1
|
Wang Q, Gao Y, Chen Y, Wang X, Pei Q, Zhang T, Wang C, Pan J. Synergistic Enhancement of Antibacterial and Osteo-Immunomodulatory Activities of Titanium Implants via Dual-Responsive Multifunctional Surfaces. Adv Healthc Mater 2025; 14:e2404260. [PMID: 39690750 DOI: 10.1002/adhm.202404260] [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] [Academic Contribution Register] [Received: 10/31/2024] [Revised: 12/04/2024] [Indexed: 12/19/2024]
Abstract
Bone implant-associated infections and inflammations, primarily caused by bacteria colonization, frequently result in unsuccessful procedures and pose significant health risks to patients. To mitigate these challenges, the development of engineered implants with spatiotemporal regulation capabilities, designed to inhibit bacterial survival and modulate immune responses in the early stage, while promoting bone defect healing in the late stage is proposed. The implants are functionalized with ε-poly-l-lysine-phenylboronic acid (PP) via dynamic boronic ester bonds, which facilitate its release through a reactive oxygen species (ROS) and pH-responsive strategy, thereby establishing an antibacterial microenvironment on and around the implants. Additionally, the dynamic metal coordination interaction facilitates the loading and sustained release of Sr2+ under an acidic environment, providing immunomodulatory and osteogenic effects. The ROS/pH-responsive feature, coupled with the implant-bone tissue integration process, affords precise spatiotemporal regulation of the Ti-TA-Sr-PP implants. This strategy represents a promising approach for the preparation of advanced bone implants.
Collapse
Affiliation(s)
- Qing Wang
- Department of Stomatology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China
| | - Ya Gao
- Department of Stomatology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China
| | - Yanzheng Chen
- School of Chemistry and Chemical Engineering, Shanghai Jiao Tong University, Shanghai, 201100, China
| | - Xuan Wang
- Department of Stomatology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China
| | - Qingguo Pei
- Department of Stomatology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China
| | - Taiyu Zhang
- Precision Research Center for Refractory Diseases, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 201620, China
| | - Changping Wang
- Department of Orthopaedics, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China
| | - Jinsong Pan
- Department of Stomatology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China
| |
Collapse
|
2
|
Zhang S, Chen T, Lu W, Lin Y, Zhou M, Cai X. Hybrid Cell Membrane-Engineered Nanocarrier for Triple-Action Strategy to Address Pseudomonas aeruginosa Infection. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2411261. [PMID: 39721013 PMCID: PMC11809413 DOI: 10.1002/advs.202411261] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Academic Contribution Register] [Received: 09/13/2024] [Revised: 11/29/2024] [Indexed: 12/28/2024]
Abstract
Bacterial infections resistant to antimicrobial treatments, particularly those caused by Pseudomonas aeruginosa (P. aeruginosa), frequently lead to elevated mortality rates. Tackling this resistance using therapeutic combinations with varied mechanisms has shown considerable promise. In this study, a bioinspired nanocarrier is successfully designed and engineered for targeted antibiotic delivery and toxin/bacteria clearance. This is achieved by encapsulating antibiotic-loaded framework nucleic acids with hybrid cell membranes acquired from neutrophils and platelets. By coating the hybrid membrane outside the shell, nanocarriers are endowed with the function of neutrophil-like chemotaxis and platelet-like bacteria adhesion to achieve the first stage of inflammation targeting. Based on the specific binding of bacteria toxin to the hybrid membrane, the release of antibiotic-loaded framework nucleic acids is triggered by toxin-mediated membrane lysis to fulfill the second stage of toxin neutralization and bacteria killing. Meanwhile, the immunomodulation potential of framework nucleic acids enables nanocarriers to accomplish the third stage of reversing the immunosuppressive microenvironment. In mouse models of acute and chronic P. aeruginosa pneumonia, the nanocarriers can reduce bacterial burden at a low dosage and decrease mortality with negligible toxicity. In sum, these findings have illustrated the remarkable capability of nanocarriers in treating recalcitrant bacterial infections.
Collapse
Affiliation(s)
- Shunhao Zhang
- State Key Laboratory of Oral DiseasesNational Center for StomatologyNational Clinical Research Center for Oral DiseasesWest China Hospital of StomatologySichuan UniversityChengduSichuan610041China
| | - Tianyu Chen
- State Key Laboratory of Oral DiseasesNational Center for StomatologyNational Clinical Research Center for Oral DiseasesWest China Hospital of StomatologySichuan UniversityChengduSichuan610041China
| | - Weitong Lu
- State Key Laboratory of Oral DiseasesNational Center for StomatologyNational Clinical Research Center for Oral DiseasesWest China Hospital of StomatologySichuan UniversityChengduSichuan610041China
| | - Yunfeng Lin
- State Key Laboratory of Oral DiseasesNational Center for StomatologyNational Clinical Research Center for Oral DiseasesWest China Hospital of StomatologySichuan UniversityChengduSichuan610041China
- Sichuan Provincial Engineering Research Center of Oral BiomaterialsChengduSichuan610041China
- National Center for Translational MedicineShanghai Jiao Tong UniversityShanghai200240China
| | - Mi Zhou
- State Key Laboratory of Oral DiseasesNational Center for StomatologyNational Clinical Research Center for Oral DiseasesWest China Hospital of StomatologySichuan UniversityChengduSichuan610041China
| | - Xiaoxiao Cai
- State Key Laboratory of Oral DiseasesNational Center for StomatologyNational Clinical Research Center for Oral DiseasesWest China Hospital of StomatologySichuan UniversityChengduSichuan610041China
- Sichuan Provincial Engineering Research Center of Oral BiomaterialsChengduSichuan610041China
| |
Collapse
|
3
|
Sharma V, Das R, Mehta DK, Sharma D, Aman S, Khan MU. Quinolone scaffolds as potential drug candidates against infectious microbes: a review. Mol Divers 2025; 29:711-737. [PMID: 38683488 DOI: 10.1007/s11030-024-10862-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 02/16/2024] [Accepted: 03/26/2024] [Indexed: 05/01/2024]
Abstract
Prevalence of microbial infections and new rising pathogens are signified as causative agent for variety of serious and lethal health crisis in past years. Despite medical advances, bacterial and fungal infections continue to be a rising problem in the health care system. As more bacteria develop resistance to antibiotics used in therapy, and as more invasive microbial species develop resistance to conventional antimicrobial drugs. Relevant published publications from the last two decades, up to 2024, were systematically retrieved from the MEDLINE/PubMed, SCOPUS, EMBASE, and WOS databases using keywords such as quinolones, anti-infective, antibacterial, antimicrobial resistance and patents on quinolone derivatives. With an approach of considerable interest towards novel heterocyclic derivatives as novel anti-infective agents, researchers have explored these as essential tools in vistas of drug design and development. Among heterocycles, quinolones have been regarded extremely essential for the development of novel derivatives, even able to tackle the associated resistance issues. The quinolone scaffold with its bicyclic structure and specific functional groups such as the carbonyl and acidic groups, is indeed considered a valuable functionalities for further lead generation and optimization in drug discovery. Besides, the substitution at N-1, C-3 and C-7 positions also subjected to be having a significant role in anti-infective potential. In this article, we intend to highlight recent quinolone derivatives based on the SAR approach and anti-infective potential such as antibacterial, antifungal, antimalarial, antitubercular, antitrypanosomal and antiviral activities. Moreover, some recent patents granted on quinolone-containing derivatives as anti-infective agents have also been highlighted in tabular form. Due consideration of this, future research in this scaffold is expected to be useful for aspiring scientists to get pharmacologically significant leads.
Collapse
Affiliation(s)
- Vishal Sharma
- Department of Pharmaceutical Chemistry, MM College of Pharmacy, Maharishi Markandeshwar (Deemed to be University), Mullana, Ambala, 133207, India
| | - Rina Das
- Department of Pharmaceutical Chemistry, MM College of Pharmacy, Maharishi Markandeshwar (Deemed to be University), Mullana, Ambala, 133207, India
| | - Dinesh Kumar Mehta
- Department of Pharmaceutical Chemistry, MM College of Pharmacy, Maharishi Markandeshwar (Deemed to be University), Mullana, Ambala, 133207, India.
| | - Diksha Sharma
- Swami Devidyal College of Pharmacy, Barwala, 134118, India
| | - Shahbaz Aman
- Department of Microbiology, Maharishi Markandeshwar (Deemed to be University), Mullana, Ambala, 133207, India
| | - M U Khan
- Department of pharmaceutical Chemistry & Pharmacognosy, Unaizah College of Pharmacy, Qassim University, Uniazah, Al Qassim, Saudi Arabia
| |
Collapse
|
4
|
Soliman MKY, Amin MAA, Nowwar AI, Hendy MH, Salem SS. Green synthesis of selenium nanoparticles from Cassia javanica flowers extract and their medical and agricultural applications. Sci Rep 2024; 14:26775. [PMID: 39500933 PMCID: PMC11538282 DOI: 10.1038/s41598-024-77353-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 07/24/2024] [Accepted: 10/22/2024] [Indexed: 11/08/2024] Open
Abstract
Nanostructured materials are advantageous within numerous fields of medicine owing to their intriguing qualities, which include their size, reactive surface, bioactivity, potential for modification, and optical characteristics. Cassia javanica flower extract was used as a chelating agent in an environmentally friendly process to create SeNPs FTIR, XRD, and TEM, SAED were utilized to analyze and characterize the synthesized. The findings showed that the MIC of Se NPs against B. subtilis and S. aureus was 500 µg/ml. Conversely, the MIC for P. aeruginosa, E. coli, and C. albicans were 125, 250, and 62.5 µg/ml, respectively. Hence, SeNPs considerably reduced the activity; the inhibition peaked at 77.6% at 250 µg/ml to reach 49.04% at 7.8 µg/ml. Which showed the greatest suppression of MRSA biofilm formation without affecting bacterial growth. SeNPs showed an intriguing antioxidant capacity, achieving an IC50 of 53.34 µg/ml. This study looked how soaking seeds before sowing them with Se NPs at 50, 100, and 200 ppm affected the plants' development in different parameters, as well as their yield of Vicia faba L. The growth conditions were effectively increased by soaking application of various quantities of Se NPs. The highest values of dry weight/pod (g), number of seeds/plant, weight of 100 seeds (g), and number of pods/plant were caused by high concentrations of Se NPs, by 28.43, 89.60, 18.20, and 94.11%, respectively.
Collapse
Affiliation(s)
- Mohamed K Y Soliman
- Botany and Microbiology Department, Faculty of Science, Al-Azhar University, Nasr City, Cairo, 11884, Egypt
| | - Mohamed Abdel-Aal Amin
- Botany and Microbiology Department, Faculty of Science, Al-Azhar University, Nasr City, Cairo, 11884, Egypt
| | - Abdelatti Ibrahim Nowwar
- Botany and Microbiology Department, Faculty of Science, Al-Azhar University, Nasr City, Cairo, 11884, Egypt
| | - Mahmoud H Hendy
- Botany and Microbiology Department, Faculty of Science, Al-Azhar University, Nasr City, Cairo, 11884, Egypt
| | - Salem S Salem
- Botany and Microbiology Department, Faculty of Science, Al-Azhar University, Nasr City, Cairo, 11884, Egypt.
| |
Collapse
|
5
|
Ndiaye I, Debarbieux L, Sow O, Ba BS, Diagne MM, Cissé A, Fall C, Dieye Y, Dia N, de Magny GC, Seck A. Characterization of two Friunavirus phages and their inhibitory effects on biofilms of extremely drug resistant Acinetobacter baumannii in Dakar, Senegal. BMC Microbiol 2024; 24:449. [PMID: 39501140 PMCID: PMC11536776 DOI: 10.1186/s12866-024-03608-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 06/25/2024] [Accepted: 10/24/2024] [Indexed: 11/09/2024] Open
Abstract
BACKGROUND Acinetobacter baumannii is a gram-negative, opportunistic pathogen, that is responsible for a wide variety of infections and is a significant cause of hospital-acquired infections. A. baumannii is listed by the World Health Organization (WHO) as a critical priority pathogen because of its high level of antibiotic resistance and the urgent need for alternative treatment solutions. To address this challenge, bacteriophages have been used to combat bacterial infections for more than a century, and phage research has regained interest in recent years due to antimicrobial resistance (AMR). However, although the vast majority of deaths from the AMR crisis will occur in developing countries in Africa and Asia, few phages' studies have been conducted in these regions. In this study, we present a comprehensive characterization of the bacteriophages vAbBal23 and vAbAbd25, actives against extremely drug-resistant (XDR) A. baumannii. METHODS Phages were isolated from environmental wastewaters in Dakar, Senegal. The host-range, thermal and pH stabilities, infection kinetics, one step growth assay, antibiofilm activity assay, sequencing, and genomic analysis, were performed to characterize the isolated phages. RESULTS Comparative genomic and phylogenetic analyses revealed that vAbBal23 and vAbAbd25 belong to the Caudoviricetes class, Autographiviridae family and Friunavirus genus. Both phages demonstrated activity against strains with capsular type KL230. They were stable over a wide pH range (pH 3 to 9) and at temperatures ranging from 25 °C to 40 °C. Additionally, the phages exhibited notable activity against both planktonic and biofilm cells of targeted extremely drug resistant A. baumannii. The results presented here indicate the lytic nature of vAbBal23 and vAbAbd25. This is further supported by the absence of genes encoding toxins, resistance genes and bacterial virulence factors, highlighting their potential for future phage applications. CONCLUSION Phages vAbBal23 and vAbAbd25 are promising biological agents that can infect A. baumannii, making them suitable candidates for use in phage therapies.
Collapse
Affiliation(s)
- Issa Ndiaye
- Pole de Microbiologie, Institut Pasteur de Dakar, 36 Avenue Pasteur, Dakar, BP 220, Senegal.
- Faculté de Médecine, Pharmacie et Odontostomatologie, Université Cheikh Anta Diop, Dakar, Sénégal.
| | - Laurent Debarbieux
- Laboratoire de Bactériophage, Bactérie, Hôte, Département de Microbiologie, Institut Pasteur, Paris, France
| | - Ousmane Sow
- Pole de Microbiologie, Institut Pasteur de Dakar, 36 Avenue Pasteur, Dakar, BP 220, Senegal
| | | | | | - Abdoulaye Cissé
- Pole de Microbiologie, Institut Pasteur de Dakar, 36 Avenue Pasteur, Dakar, BP 220, Senegal
| | - Cheikh Fall
- Pole de Microbiologie, Institut Pasteur de Dakar, 36 Avenue Pasteur, Dakar, BP 220, Senegal
| | - Yakhya Dieye
- Pole de Microbiologie, Institut Pasteur de Dakar, 36 Avenue Pasteur, Dakar, BP 220, Senegal
| | - Ndongo Dia
- Département de Virologie, Institut Pasteur de Dakar, Dakar, Sénégal
| | - Guillaume Constantin de Magny
- MIVEGEC, Univ. Montpellier, CNRS, IRD, Montpellier, France
- MEEDiN, Montpellier Ecology and Evolution of Disease Network, Montpellier, France
| | - Abdoulaye Seck
- Pole de Microbiologie, Institut Pasteur de Dakar, 36 Avenue Pasteur, Dakar, BP 220, Senegal
- Faculté de Médecine, Pharmacie et Odontostomatologie, Université Cheikh Anta Diop, Dakar, Sénégal
| |
Collapse
|
6
|
Xue Y, Wang C, Zhao Y, Zhao Z, Cui R, Du B, Fang L, Wang J, Zhu B. Mixed-charge hyperbranched polymer nanoparticles with selective antibacterial action for fighting antimicrobial resistance. Acta Biomater 2024; 189:545-558. [PMID: 39222706 DOI: 10.1016/j.actbio.2024.08.044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 04/12/2024] [Revised: 08/05/2024] [Accepted: 08/26/2024] [Indexed: 09/04/2024]
Abstract
The escalating menace of antimicrobial resistance (AMR) presents a profound global threat to life and assets. However, the incapacity of metal ions/reactive oxygen species (ROS) or the indiscriminate intrinsic interaction of cationic groups to distinguish between bacteria and mammalian cells undermines the essential selectivity required in these nanomaterials for an ideal antimicrobial agent. Hence, we devised and synthesized a range of biocompatible mixed-charge hyperbranched polymer nanoparticles (MCHPNs) incorporating cationic, anionic, and neutral alkyl groups to effectively combat multidrug-resistant bacteria and mitigate AMR. This outcome stemmed from the structural, antibacterial activity, and biocompatibility analysis of seven MCHPNs, among which MCHPN7, with a ratio of cationic groups, anionic groups, and long alkyl chains at 27:59:14, emerged as the lead candidate. Importantly, owing to inherent differences in membrane potential among diverse species, alongside its nano-size (6-15 nm) and high hydrophilicity (Kow = 0.04), MCHPN7 exhibited exceptional selective bactericidal effects over mammalian cells (selectivity index > 564) in vitro and in vivo. By inducing physical membrane disruption, MCHPN7 effectively eradicated antibiotic-resistant bacteria and significantly delayed the emergence of bacterial resistance. Utilized as a coating, MCHPN7 endowed initially inert surfaces with the ability to impede biofilm formation and mitigate infection-related immune responses in mouse models. This research heralds the advent of biocompatible polymer nanoparticles and harbors significant implications in our ongoing combat against AMR. STATEMENT OF SIGNIFICANCE: The escalating prevalence of antimicrobial resistance (AMR) has been acknowledged as one of the most significant threats to global health. Therefore, a series of mixed-charge hyperbranched polymer nanoparticles (MCHPNs) with selective antibacterial action were designed and synthesized. Owing to inherent differences in membrane potential among diverse species and high hydrophilicity (Kow = 0.04), the optimal nanoparticles exhibited exceptional selective bactericidal effects over mammalian cells (selectivity index >564) and significantly delayed the emergence of bacterial resistance. Importantly, they endowed surfaces with the ability to impede biofilm formation and mitigate infection-related immune responses. Furthermore, the above findings focus on addressing the problem of AMR in Post-Pandemic, which will for sure attract attention from both academic and industry research.
Collapse
Affiliation(s)
- Yunyun Xue
- Key Laboratory of Macromolecular Synthesis and Functionalization (Ministry of Education), Department of Polymer Science and Engineering, Zhejiang University, Hangzhou 310027, China; Center of Healthcare Materials, Shaoxing Institute, Zhejiang University, Shaoxing 312000, China
| | - Chuyao Wang
- Key Laboratory of Macromolecular Synthesis and Functionalization (Ministry of Education), Department of Polymer Science and Engineering, Zhejiang University, Hangzhou 310027, China
| | - Yu Zhao
- Key Laboratory of Macromolecular Synthesis and Functionalization (Ministry of Education), Department of Polymer Science and Engineering, Zhejiang University, Hangzhou 310027, China
| | - Zihao Zhao
- Key Laboratory of Macromolecular Synthesis and Functionalization (Ministry of Education), Department of Polymer Science and Engineering, Zhejiang University, Hangzhou 310027, China
| | - Ronglu Cui
- Key Laboratory of Macromolecular Synthesis and Functionalization (Ministry of Education), Department of Polymer Science and Engineering, Zhejiang University, Hangzhou 310027, China
| | - Bin Du
- Key Laboratory of Macromolecular Synthesis and Functionalization (Ministry of Education), Department of Polymer Science and Engineering, Zhejiang University, Hangzhou 310027, China; Center of Healthcare Materials, Shaoxing Institute, Zhejiang University, Shaoxing 312000, China
| | - Lifeng Fang
- Key Laboratory of Macromolecular Synthesis and Functionalization (Ministry of Education), Department of Polymer Science and Engineering, Zhejiang University, Hangzhou 310027, China.
| | - Jianyu Wang
- Center of Healthcare Materials, Shaoxing Institute, Zhejiang University, Shaoxing 312000, China.
| | - Baoku Zhu
- Key Laboratory of Macromolecular Synthesis and Functionalization (Ministry of Education), Department of Polymer Science and Engineering, Zhejiang University, Hangzhou 310027, China; Center of Healthcare Materials, Shaoxing Institute, Zhejiang University, Shaoxing 312000, China.
| |
Collapse
|
7
|
Ripandelli RA, van Oijen AM, Robinson A. Single-Cell Microfluidics: A Primer for Microbiologists. J Phys Chem B 2024; 128:10311-10328. [PMID: 39400277 PMCID: PMC11514030 DOI: 10.1021/acs.jpcb.4c02746] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 04/29/2024] [Revised: 08/01/2024] [Accepted: 08/05/2024] [Indexed: 10/15/2024]
Abstract
Recent advances in microfluidic technology have made it possible to image live bacterial cells with a high degree of precision and control. In particular, single-cell microfluidic designs have created new opportunities to study phenotypic variation in bacterial populations. However, the development and use of microfluidic devices require specialized resources, and these can be practical barriers to entry for microbiologists. With this review, our intentions are to help demystify the design, construction, and application of microfluidics. Our approach is to present design elements as building blocks from which a multitude of microfluidics applications can be imagined by the microbiologist.
Collapse
|
8
|
Réthi-Nagy Z, Juhász S. Microbiome's Universe: Impact on health, disease and cancer treatment. J Biotechnol 2024; 392:161-179. [PMID: 39009231 DOI: 10.1016/j.jbiotec.2024.07.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 04/23/2024] [Revised: 05/27/2024] [Accepted: 07/07/2024] [Indexed: 07/17/2024]
Abstract
The human microbiome is a diverse ecosystem of microorganisms that reside in the body and influence various aspects of health and well-being. Recent advances in sequencing technology have brought to light microbial communities in organs and tissues that were previously considered sterile. The gut microbiota plays an important role in host physiology, including metabolic functions and immune modulation. Disruptions in the balance of the microbiome, known as dysbiosis, have been linked to diseases such as cancer, inflammatory bowel disease and metabolic disorders. In addition, the administration of antibiotics can lead to dysbiosis by disrupting the structure and function of the gut microbial community. Targeting strategies are the key to rebalancing the microbiome and fighting disease, including cancer, through interventions such as probiotics, fecal microbiota transplantation (FMT), and bacteria-based therapies. Future research must focus on understanding the complex interactions between diet, the microbiome and cancer in order to optimize personalized interventions. Multidisciplinary collaborations are essential if we are going to translate microbiome research into clinical practice. This will revolutionize approaches to cancer prevention and treatment.
Collapse
Affiliation(s)
- Zsuzsánna Réthi-Nagy
- Hungarian Centre of Excellence for Molecular Medicine, Cancer Microbiome Core Group, Budapesti út 9, Szeged H-6728, Hungary
| | - Szilvia Juhász
- Hungarian Centre of Excellence for Molecular Medicine, Cancer Microbiome Core Group, Budapesti út 9, Szeged H-6728, Hungary.
| |
Collapse
|
9
|
Duarte FC, Olak APS, Cardim SL, Danelli T, Magalhães GLG, Oliveira DMLD, Pimenta JDS, Vespero EC, Yamada-Ogatta SF, Perugini MRE. Phenotypic and genotypic characteristics of mecA - positive oxacillin-sensitive Staphylococcus aureus isolated from patients with bloodstream infection in a tertiary hospital in Southern Brazil. Braz J Microbiol 2024; 55:2705-2713. [PMID: 38896343 PMCID: PMC11405550 DOI: 10.1007/s42770-024-01420-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 01/29/2024] [Accepted: 06/04/2024] [Indexed: 06/21/2024] Open
Abstract
Staphylococcus aureus are extremely important microorganisms, either from an epidemiological point of view or as a pathogen, responsible for causing a series of infectious processes, whether simple, restricted to the skin, or invasive infections such as bacteremia. The emergence of Oxacillin Sensitive-Methicillin Resistant S.aureus (OS-MRSA) isolates has imposed difficulties in the treatment of patients with staphylococcal infection, as such isolates can be mistakenly classified as sensitive and lead to failure of the therapy used. Thus, the objective of this study is to evaluate the prevalence, and genotypic and phenotypic characteristics, of OS-MRSA isolates, from bloodstream infections, collected from patients admitted to a hospital in southern Brazil, as well as to evaluate the treatment used. For this, 801 unique isolates of S. aureus, collected from blood cultures, between January 2011 and December 2020 were evaluated. Of these, 96 isolates were classified as sensitive to oxacillin. The isolates were identified and had their sensitivity profile performed by manual and automated methods. The minimum inhibitory concentration for vancomycin, daptomycin, oxacillin, linezolid and teicoplanine was performed by e-test. The mecA, vanA genes, typing of the SCCmec elements, as well as the search for the icaA, tst-1 and pvl virulence genes were performed by PCR. Biofilm formation was performed using the crystal violet technique. The Sequence Type (ST), as well as the Clonal Complex (CC) of the isolates was evaluated by the RTq -PCR. The clinical characteristics of the patients were evaluated through an active search in medical records. After investigating the mecA gene, 27.1% (26/96) of the isolates were considered OS-MRSA, with SCCmec type I being the most prevalent, 46.1% (12/26). Among the evaluated isolates, 41% (9/22) were included in CC5 and ST9. As for virulence, all isolates were positive for the icaA gene and characterized as strong biofilm formers. The pvl gene was found in 92.3% (24/26) of the isolates and the toxic shock syndrome toxin was present in 61.5% of the isolates (16/26). All isolates were negative for the presence of the van A gene. As for the clinical outcome, 73% (19/26) of the patients were discharged from the hospital and 27% (7/26) died. It was possible to observe a high frequency of OS-MRSA isolates causing bloodstream infections. Furthermore, such isolates contain several virulence genes, which may contribute to a worse clinical outcome.
Collapse
Affiliation(s)
- Felipe Crepaldi Duarte
- Departamento de Patologia,Análises Clínicas e Toxicológicas,Programa de Pós-graduação em Fisiopatologia Clínica e Laboratorial,, Universidade Estadual de Londrina,, Londrina, Paraná, Brazil.
| | - Anna Paula Silva Olak
- Departamento de Patologia,Análises Clínicas e Toxicológicas,Programa de Pós-graduação em Fisiopatologia Clínica e Laboratorial,, Universidade Estadual de Londrina,, Londrina, Paraná, Brazil
| | - Stefani Lino Cardim
- Departamento de Patologia,Análises Clínicas e Toxicológicas,Programa de Pós-graduação em Fisiopatologia Clínica e Laboratorial,, Universidade Estadual de Londrina,, Londrina, Paraná, Brazil
| | - Tiago Danelli
- Departamento de Patologia,Análises Clínicas e Toxicológicas,Programa de Pós-graduação em Fisiopatologia Clínica e Laboratorial,, Universidade Estadual de Londrina,, Londrina, Paraná, Brazil
| | - Gerusa Luciana Gomes Magalhães
- Departamento de Patologia,Análises Clínicas e Toxicológicas,Programa de Pós-graduação em Fisiopatologia Clínica e Laboratorial,, Universidade Estadual de Londrina,, Londrina, Paraná, Brazil
- Laboratório de Análises Clínicas do Hospital Universitário, Universidade Estadual de Londrina, Londrina, Paraná, Brazil
| | - Deisy Mara Lima de Oliveira
- Departamento de Patologia,Análises Clínicas e Toxicológicas,Programa de Pós-graduação em Fisiopatologia Clínica e Laboratorial,, Universidade Estadual de Londrina,, Londrina, Paraná, Brazil
| | - Julia da Silva Pimenta
- Departamento de Patologia,Análises Clínicas e Toxicológicas,Programa de Pós-graduação em Fisiopatologia Clínica e Laboratorial,, Universidade Estadual de Londrina,, Londrina, Paraná, Brazil
| | - Eliana Carolina Vespero
- Departamento de Patologia,Análises Clínicas e Toxicológicas,Programa de Pós-graduação em Fisiopatologia Clínica e Laboratorial,, Universidade Estadual de Londrina,, Londrina, Paraná, Brazil
- Laboratório de Análises Clínicas do Hospital Universitário, Universidade Estadual de Londrina, Londrina, Paraná, Brazil
| | - Sueli Fumie Yamada-Ogatta
- Departamento de Patologia,Análises Clínicas e Toxicológicas,Programa de Pós-graduação em Fisiopatologia Clínica e Laboratorial,, Universidade Estadual de Londrina,, Londrina, Paraná, Brazil
- Laboratório de Biologia Molecular de Microrganismos,Departamento de Microbiologia,Programa de Pós-graduação em Microbiologia, Universidade Estadual de Londrina, Londrina, Paraná, Brazil
| | - Marcia Regina Eches Perugini
- Departamento de Patologia,Análises Clínicas e Toxicológicas,Programa de Pós-graduação em Fisiopatologia Clínica e Laboratorial,, Universidade Estadual de Londrina,, Londrina, Paraná, Brazil
- Laboratório de Análises Clínicas do Hospital Universitário, Universidade Estadual de Londrina, Londrina, Paraná, Brazil
| |
Collapse
|
10
|
Changsan N, Atipairin A, Sakdiset P, Muenraya P, Balekar N, Srichana T, Sritharadol R, Phanapithakkun S, Sawatdee S. BrSPR-20-P1 peptide isolated from Brevibacillus sp. developed into liposomal hydrogel as a potential topical antimicrobial agent. RSC Adv 2024; 14:27394-27411. [PMID: 39205932 PMCID: PMC11351071 DOI: 10.1039/d4ra03722g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 05/20/2024] [Accepted: 08/15/2024] [Indexed: 09/04/2024] Open
Abstract
A novel BrSPR-20-P1 antimicrobial peptide (P1-AMP; NH2-VVVNVLVKVLPPPVV-COOH) isolated from Brevibacillus sp. SPR-20 was encapsulated in a liposome containing varying proportions of l-α-phosphatidylcholine (PC) and cholesterol (CH). P1-AMP liposomes were incorporated into a chitosan hydrogel to achieve a peptide concentration of 0.02%. P1-AMP has been tested for its antibacterial and in vitro wound healing activities. The physicochemical characteristics of liposomes and hydrogel were investigated, including in vitro drug release, permeability, cell toxicity, antimicrobial activities, and stability studies. P1-AMP showed higher antimicrobial and wound-healing activities than the negative control. A toxicity test of P1-AMP in keratinocyte cell lines revealed cell viability of 100% at a concentration range of 1.96-1000 μg mL-1. The empty liposomes exhibited an average particle size ranging from 324.5 ± 8.6 to 1823.7 ± 288.2 nm. The size range of P1-AMP liposomes was 378.6 ± 14.0 to 2363.0 ± 255.6 nm. The zeta potential of the blank liposome ranged from -40.43 ± 2.51 to -60.17 ± 0.93 mV and it decreased to -57.33 ± 0.72 to -70.33 ± 0.15 mV of the liposome loaded with peptide. SEM images showed liposomes were ovoid spheres with smooth surfaces. The chosen formulation, composed of PC to CH in an 18 : 1 ratio (formulation F3), had the highest entrapment effectiveness with small particle size and possessed an acceptable zeta potential. The developed P1-AMP liposome-loaded hydrogels exhibited a yellowish-clear appearance with a viscosity of 758.0 ± 149.8 cPs. The P1-AMP was rapidly released from the P1-AMP-loaded liposome hydrogel formulation. The P1-AMP-loaded liposome showed high permeability compared to P1-AMP alone or P1-AMP in hydrogel without the incorporation of liposomes. The minimum inhibitory concentration (MIC) against Staphylococcus aureus and methicillin-resistant S. aureus (MRSA) of P1-AMP-loaded liposome hydrogel was 2 μg mL-1, equivalent to P1-AMP. It completely killed S. aureus at 10× and 5× MIC after 6 and 12 h of incubation, respectively. The formulation did not induce cytotoxicity to the tested keratinocyte cell and remained stable for at least 6 months under the studied conditions.
Collapse
Affiliation(s)
- Narumon Changsan
- College of Pharmacy, Rangsit University Pathumtani 12000 Thailand
| | - Apichart Atipairin
- School of Pharmacy, Walailak University Thasala Nakhon Si Thammarat 80160 Thailand
- Drug and Cosmetics Excellence Center, Walailak University Thasala Nakhon Si Thammarat 80160 Thailand
| | - Pajaree Sakdiset
- School of Pharmacy, Walailak University Thasala Nakhon Si Thammarat 80160 Thailand
- Drug and Cosmetics Excellence Center, Walailak University Thasala Nakhon Si Thammarat 80160 Thailand
| | - Poowadon Muenraya
- School of Pharmacy, Walailak University Thasala Nakhon Si Thammarat 80160 Thailand
- Drug and Cosmetics Excellence Center, Walailak University Thasala Nakhon Si Thammarat 80160 Thailand
| | - Neelam Balekar
- College of Pharmacy, IPS Academy Indore Madhya Pradesh 452012 India
| | - Teerapol Srichana
- Drug Delivery System Excellence Center, Department of Pharmaceutical Technology Faculty of Pharmaceutical Sciences, Prince of Songkla University Hat Yai Songkhla 90112 Thailand
| | - Rutthapol Sritharadol
- Faculty of Pharmaceutical Sciences, Chulalongkorn University Phaya Thai Road, Pathum Wan Bangkok 10330 Thailand
| | - Suranate Phanapithakkun
- School of Pharmacy, Walailak University Thasala Nakhon Si Thammarat 80160 Thailand
- Drug and Cosmetics Excellence Center, Walailak University Thasala Nakhon Si Thammarat 80160 Thailand
| | - Somchai Sawatdee
- School of Pharmacy, Walailak University Thasala Nakhon Si Thammarat 80160 Thailand
- Drug and Cosmetics Excellence Center, Walailak University Thasala Nakhon Si Thammarat 80160 Thailand
| |
Collapse
|
11
|
Kaplan JB, Sukhishvili SA, Sailer M, Kridin K, Ramasubbu N. Aggregatibacter actinomycetemcomitans Dispersin B: The Quintessential Antibiofilm Enzyme. Pathogens 2024; 13:668. [PMID: 39204268 PMCID: PMC11357414 DOI: 10.3390/pathogens13080668] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 07/19/2024] [Revised: 07/30/2024] [Accepted: 08/06/2024] [Indexed: 09/03/2024] Open
Abstract
The extracellular matrix of most bacterial biofilms contains polysaccharides, proteins, and nucleic acids. These biopolymers have been shown to mediate fundamental biofilm-related phenotypes including surface attachment, intercellular adhesion, and biocide resistance. Enzymes that degrade polymeric biofilm matrix components, including glycoside hydrolases, proteases, and nucleases, are useful tools for studying the structure and function of biofilm matrix components and are also being investigated as potential antibiofilm agents for clinical use. Dispersin B is a well-studied, broad-spectrum antibiofilm glycoside hydrolase produced by Aggregatibacter actinomycetemcomitans. Dispersin B degrades poly-N-acetylglucosamine, a biofilm matrix polysaccharide that mediates biofilm formation, stress tolerance, and biocide resistance in numerous Gram-negative and Gram-positive pathogens. Dispersin B has been shown to inhibit biofilm and pellicle formation; detach preformed biofilms; disaggregate bacterial flocs; sensitize preformed biofilms to detachment by enzymes, detergents, and metal chelators; and sensitize preformed biofilms to killing by antiseptics, antibiotics, bacteriophages, macrophages, and predatory bacteria. This review summarizes the results of nearly 100 in vitro and in vivo studies that have been carried out on dispersin B since its discovery 20 years ago. These include investigations into the biological function of the enzyme, its structure and mechanism of action, and its in vitro and in vivo antibiofilm activities against numerous bacterial species. Also discussed are potential clinical applications of dispersin B.
Collapse
Affiliation(s)
- Jeffrey B. Kaplan
- Laboratory for Skin Research, Institute for Medical Research, Galilee Medical Center, Nahariya 2210001, Israel;
| | - Svetlana A. Sukhishvili
- Department of Materials Science and Engineering, Texas A&M University, College Station, TX 77843, USA;
| | | | - Khalaf Kridin
- Laboratory for Skin Research, Institute for Medical Research, Galilee Medical Center, Nahariya 2210001, Israel;
- The Azrieli Faculty of Medicine, Bar-Ilan University, Safed 1311502, Israel
| | - Narayanan Ramasubbu
- Department of Oral Biology, Rutgers School of Dental Medicine, Newark, NJ 07103, USA;
| |
Collapse
|
12
|
Al-Momani H, Aolymat I, Ibrahim L, Albalawi H, Al Balawi D, Albiss BA, Almasri M, Alghweiri S. Low-dose zinc oxide nanoparticles trigger the growth and biofilm formation of Pseudomonas aeruginosa: a hormetic response. BMC Microbiol 2024; 24:290. [PMID: 39095741 PMCID: PMC11297655 DOI: 10.1186/s12866-024-03441-y] [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] [Academic Contribution Register] [Received: 02/22/2024] [Accepted: 07/25/2024] [Indexed: 08/04/2024] Open
Abstract
INTRODUCTION Hormesis describes an inverse dose-response relationship, whereby a high dose of a toxic compound is inhibitory, and a low dose is stimulatory. This study explores the hormetic response of low concentrations of zinc oxide nanoparticles (ZnO NPs) toward Pseudomonas aeruginosa. METHOD Samples of P. aeruginosa, i.e. the reference strain, ATCC 27,853, together with six strains recovered from patients with cystic fibrosis, were exposed to ten decreasing ZnO NPs doses (0.78-400 µg/mL). The ZnO NPs were manufactured from Peganum harmala using a chemical green synthesis approach, and their properties were verified utilizing X-ray diffraction and scanning electron microscopy. A microtiter plate technique was employed to investigate the impact of ZnO NPs on the growth, biofilm formation and metabolic activity of P. aeruginosa. Real-time polymerase chain reactions were performed to determine the effect of ZnO NPs on the expression of seven biofilm-encoding genes. RESULT The ZnO NPs demonstrated concentration-dependent bactericidal and antibiofilm efficiency at concentrations of 100-400 µg/mL. However, growth was significantly stimulated at ZnO NPs concentration of 25 µg/mL (ATCC 27853, Pa 3 and Pa 4) and at 12.5 µg/mL and 6.25 µg/mL (ATCC 27853, Pa 2, Pa 4 and Pa 5). No significant positive growth was detected at dilutions < 6.25 µg/mL. similarly, biofilm formation was stimulated at concentration of 12.5 µg/mL (ATCC 27853 and Pa 1) and at 6.25 µg/mL (Pa 4). At concentration of 12.5 µg/mL, ZnO NPs upregulated the expression of LasB ( ATCC 27853, Pa 1 and Pa 4) and LasR and LasI (ATCC 27853 and Pa 1) as well as RhII expression (ATCC 27853, Pa 2 and Pa 4). CONCLUSION When exposed to low ZnO NPs concentrations, P. aeruginosa behaves in a hormetic manner, undergoing positive growth and biofilm formation. These results highlight the importance of understanding the response of P. aeruginosa following exposure to low ZnO NPs concentrations.
Collapse
Affiliation(s)
- Hafez Al-Momani
- Department of Microbiology, Pathology and Forensic Medicine, Faculty of Medicine, The Hashemite University, P.O box 330127, Zarqa, 13133, Jordan.
| | - Iman Aolymat
- Department of Anatomy, Physiology and Biochemistry, Faculty of Medicine, The Hashemite University, P.O box 330127, Zarqa, 13133, Jordan
| | - Lujain Ibrahim
- Faculty of Applied Medical Sciences, The Hashemite University, Zarqa, 13133, Jordan
| | - Hadeel Albalawi
- Faculty of Applied Medical Sciences, The Hashemite University, Zarqa, 13133, Jordan
| | - Dua'a Al Balawi
- Faculty of Applied Medical Sciences, The Hashemite University, Zarqa, 13133, Jordan
| | - Borhan Aldeen Albiss
- Nanotechnology Institute, Jordan University of Science & Technology, P.O. Box 3030, Irbid, 22110, Jordan
| | - Muna Almasri
- Faculty of Applied Medical Sciences, The Hashemite University, Zarqa, 13133, Jordan
| | - Sahar Alghweiri
- Medical Laboratory Department, Prince Hashem Military Hospital, Zarqa, 13133, Jordan
| |
Collapse
|
13
|
Wang H, Hu S, Pei Y, Sun H. Nordalbergin Synergizes with Novel β-Lactam Antibiotics against MRSA Infection. Int J Mol Sci 2024; 25:7704. [PMID: 39062947 PMCID: PMC11277203 DOI: 10.3390/ijms25147704] [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] [Academic Contribution Register] [Received: 06/23/2024] [Revised: 07/11/2024] [Accepted: 07/11/2024] [Indexed: 07/28/2024] Open
Abstract
The synergetic strategy has created tremendous advantages in drug-resistance bacterial infection treatment, whereas challenges related to novel compound discovery and identifying drug-binding targets still remain. The mechanisms of antimicrobial resistance involving β-lactamase catalysis and the degradation of β-lactam antibiotics are being revealed, with relevant therapies promising to improve the efficacy of existing major classes of antibiotics in the foreseeable future. In this study, it is demonstrated that nordalbergin, a coumarin isolated from the wood bark of Dalbergia sissoo, efficiently potentiated the activities of β-lactam antibiotics against methicillin-resistant Staphylococcus aureus (MRSA) by suppressing β-lactamase performance and improving the bacterial biofilm susceptibility to antibiotics. Nordalbergin was found to destabilize the cell membrane and promote its permeabilization. Moreover, nordalbergin efficiently improved the therapeutic efficacy of amoxicillin against MRSA pneumonia in mice, as supported by the lower bacterial load, attenuated pathological damage, and decreased inflammation level. These results demonstrate that nordalbergin might be a promising synergist of amoxicillin against MRSA infections. This study provided a new approach for developing potentiators for β-lactam antibiotics against MRSA infections.
Collapse
Affiliation(s)
- Haiting Wang
- College of Animal Sciences, Zhejiang University, Hangzhou 310058, China;
| | - Sangyu Hu
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Institute of Zoonosis, College of Veterinary Medicine, Jilin University, Changchun 130062, China; (S.H.); (Y.P.)
- Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, College of Veterinary Medicine, Jilin University, Changchun 130062, China
| | - Yuzhu Pei
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Institute of Zoonosis, College of Veterinary Medicine, Jilin University, Changchun 130062, China; (S.H.); (Y.P.)
- Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, College of Veterinary Medicine, Jilin University, Changchun 130062, China
| | - Hongxiang Sun
- College of Animal Sciences, Zhejiang University, Hangzhou 310058, China;
| |
Collapse
|
14
|
Srivastava P, Shukla A, Singh R, Kant R, Mishra N, Behera SP, Dwivedi GR, Yadav DK. Orientia tsutsugamushi: An Unusual Intracellular Bacteria-Adaptation Strategies, Available Antibiotics, and Alternatives for Treatment. Curr Microbiol 2024; 81:236. [PMID: 38907107 DOI: 10.1007/s00284-024-03754-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 11/16/2023] [Accepted: 05/28/2024] [Indexed: 06/23/2024]
Abstract
During evolution Orientia tsutsugamushi became a smarter obligate bacterium to establish as intracellular pathogens. O. tsutsugamushi is a human pathogenic bacterium responsible for 1 billion infections of scrub typhus. Several novel mechanisms make this bacterium unique (cell wall, genetic constitutions, secretion system, etc.). In 2007, O. tsutsugamushi Boryong was pioneer strain for whole-genome sequencing. But the fundamental biology of this bacterial cell is a mystery till date. The unusual biology makes this organism as model for host cell interaction. Only a few antibiotics are effective against this intracellular pathogen but emergence of less susceptibility toward antibiotics make the situation alarming. The review was captivated to highlight the unusual aspects of adaptation, antibiotics, and drugs beyond antibiotics.
Collapse
Affiliation(s)
- Prashansha Srivastava
- ICMR-Regional Medical Research Centre, BRD Medical College Campus, Gorakhpur, 273013, India
| | - Aishwarya Shukla
- ICMR-Regional Medical Research Centre, BRD Medical College Campus, Gorakhpur, 273013, India
| | - Rajeev Singh
- ICMR-Regional Medical Research Centre, BRD Medical College Campus, Gorakhpur, 273013, India
| | - Rajni Kant
- ICMR-Regional Medical Research Centre, BRD Medical College Campus, Gorakhpur, 273013, India
| | - Nalini Mishra
- ICMR-Regional Medical Research Centre, BRD Medical College Campus, Gorakhpur, 273013, India
| | - Sthita P Behera
- ICMR-Regional Medical Research Centre, BRD Medical College Campus, Gorakhpur, 273013, India
| | - Gaurav R Dwivedi
- ICMR-Regional Medical Research Centre, BRD Medical College Campus, Gorakhpur, 273013, India.
| | - Dharmendra K Yadav
- College of Pharmacy, Gachon University of Medicine and Science, Hambakmoeiro 191, Yeonsu-Gu, Incheon, 21924, Korea
| |
Collapse
|
15
|
Xiu W, Dong H, Chen X, Wan L, Lu L, Yang K, Yuwen L, Li Q, Ding M, Zhang Y, Mou Y, Wang L. Metabolic Modulation-Mediated Antibiotic and Immune Activation for Treatment of Chronic Lung Infections. ACS NANO 2024; 18:15204-15217. [PMID: 38803167 DOI: 10.1021/acsnano.4c03527] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Academic Contribution Register] [Indexed: 05/29/2024]
Abstract
The Pseudomonas aeruginosa biofilm in recalcitrant chronic lung infections not only develops high antimicrobial tolerance but also induces an aberrant host inflammatory response. The metabolic condition plays a vital role in both the antimicrobial susceptibility of bacteria and the inflammatory response of immune cells, thereby offering a potential therapeutic target. Herein, we described a metabolic modulation strategy by using ultrasound-responsive liposomal nanoparticles containing a sonosensitizer and a hypoxia-activated prodrug against biofilm-associated chronic lung infections. Under ultrasound stimulation, the sonosensitizer generates antibacterial reactive oxygen species by oxygen consumption. Subsequently, the oxygen consumption-mediated hypoxia not only induces the anaerobic metabolism of bacteria for antibiotic activation but also triggers the glycolysis pathway of immune cells for inflammatory activation. Such metabolic modulation strategy demonstrated efficient therapeutic efficacy for P. aeruginosa biofilm-induced chronic lung infections in mice models and provides a promising way for combating biofilm-associated chronic infections.
Collapse
Affiliation(s)
- Weijun Xiu
- State Key Laboratory of Organic Electronics and Information Displays & Jiangsu Key Laboratory for Biosensors, Institute of Advanced Materials (IAM), Nanjing University of Posts and Telecommunications, Nanjing210023, China
| | - Heng Dong
- Nanjing Stomatological Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing 210008, China
| | - Xiaolong Chen
- State Key Laboratory of Organic Electronics and Information Displays & Jiangsu Key Laboratory for Biosensors, Institute of Advanced Materials (IAM), Nanjing University of Posts and Telecommunications, Nanjing210023, China
| | - Ling Wan
- State Key Laboratory of Organic Electronics and Information Displays & Jiangsu Key Laboratory for Biosensors, Institute of Advanced Materials (IAM), Nanjing University of Posts and Telecommunications, Nanjing210023, China
| | - Liang Lu
- State Key Laboratory of Organic Electronics and Information Displays & Jiangsu Key Laboratory for Biosensors, Institute of Advanced Materials (IAM), Nanjing University of Posts and Telecommunications, Nanjing210023, China
| | - Kaili Yang
- State Key Laboratory of Organic Electronics and Information Displays & Jiangsu Key Laboratory for Biosensors, Institute of Advanced Materials (IAM), Nanjing University of Posts and Telecommunications, Nanjing210023, China
| | - Lihui Yuwen
- State Key Laboratory of Organic Electronics and Information Displays & Jiangsu Key Laboratory for Biosensors, Institute of Advanced Materials (IAM), Nanjing University of Posts and Telecommunications, Nanjing210023, China
| | - Qiang Li
- Nanjing Stomatological Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing 210008, China
| | - Meng Ding
- Nanjing Stomatological Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing 210008, China
| | - Yu Zhang
- Nanjing Stomatological Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing 210008, China
| | - Yongbin Mou
- Nanjing Stomatological Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing 210008, China
| | - Lianhui Wang
- State Key Laboratory of Organic Electronics and Information Displays & Jiangsu Key Laboratory for Biosensors, Institute of Advanced Materials (IAM), Nanjing University of Posts and Telecommunications, Nanjing210023, China
| |
Collapse
|
16
|
Brar NK, Dhariwal A, Åmdal HA, Junges R, Salvadori G, Baker JL, Edlund A, Petersen FC. Exploring ex vivo biofilm dynamics: consequences of low ampicillin concentrations on the human oral microbiome. NPJ Biofilms Microbiomes 2024; 10:37. [PMID: 38565843 PMCID: PMC10987642 DOI: 10.1038/s41522-024-00507-7] [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] [Academic Contribution Register] [Received: 08/28/2023] [Accepted: 03/20/2024] [Indexed: 04/04/2024] Open
Abstract
Prolonged exposure to antibiotics at low concentration can promote processes associated with bacterial biofilm formation, virulence and antibiotic resistance. This can be of high relevance in microbial communities like the oral microbiome, where commensals and pathogens share a common habitat and where the total abundance of antibiotic resistance genes surpasses the abundance in the gut. Here, we used an ex vivo model of human oral biofilms to investigate the impact of ampicillin on biofilm viability. The ecological impact on the microbiome and resistome was investigated using shotgun metagenomics. The results showed that low concentrations promoted significant shifts in microbial taxonomic profile and could enhance biofilm viability by up to 1 to 2-log. For the resistome, low concentrations had no significant impact on antibiotic resistance gene (ARG) diversity, while ARG abundance decreased by up to 84%. A positive correlation was observed between reduced microbial diversity and reduced ARG abundance. The WHO priority pathogens Streptococcus pneumoniae and Staphylococcus aureus were identified in some of the samples, but their abundance was not significantly altered by ampicillin. Most of the antibiotic resistance genes that increased in abundance in the ampicillin group were associated with streptococci, including Streptococcus mitis, a well-known potential donor of ARGs to S. pneumoniae. Overall, the results highlight the potential of using the model to further our understanding of ecological and evolutionary forces driving antimicrobial resistance in oral microbiomes.
Collapse
Affiliation(s)
- N K Brar
- Institute of Oral Biology, Faculty of Dentistry, University of Oslo, Oslo, Norway
| | - A Dhariwal
- Institute of Oral Biology, Faculty of Dentistry, University of Oslo, Oslo, Norway
| | - H A Åmdal
- Institute of Oral Biology, Faculty of Dentistry, University of Oslo, Oslo, Norway
| | - R Junges
- Institute of Oral Biology, Faculty of Dentistry, University of Oslo, Oslo, Norway
| | - G Salvadori
- Institute of Oral Biology, Faculty of Dentistry, University of Oslo, Oslo, Norway
| | - J L Baker
- Department of Oral Rehabilitation & Biosciences, Oregon Health & Science University, Portland, OR, USA
- Microbial and Environmental Genomics, J. Craig Venter Institute, La Jolla, CA, USA
- Department of Pediatrics, UC San Diego School of Medicine, La Jolla, CA, USA
| | - A Edlund
- Microbial and Environmental Genomics, J. Craig Venter Institute, La Jolla, CA, USA
- Department of Pediatrics, UC San Diego School of Medicine, La Jolla, CA, USA
| | - F C Petersen
- Institute of Oral Biology, Faculty of Dentistry, University of Oslo, Oslo, Norway.
| |
Collapse
|
17
|
Jiang L, Xu Q, Wu Y, Zhou X, Chen Z, Sun Q, Wen J. Characterization of a Straboviridae phage vB_AbaM-SHI and its inhibition effect on biofilms of Acinetobacter baumannii. Front Cell Infect Microbiol 2024; 14:1351993. [PMID: 38524182 PMCID: PMC10958429 DOI: 10.3389/fcimb.2024.1351993] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 12/15/2023] [Accepted: 02/22/2024] [Indexed: 03/26/2024] Open
Abstract
Acinetobacter baumannii (A. baumannii) is a popular clinical pathogen worldwide. Biofilm-associated antibiotic-resistant A. baumannii infection poses a great threat to human health. Bacteria in biofilms are highly resistant to antibiotics and disinfectants. Furthermore, inhibition or eradication of biofilms in husbandry, the food industry and clinics are almost impossible. Phages can move across the biofilm matrix and promote antibiotic penetration. In the present study, a lytic A. baumannii phage vB_AbaM-SHI, belonging to family Straboviridae, was isolated from sauce chop factory drain outlet in Wuxi, China. The DNA genome consists of 44,180 bp which contain 93 open reading frames, and genes encoding products morphogenesis are located at the end of the genome. The amino acid sequence of vB_AbaM-SHI endolysin is different from those of previously reported A. baumannii phages in NCBI. Phage vB_AbaM-SHI endolysin has two additional β strands due to the replacement of a lysine (K) (in KU510289.1, NC_041857.1, JX976549.1 and MH853786.1) with an arginine (R) (SHI) at position 21 of A. baumannii phage endolysin. Spot test showed that phage vB_AbaM-SHI is able to lyse some antibiotic-resistant bacteria, such as A. baumannii (SL, SL1, and SG strains) and E. coli BL21 strain. Additionally, phage vB_AbaM-SHI independently killed bacteria and inhibited bacterial biofilm formation, and synergistically exerted strong antibacterial effects with antibiotics. This study provided a new perspective into the potential application value of phage vB_AbaM-SHI as an antimicrobial agent.
Collapse
Affiliation(s)
- Liming Jiang
- School of Basic Medical Sciences, Health Science Center, Ningbo University, Ningbo, Zhejiang, China
| | - Qian Xu
- Department of Blood Transfusion, Hubei No. 3 People’s Hospital of Jianghan University, Wuhan, Hubei, China
| | - Ying Wu
- Department of Rheumatology Immunology, The First People’s Hospital of Hefei, Hefei, Anhui, China
| | - Xianglian Zhou
- Department of Rheumatology Immunology, The First People’s Hospital of Hefei, Hefei, Anhui, China
| | - Zhu Chen
- Department of Laboratory, Ningbo No.2 Hospital, Ningbo, Zhejiang, China
| | - Qiangming Sun
- National Kunming High-level Biosafety Primate Research Center, Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming, Yunnan, China
| | - Jinsheng Wen
- School of Basic Medical Sciences, Health Science Center, Ningbo University, Ningbo, Zhejiang, China
| |
Collapse
|
18
|
Marín EM, Reyes MG, Audisio MC, Nicotra VE, Uriburu ML. Antibacterial Sesquiterpene Lactones from Kaunia lasiophthalma. Chem Biodivers 2024; 21:e202301379. [PMID: 38345295 DOI: 10.1002/cbdv.202301379] [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] [Academic Contribution Register] [Received: 09/06/2023] [Accepted: 01/24/2024] [Indexed: 03/16/2024]
Abstract
Phytochemical study of the species Kaunia lasiophthalma enabled the isolation of three undescribed and three known guaianolide-type sesquiterpene lactones, and one new benzofuran. The bioguided fractionation methodology was successful in detecting antimicrobial metabolites against Staphylococcus aureus strains and permitted the description of undescribed guaianolide-type sesquiterpene lactones with substitution patterns matching all those described in the Oxylobinae subtribe.
Collapse
Affiliation(s)
- Edith M Marín
- Instituto de Investigaciones para la Industria Química (INIQUI-CONICET), Universidad Nacional de Salta, Av. Bolivia 5150, 4400, Salta, Argentina
| | - María G Reyes
- Instituto de Investigaciones para la Industria Química (INIQUI-CONICET), Universidad Nacional de Salta, Av. Bolivia 5150, 4400, Salta, Argentina
| | - Marcela C Audisio
- Instituto de Investigaciones para la Industria Química (INIQUI-CONICET), Universidad Nacional de Salta, Av. Bolivia 5150, 4400, Salta, Argentina
| | - Viviana E Nicotra
- Facultad de Ciencias Químicas, Instituto Multidisciplinario de Biología Vegetal (IMBIV-CONICET), Universidad Nacional de Córdoba, Casilla de Correo 495, 5000, Córdoba, Argentina
| | - María L Uriburu
- Instituto de Investigaciones para la Industria Química (INIQUI-CONICET), Universidad Nacional de Salta, Av. Bolivia 5150, 4400, Salta, Argentina
| |
Collapse
|
19
|
Xiu W, Li X, Li Q, Ding M, Zhang Y, Wan L, Wang S, Gao Y, Mou Y, Wang L, Dong H. Ultrasound-Stimulated "Exocytosis" by Cell-Like Microbubbles Enhances Antibacterial Species Penetration and Immune Activation Against Implant Infection. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2307048. [PMID: 38109089 PMCID: PMC10933665 DOI: 10.1002/advs.202307048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Academic Contribution Register] [Received: 09/25/2023] [Revised: 11/18/2023] [Indexed: 12/19/2023]
Abstract
Host immune systems serving as crucial defense lines are vital resisting mechanisms against biofilm-associated implant infections. Nevertheless, biofilms hinder the penetration of anti-bacterial species, inhibit phagocytosis of immune cells, and frustrate host inflammatory responses, ultimately resulting in the weakness of the host immune system for biofilm elimination. Herein, a cell-like construct is developed through encapsulation of erythrocyte membrane fragments on the surface of Fe3 O4 nanoparticle-fabricated microbubbles and then loaded with hydroxyurea (EMB-Hu). Under ultrasound (US) stimulation, EMB-Hu undergoes a stable oscillation manner to act in an "exocytosis" mechanism for disrupting biofilm, releasing agents, and enhancing penetration of catalytically generated anti-bacterial species within biofilms. Additionally, the US-stimulated "exocytosis" by EMB-Hu can activate pro-inflammatory macrophage polarization and enhance macrophage phagocytosis for clearance of disrupted biofilms. Collectively, this work has exhibited cell-like microbubbles with US-stimulated "exocytosis" mechanisms to overcome the biofilm barrier and signal macrophages for inflammatory activation, finally achieving favorable therapeutic effects against implant infections caused by methicillin-resistant Staphylococcus aureus (MRSA) biofilms.
Collapse
Affiliation(s)
- Weijun Xiu
- Nanjing Stomatological HospitalAffiliated Hospital of Medical SchoolNanjing University30 Zhongyang RoadNanjing210008P. R. China
- Key Laboratory for Organic Electronics and Information DisplaysJiangsu Key Laboratory for BiosensorsInstitute of Advanced MaterialsJiangsu National Synergetic Innovation Centre for Advanced MaterialsNanjing University of Posts and Telecommunications9 Wenyuan RoadNanjing210023P. R. China
| | - Xiaoye Li
- Nanjing Stomatological HospitalAffiliated Hospital of Medical SchoolNanjing University30 Zhongyang RoadNanjing210008P. R. China
| | - Qiang Li
- Nanjing Stomatological HospitalAffiliated Hospital of Medical SchoolNanjing University30 Zhongyang RoadNanjing210008P. R. China
| | - Meng Ding
- Nanjing Stomatological HospitalAffiliated Hospital of Medical SchoolNanjing University30 Zhongyang RoadNanjing210008P. R. China
| | - Yu Zhang
- Nanjing Stomatological HospitalAffiliated Hospital of Medical SchoolNanjing University30 Zhongyang RoadNanjing210008P. R. China
| | - Ling Wan
- Key Laboratory for Organic Electronics and Information DisplaysJiangsu Key Laboratory for BiosensorsInstitute of Advanced MaterialsJiangsu National Synergetic Innovation Centre for Advanced MaterialsNanjing University of Posts and Telecommunications9 Wenyuan RoadNanjing210023P. R. China
| | - Siyu Wang
- Key Laboratory for Organic Electronics and Information DisplaysJiangsu Key Laboratory for BiosensorsInstitute of Advanced MaterialsJiangsu National Synergetic Innovation Centre for Advanced MaterialsNanjing University of Posts and Telecommunications9 Wenyuan RoadNanjing210023P. R. China
| | - Yu Gao
- Key Laboratory for Organic Electronics and Information DisplaysJiangsu Key Laboratory for BiosensorsInstitute of Advanced MaterialsJiangsu National Synergetic Innovation Centre for Advanced MaterialsNanjing University of Posts and Telecommunications9 Wenyuan RoadNanjing210023P. R. China
| | - Yongbin Mou
- Nanjing Stomatological HospitalAffiliated Hospital of Medical SchoolNanjing University30 Zhongyang RoadNanjing210008P. R. China
| | - Lianhui Wang
- Key Laboratory for Organic Electronics and Information DisplaysJiangsu Key Laboratory for BiosensorsInstitute of Advanced MaterialsJiangsu National Synergetic Innovation Centre for Advanced MaterialsNanjing University of Posts and Telecommunications9 Wenyuan RoadNanjing210023P. R. China
| | - Heng Dong
- Nanjing Stomatological HospitalAffiliated Hospital of Medical SchoolNanjing University30 Zhongyang RoadNanjing210008P. R. China
| |
Collapse
|
20
|
Zou Y, Li X, Mao Y, Song W, Liu Q. Enhanced Biofilm Formation by Tetracycline in a Staphylococcus aureus Naturally Lacking ica Operon and atl. Microb Drug Resist 2024; 30:82-90. [PMID: 38252794 DOI: 10.1089/mdr.2023.0186] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 01/24/2024] Open
Abstract
Staphylococcus aureus is a major, widespread pathogen, and its biofilm-forming characteristics make it even more difficult to eliminate by biocides. Tetracycline (TCY) is a major broad-spectrum antibiotic, the residues of which can cause deleterious health impacts, and subinhibitory concentrations of TCY have the potential to increase biofilm formation in S. aureus. In this study, we showed how the biofilm formation of S. aureus 123786 is enhanced in the presence of TCY at specific subinhibitory concentrations. S. aureus 123786 used in this study was identified as Staphylococcal Cassette Chromosome mec III, sequence type239 and naturally lacking ica operon and atl gene. Two assays were performed to quantify the formation of S. aureus biofilm. In the crystal violet (CV) assay, the absorbance values of biofilm stained with CV at optical density (OD)540 nm increased after 8 and 16 hr of incubation when the concentration of TCY was 1/2 minimum inhibitory concentration (MIC), whereas at the concentration of 1/16 MIC, the absorbance values increased after 16 and 24 hr of incubation. In tetrazolium salt reduction assay, the absorbance value at OD490 nm of S. aureus 123786 biofilms mixed with 3-(4,5-dimethylthiazol-2-yl)-5-(3-carboxymethoxyphenyl)-2-(4-sulfophenyl)-2H-tetrazolium solution increased after 8 hr when the concentration of TCY was 1/4 MIC, which may be correlated with the higher proliferation and maturation of biofilm. In conclusion, the biofilm formation of S. aureus 123786 could be enhanced in the presence of TCY at specific subinhibitory concentrations.
Collapse
Affiliation(s)
- Yimin Zou
- Key Laboratory of Respiratory Disease of Zhejiang Province, Department of Respiratory and Critical Care Medicine, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Xuejie Li
- Guangdong Province Key Laboratory for Green Processing of Natural Products and Product Safety, Engineering Research Center of Starch and Vegetable Protein Processing Ministry of Education, School of Food Science and Engineering, South China University of Technology, Guangzhou, China
| | - Yanxiong Mao
- Key Laboratory of Respiratory Disease of Zhejiang Province, Department of Respiratory and Critical Care Medicine, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Wenjuan Song
- Department of Economics, School of Economics and Management, Zhejiang Sci-Tech University, Hangzhou, China
| | - Qing Liu
- Key Laboratory of Respiratory Disease of Zhejiang Province, Department of Respiratory and Critical Care Medicine, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
21
|
Alfei S. Shifting from Ammonium to Phosphonium Salts: A Promising Strategy to Develop Next-Generation Weapons against Biofilms. Pharmaceutics 2024; 16:80. [PMID: 38258091 PMCID: PMC10819902 DOI: 10.3390/pharmaceutics16010080] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 11/24/2023] [Revised: 01/03/2024] [Accepted: 01/04/2024] [Indexed: 01/24/2024] Open
Abstract
Since they are difficult and sometimes impossible to treat, infections sustained by multidrug-resistant (MDR) pathogens, emerging especially in nosocomial environments, are an increasing global public health concern, translating into high mortality and healthcare costs. In addition to having acquired intrinsic abilities to resist available antibiotic treatments, MDR bacteria can transmit genetic material encoding for resistance to non-mutated bacteria, thus strongly decreasing the number of available effective antibiotics. Moreover, several pathogens develop resistance by forming biofilms (BFs), a safe and antibiotic-resistant home for microorganisms. BFs are made of well-organized bacterial communities, encased and protected in a self-produced extracellular polymeric matrix, which impedes antibiotics' ability to reach bacteria, thus causing them to lose efficacy. By adhering to living or abiotic surfaces in healthcare settings, especially in intensive care units where immunocompromised older patients with several comorbidities are hospitalized BFs cause the onset of difficult-to-eradicate infections. In this context, recent studies have demonstrated that quaternary ammonium compounds (QACs), acting as membrane disruptors and initially with a low tendency to develop resistance, have demonstrated anti-BF potentialities. However, a paucity of innovation in this space has driven the emergence of QAC resistance. More recently, quaternary phosphonium salts (QPSs), including tri-phenyl alkyl phosphonium derivatives, achievable by easy one-step reactions and well known as intermediates of the Wittig reaction, have shown promising anti-BF effects in vitro. Here, after an overview of pathogen resistance, BFs, and QACs, we have reviewed the QPSs developed and assayed to this end, so far. Finally, the synthetic strategies used to prepare QPSs have also been provided and discussed to spur the synthesis of novel compounds of this class. We think that the extension of the knowledge about these materials by this review could be a successful approach to finding effective weapons for treating chronic infections and device-associated diseases sustained by BF-producing MDR bacteria.
Collapse
Affiliation(s)
- Silvana Alfei
- Department of Pharmacy, University of Genoa, Viale Cembrano, 4, 16148 Genova, Italy
| |
Collapse
|
22
|
Aburayyan WS, Seder N, Al-fawares O, Fararjeh A, Majali IS, Al-Hajaya Y. Characterization of Antibiofilm and Antimicrobial Effects of Trigona Stingless Bee Honey Compared to Stinging Bee Centaurea hyalolepis and Citrus Honeys. J Evid Based Integr Med 2024; 29:2515690X241271978. [PMID: 39118572 PMCID: PMC11311187 DOI: 10.1177/2515690x241271978] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 06/11/2023] [Revised: 05/15/2024] [Accepted: 06/26/2024] [Indexed: 08/10/2024] Open
Abstract
The antibiofilm and antimicrobial properties of tropical honey types including Malaysian stingless bee honey remain explicitly unexplored when compared with Apies honey. The antibiofilm and antimicrobial activities of the Malaysian Trigona honey were characterized with two stinging bee honey types (Centaurea hyalolepis and Citrus honeys) from Jordan. The antibiofilm and antimicrobial investigations were conducted on a set of seven microbial strains; five bacterial species of Pseudomonas aeruginosa ATCC 10145, Streptococcus pyogenes ATCC 19615, Staphylococcus aureus ATCC 25923, Escherichia coli ATCC 25922, Klebsiella pneumoniae ATCC 13883, and two fungal strains Candida albicans ATCC 10231 and Candida krusei ATCC 14243. The antimicrobial investigations revealed a broad spectrum activity for Trigona honey against Gram-positive, Gram-negative, and fungal strains over the two honey types. One-way ANOVA showed a significant difference (p < 0.001) in the zone of inhibition ranging from 9 to 25 mm and minimum inhibition activity (MIC) ranged from 9.4-29.6% (w/v) against the microbial strains. Moreover, the addition of honey to established biofilms has induced a degradation activity in the biofilm mass. Two-way ANOVA showed a significant biofilm degradation proportion (p < 0.001) ranging from 1.3% to 91.3% following treatment with Trigona honey and the other honey types in relevance to the concentration ranging from 10% to 50% (w/v). Moreover, the antibiofilm activity was highly consistent with MIC affecting bacterial growth inhibition. In conclusion, a robust antimicrobial and antibiofilm activity for Trigona stingless bee honey over the stinging bee Centaurea hyalolepis and Citrus honeys is noticed which endows the usage of Trigona honey in the antimicrobial industry.
Collapse
Affiliation(s)
- Walid Salem Aburayyan
- Department of Medical Laboratory Sciences, Faculty of Science, Al-Balqa Applied University, Al-Salt, Jordan
| | - Nesrin Seder
- Department of Pharmaceutical Chemistry and Pharmacognosy, Faculty of Pharmacy, Applied Science Private University, Amman, Jordan
| | - O’la Al-fawares
- Department of Medical Laboratory Sciences, Faculty of Science, Al-Balqa Applied University, Al-Salt, Jordan
| | - AbdulFattah Fararjeh
- Department of Medical Laboratory Sciences, Faculty of Science, Al-Balqa Applied University, Al-Salt, Jordan
| | - Ibrahim S. Majali
- Department of Medical Laboratory Sciences, Mutah University, Karak, Jordan
| | - Yousef Al-Hajaya
- Department of Biological Sciences, Mutah University, Karak, Jordan
| |
Collapse
|
23
|
Lazarus E, Meyer AS, Ikuma K, Rivero IV. Three dimensional printed biofilms: Fabrication, design and future biomedical and environmental applications. Microb Biotechnol 2024; 17:e14360. [PMID: 38041693 PMCID: PMC10832517 DOI: 10.1111/1751-7915.14360] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 07/01/2023] [Revised: 10/02/2023] [Accepted: 10/11/2023] [Indexed: 12/03/2023] Open
Abstract
Three dimensional printing has emerged as a widely acceptable strategy for the fabrication of mammalian cell laden constructs with complex microenvironments for tissue engineering and regenerative medicine. More recently 3D printed living materials containing microorganisms have been developed and matured into living biofilms. The potential for engineered 3D biofilms as in vitro models for biomedical applications, such as antimicrobial susceptibility testing, and environmental applications, such as bioleaching, bioremediation, and wastewater purification, is extensive but the need for an in-depth understanding of the structure-function relationship between the complex construct and the microorganism response still exists. This review discusses 3D printing fabrication methods for engineered biofilms with specific structural features. Next, it highlights the importance of bioink compositions and 3D bioarchitecture design. Finally, a brief overview of current and potential applications of 3D printed biofilms in environmental and biomedical fields is discussed.
Collapse
Affiliation(s)
- Emily Lazarus
- Department Industrial and Systems EngineeringRochester Institute of TechnologyRochesterNew YorkUSA
| | - Anne S. Meyer
- Department of BiologyUniversity of RochesterRochesterNew YorkUSA
| | - Kaoru Ikuma
- Department of Civil, Construction, and Environmental EngineeringIowa State UniversityAmesIowaUSA
| | - Iris V. Rivero
- Department Industrial and Systems EngineeringRochester Institute of TechnologyRochesterNew YorkUSA
- Department of Biomedical EngineeringRochester Institute of TechnologyRochesterNew YorkUSA
- Department of Industrial and Systems EngineeringUniversity of FloridaGainesvilleFloridaUSA
| |
Collapse
|
24
|
Song J, Ali A, Ma Y, Li Y. A graphene microelectrode array based microfluidic device for in situ continuous monitoring of biofilms. NANOSCALE ADVANCES 2023; 5:4681-4686. [PMID: 37705780 PMCID: PMC10496883 DOI: 10.1039/d3na00482a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Academic Contribution Register] [Received: 07/03/2023] [Accepted: 08/10/2023] [Indexed: 09/15/2023]
Abstract
In situ continuous monitoring of bacterial biofilms has been a challenging job so far, but it is fundamental to the screening of novel anti-biofilm reagents. In this work, a microfluidic system utilizing a graphene-modified microelectrode array sensor was proposed to realize the dynamic state of bacterial biofilm monitoring by electrochemical impedance. The results illustrated that the observation window period of the biofilm state is significantly prolonged due to the increment of bacterial cell load on the sensing interface, thereby greatly improving the sensing signal quality. Simulation of anti-biofilm drug screening demonstrated that the performance of this method manifestly exceeded that of its endpoint counterparts.
Collapse
Affiliation(s)
- Jin Song
- Biology Institute, Qilu University of Technology (Shandong Academy of Sciences) Jinan 250103 China
- Shandong Provincial Key Laboratory of Biosensors Jinan 250103 China
| | - Ashaq Ali
- Center of Excellence in Science & Applied Technologies (CESAT) Islamabad 75000 Pakistan
| | - Yaohong Ma
- Biology Institute, Qilu University of Technology (Shandong Academy of Sciences) Jinan 250103 China
- Shandong Provincial Key Laboratory of Biosensors Jinan 250103 China
| | - Yiwei Li
- Biology Institute, Qilu University of Technology (Shandong Academy of Sciences) Jinan 250103 China
- Shandong Provincial Key Laboratory of Biosensors Jinan 250103 China
| |
Collapse
|
25
|
Stryja J, Teplá K, Routek M, Pavlík V, Perutková D. Octenidine with hyaluronan dressing versus a silver dressing in hard-to-heal wounds: a post-marketing study. J Wound Care 2023; 32:480-491. [PMID: 37572339 DOI: 10.12968/jowc.2023.32.8.480] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 08/14/2023]
Abstract
OBJECTIVE This study was conducted to provide comparative data on the clinical efficacy and safety of a novel hard-to-heal wound dressing (Sorelex; Contipro a.s., Czech Republic) that combines octenidine and hyaluronan, compared with a silver-based dressing. METHOD This multicentre, open-label, randomised, post-market, clinical follow-up study provides a comparison of a octenidine and hyaluronan combination with a standard-of-care (SoC) silver-containing dressing. The investigators evaluated the management of infection in the hard-to-heal wounds based on recorded changes in the appearance of the wound bed tissue and the presence of clinical signs of infection after three weeks of treatment. Other relevant assessed parameters of wound healing were: wound size; exudation level; wound pain level; and surrounding skin appearance. RESULTS The study cohort included 48 patients in the Sorelex arm and 39 patients in the SoC arm. Both products evinced numerous parameters of wound infection management: reductions in the wound bed slough; marked decreases in wound size; the formation of re-epithelialisation and granulation tissue; and improved pain management. Sorelex significantly improved the condition of surrounding skin after three weeks of treatment, unlike SoC. Sorelex reduced wound area significantly more than SoC (p=0.04). No statistically significant differences were detected in other assessed parameters between the two study arms. All the participating investigators expressed their satisfaction with both products. No adverse reactions to Sorelex were recorded over the mean treatment period of 53 days. CONCLUSION The octenidine and hyaluronan combination provides a new alternative choice of dressing for the treatment of infected hard-to-heal wounds when compared with a silver-based product.
Collapse
Affiliation(s)
- Jan Stryja
- Centre of Vascular and Miniinvasive Surgery, Hospital Agel, Trinec Podlesi, The Czech Republic; Salvatella Ltd., Centre of Non-healing Wounds Treatment, Podiatric Outpatients' Department, Trinec, The Czech Republic
| | | | - Miroslav Routek
- Ambulance for Treatment of Chronic Wounds, Regional Hospital Nachod, Broumov, The Czech Republic
| | - Vojtech Pavlík
- Department of Dermatology, Third Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Darja Perutková
- Department of dermatology, Military hospital, Olomouc, The Czech Republic
| |
Collapse
|
26
|
Sushmitha TJ, Rajeev M, Kathirkaman V, Shivam S, Rao TS, Pandian SK. 3-Hydroxy coumarin demonstrates anti-biofilm and anti-hyphal efficacy against Candida albicans via inhibition of cell-adhesion, morphogenesis, and virulent genes regulation. Sci Rep 2023; 13:11687. [PMID: 37468600 DOI: 10.1038/s41598-023-37851-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 04/03/2023] [Accepted: 06/28/2023] [Indexed: 07/21/2023] Open
Abstract
Candida albicans, a common fungus of human flora, can become an opportunistic pathogen and causes invasive candidiasis in immunocompromised individuals. Biofilm formation is the prime cause of antibiotic resistance during C. albicans infections and treating biofilm-forming cells is challenging due to their intractable and persistent nature. The study intends to explore the therapeutic potential of naturally produced compounds by competitive marine bacteria residing in marine biofilms against C. albicans biofilm. To this end, 3-hydroxy coumarin (3HC), a compound identified from the cell-free culture supernatant of the marine bacterium Brevundimonas abyssalis, was found to exhibit anti-biofilm and anti-hyphal activity against both reference and clinical isolates of C. albicans. The compound demonstrated significant inhibitory effects on biofilms and impaired the yeast-to-hyphal transition, wrinkle, and filament morphology at the minimal biofilm inhibitory concentration (MBIC) of 250 µg mL-1. Intriguingly, quantitative PCR analysis of 3HC-treated C. albicans biofilm revealed significant downregulation of virulence genes (hst7, ume6, efg1, cph1, ras1, als1) associated with adhesion and morphogenesis. Moreover, 3HC displayed non-fungicidal and non-toxic characteristics against human erythrocytes and buccal cells. In conclusion, this study showed that marine biofilms are a hidden source of diverse therapeutic drugs, and 3HC could be a potent drug to treat C. albicans infections.
Collapse
Affiliation(s)
- T J Sushmitha
- Department of Biotechnology, Alagappa University, Science Campus, Karaikudi, Tamil Nadu, 630 003, India
| | - Meora Rajeev
- Department of Biotechnology, Alagappa University, Science Campus, Karaikudi, Tamil Nadu, 630 003, India
- Department of Biological Sciences and Bioengineering, Inha University, Inharo 100, Incheon, 22212, Republic of Korea
| | - Vellaisamy Kathirkaman
- Department of Biotechnology, Alagappa University, Science Campus, Karaikudi, Tamil Nadu, 630 003, India
| | - Singh Shivam
- Department of Biotechnology, Alagappa University, Science Campus, Karaikudi, Tamil Nadu, 630 003, India
| | - Toleti Subba Rao
- School of Arts and Sciences, Sai University, OMR, Paiyanur, Tamil Nadu, 603105, India
| | - Shunmugiah Karutha Pandian
- Department of Biotechnology, Alagappa University, Science Campus, Karaikudi, Tamil Nadu, 630 003, India.
| |
Collapse
|
27
|
Shamim A, Ansari MJ, Aodah A, Iqbal M, Aqil M, Mirza MA, Iqbal Z, Ali A. QbD-Engineered Development and Validation of a RP-HPLC Method for Simultaneous Estimation of Rutin and Ciprofloxacin HCl in Bilosomal Nanoformulation. ACS OMEGA 2023; 8:21618-21627. [PMID: 37360463 PMCID: PMC10286274 DOI: 10.1021/acsomega.3c00956] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Academic Contribution Register] [Received: 02/13/2023] [Accepted: 05/23/2023] [Indexed: 06/28/2023]
Abstract
In the given study, a new reverse-phase high-performance liquid chromatography (RP-HPLC) method has been reported for the simultaneous estimation of ciprofloxacin hydrochloride (CPX) and rutin (RUT) using quality by design (QbD) approach. The analysis was carried out by applying the Box-Behnken design having fewer design points and less experimental runs. It relates between factors and responses and gives statistically significant values, along with enhancing the quality of the analysis. CPX and RUT were separated on the Kromasil C18 column (4.6 × 150 mm, 5 μm) using an isocratic mobile phase combination of phosphoric acid buffer (pH 3.0) and acetonitrile with the ratio of 87:13% v/v at a flow rate of 1.0 mL/min. CPX and RUT were detected at their respective wavelengths of 278 and 368 nm using a photodiode array detector. The developed method was validated according to guideline ICH Q2 R (1). The validation parameters taken were linearity, system suitability, accuracy, precision, robustness, sensitivity, and solution stability which were in the acceptable range. The findings suggest that the developed RP-HPLC method can be successfully applied to analyze novel CPX-RUT-loaded bilosomal nanoformulation prepared by thin-film hydration technique.
Collapse
Affiliation(s)
- Athar Shamim
- Department
of Pharmaceutics, School of Pharmaceutical
Education and Research, Jamia Hamdard, New Delhi 110062, India
| | - Mohammad Javed Ansari
- Department
of Pharmaceutics, College of Pharmacy, Prince
Sattam Bin Abdulaziz University, Al-Kharj 11942, Saudi Arabia
| | - Alhussain Aodah
- Department
of Pharmaceutics, College of Pharmacy, Prince
Sattam Bin Abdulaziz University, Al-Kharj 11942, Saudi Arabia
| | - Muzaffar Iqbal
- Department
of Pharmaceutical Chemistry, College of Pharmacy, King Saud University, Riyadh 11451,Saudi Arabia
| | - Mohd. Aqil
- Department
of Pharmaceutics, School of Pharmaceutical
Education and Research, Jamia Hamdard, New Delhi 110062, India
| | - Mohd. Aamir Mirza
- Department
of Pharmaceutics, School of Pharmaceutical
Education and Research, Jamia Hamdard, New Delhi 110062, India
| | - Zeenat Iqbal
- Department
of Pharmaceutics, School of Pharmaceutical
Education and Research, Jamia Hamdard, New Delhi 110062, India
| | - Asgar Ali
- Department
of Pharmaceutics, School of Pharmaceutical
Education and Research, Jamia Hamdard, New Delhi 110062, India
| |
Collapse
|
28
|
Drumond MM, Tapia-Costa AP, Neumann E, Nunes ÁC, Barbosa JW, Kassuha DE, Mancha-Agresti P. Cell-free supernatant of probiotic bacteria exerted antibiofilm and antibacterial activities against Pseudomonas aeruginosa: A novel biotic therapy. Front Pharmacol 2023; 14:1152588. [PMID: 37397469 PMCID: PMC10311102 DOI: 10.3389/fphar.2023.1152588] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 01/27/2023] [Accepted: 05/22/2023] [Indexed: 07/04/2023] Open
Abstract
Aim: This study aims to verify the antibacterial and antibiofilm action of cell-free spent medium (CFSM) from four lactic acid bacteria with potential probiotic characteristics (Lactiplantibacillus plantarum, Lactobacillus acidophilus, Lactobacillus johnsonii, and Lactobacillus delbrueckii) against two Pseudomonas aeruginosa strains. Main methods: The minimum inhibitory concentration (MIC) and minimum bactericidal concentration (MBC) of the CFSM, antibacterial activity by analysing the formation of inhibition zones, and inhibition of planktonic cultures were determined. Whether an increase in the concentration of CFSM influenced the growth of pathogenic strains and the anti-adhesive activity of the CFSM in biofilm formation (crystal violet and MTT assays) were determined, which were all corroborated by using scanning electron microscopy. Key findings: The relationship between the MIC and MBC values showed a bactericidal or bacteriostatic effect for all the cell-free spent media (CFSMs) tested for P. aeruginosa 9027™ and 27853™ strains. The CFSM supplemental doses of 18 or 22%, 20 or 22%, 46 or 48%, and 50 or 54% of L. acidophilus, L. delbrueckii, L. plantarum, and L. johnsonii, respectively, could completely inhibit the growth of both pathogen strains. The antibiofilm activity of the CFSM in three biofilm conditions (pre-coated, co-incubated, and preformed) demonstrated values ranging between 40% and 80% for biofilm inhibition, and similar results were observed for cell viability. Significance: This work provides strong evidence that the postbiotic derived from different Lactobacilli could be practical as an adjuvant therapy for reducing the use of antibiotics, being a good candidate to overcome the growing challenge of hospital infections due to this pathogen.
Collapse
Affiliation(s)
- Mariana Martins Drumond
- Centro Federal de Educação Tecnológica de Minas Gerais (CEFET/MG), Departamento de Ciências Biológicas, Belo Horizonte, Minas Gerais, Brazil
- Centro Federal de Educação Tecnológica de Minas Gerais (CEFET/MG), Programa de Pós Graduação em Engenharia de Materiais, Belo Horizonte, Minas Gerais, Brazil
| | - Ana Paula Tapia-Costa
- Instituto de Investigaciones en Ciencias Químicas, Facultad de Ciencias Químicas y Tecnológicas, Universidad Católica de Cuyo, San Juan, Argentina
| | - Elisabeth Neumann
- Departamento de Microbiologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Álvaro Cantini Nunes
- Laboratório de Genética Molecular de Protozoários Parasitas, Departamento de Genética, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, Minas Gerais, Brazil
| | - Jorge Wanderson Barbosa
- Centro Federal de Educação Tecnológica de Minas Gerais (CEFET/MG), Departamento de Engenharia de Materiais, Belo Horizonte, Minas Gerais, Brazil
| | - Diego E. Kassuha
- Instituto de Investigaciones en Ciencias Químicas, Facultad de Ciencias Químicas y Tecnológicas, Universidad Católica de Cuyo, San Juan, Argentina
| | - Pamela Mancha-Agresti
- Centro Federal de Educação Tecnológica de Minas Gerais (CEFET/MG), Programa de Pós Graduação em Engenharia de Materiais, Belo Horizonte, Minas Gerais, Brazil
| |
Collapse
|
29
|
Kang T, Zhou M, Yan X, Song S, Yuan S, Yang H, Ding H, Jiang H, Zhang D, Bai Y, Zhang N. Biofilm formation and correlations with drug resistance in Mycoplasma synoviae. Vet Microbiol 2023; 283:109777. [PMID: 37267807 DOI: 10.1016/j.vetmic.2023.109777] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 07/02/2022] [Revised: 05/14/2023] [Accepted: 05/15/2023] [Indexed: 06/04/2023]
Abstract
Infectious synovitis in chickens caused by Mycoplasma synoviae infections are characterized by exudative synovial joint membranes and tenosynovitis. We isolated M. synoviae from chickens on farms in Guangdong, China and identifed 29 K-type and 3 A-type strains using vlhA genotyping and all displayed decreased susceptibilities to enrofloxacin, doxycycline, tiamulin and tylosin compared with the type strain WVU1853 (ATCC 25204). M. synoviae biofilms were present after staining as block or continuous dot shape morphologies and these appeared as tower-like and mushroom-like structures in scanning electron micrographs. The optimal temperature for biofilm formation was 33 °C and these biofilms enhanced the resistance of M. synoviae to all 4 antibiotics we tested and minimum biofilm inhibitory concentration for enrofloxacin and biofilm biomass were significantly negatively correlated (r < 0, 0.3 ≤|r|<0.5, P < 0.05). This work is the first study of the biofilm formation ability of M. synoviae and provides the foundation for further investigations.
Collapse
Affiliation(s)
- Tianhao Kang
- School of life science and engineering, Foshan University, Foshan 528225, China
| | - Minghu Zhou
- School of life science and engineering, Foshan University, Foshan 528225, China
| | - Xiu Yan
- School of life science and engineering, Foshan University, Foshan 528225, China
| | - Shuti Song
- School of life science and engineering, Foshan University, Foshan 528225, China
| | - Sheng Yuan
- School of life science and engineering, Foshan University, Foshan 528225, China
| | - Hong Yang
- School of life science and engineering, Foshan University, Foshan 528225, China
| | - Huanzhong Ding
- Guangdong Key Laboratory for Veterinary Drug Development and Safety Evaluation, South China Agricultural University, Guangzhou, China
| | - Hongxia Jiang
- Guangdong Key Laboratory for Veterinary Drug Development and Safety Evaluation, South China Agricultural University, Guangzhou, China
| | - Dexian Zhang
- School of life science and engineering, Foshan University, Foshan 528225, China
| | - Yinshan Bai
- School of life science and engineering, Foshan University, Foshan 528225, China
| | - Nan Zhang
- School of life science and engineering, Foshan University, Foshan 528225, China.
| |
Collapse
|
30
|
Ghorab MM, M Soliman A, El-Sayyad GS, Abdel-Kader MS, El-Batal AI. Synthesis, Antimicrobial, and Antibiofilm Activities of Some Novel 7-Methoxyquinoline Derivatives Bearing Sulfonamide Moiety against Urinary Tract Infection-Causing Pathogenic Microbes. Int J Mol Sci 2023; 24:ijms24108933. [PMID: 37240275 DOI: 10.3390/ijms24108933] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 04/06/2023] [Revised: 05/07/2023] [Accepted: 05/16/2023] [Indexed: 05/28/2023] Open
Abstract
A new series of 4-((7-methoxyquinolin-4-yl) amino)-N-(substituted) benzenesulfonamide 3(a-s) was synthesized via the reaction of 4-chloro-7-methoxyquinoline 1 with various sulfa drugs. The structural elucidation was verified based on spectroscopic data analysis. All the target compounds were screened for their antimicrobial activity against Gram-positive bacteria, Gram-negative bacteria, and unicellular fungi. The results revealed that compound 3l has the highest effect on most tested bacterial and unicellular fungal strains. The highest effect of compound 3l was observed against E. coli and C. albicans with MIC = 7.812 and 31.125 µg/mL, respectively. Compounds 3c and 3d showed broad-spectrum antimicrobial activity, but the activity was lower than that of 3l. The antibiofilm activity of compound 3l was measured against different pathogenic microbes isolated from the urinary tract. Compound 3l could achieve biofilm extension at its adhesion strength. After adding 10.0 µg/mL of compound 3l, the highest percentage was 94.60% for E. coli, 91.74% for P. aeruginosa, and 98.03% for C. neoformans. Moreover, in the protein leakage assay, the quantity of cellular protein discharged from E. coli was 180.25 µg/mL after treatment with 1.0 mg/mL of compound 3l, which explains the creation of holes in the cell membrane of E. coli and proves compound 3l's antibacterial and antibiofilm properties. Additionally, in silico ADME prediction analyses of compounds 3c, 3d, and 3l revealed promising results, indicating the presence of drug-like properties.
Collapse
Affiliation(s)
- Mostafa M Ghorab
- Drug Chemistry Laboratory, Drug Radiation Research Department, National Center for Radiation Research and Technology (NCRRT), Egyptian Atomic Energy Authority (EAEA), Cairo 11787, Egypt
| | - Aiten M Soliman
- Drug Chemistry Laboratory, Drug Radiation Research Department, National Center for Radiation Research and Technology (NCRRT), Egyptian Atomic Energy Authority (EAEA), Cairo 11787, Egypt
| | - Gharieb S El-Sayyad
- Drug Microbiology Laboratory, Drug Radiation Research Department, National Center for Radiation Research and Technology (NCRRT), Egyptian Atomic Energy Authority (EAEA), Cairo 11787, Egypt
| | - Maged S Abdel-Kader
- Department of Pharmacognosy, College of Pharmacy, Prince Sattam Bin Abdulaziz University, Al-Kharj 11942, Saudi Arabia
- Department of Pharmacognosy, College of Pharmacy, Alexandria University, Alexandria 21215, Egypt
| | - Ahmed I El-Batal
- Drug Microbiology Laboratory, Drug Radiation Research Department, National Center for Radiation Research and Technology (NCRRT), Egyptian Atomic Energy Authority (EAEA), Cairo 11787, Egypt
| |
Collapse
|
31
|
Valentin JDP, Altenried S, Varadarajan AR, Ahrens CH, Schreiber F, Webb JS, van der Mei HC, Ren Q. Identification of Potential Antimicrobial Targets of Pseudomonas aeruginosa Biofilms through a Novel Screening Approach. Microbiol Spectr 2023; 11:e0309922. [PMID: 36779712 PMCID: PMC10100978 DOI: 10.1128/spectrum.03099-22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 08/08/2022] [Accepted: 01/15/2023] [Indexed: 02/14/2023] Open
Abstract
Pseudomonas aeruginosa is an opportunistic pathogen of considerable medical importance, owing to its pronounced antibiotic tolerance and association with cystic fibrosis and other life-threatening diseases. The aim of this study was to highlight the genes responsible for P. aeruginosa biofilm tolerance to antibiotics and thereby identify potential new targets for the development of drugs against biofilm-related infections. By developing a novel screening approach and utilizing a public P. aeruginosa transposon insertion library, several biofilm-relevant genes were identified. The Pf phage gene (PA0720) and flagellin gene (fliC) conferred biofilm-specific tolerance to gentamicin. Compared with the reference biofilms, the biofilms formed by PA0720 and fliC mutants were completely eliminated with a 4-fold-lower gentamicin concentration. Furthermore, the mreC, pprB, coxC, and PA3785 genes were demonstrated to play major roles in enhancing biofilm tolerance to gentamicin. The analysis of biofilm-relevant genes performed in this study provides important novel insights into the understanding of P. aeruginosa antibiotic tolerance, which will facilitate the detection of antibiotic resistance and the development of antibiofilm strategies against P. aeruginosa. IMPORTANCE Pseudomonas aeruginosa is an opportunistic pathogen of high medical importance and is one of the main pathogens responsible for the mortality of patients with cystic fibrosis. In addition to inherited antibiotic resistance, P. aeruginosa can form biofilms, defined as communities of microorganisms embedded in a self-produced matrix of extracellular polymeric substances adhering to each other and/or to a surface. Biofilms protect bacteria from antibiotic treatments and represent a major reason for antibiotic failure in the treatment of chronic infections caused by cystic fibrosis. Therefore, it is crucial to develop new therapeutic strategies aimed at specifically eradicating biofilms. The aim of this study was to generalize a novel screening method for biofilm research and to identify the possible genes involved in P. aeruginosa biofilm tolerance to antibiotics, both of which could improve the understanding of biofilm-related infections and allow for the identification of relevant therapeutic targets for drug development.
Collapse
Affiliation(s)
- Jules D. P. Valentin
- Laboratory for Biointerfaces, Empa, Swiss Federal Laboratories for Materials Science and Technology, St. Gallen, Switzerland
- University of Groningen and University Medical Center Groningen, Department of BioMedical Engineering, Groningen, Netherlands
| | - Stefanie Altenried
- Laboratory for Biointerfaces, Empa, Swiss Federal Laboratories for Materials Science and Technology, St. Gallen, Switzerland
| | - Adithi R. Varadarajan
- Molecular Ecology, Agroscope and Swiss Institute of Bioinformatics, Zurich, Switzerland
| | - Christian H. Ahrens
- Molecular Ecology, Agroscope and Swiss Institute of Bioinformatics, Zurich, Switzerland
| | - Frank Schreiber
- Division of Biodeterioration and Reference Organisms (4.1), Department of Materials and the Environment, Federal Institute for Materials Research and Testing (BAM), Berlin, Germany
| | - Jeremy S. Webb
- Institute for Life Sciences, University of Southampton, Southampton, United Kingdom
- National Biofilms Innovation Centre, University of Southampton, Southampton, United Kingdom
| | - Henny C. van der Mei
- University of Groningen and University Medical Center Groningen, Department of BioMedical Engineering, Groningen, Netherlands
| | - Qun Ren
- Laboratory for Biointerfaces, Empa, Swiss Federal Laboratories for Materials Science and Technology, St. Gallen, Switzerland
| |
Collapse
|
32
|
Xiu W, Ren L, Xiao H, Zhang Y, Wang D, Yang K, Wang S, Yuwen L, Li X, Dong H, Li Q, Mou Y, Zhang Y, Yin Z, Liang B, Gao Y, Wang L. Ultrasound-responsive catalytic microbubbles enhance biofilm elimination and immune activation to treat chronic lung infections. SCIENCE ADVANCES 2023; 9:eade5446. [PMID: 36696490 DOI: 10.1126/sciadv.ade5446] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Academic Contribution Register] [Received: 08/23/2022] [Accepted: 12/27/2022] [Indexed: 06/17/2023]
Abstract
Efficient treatment of chronic lung infections caused by Pseudomonas aeruginosa biofilms is a great challenge because of drug tolerance and immune evasion issues. Here, we develop ultrasound-responsive catalytic microbubbles with biofilm elimination and immune activation properties to combat chronic lung infection induced by P. aeruginosa biofilms. In these microbubbles, piperacillin and Fe3O4 nanoparticles form a drug-loaded shell surrounding the air core. Under ultrasound stimulation, the microbubbles can physically disrupt the structure of biofilms and enhance the penetration of both Fe3O4 nanoparticles and piperacillin into the biofilm. Then, Fe3O4 nanoparticles chemically degrade the biofilm matrix and kill the bacteria with the assistance of piperacillin. Fe3O4 nanoparticles can activate the immune response for biofilm elimination by polarizing macrophages into a pro-inflammatory phenotype. These ultrasound-responsive catalytic microbubbles efficiently treat chronic lung infections in a mouse model by combining physical/chemical/antibiotic biofilm elimination and immune activation, thus providing a promising strategy for combating bacterial biofilm infections.
Collapse
Affiliation(s)
- Weijun Xiu
- State Key Laboratory of Organic Electronics and Information Displays & Jiangsu Key Laboratory for Biosensors, Institute of Advanced Materials (IAM), Nanjing University of Posts and Telecommunications, Nanjing 210023, China
| | - Lili Ren
- State Key Laboratory of Organic Electronics and Information Displays & Jiangsu Key Laboratory for Biosensors, Institute of Advanced Materials (IAM), Nanjing University of Posts and Telecommunications, Nanjing 210023, China
| | - Huayu Xiao
- State Key Laboratory of Organic Electronics and Information Displays & Jiangsu Key Laboratory for Biosensors, Institute of Advanced Materials (IAM), Nanjing University of Posts and Telecommunications, Nanjing 210023, China
| | - Yue Zhang
- State Key Laboratory of Organic Electronics and Information Displays & Jiangsu Key Laboratory for Biosensors, Institute of Advanced Materials (IAM), Nanjing University of Posts and Telecommunications, Nanjing 210023, China
| | - Dou Wang
- State Key Laboratory of Organic Electronics and Information Displays & Jiangsu Key Laboratory for Biosensors, Institute of Advanced Materials (IAM), Nanjing University of Posts and Telecommunications, Nanjing 210023, China
| | - Kaili Yang
- State Key Laboratory of Organic Electronics and Information Displays & Jiangsu Key Laboratory for Biosensors, Institute of Advanced Materials (IAM), Nanjing University of Posts and Telecommunications, Nanjing 210023, China
| | - Siyu Wang
- State Key Laboratory of Organic Electronics and Information Displays & Jiangsu Key Laboratory for Biosensors, Institute of Advanced Materials (IAM), Nanjing University of Posts and Telecommunications, Nanjing 210023, China
| | - Lihui Yuwen
- State Key Laboratory of Organic Electronics and Information Displays & Jiangsu Key Laboratory for Biosensors, Institute of Advanced Materials (IAM), Nanjing University of Posts and Telecommunications, Nanjing 210023, China
| | - Xiao Li
- State Key Laboratory of Organic Electronics and Information Displays & Jiangsu Key Laboratory for Biosensors, Institute of Advanced Materials (IAM), Nanjing University of Posts and Telecommunications, Nanjing 210023, China
| | - Heng Dong
- Nanjing Stomatological Hospital, Medical School of Nanjing University, Nanjing 210008, China
| | - Qiang Li
- Nanjing Stomatological Hospital, Medical School of Nanjing University, Nanjing 210008, China
| | - Yongbin Mou
- Nanjing Stomatological Hospital, Medical School of Nanjing University, Nanjing 210008, China
| | - Yuqian Zhang
- CAS Key Laboratory of Quantitative Engineering Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Zhaowei Yin
- Department of Orthopedics, Nanjing First Hospital, Nanjing Medical University, Nanjing 210006, China
| | - Bin Liang
- Department of Orthopedics, Nanjing First Hospital, Nanjing Medical University, Nanjing 210006, China
| | - Yu Gao
- State Key Laboratory of Organic Electronics and Information Displays & Jiangsu Key Laboratory for Biosensors, Institute of Advanced Materials (IAM), Nanjing University of Posts and Telecommunications, Nanjing 210023, China
| | - Lianhui Wang
- State Key Laboratory of Organic Electronics and Information Displays & Jiangsu Key Laboratory for Biosensors, Institute of Advanced Materials (IAM), Nanjing University of Posts and Telecommunications, Nanjing 210023, China
| |
Collapse
|
33
|
Lu P, Zhang X, Li F, Xu KF, Li YH, Liu X, Yang J, Zhu B, Wu FG. Cationic Liposomes with Different Lipid Ratios: Antibacterial Activity, Antibacterial Mechanism, and Cytotoxicity Evaluations. Pharmaceuticals (Basel) 2022; 15:ph15121556. [PMID: 36559007 PMCID: PMC9783835 DOI: 10.3390/ph15121556] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 10/20/2022] [Revised: 12/05/2022] [Accepted: 12/06/2022] [Indexed: 12/15/2022] Open
Abstract
Due to their strong bacterial binding and bacterial toxicity, cationic liposomes have been utilized as effective antibacterial materials in many studies. However, few researchers have systematically compared their antibacterial activity with their mammalian cell cytotoxicity or have deeply explored their antibacterial and cytotoxicity mechanisms. Here, we prepared a series of cationic liposomes (termed CLs) using dimethyldioctadecylammonium chloride (DODAC) and lecithin at different molar ratios. CLs have the ability to effectively bind with Gram-positive and Gram-negative bacteria through electrostatic and hydrophobic interactions. Further, the CLs with high molar ratios of DODAC (30 and 40 mol%) can disrupt the bacterial wall/membrane, efficiently inducing the production of reactive oxygen species (ROS). More importantly, we carefully compared the antibacterial activity and the mammalian cell cytotoxicity of various CLs differing in DODAC contents and liposomal concentrations and revealed that, whether they are bacterial or mammalian cells, an increasing DODAC content in CLs can lead to an elevated cytotoxicity level. Further, there exists a critical DODAC contents (>20 mol%) in CLs to endow them with effective antibacterial ability. However, the variation in the DODAC content and liposomal concentration of CLs has different degrees of influence on the antibacterial activity or cytotoxicity. For example, CLs at high DODAC content (i.e., CL0.3 and CL0.4) could effectively kill both types of bacterial cells but only cause negligible toxicity to mammalian cells. We believe that a systematic comparison between the antibacterial activity and the cytotoxicity of CLs with different DODAC contents will provide an important reference for the potential clinical applications of cationic liposomes.
Collapse
Affiliation(s)
- Pengpeng Lu
- Department of Emergency, The Second Affiliated Hospital of Nantong University, 6 North Hai’erxiang Road, Nantong 226001, China
| | - Xinping Zhang
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, 2 Sipailou Road, Nanjing 210096, China
| | - Feng Li
- Department of Emergency, The Second Affiliated Hospital of Nantong University, 6 North Hai’erxiang Road, Nantong 226001, China
| | - Ke-Fei Xu
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, 2 Sipailou Road, Nanjing 210096, China
| | - Yan-Hong Li
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, 2 Sipailou Road, Nanjing 210096, China
| | - Xiaoyang Liu
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, 2 Sipailou Road, Nanjing 210096, China
| | - Jing Yang
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, 2 Sipailou Road, Nanjing 210096, China
| | - Baofeng Zhu
- Department of Emergency, The Second Affiliated Hospital of Nantong University, 6 North Hai’erxiang Road, Nantong 226001, China
- Correspondence: (B.Z.); (F.-G.W.)
| | - Fu-Gen Wu
- Department of Emergency, The Second Affiliated Hospital of Nantong University, 6 North Hai’erxiang Road, Nantong 226001, China
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, 2 Sipailou Road, Nanjing 210096, China
- Correspondence: (B.Z.); (F.-G.W.)
| |
Collapse
|
34
|
Songnaka N, Lertcanawanichakul M, Hutapea AM, Krobthong S, Yingchutrakul Y, Atipairin A. Purification and Characterization of Novel Anti-MRSA Peptides Produced by Brevibacillus sp. SPR-20. MOLECULES (BASEL, SWITZERLAND) 2022; 27:molecules27238452. [PMID: 36500545 PMCID: PMC9738727 DOI: 10.3390/molecules27238452] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Academic Contribution Register] [Received: 11/04/2022] [Revised: 11/28/2022] [Accepted: 11/30/2022] [Indexed: 12/11/2022]
Abstract
Methicillin-resistant Staphylococcus aureus (MRSA) is listed as a high-priority pathogen because its infection is associated with a high mortality rate. It is urgent to search for new agents to treat such an infection. Our previous study isolated a soil bacterium (Brevibacillus sp. SPR-20), showing the highest antimicrobial activity against S. aureus TISTR 517 and MRSA strains. The present study aimed to purify and characterize anti-MRSA substances produced by SPR-20. The result showed that five active substances (P1-P5) were found, and they were identified by LC-MS/MS that provided the peptide sequences of 14-15 residues. Circular dichroism showed that all peptides contained β-strand and disordered conformations as the major secondary structures. Only P1-P4 adopted more α-helix conformations when incubated with 50 mM SDS. These anti-MRSA peptides could inhibit S. aureus and MRSA in concentrations of 2-32 μg/mL. P1 (NH2-VVVNVLVKVLPPPVV-COOH) had the highest activity and was identified as a novel antimicrobial peptide (AMP). The stability study revealed that P1 was stable in response to temperature, proteolytic enzymes, surfactant, and pH. The electron micrograph showed that P1 induced bacterial membrane damage when treated at 1× MIC in the first hour of incubation. The killing kinetics of P1 was dependent on concentration and time. Mechanisms of P1 on tested pathogens involved membrane permeability, leakage of genetic material, and cell lysis. The P1 peptide at a concentration up to 32 μg/mL showed hemolysis of less than 10%, supporting its safety for human erythrocytes. This study provides promising anti-MRSA peptides that might be developed for effective antibiotics in the post-antibiotic era.
Collapse
Affiliation(s)
- Nuttapon Songnaka
- School of Pharmacy, Walailak University, Nakhon Si Thammarat 80161, Thailand
| | | | - Albert M. Hutapea
- Faculty of Science, Universitas Advent Indonesia, Bandung 40559, Indonesia
| | - Sucheewin Krobthong
- Center of Excellence in Natural Products Chemistry (CENP), Department of Chemistry, Faculty of Science, Chulalongkorn University, Bangkok 10330, Thailand
| | - Yodying Yingchutrakul
- National Omics Center, National Science and Technology Development Agency, Pathum Thani 12120, Thailand
| | - Apichart Atipairin
- School of Pharmacy, Walailak University, Nakhon Si Thammarat 80161, Thailand
- Drug and Cosmetics Excellence Center, Walailak University, Nakhon Si Thammarat 80161, Thailand
- Correspondence: ; Tel.: +66-7567-2832; Fax: +66-7567-2814
| |
Collapse
|
35
|
Bruce J, Oyedemi B, Parsons N, Harrison F. Phase 1 safety trial of a natural product cocktail with antibacterial activity in human volunteers. Sci Rep 2022; 12:19656. [PMID: 36385621 PMCID: PMC9667429 DOI: 10.1038/s41598-022-22700-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 11/02/2021] [Accepted: 10/18/2022] [Indexed: 11/17/2022] Open
Abstract
New antibiotics are urgently needed to reduce the health burden of antibiotic-resistant bacterial infection. Natural products (NPs) derived from plants and animals are a current focus of research seeking to discover new antibacterial molecules with clinical potential. A cocktail of NPs based on a medieval remedy for eye infection eliminated biofilms of several highly antibiotic-resistant bacterial species in laboratory studies, and had a promising safety profile in vitro and in a mouse model. A necessary prelude to refining this remedy into a defined, synthetic mixture suitable for testing with wound infections is to firstly establish safety when applied to healthy human skin. We aimed to assess skin-related outcomes of the preparation in a sample of healthy volunteers. This prospective, single arm, non-randomised Phase I clinical trial consisted of a single patch test intervention with 48-h follow-up. Volunteers were staff, students and members of the public recruited from the University of Warwick and surrounding locality. Adults aged 18-79 years, with no history of severe immunity-related disease, diabetes, recent infection, or known pregnancy were eligible. A 100 µl application of a filter-sterilised NP mixture, comprising ground garlic, onion, white wine and bovine bile, was applied to skin on the upper arm and covered with a dressing. The primary outcome was skin-related adverse events over 48 h. Digital photographs were captured where bothersome, salve-related events were reported. 109 volunteers, aged 18-77 years, were recruited between June and July 2021. Sample mean age was 37.6 (SD 16.1) years, and 63 (58%) participants were female. Outcome data were obtained for 106/109 (97%); two participants were lost to follow-up and one removed the skin patch after nine hours due to a bothersome garlic odour. Twenty-one (19.8%) participants reported any patch-test related sign or symptom; of these 14 (13.2%) participants reported minor events related to the salve, including itchiness, redness, or garlic odour. No serious events were reported. We found no evidence of serious skin-related adverse events related to the NP preparation.Trial registration: International Standard Randomised Controlled Trial Number (ISRCTN10773579). Date registered: 08/01/2021.
Collapse
Affiliation(s)
- Julie Bruce
- Warwick Clinical Trials Unit, Division of Health Sciences, University of Warwick, Coventry, CV4 7AL, UK.
- University Hospital Coventry and Warwickshire NHS Trust, Clifford Bridge Road, Coventry, CV2 2DX, UK.
| | - Blessing Oyedemi
- School of Life Sciences, University of Warwick, Coventry, CV4 7AL, UK
| | - Nick Parsons
- Statistics and Epidemiology Unit, Warwick Medical School, University of Warwick, Coventry, CV4 7AL, UK
| | - Freya Harrison
- School of Life Sciences, University of Warwick, Coventry, CV4 7AL, UK.
| |
Collapse
|
36
|
Rafiq MA, Shahid M, Jilani K, Aslam MA. Antibacterial, Antibiofilm, and Anti-Quorum Sensing Potential of Novel Synthetic Compounds Against Pathogenic Bacteria Isolated From Chronic Sinusitis Patients. Dose Response 2022; 20:15593258221135731. [PMID: 36311176 PMCID: PMC9597054 DOI: 10.1177/15593258221135731] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 08/11/2022] [Revised: 09/17/2022] [Accepted: 10/12/2022] [Indexed: 11/16/2022] Open
Abstract
Quorum sensing (QS) is a major controller of virulence and biofilm formation in
pathogenic bacteria. The aim of the research was to screen novel synthetic
compounds (18) from 2 series (Pyrazole and Diene dione) for quorum sensing and
biofilm inhibitory potential against resistant pathogens isolated from patients
with chronic sinusitis. Most of the compounds have documented zone of inhibition
against Gram positive strains Staphylococcus aureus,
Enterococcus faecalis and moderate activity against Gram
negative Klebseilla pneumoniae and Proteus
mirabilis in comparison with standard antibiotic. Compounds Q1 and
Q7 have given the maximum zone of inhibition 18 and 20 mm with MICs 0.312 mg/mL
and .156 mg/mL against S aureus and E
faecalis, respectively. Some compounds were equally potent at
inhibiting the formation of biofilm which later established by phase contrast
microscopy. Regarding quorum sensing inhibition, the tested concentration of
synthetic compound UA3 0.313 mg/mL inhibited violacein production without
decreasing Chromobacterium pseudoviolaceum count which was
significantly lower than determined MIC’s. It was depicted from the results that
selected compounds exhibited low level of cytotoxicity toward human red blood
cells. Hence, these findings revealed that most novel compounds were effective
antibacterial, whereas compound UA3 has shared significant anti-quorum sensing
potential against Chromobacterium pseudoviolaceum.
Collapse
Affiliation(s)
| | - Muhammad Shahid
- Department of Biochemistry, University of
Agriculture, Faisalabad, Pakistan,Muhammad Shahid, Department of
Biochemistry, University of Agriculture, Faisalabad 38040, Pakistan.
| | - Kashif Jilani
- Department of Biochemistry, University of
Agriculture, Faisalabad, Pakistan
| | | |
Collapse
|
37
|
Choudhary M, Shrivastava R, Vashistt J. Acinetobacter baumannii Biofilm Formation: Association with Antimicrobial Resistance and Prolonged Survival under Desiccation. Curr Microbiol 2022; 79:361. [PMID: 36253556 DOI: 10.1007/s00284-022-03071-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 07/19/2022] [Accepted: 09/29/2022] [Indexed: 11/30/2022]
Abstract
Biofilm-forming multidrug-resistant Acinetobacter baumannii has emerged as a global pathogen. This study investigated the impact of biofilm formation by A. baumannii on antimicrobial resistance and prolonged survival under desiccation, which is essential for effective infection control of A. baumannii in hospital settings. Seventy-eight clinical isolates of A. baumannii were identified, and antibiotic susceptibility profiles were assessed. All the isolates were investigated for their biofilm-forming abilities at 24 and 48 h. The biofilm inhibitory concentrations of antibiotics were evaluated for selected biofilm-forming isolates to determine the influence of biofilm on antibiotic tolerance. The impact of biofilm formation on desiccation tolerance was also evaluated for up to 48 days. The results revealed that out of 78 A. baumannii clinical isolates, 83% were MDR and 17% non-MDR. Overall, 79% of isolates formed high biofilm after 24 h. The extent of biofilm formation gets significantly increased after 48 h, and 87% of isolates formed high biofilm. It was observed that eradicating mature biofilm requires up to a thousandfold higher concentration of antibiotics than MICs, and biofilm-forming isolates can survive for a prolonged period under desiccation. In conclusion, our findings revealed that both MDR and non-MDR isolates of A. baumannii could form biofilms on abiotic surfaces. A. baumannii biofilms contribute to endurance in the presence of antimicrobials and desiccation conditions, which are significant trouble for hospital patient care management. The present findings may offer insights for developing preventive measures to tackle biofilm-associated A. baumannii infection.
Collapse
Affiliation(s)
- Monika Choudhary
- Department of Biotechnology & Bioinformatics, Jaypee University of Information Technology, Waknaghat, Solan, Himachal Pradesh, 173234, India
| | - Rahul Shrivastava
- Department of Biotechnology & Bioinformatics, Jaypee University of Information Technology, Waknaghat, Solan, Himachal Pradesh, 173234, India
| | - Jitendraa Vashistt
- Department of Biotechnology & Bioinformatics, Jaypee University of Information Technology, Waknaghat, Solan, Himachal Pradesh, 173234, India.
| |
Collapse
|
38
|
Jia J, Xue X, Guan Y, Fan X, Wang Z. Biofilm characteristics and transcriptomic profiling of Acinetobacter johnsonii defines signatures for planktonic and biofilm cells. ENVIRONMENTAL RESEARCH 2022; 213:113714. [PMID: 35718162 DOI: 10.1016/j.envres.2022.113714] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Academic Contribution Register] [Received: 02/08/2022] [Revised: 06/11/2022] [Accepted: 06/14/2022] [Indexed: 06/15/2023]
Abstract
Most bacteria in the natural environment have a biofilm mode of life, which is intrinsically tolerant to antibiotics. While until now, the knowledge of biofilm formation by Acinetobacter johnsonii is not well understood. In this study, the characteristics and the effect of a sub-inhibitory concentration of antibiotic on A. johnsonii biofilm and planktonic cells were determined. We discovered a positive relationship between biofilm formation and tetracycline resistance, and biofilms rapidly evolve resistance to tetracycline they are treated with. Persister cells commonly exist in both planktonic and biofilm cells, with a higher frequency in the latter. Further transcriptomic analysis speculates that the overexpression of multidrug resistance genes and stress genes were mainly answered to sub lethal concentration of tetracycline in planktonic cells, and the lower metabolic levels after biofilm formation result in high resistance level of biofilm cells to tetracycline. Altogether, these data suggest that A. johnsonii can adjust its phenotype when grown as biofilm and change its metabolism under antibiotic stress, and provide implications for subsequent biofilm control.
Collapse
Affiliation(s)
- Jia Jia
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, 712100, China
| | - Xue Xue
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, 712100, China
| | - Yongjing Guan
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, 712100, China
| | - Xiaoteng Fan
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, 712100, China
| | - Zaizhao Wang
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, 712100, China.
| |
Collapse
|
39
|
Development and Evaluation of Duplex MIRA-qPCR Assay for Simultaneous Detection of Staphylococcus aureus and non-aureus Staphylococci. Microorganisms 2022; 10:microorganisms10091734. [PMID: 36144336 PMCID: PMC9502308 DOI: 10.3390/microorganisms10091734] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 07/21/2022] [Revised: 08/22/2022] [Accepted: 08/24/2022] [Indexed: 11/24/2022] Open
Abstract
Staphylococcus spp., especially Staphylococcus aureus (S. aureus), is an important pathogen in hospital-acquired infection and food poisoning. Here, we developed a multienzyme isothermal rapid amplification combined with duplex quantitative PCR (duplex MIRA-qPCR) method, which can simultaneously detect the S. aureus species-specific conserved gene FMN-bgsfp and the Staphylococcus genus-specific conserved gene tuf. This assay enabled the amplification of DNA within 20 min at a constant temperature of 39 °C. Specificity analysis indicated that all nine common Staphylococcus species were positive and non-Staphylococcus spp. were negative for tuf gene, whereas S. aureus was positive, non-aureus Staphylococci species and non-Staphylococcus spp. were negative for FMN-bgsfp gene, suggesting that duplex MIRA-qPCR exhibited high specificity. Meanwhile, the sensitivity was tested and the limit of detection (LoD) was 3 × 102 CFU/mL. The coefficient variation values ranged from 0.13% to 2.09%, indicating that the assay had good repeatability. Furthermore, all the nine common Staphylococcus species (including S. aureus) could be detected from four kinds of simulated samples and the LoD of S. aureus was 8.56 × 103 CFU/mL. In conclusion, the duplex MIRA-qPCR has advantages of stronger specificity, lower detection threshold, shorter detection time, and simpler operation, which is an effective tool to detect S. aureus and non-aureus Staphylococci spp. infections rapidly.
Collapse
|
40
|
Wei G, He Y. Antibacterial and Antibiofilm Activities of Novel Cyclic Peptides against Methicillin-Resistant Staphylococcus aureus. Int J Mol Sci 2022; 23:8029. [PMID: 35887376 PMCID: PMC9321466 DOI: 10.3390/ijms23148029] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 05/19/2022] [Revised: 06/10/2022] [Accepted: 06/27/2022] [Indexed: 02/04/2023] Open
Abstract
Methicillin-resistant Staphylococcus aureus (MRSA) has led to serious infections, especially in hospitals and clinics, where treatment and prevention have become more difficult due to the formation of biofilms. Owing to biofilm-derived antibiotic tolerance, the currently available traditional antibiotics have failed to treat MRSA infections. Hence, there is a urgent need to develop novel antibiotics for treating life-threatening MRSA infections. Lugdunin (cyclic peptide-1), a nonribosomal cyclic peptide produced by Staphylococcus lugdunensis, exhibits potent antimicrobial activity against MRSA. Amazingly, cyclic peptide-1 and its analogues cyclic peptide-11 and cyclic peptide-14 have the ability to disperse mature MRSA biofilms and show anti-clinical MRSA activity, including MRSA persister cells. In addition, these three cyclic peptide compounds have non-toxicity, lower hemolytic activity and lack of resistance development. Our results indicate that cyclic peptide-1, cyclic peptide-11, and cyclic peptide-14 have great potential as new antimicrobial drug candidates for the treatment of clinical MRSA infections.
Collapse
Affiliation(s)
- Guoxing Wei
- Chongqing Key Laboratory of Natural Product Synthesis and Drug Research, School of Pharmaceutical Sciences, Chongqing University, Chongqing 401331, China
| | - Yun He
- Chongqing Key Laboratory of Natural Product Synthesis and Drug Research, School of Pharmaceutical Sciences, Chongqing University, Chongqing 401331, China
| |
Collapse
|
41
|
Coraça-Huber DC, Steixner SJM, Najman S, Stojanovic S, Finze R, Rimashevskiy D, Saginova D, Barbeck M, Schnettler R. Lyophilized Human Bone Allograft as an Antibiotic Carrier: An In Vitro and In Vivo Study. Antibiotics (Basel) 2022; 11:antibiotics11070969. [PMID: 35884224 PMCID: PMC9312243 DOI: 10.3390/antibiotics11070969] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 06/14/2022] [Revised: 07/04/2022] [Accepted: 07/13/2022] [Indexed: 02/04/2023] Open
Abstract
Background: Antibiotics delivered from implanted bone substitute materials (BSM) can potentially be used to prevent acute infections and biofilm formation, providing high concentrations of antibiotics at the surgical site without systemic toxicity. In addition, BSM should allow osteoconductivity supporting bone healing without further surgery. Promising results have been achieved using lyophilized bone allografts mixed with antibiotics. Methods: In this study specially prepared human bone allografts were evaluated as an antibiotic carrier in vitro and in vivo. The efficacy of different antibiotic-impregnated bone allografts was measured by drug release tests in vitro and in vivo and bacterial susceptibility tests using four bacterial species usually responsible for implant-associated infections. Results: The loading procedures of allograft bone substitutes with antibiotics were successful. Some of the antibiotic concentrations exceeded the MIC90 for up to 7 days in vitro and for up to 72 h in vivo. The susceptibility tests showed that S. epidermidis ATCC 12228 was the most susceptible bacterial species in comparison to the other strains tested for all antibiotic substances. Vancomycin and rifampicin showed the best results against standard and patient-isolated strains in vitro. In vivo, new bone formation was comparable in all study groups including the control group without antibiotic loading. Conclusions: Human bone allografts showed the capacity to act as customized loaded antibiotic carriers to prevent acute infections and should be considered in the management of bone infections in combination with systemic antimicrobial therapy.
Collapse
Affiliation(s)
- Débora C. Coraça-Huber
- Research Laboratory for Biofilms and Implant Associated Infections (BIOFILM LAB), Experimental Orthopaedics, University Hospital for Orthopaedics and Traumatology, Medical University of Innsbruck, Peter-Mayr-Strasse 4b, Room 204, 6020 Innsbruck, Austria;
- Correspondence: ; Tel.: +43-512-9003-71697; Fax: +43-512-9003-73691
| | - Stephan J. M. Steixner
- Research Laboratory for Biofilms and Implant Associated Infections (BIOFILM LAB), Experimental Orthopaedics, University Hospital for Orthopaedics and Traumatology, Medical University of Innsbruck, Peter-Mayr-Strasse 4b, Room 204, 6020 Innsbruck, Austria;
| | - Stevo Najman
- Department of Cell and Tissue Engineering, Scientific Research Center for Biomedicine, Faculty of Medicine, University of Niš, Blvd. Dr Zorana Djindjica, 18108 Niš, Serbia; (S.N.); (S.S.)
- Department of Biology and Human Genetics, Faculty of Medicine, University of Niš, Blvd. Dr Zorana Djindjica, 18108 Niš, Serbia
| | - Sanja Stojanovic
- Department of Cell and Tissue Engineering, Scientific Research Center for Biomedicine, Faculty of Medicine, University of Niš, Blvd. Dr Zorana Djindjica, 18108 Niš, Serbia; (S.N.); (S.S.)
- Department of Biology and Human Genetics, Faculty of Medicine, University of Niš, Blvd. Dr Zorana Djindjica, 18108 Niš, Serbia
| | - Ronja Finze
- University Medical Centre, Justus Liebig University of Giessen, 35390 Giessen, Germany; (R.F.); (R.S.)
| | - Denis Rimashevskiy
- Department of Traumatology and Orthopedics, Peoples Friendship University of Russia, Miklukho-Maklaya Street 6, 117198 Moscow, Russia;
| | - Dina Saginova
- National Scientific Center of Traumatology and Orthopedics Named after Academician N. D. Batpenov, 15a Abylay khan Ave., Nur-Sultan 01000, Kazakhstan;
| | - Mike Barbeck
- Clinic and Policlinic for Dermatology and Venereology, University Medical Center Rostock, 18057 Rostock, Germany;
- BerlinAnalytix GmbH, 12109 Berlin, Germany
| | - Reinhard Schnettler
- University Medical Centre, Justus Liebig University of Giessen, 35390 Giessen, Germany; (R.F.); (R.S.)
| |
Collapse
|
42
|
Xiu W, Wan L, Yang K, Li X, Yuwen L, Dong H, Mou Y, Yang D, Wang L. Potentiating hypoxic microenvironment for antibiotic activation by photodynamic therapy to combat bacterial biofilm infections. Nat Commun 2022; 13:3875. [PMID: 35790729 PMCID: PMC9256606 DOI: 10.1038/s41467-022-31479-x] [Citation(s) in RCA: 105] [Impact Index Per Article: 35.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 11/08/2021] [Accepted: 06/20/2022] [Indexed: 12/20/2022] Open
Abstract
Traditional antibiotic treatment has limited efficacy for the drug-tolerant bacteria present in biofilms because of their unique metabolic conditions in the biofilm infection microenvironment. Modulating the biofilm infection microenvironment may influence the metabolic state of the bacteria and provide alternative therapeutic routes. In this study, photodynamic therapy is used not only to eradicate methicillin-resistant Staphylococcus aureus biofilms in the normoxic condition, but also to potentiate the hypoxic microenvironment, which induces the anaerobic metabolism of methicillin-resistant Staphylococcus aureus and activates the antibacterial activity of metronidazole. Moreover, the photodynamic therapy-activated chemotherapy can polarize the macrophages to a M2-like phenotype and promote the repair of the biofilm infected wounds in mice. This biofilm infection microenvironment modulation strategy, whereby the hypoxic microenvironment is potentiated to synergize photodynamic therapy with chemotherapy, provides an alternative pathway for efficient treatment of biofilm-associated infections. Bacteria in biofilms present unique metabolic conditions that limit the traditional antibiotic treatment. Here, the authors show a photodynamic therapy-activated chemotherapy potentiating the hypoxia of biofilms of methicillin-resistant Staphylococcus aureus, by developing hyaluronic acid nanoparticles functionalized with chlorin e6 and metronidazole.
Collapse
Affiliation(s)
- Weijun Xiu
- State Key Laboratory for Organic Electronics and Information Displays & Jiangsu Key Laboratory for Biosensors, Institute of Advanced Materials (IAM), Jiangsu National Synergetic Innovation Centre for Advanced Materials (SICAM), Nanjing University of Posts and Telecommunications, Nanjing, 210023, China
| | - Ling Wan
- State Key Laboratory for Organic Electronics and Information Displays & Jiangsu Key Laboratory for Biosensors, Institute of Advanced Materials (IAM), Jiangsu National Synergetic Innovation Centre for Advanced Materials (SICAM), Nanjing University of Posts and Telecommunications, Nanjing, 210023, China
| | - Kaili Yang
- State Key Laboratory for Organic Electronics and Information Displays & Jiangsu Key Laboratory for Biosensors, Institute of Advanced Materials (IAM), Jiangsu National Synergetic Innovation Centre for Advanced Materials (SICAM), Nanjing University of Posts and Telecommunications, Nanjing, 210023, China
| | - Xiao Li
- State Key Laboratory for Organic Electronics and Information Displays & Jiangsu Key Laboratory for Biosensors, Institute of Advanced Materials (IAM), Jiangsu National Synergetic Innovation Centre for Advanced Materials (SICAM), Nanjing University of Posts and Telecommunications, Nanjing, 210023, China
| | - Lihui Yuwen
- State Key Laboratory for Organic Electronics and Information Displays & Jiangsu Key Laboratory for Biosensors, Institute of Advanced Materials (IAM), Jiangsu National Synergetic Innovation Centre for Advanced Materials (SICAM), Nanjing University of Posts and Telecommunications, Nanjing, 210023, China.
| | - Heng Dong
- Nanjing Stomatological Hospital, Medicine School of Nanjing University, Nanjing, 210008, China
| | - Yongbin Mou
- Nanjing Stomatological Hospital, Medicine School of Nanjing University, Nanjing, 210008, China
| | - Dongliang Yang
- School of Physical and Mathematical Sciences & Institute of Advanced Materials (IAM), Nanjing Tech University, Nanjing, 211800, China
| | - Lianhui Wang
- State Key Laboratory for Organic Electronics and Information Displays & Jiangsu Key Laboratory for Biosensors, Institute of Advanced Materials (IAM), Jiangsu National Synergetic Innovation Centre for Advanced Materials (SICAM), Nanjing University of Posts and Telecommunications, Nanjing, 210023, China.
| |
Collapse
|
43
|
De Masi R, Orlando S. GANAB and N-Glycans Substrates Are Relevant in Human Physiology, Polycystic Pathology and Multiple Sclerosis: A Review. Int J Mol Sci 2022; 23:7373. [PMID: 35806376 PMCID: PMC9266668 DOI: 10.3390/ijms23137373] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 05/13/2022] [Revised: 06/22/2022] [Accepted: 06/28/2022] [Indexed: 11/29/2022] Open
Abstract
Glycans are one of the four fundamental macromolecular components of living matter, and they are highly regulated in the cell. Their functions are metabolic, structural and modulatory. In particular, ER resident N-glycans participate with the Glc3Man9GlcNAc2 highly conserved sequence, in protein folding process, where the physiological balance between glycosylation/deglycosylation on the innermost glucose residue takes place, according GANAB/UGGT concentration ratio. However, under abnormal conditions, the cell adapts to the glucose availability by adopting an aerobic or anaerobic regimen of glycolysis, or to external stimuli through internal or external recognition patterns, so it responds to pathogenic noxa with unfolded protein response (UPR). UPR can affect Multiple Sclerosis (MS) and several neurological and metabolic diseases via the BiP stress sensor, resulting in ATF6, PERK and IRE1 activation. Furthermore, the abnormal GANAB expression has been observed in MS, systemic lupus erythematous, male germinal epithelium and predisposed highly replicating cells of the kidney tubules and bile ducts. The latter is the case of Polycystic Liver Disease (PCLD) and Polycystic Kidney Disease (PCKD), where genetically induced GANAB loss affects polycystin-1 (PC1) and polycystin-2 (PC2), resulting in altered protein quality control and cyst formation phenomenon. Our topics resume the role of glycans in cell physiology, highlighting the N-glycans one, as a substrate of GANAB, which is an emerging key molecule in MS and other human pathologies.
Collapse
Affiliation(s)
- Roberto De Masi
- Complex Operative Unit of Neurology, “F. Ferrari” Hospital, Casarano, 73042 Lecce, Italy;
- Laboratory of Neuroproteomics, Multiple Sclerosis Centre, “F. Ferrari” Hospital, Casarano, 73042 Lecce, Italy
| | - Stefania Orlando
- Laboratory of Neuroproteomics, Multiple Sclerosis Centre, “F. Ferrari” Hospital, Casarano, 73042 Lecce, Italy
| |
Collapse
|
44
|
Che J, Sun L, Shan J, Shi Y, Zhou Q, Zhao Y, Sun L. Artificial Lipids and Macrophage Membranes Coassembled Biomimetic Nanovesicles for Antibacterial Treatment. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2022; 18:e2201280. [PMID: 35616035 DOI: 10.1002/smll.202201280] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Academic Contribution Register] [Received: 02/27/2022] [Revised: 04/23/2022] [Indexed: 06/15/2023]
Abstract
Tissue bacterial infections are a major pathological factor in many diseases. Effects on this aspect are in focus for the development of coordinated therapeutic strategies for bacterial killing and anti-inflammation. Here, inspired by the biodetoxification capacity of immune cells, multifunctional biomimetic nanovesicles (MϕM-LPs) that are co-assembled by macrophage membranes and artificial lipids to deliver antibiotics for treating bacterial infections, are presented. The macrophage membrane endows the MϕM-LPs with the capacity of lipopolysaccharide and inflammatory cytokine neutralization, while the artificial lipid membrane can be further engineered to increase the fluidity and anchor to bacteria. In addition, the MϕM-LPs can deliver sufficient ciprofloxacin with controllable release to accomplish an excellent antibacterial activity and biodetoxification capacity in vitro. Based on these advantages, it is demonstrated in a mouse model of Staphylococcus aureus (S. aureus) focal infection, that a single injection of the biomimetic nanovesicles can effectively anchor to and eliminate S. aureus in the infected tissue and reduce inflammatory cytokine levels. Thus, the tissue regeneration and collagen deposition can be accelerated. These results indicate the potential values of integrating natural and artificial membrane materials as a multifunctional biomimetic drug delivery system to treat bacterial infections.
Collapse
Affiliation(s)
- Junyi Che
- Department of Rheumatology and Immunology, Institute of Translational Medicine, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, 210008, China
| | - Lingyu Sun
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096, China
| | - Jingyang Shan
- Department of Neurology, Shenzhen Institute of Translational Medicine, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen, 518000, China
| | - Yong Shi
- Department of Rheumatology and Immunology, Institute of Translational Medicine, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, 210008, China
| | - Qing Zhou
- Department of Cardio-Thoracic Surgery, Institute of Translational Medicine, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, 210008, China
| | - Yuanjin Zhao
- Department of Rheumatology and Immunology, Institute of Translational Medicine, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, 210008, China
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096, China
| | - Lingyun Sun
- Department of Rheumatology and Immunology, Institute of Translational Medicine, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, 210008, China
| |
Collapse
|
45
|
Asare EO, Mun EA, Marsili E, Paunov VN. Nanotechnologies for control of pathogenic microbial biofilms. J Mater Chem B 2022; 10:5129-5153. [PMID: 35735175 DOI: 10.1039/d2tb00233g] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 12/12/2022]
Abstract
Biofilms are formed at interfaces by microorganisms, which congregate in microstructured communities embedded in a self-produced extracellular polymeric substance (EPS). Biofilm-related infections are problematic due to the high resistance towards most clinically used antimicrobials, which is associated with high mortality and morbidity, combined with increased hospital stays and overall treatment costs. Several new nanotechnology-based approaches have recently been proposed for targeting resistant bacteria and microbial biofilms. Here we discuss the impacts of biofilms on healthcare, food processing and packaging, and water filtration and distribution systems, and summarize the emerging nanotechnological strategies that are being developed for biofilm prevention, control and eradication. Combination of novel nanomaterials with conventional antimicrobial therapies has shown great potential in producing more effective platforms for controlling biofilms. Recent developments include antimicrobial nanocarriers with enzyme surface functionality that allow passive infection site targeting, degradation of the EPS and delivery of high concentrations of antimicrobials to the residing cells. Several stimuli-responsive antimicrobial formulation strategies have taken advantage of the biofilm microenvironment to enhance interaction and passive delivery into the biofilm sites. Nanoparticles of ultralow size have also been recently employed in formulations to improve the EPS penetration, enhance the carrier efficiency, and improve the cell wall permeability to antimicrobials. We also discuss antimicrobial metal and metal oxide nanoparticle formulations which provide additional mechanical factors through externally induced actuation and generate reactive oxygen species (ROS) within the biofilms. The review helps to bridge microbiology with materials science and nanotechnology, enabling a more comprehensive interdisciplinary approach towards the development of novel antimicrobial treatments and biofilm control strategies.
Collapse
Affiliation(s)
- Evans O Asare
- Department of Chemistry, School of Sciences and Humanities, Nazarbayev University, 53 Kabanbay Batyr Avenue, Nursultan city, 010000, Kazakhstan.
| | - Ellina A Mun
- Department of Chemistry, School of Sciences and Humanities, Nazarbayev University, 53 Kabanbay Batyr Avenue, Nursultan city, 010000, Kazakhstan.
| | - Enrico Marsili
- Department of Chemical Engineering, School of Engineering and Digital Sciences, Nazarbayev University, 53 Kabanbay Batyr Avenue, Nursultan city, 010000, Kazakhstan
| | - Vesselin N Paunov
- Department of Chemistry, School of Sciences and Humanities, Nazarbayev University, 53 Kabanbay Batyr Avenue, Nursultan city, 010000, Kazakhstan.
| |
Collapse
|
46
|
Salem SS, Badawy MSEM, Al-Askar AA, Arishi AA, Elkady FM, Hashem AH. Green Biosynthesis of Selenium Nanoparticles Using Orange Peel Waste: Characterization, Antibacterial and Antibiofilm Activities against Multidrug-Resistant Bacteria. LIFE (BASEL, SWITZERLAND) 2022; 12:life12060893. [PMID: 35743924 PMCID: PMC9227136 DOI: 10.3390/life12060893] [Citation(s) in RCA: 42] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Academic Contribution Register] [Received: 05/16/2022] [Revised: 06/10/2022] [Accepted: 06/11/2022] [Indexed: 12/13/2022]
Abstract
There is an increase of pathogenic multidrug-resistant bacteria globally due to the misuse of antibiotics. Recently, more scientists used metal nanoparticles to counteract antibacterial resistance. In this study, orange peel waste (OPW) was used for selenium nanoparticles’ (Se-NPs) biosynthesis through the green and ecofriendly method, and their applications as antibacterial and antibiofilm agents. Green biosynthesized Se-NPs were characterized using FTIR, XRD, SEM, EDAX, and TEM. Characterization results revealed that biosynthesized Se-NPs were highly crystalline, spherical, and polydisperse, and had sizes in the range of 16–95 nm. The biosynthesized Se-NPs were evaluated as antibacterial and antibiofilm activities against multidrug-resistant bacteria. Results illustrated that Se-NPs exhibited potential antibacterial activity against Gram-positive bacteria (S. aureus ATCC 29213 and biofilm-producing clinical isolates of S. aureus) and Gram-negative bacteria (Pseudomonas aeruginosa PAO1, MDR, biofilm, and quorum-sensing and producing clinical isolates of MDR P. aeruginosa, MDR E. coli, and K. pneumonia). Moreover, results illustrated that S. aureus ATCC 29213 was the most sensitive bacteria to Se-NPs at 1000 µg/mL, where the inhibition zone was 35 mm and MIC was 25 µg/mL. Furthermore, Se-NPs at 0.25 and 0.5 MIC decreased the biofilm significantly. The largest inhibition of biofilm was noticed in MDR K. pneumonia, which was 62% and 92% at 0.25 and 0.5 MIC, respectively. In conclusion, Se-NPs were successfully biosynthesized using OPW through the green method and had promising antibacterial and antibiofilm activity against multidrug-resistant bacteria, which can be used later in fighting resistant bacteria.
Collapse
Affiliation(s)
- Salem S. Salem
- Botany and Microbiology Department, Faculty of Science, Al-Azhar University, Nasr City, Cairo 11884, Egypt
- Correspondence: (S.S.S.); (A.A.A.-A.); (A.H.H.)
| | - Mona Shaban E. M. Badawy
- Department of Microbiology and Immunology, Faculty of Pharmacy (Girls), Al-Azhar University, Nasr City, Cairo 11884, Egypt;
| | - Abdulaziz A. Al-Askar
- Department of Botany and Microbiology, Faculty of Science, King Saud University, Riyadh 12372, Saudi Arabia
- Correspondence: (S.S.S.); (A.A.A.-A.); (A.H.H.)
| | - Amr Abker Arishi
- School of Molecular Sciences, The University of Western Australia, Perth, WA 6009, Australia;
| | - Fathy M. Elkady
- Microbiology and Immunology Department, Faculty of Pharmacy (Boys), Al-Azhar University, Nasr City, Cairo 11884, Egypt;
| | - Amr H. Hashem
- Botany and Microbiology Department, Faculty of Science, Al-Azhar University, Nasr City, Cairo 11884, Egypt
- Correspondence: (S.S.S.); (A.A.A.-A.); (A.H.H.)
| |
Collapse
|
47
|
Li P, Yu M, Ke X, Gong X, Li Z, Xing X. Cytocompatible Amphipathic Carbon Quantum Dots as Potent Membrane-Active Antibacterial Agents with Low Drug Resistance and Effective Inhibition of Biofilm Formation. ACS APPLIED BIO MATERIALS 2022; 5:3290-3299. [PMID: 35700313 DOI: 10.1021/acsabm.2c00292] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 11/29/2022]
Abstract
It is very challenging to design nanomaterials with both excellent antibacterial activity and cytocompatibility when facing bacterial infection. Here, inspired by antimicrobial peptides (AMPs), we fabricate carbon quantum dots (CQDs) derived from hydrophobic tryptophan and hydrophilic lysine or arginine (Lys/Trp-CQDs and Arg/Trp-CQDs), which possess amphipathic properties. These CQDs could effectively destroy bacterial membranes without developing resistance, inhibit biofilms formed by Staphylococcus aureus, and exhibit good in vitro biocompatibility. The antibacterial activities are caused by not only surface cationic structures and excess intracellular reactive oxygen species (ROS) generated by the CQDs but also the effects of the surface hydrophobic groups. These combined mechanisms of actions lead to bacterial membrane disruption, which raises the hope for combating bacterial infection without concern about drug resistance. What's more, the effect of amphiphilicity on balancing sterilization with biocompatibility expands the research ideas for developing available antibacterial nanomaterials.
Collapse
Affiliation(s)
- Peili Li
- College of Chemistry and Materials Engineering, Anhui Science and Technology University, Bengbu 233000, China
| | - Meizhe Yu
- School of Chemistry and Chemical Engineering, Nanjing University of Science and Technology, Nanjing 210094, China
| | - Xiang Ke
- College of Chemistry and Materials Engineering, Anhui Science and Technology University, Bengbu 233000, China
| | - Xuedong Gong
- School of Chemistry and Chemical Engineering, Nanjing University of Science and Technology, Nanjing 210094, China
| | - Zirong Li
- College of Chemistry and Materials Engineering, Anhui Science and Technology University, Bengbu 233000, China
| | - Xiaodong Xing
- School of Chemistry and Chemical Engineering, Nanjing University of Science and Technology, Nanjing 210094, China
| |
Collapse
|
48
|
Probing the growth and mechanical properties of Bacillus subtilis biofilms through genetic mutation strategies. Synth Syst Biotechnol 2022; 7:965-971. [PMID: 35756965 PMCID: PMC9194759 DOI: 10.1016/j.synbio.2022.05.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 03/12/2022] [Revised: 05/18/2022] [Accepted: 05/20/2022] [Indexed: 12/02/2022] Open
Abstract
Bacterial communities form biofilms on various surfaces by synthesizing a cohesive and protective extracellular matrix, and these biofilms protect microorganisms against harsh environmental conditions. Bacillus subtilis is a widely used experimental species, and its biofilms are used as representative models of beneficial biofilms. Specifically, B. subtilis biofilms are known to be rich in extracellular polymeric substances (EPS) and other biopolymers such as DNA and proteins like the amyloid protein TasA and the hydrophobic protein BslA. These materials, which form an interconnected, cohesive, three-dimensional polymer network, provide the mechanical stability of biofilms and mediate their adherence to surfaces among other functional contributions. Here, we explored how genetically-encoded components specifically contribute to regulate the growth status, mechanical properties, and antibiotic resistance of B. subtilis biofilms, thereby establishing a solid empirical basis for understanding how various genetic engineering efforts are likely to affect the structure and function of biofilms. We noted discrete contributions to biofilm morphology, mechanical properties, and survival from major biofilm components such as EPS, TasA and BslA. For example, EPS plays an important role in maintaining the stability of the mechanical properties and the antibiotic resistance of biofilms, whereas BslA has a significant impact on the resolution that can be obtained for printing applications. This work provides a deeper understanding of the internal interactions of biofilm components through systematic genetic manipulations. It thus not only broadens the application prospects of beneficial biofilms, but also serves as the basis of future strategies for targeting and effectively removing harmful biofilms.
Collapse
|
49
|
El-Sayed DS, El-Faham A, Soliman SM. Synthesis, molecular and supramolecular structure aspects and biological evaluations of a novel [Ag2(phthalazine)(NO3)2]n 3D coordination polymer. J Mol Struct 2022. [DOI: 10.1016/j.molstruc.2022.132592] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 10/19/2022]
|
50
|
Ndukwe ARN, Wiedbrauk S, Boase NRB, Fairfull‐Smith KE. Strategies to Improve the Potency of Oxazolidinones towards Bacterial Biofilms. Chem Asian J 2022; 17:e202200201. [PMID: 35352479 PMCID: PMC9321984 DOI: 10.1002/asia.202200201] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 02/28/2022] [Revised: 03/28/2022] [Indexed: 11/29/2022]
Abstract
Biofilms are part of the natural lifecycle of bacteria and are known to cause chronic infections that are difficult to treat. Most antibiotics are developed and tested against bacteria in the planktonic state and are ineffective against bacterial biofilms. The oxazolidinones, including the last resort drug linezolid, are one of the main classes of synthetic antibiotics progressed to clinical use in the last 50 years. They have a unique mechanism of action and only develop low levels of resistance in the clinical setting. With the aim of providing insight into strategies to design more potent antibiotic compounds with activity against bacterial biofilms, we review the biofilm activity of clinically approved oxazolidinones and report on structural modifications to oxazolidinones and their delivery systems which lead to enhanced anti-biofilm activity.
Collapse
Affiliation(s)
- Audrey R. N. Ndukwe
- School of Chemistry and Physics, Faculty of ScienceQueensland University of TechnologyBrisbaneQueensland4001Australia
- Centre for Materials ScienceQueensland University of TechnologyBrisbaneQueensland4001Australia
| | - Sandra Wiedbrauk
- School of Chemistry and Physics, Faculty of ScienceQueensland University of TechnologyBrisbaneQueensland4001Australia
- Centre for Materials ScienceQueensland University of TechnologyBrisbaneQueensland4001Australia
| | - Nathan R. B. Boase
- School of Chemistry and Physics, Faculty of ScienceQueensland University of TechnologyBrisbaneQueensland4001Australia
- Centre for Materials ScienceQueensland University of TechnologyBrisbaneQueensland4001Australia
| | - Kathryn E. Fairfull‐Smith
- School of Chemistry and Physics, Faculty of ScienceQueensland University of TechnologyBrisbaneQueensland4001Australia
- Centre for Materials ScienceQueensland University of TechnologyBrisbaneQueensland4001Australia
| |
Collapse
|