the clinical translation of this approach worldwide, especially where the chemicals and synthetic drugs are more expensive or unavailable in the healthcare system. MLT-nanoconjugation may be one of the best-optimized strategies for improving peptide delivery, increasing its therapeutic efficacy, and providing minimal nonspecific cellular lytic activity. [Figure: see text].
Collapse
Affiliation(s)
- Mostafa Akbarzadeh-Khiavi
- Liver and Gastrointestinal Diseases Research Center Tabriz University of Medical Sciences, Tabriz, Iran.,Research Center for Pharmaceutical Nanotechnology, Biomedicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mitra Torabi
- Research Center for Pharmaceutical Nanotechnology, Biomedicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Amir-Hossein Olfati
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Leila Rahbarnia
- Infectious and Tropical Diseases Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Azam Safary
- Connective Tissue Diseases Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
32
|
Donadu MG, Ferrari M, Mazzarello V, Zanetti S, Kushkevych I, Rittmann SKMR, Stájer A, Baráth Z, Szabó D, Urbán E, Gajdács M. No Correlation between Biofilm-Forming Capacity and Antibiotic Resistance in Environmental Staphylococcus spp.: In Vitro Results. Pathogens 2022; 11:pathogens11040471. [PMID: 35456146 PMCID: PMC9031815 DOI: 10.3390/pathogens11040471] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Revised: 04/08/2022] [Accepted: 04/13/2022] [Indexed: 01/18/2023] Open
Abstract
The production of biofilms is a critical factor in facilitating the survival of Staphylococcus spp. in vivo and in protecting against various environmental noxa. The possible relationship between the antibiotic-resistant phenotype and biofilm-forming capacity has raised considerable interest. The purpose of the study was to assess the interdependence between biofilm-forming capacity and the antibiotic-resistant phenotype in 299 Staphylococcus spp. (S. aureus n = 143, non-aureus staphylococci [NAS] n = 156) of environmental origin. Antimicrobial susceptibility testing and detection of methicillin resistance (MR) was performed. The capacity of isolates to produce biofilms was assessed using Congo red agar (CRA) plates and a crystal violet microtiter-plate-based (CV-MTP) method. MR was identified in 46.9% of S. aureus and 53.8% of NAS isolates (p > 0.05), with resistance to most commonly used drugs being significantly higher in MR isolates compared to methicillin-susceptible isolates. Resistance rates were highest for clindamycin (57.9%), erythromycin (52.2%) and trimethoprim-sulfamethoxazole (51.1%), while susceptibility was retained for most last-resort drugs. Based on the CRA plates, biofilm was produced by 30.8% of S. aureus and 44.9% of NAS (p = 0.014), while based on the CV-MTP method, 51.7% of S. aureus and 62.8% of NAS were identified as strong biofilm producers, respectively (mean OD570 values: S. aureus: 0.779±0.471 vs. NAS: 1.053±0.551; p < 0.001). No significant differences in biofilm formation were observed based on MR (susceptible: 0.824 ± 0.325 vs. resistant: 0.896 ± 0.367; p = 0.101). However, pronounced differences in biofilm formation were identified based on rifampicin susceptibility (S: 0.784 ± 0.281 vs. R: 1.239 ± 0.286; p = 0.011). The mechanistic understanding of the mechanisms Staphylococcus spp. use to withstand harsh environmental and in vivo conditions is crucial to appropriately address the therapy and eradication of these pathogens.
Collapse
Affiliation(s)
- Matthew Gavino Donadu
- Hospital Pharmacy, Azienda Ospedaliero Universitaria di Sassari, 07100 Sassari, Italy;
- Department of Biomedical Sciences, University of Sassari, 07100 Sassari, Italy; (V.M.); (S.Z.)
| | - Marco Ferrari
- Department of Biomedical Sciences, University of Sassari, 07100 Sassari, Italy; (V.M.); (S.Z.)
- Correspondence:
| | - Vittorio Mazzarello
- Department of Biomedical Sciences, University of Sassari, 07100 Sassari, Italy; (V.M.); (S.Z.)
| | - Stefania Zanetti
- Department of Biomedical Sciences, University of Sassari, 07100 Sassari, Italy; (V.M.); (S.Z.)
| | - Ivan Kushkevych
- Department of Experimental Biology, Faculty of Science, Masaryk University, 62500 Brno, Czech Republic;
| | - Simon K.-M. R. Rittmann
- Archaea Physiology & Biotechnology Group, Department of Functional and Evolutionary Ecology, Universität Wien, 1090 Wien, Austria;
| | - Anette Stájer
- Department of Periodontology, Faculty of Dentistry, University of Szeged, Tisza Lajos körút 62-64, 6720 Szeged, Hungary;
| | - Zoltán Baráth
- Department of Prosthodontics, Faculty of Dentistry, University of Szeged, Tisza Lajos körút 62–64, 6720 Szeged, Hungary;
| | - Dóra Szabó
- Institute of Medical Microbiology, Faculty of Medicine, Semmelweis University, Nagyvárad tér 4, 1089 Budapest, Hungary;
| | - Edit Urbán
- Department of Medical Microbiology and Immunology, University of Pécs Medical School, Szigeti út 12, 7624 Pécs, Hungary;
| | - Márió Gajdács
- Department of Oral Biology and Experimental Dental Research, Faculty of Dentistry, University of Szeged, Tisza Lajos krt. 63, 6720 Szeged, Hungary;
| |
Collapse
|