1
|
Obeid MA, Alyamani H, Alenaizat A, Tunç T, Aljabali AAA, Alsaadi MM. Nanomaterial-based drug delivery systems in overcoming bacterial resistance: Current review. Microb Pathog 2025; 203:107455. [PMID: 40057006 DOI: 10.1016/j.micpath.2025.107455] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2024] [Revised: 03/02/2025] [Accepted: 03/06/2025] [Indexed: 03/14/2025]
Abstract
Antimicrobial resistance is one of the most serious contemporary global health concerns, threatening the effectiveness of existing antibiotics and resulting in morbidity, mortality, and economic burdens. This review examines the contribution of nanomaterial-based drug delivery systems to solving the problems associated with bacterial resistance and provides a thorough overview of their mechanisms of action, efficiency, and perspectives for the future. Owing to their unique physicochemical properties, nanomaterials reveal new ways of passing through the traditional mechanisms of bacterial defence connected to the permeability barrier of membranes, efflux pumps, and biofilm formation. This review addresses the different types of nanomaterials, including metallic nanoparticles, liposomes, and polymeric nanoparticles, in terms of their antimicrobial properties and modes of action. More emphasis has been placed on the critical discussion of recent studies on such active systems. Both in vitro and in vivo models are discussed, with particular attention paid to multidrug-resistant bacteria. This review begins by reviewing the urgency for antimicrobial resistance (AMR) by citing recent statistics, which indicate that the number of deaths and reasons for financial losses continue to increase. A background is then provided on the limitations of existing antibiotic therapies and the pressing need to develop innovative approaches. Nanomaterial-based drug delivery systems have been proposed as promising solutions because of their potential to improve drug solubility, stability, and targeted delivery, although side effects can also be mitigated. In addition to established knowledge, this review also covers ongoing debates on the continuous risks associated with the use of nanomaterials, such as toxicity and environmental impact. This discussion emphasizes the optimization of nanomaterial design to target specific bacteria, and rigorous clinical trials to establish safety and efficacy in humans. It concludes with reflections on the future directions of nanomaterial-based drug delivery systems in fighting AMR, underlining the need for an interdisciplinary approach, along with continuous research efforts to translate these promising technologies into clinical practice. As the fight against bacterial resistance reaches its peak, nanomaterials may be the key to developing next-generation antimicrobial therapies.
Collapse
Affiliation(s)
- Mohammad A Obeid
- Department of Pharmaceutics and Pharmaceutical Technology, Faculty of Pharmacy, Yarmouk University, P.O.BOX 566, Irbid, 21163, Jordan.
| | - Hanin Alyamani
- William Harvey Research Institute, Center for Microvascular Research, Queen Mary University of London, London, United Kingdom
| | | | - Tutku Tunç
- Department of Pharmaceutical Microbiology, Faculty of Pharmacy, Sivas Cumhuriyet University, Sivas, Turkey
| | - Alaa A A Aljabali
- Department of Pharmaceutics and Pharmaceutical Technology, Faculty of Pharmacy, Yarmouk University, P.O.BOX 566, Irbid, 21163, Jordan
| | - Manal M Alsaadi
- Department of Industrial Pharmacy, Faculty of Pharmacy, University of Tripoli, PO Box, Tripoli, 13645, Libya
| |
Collapse
|
2
|
Kharga K, Jha S, Vishwakarma T, Kumar L. Current developments and prospects of the antibiotic delivery systems. Crit Rev Microbiol 2025; 51:44-83. [PMID: 38425122 DOI: 10.1080/1040841x.2024.2321480] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Revised: 02/11/2024] [Accepted: 02/16/2024] [Indexed: 03/02/2024]
Abstract
Antibiotics have remained the cornerstone for the treatment of bacterial infections ever since their discovery in the twentieth century. The uproar over antibiotic resistance among bacteria arising from genome plasticity and biofilm development has rendered current antibiotic therapies ineffective, urging the development of innovative therapeutic approaches. The development of antibiotic resistance among bacteria has further heightened the clinical failure of antibiotic therapy, which is often linked to its low bioavailability, side effects, and poor penetration and accumulation at the site of infection. In this review, we highlight the potential use of siderophores, antibodies, cell-penetrating peptides, antimicrobial peptides, bacteriophages, and nanoparticles to smuggle antibiotics across impermeable biological membranes to achieve therapeutically relevant concentrations of antibiotics and combat antimicrobial resistance (AMR). We will discuss the general mechanisms via which each delivery system functions and how it can be tailored to deliver antibiotics against the paradigm of mechanisms underlying antibiotic resistance.
Collapse
Affiliation(s)
- Kusum Kharga
- School of Biotechnology, Faculty of Applied Sciences and Biotechnology, Shoolini University, Himachal Pradesh, India
| | - Shubhang Jha
- School of Bioengineering and Food Technology, Faculty of Applied Sciences and Biotechnology, Shoolini University, Himachal Pradesh, India
| | - Tanvi Vishwakarma
- School of Bioengineering and Food Technology, Faculty of Applied Sciences and Biotechnology, Shoolini University, Himachal Pradesh, India
| | - Lokender Kumar
- School of Biotechnology, Faculty of Applied Sciences and Biotechnology, Shoolini University, Himachal Pradesh, India
| |
Collapse
|
3
|
Amer AM, Charnock C, Nguyen S. The impact of surface charge on the interaction of cholesterol-free fusogenic liposomes with planktonic microbial cells and biofilms. Int J Pharm 2025; 669:125088. [PMID: 39706381 DOI: 10.1016/j.ijpharm.2024.125088] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2024] [Revised: 12/05/2024] [Accepted: 12/13/2024] [Indexed: 12/23/2024]
Abstract
This study focused on the development of cholesterol-free fusogenic liposomes with different surface charge with the aim of improving biofilm penetration. In vitro assessments of the liposomes included physical stability, biocompatibility, fusion with microbial cells, and the ability to penetrate established biofilms. Using dynamic light scattering, cholesterol-free, fusogenic liposomes were found to be < 200 nm in size with small size distribution (PDI < 0.1) and physically stable for a year when stored at 4 °C. Transmission electron microscopy (TEM) images confirmed vesicular sizes for selected liposomal formulations. Liposomal ability to fuse with microbial cells was assessed using lipid mixing and flow cytometer assays. Fusion levels were found to be higher with Escherichia coli compared to Staphylococcus aureus and Candida albicans regardless of the liposomal charge. Neutral liposomes exhibited highest fusion, followed by cationic and anionic liposomes, respectively. Our investigations demonstrated that fusion is a multifactorial process influenced by the chemical composition of the liposomes, the liposomal surface charge, and components of the microbial cell envelope. Penetration and retention within preformed S. aureus biofilms were assessed for liposomes with various surface charges. All liposomes, regardless of surface charge, were capable of penetrating and diffusing through the biofilm matrix. However, cationic liposomes displayed greatest interaction and retention. Biocompatibility was confirmed through haemolysis and cytotoxicity studies. The cholesterol-free fusogenic liposomes developed in this study demonstrated promising potential as drug delivery systems for incorporating antimicrobial agents for biofilm treatment.
Collapse
Affiliation(s)
- Ahmed M Amer
- Department of Life Sciences and Health, Oslo Metropolitan University (OsloMet), Oslo, Norway.
| | - Colin Charnock
- Department of Life Sciences and Health, Oslo Metropolitan University (OsloMet), Oslo, Norway
| | - Sanko Nguyen
- Department of Life Sciences and Health, Oslo Metropolitan University (OsloMet), Oslo, Norway
| |
Collapse
|
4
|
Panda S, Rout L, Mohanty N, Satpathy A, Sankar Satapathy B, Rath S, Gopinath D. Exploring the photosensitizing potential of Nanoliposome Loaded Improved Toluidine Blue O (NLITBO) Against Streptococcus mutans: An in-vitro feasibility study. PLoS One 2024; 19:e0312521. [PMID: 39475963 PMCID: PMC11524459 DOI: 10.1371/journal.pone.0312521] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2024] [Accepted: 10/08/2024] [Indexed: 11/02/2024] Open
Abstract
BACKGROUND Streptococcus mutans is a major contributor to dental caries due to its ability to produce acid and survive in biofilms. Microbial resistance towards common antimicrobial agents like chlorhexidine and triclosan has shifted the research towards antimicrobial Photodynamic therapy (PDT). In this context, Toluidine Blue O (TBO) is being explored for its photosensitizing properties against Streptococcus mutans. There is a huge variation in the effective concentration of TBO among the current studies owing to the differences in source of and delivery system TBO as well as the time, power and energy densities of light. OBJECTIVE The primary objectives of this study are to encapsulate improved Toluidine Blue O (ITBO) in nanoliposomes (NLITBO), characterize it, and evaluate its antibacterial photosensitizing potential against Streptococcus mutans suspensions in vitro. METHOD ITBO was synthesised as per Indian patent (number -543908). NLITBO was prepared using the thin-film hydration method. Dynamic light scattering experiment determined the vesicle size, polydispersity index (PDI), and zeta potential. Surface features were characterized by Scanning and Transmission Electron microscopy. ITBO release from NLITBO was assessed using the extrapolation method. The antibacterial activity of the NLITBO was determined by evaluating the zone of inhibition (ZOI) in the Streptococcus mutans culture and comparing with 2% chlorhexidine gluconate. The minimum inhibitory concentration (MIC) of NLITBO as a photosensitizer with red light (wavelength 650nm, power density 0.1 W/cm2, energy density 9-9.1 J/ cm2, 90seconds time) was evaluated against Streptococcus mutans cells by colorimetric method in 96 well plate. RESULTS Percentage drug loading, loading efficiency, yield percentage, vesicle size, PDI, Zeta potential of NLTBO was reported as 9.3±0.4%, 84.4±7.6%, 73.5%, 123.52 nm, 0.57, -39.54mV respectively. Clusters of uni-lamellar nanovesicles with smooth non-perforated surfaces were observed in SEM and TEM. The size of the vesicle was within 100 nm. At 24 hours, a cumulative 79.81% of ITBO was released from NLITBO. Mean ZOI and MIC of NLITBO (1 μg /ml) were found to be 0.7±0.2 mm, 0.6μg/ml respectively. CONCLUSION We have synthesized and encapsulated improved Toluidine Blue O (ITBO) in nanoliposomes (NLITBO) and thoroughly characterized the formulation. The antibacterial efficacy of NLITBO without light was demonstrated by ZOI which is similar to 2% chlorhexidine gluconate. MIC of NLITBO as a photosensitiser along with the optimal light parameter was also proposed in this study. These findings suggested that NLITBO could serve as an effective alternative to conventional antibacterial treatments in managing Streptococcus mutans rich biofilms. It can have potential pharmaceutical application in oral health care.
Collapse
Affiliation(s)
- Swagatika Panda
- Department of Oral Pathology and Microbiology, Institute of Dental Sciences, Siksha’O’Anusandhan Deemed to be University, Bhubaneswar, Odisha, India
| | - Lipsa Rout
- Institute of Dental Sciences, Siksha’O’Anusandhan Deemed to be University. Bhubaneswar, Odisha, India
| | - Neeta Mohanty
- Department of Oral Pathology and Microbiology, Institute of Dental Sciences, Siksha’O’Anusandhan Deemed to be University, Bhubaneswar, Odisha, India
| | - Anurag Satpathy
- Department of Periodontics and Implantology, Institute of Dental Sciences, Siksha’O’Anusandhan Deemed to be University, Bhubaneswar, Odisha, India
| | | | - Shakti Rath
- Department of Microbiology & Research, Institute of Dental Sciences, Siksha’O’Anusandhan Deemed to be University, Bhubaneswar, Odisha, India
| | - Divya Gopinath
- Basic Medical and Dental Sciences Dept, College of Dentistry, Ajman University, Ajman, United Arab Emirates
- Centre of Medical and Bio-Allied Health Sciences and Research, Ajman University, Ajman, United Arab Emirates
| |
Collapse
|
5
|
Alzahrani RR, Alkhulaifi MM, Al Jeraisy M, Albekairy AM, Ali R, Alrfaei BM, Ehaideb SN, Al-Asmari AI, Qahtani SA, Halwani A, Yassin AEB, Halwani MA. Enhancing Gentamicin Antibacterial Activity by Co-Encapsulation with Thymoquinone in Liposomal Formulation. Pharmaceutics 2024; 16:1330. [PMID: 39458659 PMCID: PMC11510464 DOI: 10.3390/pharmaceutics16101330] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2024] [Revised: 09/16/2024] [Accepted: 09/23/2024] [Indexed: 10/28/2024] Open
Abstract
BACKGROUND AND PURPOSE Gentamicin (GEN) is a broad-spectrum antibiotic that cannot be prescribed freely because of its toxicity. Thymoquinone (THQ), a phytochemical, has antibacterial, antioxidant, and toxicity-reducing properties. However, its hydrophobicity and light sensitivity make it challenging to utilize. This incited the idea of co-encapsulating GEN and THQ in liposomes (Lipo-GEN-THQ). METHOD Lipo-GEN-THQ were characterized using the zeta-potential, dynamic light scattering, Fourier transform infrared spectroscopy, and transmission electron microscope (TEM). The liposomes' stability was evaluated under different storage and biological conditions. Lipo-GEN-THQ's efficacy was investigated by the minimum inhibitory/bactericidal concentrations (MICs-MBCs), time-kill curves, and antibiofilm and antiadhesion assays. Bacterial interactions with the empty and GEN-THQ-loaded liposomes were evaluated using TEM. RESULTS The Lipo-GEN-THQ were spherical, monodispersed, and negatively charged. The Lipo-GEN-THQ were relatively stable and released GEN sustainably over 24 h. The liposomes exhibited significantly higher antibacterial activity than free GEN, as evidenced by the four-fold lower MIC and biofilm eradication in resistant E. coli strain (EC-219). TEM images display how the empty liposomes fused closely to the tested bacteria and how the loaded liposomes caused ultrastructure damage and intracellular component release. An antiadhesion assay showed that the Lipo-GEN-THQ and free GEN (0.125 mg/L) similarly inhibited Escherichia coli (EC-157) adhesion to the A549 cells (68% vs. 64%). CONCLUSIONS The Lipo-THQ-GEN enhanced GEN by combining it with THQ within the liposomes, reducing the effective dose. The reduction in the GEN dose after adding THQ may indirectly reduce the toxicity and aid in developing an enhanced and safer form of GEN.
Collapse
Affiliation(s)
- Raghad R. Alzahrani
- Department of Botany and Microbiology, College of Science, King Saud University, Riyadh 11451, Saudi Arabia; (R.R.A.); (M.M.A.)
- Nanomedicine Department, King Abdullah International Medical Research Center, King Saud bin Abdulaziz University for Health Sciences, Riyadh 11481, Saudi Arabia
| | - Manal M. Alkhulaifi
- Department of Botany and Microbiology, College of Science, King Saud University, Riyadh 11451, Saudi Arabia; (R.R.A.); (M.M.A.)
| | - Majed Al Jeraisy
- King Abdullah International Medical Research Center, King Saud bin Abdulaziz University for Health Sciences, Riyadh 11481, Saudi Arabia; (M.A.J.); (A.H.)
| | - Abdulkareem M. Albekairy
- Department of Pharmacy Practice, College of Pharmacy, King Abdullah International Medical Research Center, King Saud bin Abdulaziz University for Health Sciences, Riyadh 11481, Saudi Arabia;
- Pharmaceutical Care Department, King Abdulaziz Medical City, National Guard Health Affairs, Riyadh 11481, Saudi Arabia
| | - Rizwan Ali
- Medical Research Core Facility and Platforms, King Abdullah International Medical Research Center, King Saud bin Abdulaziz University for Health Sciences, Riyadh 11481, Saudi Arabia;
| | - Bahauddeen M. Alrfaei
- Stem Cells and Regenerative Medicine, King Abdullah International Medical Research Center, King Saud bin Abdulaziz University for Health Sciences, National Guard Health Affairs, Riyadh 11481, Saudi Arabia;
| | - Salleh N. Ehaideb
- Experimental Medicine Department, King Abdullah International Medical Research Center, King Saud bin Abdulaziz University for Health Sciences, Ministry of National Guard—Health Affairs, Riyadh 11481, Saudi Arabia;
| | - Ahmed I. Al-Asmari
- Department of Pathology and Laboratory Medicine, King Faisal Specialist Hospital and Research Centre, Riyadh 11211, Saudi Arabia
| | - Sultan Al Qahtani
- Department of Basic Medical Sciences, College of Medicine, King Abdullah International Medical Research Center, King Saud bin Abdulaziz University for Health Sciences, Riyadh 11481, Saudi Arabia;
| | - Abdulaziz Halwani
- King Abdullah International Medical Research Center, King Saud bin Abdulaziz University for Health Sciences, Riyadh 11481, Saudi Arabia; (M.A.J.); (A.H.)
- College of Dentistry, King Saud bin Abdul Aziz University for Health Sciences, Riyadh 11481, Saudi Arabia
| | - Alaa Eldeen B. Yassin
- College of Pharmacy, King Abdullah International Medical Research Center, King Saud bin Abdulaziz University for Health Sciences, Riyadh 11481, Saudi Arabia
| | - Majed A. Halwani
- Nanomedicine Department, King Abdullah International Medical Research Center, King Saud bin Abdulaziz University for Health Sciences, Riyadh 11481, Saudi Arabia
| |
Collapse
|
6
|
Singh I, Kumar S, Singh S, Wani MY. Overcoming resistance: Chitosan-modified liposomes as targeted drug carriers in the fight against multidrug resistant bacteria-a review. Int J Biol Macromol 2024; 278:135022. [PMID: 39182895 DOI: 10.1016/j.ijbiomac.2024.135022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2024] [Revised: 07/21/2024] [Accepted: 08/22/2024] [Indexed: 08/27/2024]
Abstract
Antimicrobial resistance (AMR) poses a significant global health threat, rendering standard antibiotics ineffective against multi-drug resistant bacteria. To tackle this urgent issue, innovative approaches are essential. Liposomes, small spherical vesicles made of a phospholipid bilayer, present a promising solution. These vesicles can encapsulate various medicines and are both biocompatible and biodegradable. Their ability to be modified for targeted tissue or cell uptake makes them an ideal drug delivery system. By delivering antibiotics directly to infection sites, liposomes minimize side effects and reduce the development of resistance. However, challenges such as poor stability and rapid drug leakage limit their biological application. Chitosan, a biocompatible polymer, enhances liposome interaction with specific tissues or cells, enabling selective drug release at infection sites. Incorporating chitosan into liposome formulations alters and diversifies their surface characteristics through electrostatic interactions, resulting in improved stability and pH-sensitive drug release. These interactions are crucial for enhancing drug retention and targeted delivery, especially in varying pH environments like tumor sites or infection areas, thereby improving therapeutic outcomes and reducing systemic side effects. This review discusses recent advancements, challenges, and the need for further research to optimize liposome formulations and enhance targeted drug delivery for effective AMR treatment. Chitosan-modified liposomes offer a promising strategy to overcome AMR and improve antimicrobial therapies.
Collapse
Affiliation(s)
- Ira Singh
- Functional Polymer Material Lab, Department of Chemistry, Harcourt Butler Technical University, Kanpur 208002, Uttar Pradesh, India
| | - Santosh Kumar
- Functional Polymer Material Lab, Department of Chemistry, Harcourt Butler Technical University, Kanpur 208002, Uttar Pradesh, India.
| | - Shalinee Singh
- Functional Polymer Material Lab, Department of Chemistry, Harcourt Butler Technical University, Kanpur 208002, Uttar Pradesh, India
| | - Mohmmad Younus Wani
- Department of Chemistry, College of Science, University of Jeddah, 21589 Jeddah, Saudi Arabia
| |
Collapse
|
7
|
Hemmati J, Chiani M, Chegini Z, Seifalian A, Arabestani MR. Surface modified niosomal quercetin with cationic lipid: an appropriate drug delivery system against Pseudomonas aeruginosa Infections. Sci Rep 2024; 14:13362. [PMID: 38862754 PMCID: PMC11167023 DOI: 10.1038/s41598-024-64416-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Accepted: 06/08/2024] [Indexed: 06/13/2024] Open
Abstract
The Increase in infections caused by resistant strains of Pseudomonas aeruginosa poses a formidable challenge to global healthcare systems. P. aeruginosa is capable of causing severe human infections across diverse anatomical sites, presenting considerable therapeutic obstacles due to its heightened drug resistance. Niosomal drug delivery systems offer enhanced pharmaceutical potential for loaded contents due to their desirable properties, mainly providing a controlled-release profile. This study aimed to formulate an optimized niosomal drug delivery system incorporating stearylamine (SA) to augment the anti-bacterial and anti-biofilm activities of quercetin (QCT) against both standard and clinical strains of P. aeruginosa. QCT-loaded niosome (QCT-niosome) and QCT-loaded SA- niosome (QCT-SA- niosome) were synthesized by the thin-film hydration technique, and their physicochemical characteristics were evaluated by field emission scanning electron microscopy (FE-SEM), zeta potential measurement, entrapment efficacy (EE%), and in vitro release profile. The anti-P. aeruginosa activity of synthesized niosomes was assessed using minimum inhibitory and bactericidal concentrations (MICs/MBCs) and compared with free QCT. Additionally, the minimum biofilm inhibitory and eradication concentrations (MBICs/MBECs) were carried out to analyze the ability of QCT-niosome and QCT-SA-niosome against P. aeruginosa biofilms. Furthermore, the cytotoxicity assay was conducted on the L929 mouse fibroblasts cell line to evaluate the biocompatibility of the formulated niosomes. FE-SEM analysis revealed that both synthesized niosomal formulations exhibited spherical morphology with different sizes (57.4 nm for QCT-niosome and 178.9 nm for QCT-SA-niosome). The EE% for cationic and standard niosomal formulations was reported at 75.9% and 59.6%, respectively. Both formulations showed an in vitro sustained-release profile, and QCT-SA-niosome exhibited greater stability during a 4-month storage time compared to QCT-niosome. Microbial experiments indicated that both prepared formulations had higher anti-bacterial and anti-biofilm activities than free QCT. Also, the QCT-SA-niosome exhibited greater reductions in MIC, MBC, MBIC, and MBEC values compared to the QCT-niosome at equivalent concentrations. This study supports the potential of QCT-niosome and QCT-SA-niosome as effective agents against P. aeruginosa infections, manifesting significant anti-bacterial and anti-biofilm efficacy alongside biocompatibility with L929 cell lines. Furthermore, our results suggest that optimized QCT-niosome with cationic lipids could efficiently target P. aeruginosa cells with negligible cytotoxic effect.
Collapse
Affiliation(s)
- Jaber Hemmati
- Department of Microbiology, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
- Student Research Committee, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Mohsen Chiani
- Department of NanoBiotechnology, Pasteur Institute of Iran, Tehran, Iran
| | - Zahra Chegini
- Department of Microbiology, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Alexander Seifalian
- Nanotechnology & Regenerative Medicine Commercialization Centre (NanoRegMed Ltd, Nanoloom Ltd & Liberum Health Ltd), LBIC, University of London, London, UK
| | - Mohammad Reza Arabestani
- Department of Microbiology, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran.
- Infectious Disease Research Center, Hamadan University of Medical Sciences, Hamadan, Iran.
| |
Collapse
|
8
|
Panthi VK, Fairfull-Smith KE, Islam N. Liposomal drug delivery strategies to eradicate bacterial biofilms: Challenges, recent advances, and future perspectives. Int J Pharm 2024; 655:124046. [PMID: 38554739 DOI: 10.1016/j.ijpharm.2024.124046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2023] [Revised: 03/08/2024] [Accepted: 03/23/2024] [Indexed: 04/02/2024]
Abstract
Typical antibiotic treatments are often ineffectual against biofilm-related infections since bacteria residing within biofilms have developed various mechanisms to resist antibiotics. To overcome these limitations, antimicrobial-loaded liposomal nanoparticles are a promising anti-biofilm strategy as they have demonstrated improved antibiotic delivery and eradication of bacteria residing in biofilms. Antibiotic-loaded liposomal nanoparticles revealed remarkably higher antibacterial and anti-biofilm activities than free drugs in experimental settings. Moreover, liposomal nanoparticles can be used efficaciously for the combinational delivery of antibiotics and other antimicrobial compounds/peptide which facilitate, for instance, significant breakdown of the biofilm matrix, increased bacterial elimination from biofilms and depletion of metabolic activity of various pathogens. Drug-loaded liposomes have mitigated recurrent infections and are considered a promising tool to address challenges associated to antibiotic resistance. Furthermore, it has been demonstrated that surface charge and polyethylene glycol modification of liposomes have a notable impact on their antibacterial biofilm activity. Future investigations should tackle the persistent hurdles associated with development of safe and effective liposomes for clinical application and investigate novel antibacterial treatments, including CRISPR-Cas gene editing, natural compounds, phages, and nano-mediated approaches. Herein, we emphasize the significance of liposomes in inhibition and eradication of various bacterial biofilms, their challenges, recent advances, and future perspectives.
Collapse
Affiliation(s)
- Vijay Kumar Panthi
- Pharmacy Discipline, School of Clinical Sciences, Queensland University of Technology (QUT), Brisbane, QLD, Australia
| | - Kathryn E Fairfull-Smith
- School of Chemistry and Physics, Faculty of Science, Queensland University of Technology, Brisbane, Queensland 4001, Australia; Centre for Materials Science, Queensland University of Technology, Brisbane, Queensland 4001, Australia
| | - Nazrul Islam
- Pharmacy Discipline, School of Clinical Sciences, Queensland University of Technology (QUT), Brisbane, QLD, Australia; Institute of Health and Biomedical Innovation (IHBI), Queensland University of Technology (QUT), Brisbane, QLD, Australia; Centre for Immunology and Infection Control (CIIC), Queensland University of Technology (QUT), Brisbane, QLD, Australia.
| |
Collapse
|
9
|
Ong HX, Traini D, Young PM. Liposomes for Inhalation. J Aerosol Med Pulm Drug Deliv 2024; 37:100-110. [PMID: 38640446 DOI: 10.1089/jamp.2024.29112.hxo] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/21/2024] Open
Abstract
Inhalation of liposomes formulated with phospholipids similar to endogenous lung surfactants and lipids offers biocompatibility and versatility within the pulmonary medicine field to treat a range of diseases such as lung cancer, cystic fibrosis and lung infections. Manipulation of the physicochemical properties of liposomes enables innovative design of the carrier to meet specific delivery, release and targeting requirements. This delivery system offers several benefits: improved pharmacokinetics with reduced toxicity, enhanced therapeutic efficacy, increased delivery of poorly soluble drugs, taste masking, biopharmaceutics degradation protection and targeted cellular therapy. This section provides an overview of liposomal formulation and delivery, together with their applications for different disease states in the lung.
Collapse
Affiliation(s)
- Hui Xin Ong
- Woolcock Institute for Medical Research, University of Sydney, Sydney, NSW, Australia
| | - Daniela Traini
- Woolcock Institute for Medical Research, University of Sydney, Sydney, NSW, Australia
| | - Paul M Young
- CEO, Ab Inito Pharma, Macquarie Park, NSW, Australia
| |
Collapse
|
10
|
Gkartziou F, Plota M, Kypraiou C, Gauttam I, Kolonitsiou F, Klepetsanis P, Spiliopoulou I, Antimisiaris SG. Daptomycin Liposomes Exhibit Enhanced Activity against Staphylococci Biofilms Compared to Free Drug. Pharmaceutics 2024; 16:459. [PMID: 38675120 PMCID: PMC11054717 DOI: 10.3390/pharmaceutics16040459] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2024] [Revised: 03/21/2024] [Accepted: 03/22/2024] [Indexed: 04/28/2024] Open
Abstract
The purpose of the present study was to investigate the anti-staphylococcal activity of liposomal daptomycin against four biofilm-producing S. aureus and S. epidermidis clinical strains, three of which are methicillin-resistant. Neutral and negatively charged daptomycin-loaded liposomes were prepared using three methods, namely, thin-film hydration (TFH), a dehydration-rehydration vesicle (DRV) method, and microfluidic mixing (MM); moreover, they were characterized for drug encapsulation (EE%), size distribution, zeta-potential, vesicle stability, drug release, and drug integrity. Interestingly, whilst drug loading in THF and DRV nanosized (by extrusion) vesicles was around 30-35, very low loading (~4%) was possible in MM vesicles, requiring further explanatory investigations. Liposomal encapsulation protected daptomycin from degradation and preserved its bioactivity. Biofilm mass (crystal violet, CV), biofilm viability (MTT), and growth curve (GC) assays evaluated the antimicrobial activity of neutral and negatively charged daptomycin-liposomes towards planktonic bacteria and biofilms. Neutral liposomes exhibited dramatically enhanced inhibition of bacterial growth (compared to the free drug) for all species studied, while negatively charged liposomes were totally inactive. Biofilm prevention and treatment studies revealed high antibiofilm activity of liposomal daptomycin. Neutral liposomes were more active for prevention and negative charge ones for treating established biofilms. Planktonic bacteria as well as the matured biofilms of low daptomycin-susceptible, methicillin-resistant Staphylococcus aureus (MRSA) and Staphylococcus epidermidis (MRSE) strains were almost completely eradicated by liposomal-daptomycin, indicating the need for their further exploration as antimicrobial therapeutics.
Collapse
Affiliation(s)
- Foteini Gkartziou
- Department of Pharmacy, School of Health Sciences, University of Patras, 26504 Patras, Greece; (C.K.)
- Institute of Chemical Engineering Sciences, FORTH/ICE-HT, Platani, 26504 Patras, Greece
| | - Maria Plota
- Department of Microbiology, School of Medicine, University of Patras, 26504 Patras, Greece; (M.P.); (F.K.)
- National Reference Centre for Staphylococci, School of Medicine, University of Patras, 26504 Patras, Greece;
| | - Charikleia Kypraiou
- Department of Pharmacy, School of Health Sciences, University of Patras, 26504 Patras, Greece; (C.K.)
| | - Iti Gauttam
- Department of Pharmacy, School of Health Sciences, University of Patras, 26504 Patras, Greece; (C.K.)
| | - Fevronia Kolonitsiou
- Department of Microbiology, School of Medicine, University of Patras, 26504 Patras, Greece; (M.P.); (F.K.)
- National Reference Centre for Staphylococci, School of Medicine, University of Patras, 26504 Patras, Greece;
| | - Pavlos Klepetsanis
- Department of Pharmacy, School of Health Sciences, University of Patras, 26504 Patras, Greece; (C.K.)
- Institute of Chemical Engineering Sciences, FORTH/ICE-HT, Platani, 26504 Patras, Greece
| | - Iris Spiliopoulou
- National Reference Centre for Staphylococci, School of Medicine, University of Patras, 26504 Patras, Greece;
| | - Sophia G. Antimisiaris
- Department of Pharmacy, School of Health Sciences, University of Patras, 26504 Patras, Greece; (C.K.)
- Institute of Chemical Engineering Sciences, FORTH/ICE-HT, Platani, 26504 Patras, Greece
| |
Collapse
|
11
|
Bharathi D, Lee JH, Lee J. Enhancement of antimicrobial and antibiofilm activities of liposomal fatty acids. Colloids Surf B Biointerfaces 2024; 234:113698. [PMID: 38070368 DOI: 10.1016/j.colsurfb.2023.113698] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2023] [Revised: 11/27/2023] [Accepted: 12/04/2023] [Indexed: 02/09/2024]
Abstract
Microbial biofilms are protected surface-attached communities of bacteria or fungi with high drug tolerance that typically cause persistent infections. Smart drug carriers are being explored as a promising platform of antimicrobials to address their recalcitrance to antibiotic agents and minimize the side effects of current therapies. In this study, soy lecithin liposomes loaded with lauric acid (LA) and myristoleic acid (MA) were formulated using an emulsification method, and their antibiofilm properties were evaluated. The physio-chemical properties of the most potent liposome were characterized using a zeta sizer, transmission electron microscopy (TEM), fourier transform infrared spectroscopy, and nuclear magnetic resonance spectroscopy. TEM and zeta sizer analysis of the liposome revealed a homogeneous spherical structure with an average size of 159.2 nm and zeta potential of - 5.4 mV. The unilamellar liposomes loaded with LA at 0.1-0.5 µg/mL achieved obvious antibiofilm efficiency against Staphylococcus aureus and Candida albicans and their dual biofilms. Also, LA-loaded liposome formulation efficiently disrupted preformed biofilms of S. aureus and C. albicans. Furthermore, formulated liposomal LA (0.1 µg/mL) exhibited 100-fold increased dual biofilm inhibition compared to LA alone. The single biofilms and dual biofilm formation on polystyrene were reduced as determined by 3D-bright field and scanning electron microscopy. Zeta potential measurements exhibited neutralized surface charge of S. aureus, and the liposomes inhibited hyphae formation in C. albicans. These findings demonstrated that the LA-incorporated liposomes have great potential to become a new, effective, and good antibiofilm agent for treating S. aureus and C. albicans infections.
Collapse
Affiliation(s)
- Devaraj Bharathi
- School of Chemical Engineering, Yeungnam University, 280 Daehak-Ro, Gyeongsan 38541, Republic of Korea
| | - Jin-Hyung Lee
- School of Chemical Engineering, Yeungnam University, 280 Daehak-Ro, Gyeongsan 38541, Republic of Korea
| | - Jintae Lee
- School of Chemical Engineering, Yeungnam University, 280 Daehak-Ro, Gyeongsan 38541, Republic of Korea.
| |
Collapse
|
12
|
Scheeder A, Brockhoff M, Ward EN, Kaminski Schierle GS, Mela I, Kaminski CF. Molecular Mechanisms of Cationic Fusogenic Liposome Interactions with Bacterial Envelopes. J Am Chem Soc 2023; 145:28240-28250. [PMID: 38085801 PMCID: PMC10755748 DOI: 10.1021/jacs.3c11463] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Revised: 11/22/2023] [Accepted: 11/27/2023] [Indexed: 12/28/2023]
Abstract
Although fusogenic liposomes offer a promising approach for the delivery of antibiotic payloads across the cell envelope of Gram-negative bacteria, there is still a limited understanding of the individual nanocarrier interactions with the bacterial target. Using super-resolution microscopy, we characterize the interaction dynamics of positively charged fusogenic liposomes with Gram-negative (Escherichia coli) and Gram-positive (Bacillus subtilis) bacteria. The liposomes merge with the outer membrane (OM) of Gram-negative bacteria, while attachment or lipid internalization is observed in Gram-positive cells. Employing total internal reflection fluorescence microscopy, we demonstrated liposome fusion with model supported lipid bilayers. For whole E. coli cells, however, we observed heterogeneous membrane integrations, primarily involving liposome attachment and hemifusion events. With increasing lipopolysaccharide length, the likelihood of full-fusion events was reduced. The integration of artificial lipids into the OM of Gram-negative cells led to membrane destabilization, resulting in decreased bacterial vitality, membrane detachment, and improved codelivery of vancomycin─an effective antibiotic against Gram-positive cells. These findings provide significant insights into the interactions of individual nanocarriers with bacterial envelopes at the single-cell level, uncovering effects that would be missed in bulk measurements. This highlights the importance of conducting single-particle and single-cell investigations to assess the performance of next-generation drug delivery platforms.
Collapse
Affiliation(s)
- Anna Scheeder
- Department
of Chemical Engineering and Biotechnology, University of Cambridge, Philippa Fawcett Drive, Cambridge CB3 0AS, U.K.
| | - Marius Brockhoff
- Department
of Chemical Engineering and Biotechnology, University of Cambridge, Philippa Fawcett Drive, Cambridge CB3 0AS, U.K.
| | - Edward N. Ward
- Department
of Chemical Engineering and Biotechnology, University of Cambridge, Philippa Fawcett Drive, Cambridge CB3 0AS, U.K.
| | - Gabriele S. Kaminski Schierle
- Department
of Chemical Engineering and Biotechnology, University of Cambridge, Philippa Fawcett Drive, Cambridge CB3 0AS, U.K.
| | - Ioanna Mela
- Department
of Pharmacology, University of Cambridge, Tennis Court Road, Cambridge CB2 1PD, U.K.
| | - Clemens F. Kaminski
- Department
of Chemical Engineering and Biotechnology, University of Cambridge, Philippa Fawcett Drive, Cambridge CB3 0AS, U.K.
| |
Collapse
|
13
|
Saxena D, Maitra R, Bormon R, Czekanska M, Meiers J, Titz A, Verma S, Chopra S. Tackling the outer membrane: facilitating compound entry into Gram-negative bacterial pathogens. NPJ ANTIMICROBIALS AND RESISTANCE 2023; 1:17. [PMID: 39843585 PMCID: PMC11721184 DOI: 10.1038/s44259-023-00016-1] [Citation(s) in RCA: 31] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/28/2023] [Accepted: 10/23/2023] [Indexed: 01/17/2025]
Abstract
Emerging resistance to all available antibiotics highlights the need to develop new antibiotics with novel mechanisms of action. Most of the currently used antibiotics target Gram-positive bacteria while Gram-negative bacteria easily bypass the action of most drug molecules because of their unique outer membrane. This additional layer acts as a potent barrier restricting the entry of compounds into the cell. In this scenario, several approaches have been elucidated to increase the accumulation of compounds into Gram-negative bacteria. This review includes a brief description of the physicochemical properties that can aid compounds to enter and accumulate in Gram-negative bacteria and covers different strategies to target or bypass the outer membrane-mediated barrier in Gram-negative bacterial pathogens.
Collapse
Affiliation(s)
- Deepanshi Saxena
- Department of Molecular Microbiology and Immunology, CSIR-Central Drug Research Institute, Jankipuram Extension, Sitapur Road, Lucknow, 226031, UP, India
| | - Rahul Maitra
- Department of Molecular Microbiology and Immunology, CSIR-Central Drug Research Institute, Jankipuram Extension, Sitapur Road, Lucknow, 226031, UP, India
| | - Rakhi Bormon
- Department of Chemistry, IIT Kanpur, Kanpur, 208016, UP, India
| | - Marta Czekanska
- Chemical Biology of Carbohydrates (CBCH), Helmholtz-Institute for Pharmaceutical Research Saarland (HIPS), Helmholtz Centre for Infection Research, 66123, Saarbrücken, Germany
- Department of Chemistry, Saarland University, 66123, Saarbrücken, Germany
- Deutsches Zentrum für Infektionsforschung (DZIF), 38124, Standort Hannover-Braunschweig, Germany
| | - Joscha Meiers
- Chemical Biology of Carbohydrates (CBCH), Helmholtz-Institute for Pharmaceutical Research Saarland (HIPS), Helmholtz Centre for Infection Research, 66123, Saarbrücken, Germany
- Department of Chemistry, Saarland University, 66123, Saarbrücken, Germany
- Deutsches Zentrum für Infektionsforschung (DZIF), 38124, Standort Hannover-Braunschweig, Germany
| | - Alexander Titz
- Chemical Biology of Carbohydrates (CBCH), Helmholtz-Institute for Pharmaceutical Research Saarland (HIPS), Helmholtz Centre for Infection Research, 66123, Saarbrücken, Germany.
- Department of Chemistry, Saarland University, 66123, Saarbrücken, Germany.
- Deutsches Zentrum für Infektionsforschung (DZIF), 38124, Standort Hannover-Braunschweig, Germany.
| | - Sandeep Verma
- Department of Chemistry, IIT Kanpur, Kanpur, 208016, UP, India.
- Center for Nanoscience, IIT Kanpur, Kanpur, 208016, UP, India.
| | - Sidharth Chopra
- Department of Molecular Microbiology and Immunology, CSIR-Central Drug Research Institute, Jankipuram Extension, Sitapur Road, Lucknow, 226031, UP, India.
- AcSIR: Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India.
| |
Collapse
|
14
|
Chen X, Li W, Li X, Li K, Zhang G, Hong W. Photodynamic Cationic Ultrasmall Copper Oxide Nanoparticles-Loaded Liposomes for Alleviation of MRSA Biofilms. Int J Nanomedicine 2023; 18:5441-5455. [PMID: 37753066 PMCID: PMC10519346 DOI: 10.2147/ijn.s426682] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Accepted: 09/13/2023] [Indexed: 09/28/2023] Open
Abstract
Introduction As we enter the post-antibiotic era, the rise of antibiotic-resistant pathogenic bacteria is becoming a serious threat to public health. This problem is further complicated by antibiotic-resistant biofilms, for which current treatment options are limited. Methods To tackle this challenge, we propose a novel approach that involves the use of photodynamic cationic pH-sensitive liposomes loaded with ultra-small copper oxide (Ce6@Lipo/UCONs) to effectively eliminate drug-resistant bacteria and eradicate biofilms while minimizing safety concerns and the risk of resistance development. Results Our study demonstrates that Ce6@Lipo/UCONs have minimal toxicity to mammalian cells and can significantly enhance the association affinity with methicillin-resistant Staphylococcus aureus (MRSA) as confirmed by fluorescent microscope and flow cytometry, thereby greatly improving the bactericidal effect against planktonic MRSA. The cationic nature of Ce6@Lipo/UCONs also enables them to penetrate MRSA biofilms and respond to the acidic microenvironment within the biofilm, effectively releasing the loaded UCONs. Our results indicate that Ce6@Lipo/UCONs could effectively eliminate biofilms under light irradiation conditions, as evidenced by both biomass analysis and scanning electron microscopy observations. In addition, significant antibacterial effects and abscess healing were observed in MRSA-infected mice treated with Ce6@Lipo/UCONs upon light irradiation, while good biocompatibility was achieved in vivo. Conclusion Taken together, our findings suggest that photodynamic cationic ultrasmall copper oxide nanoparticles-loaded liposomes are a highly promising nano platform for combating antibiotic-resistant microbial pathogens and biofilms. The effective biofilm penetration and synergistic effect between photodynamic inactivation and metal sterilization make them a valuable tool for overcoming the challenges posed by antibiotic resistance.
Collapse
Affiliation(s)
- Xiangjun Chen
- School of Pharmacy, Shandong New Drug Loading & Release Technology and Preparation Engineering Laboratory, Binzhou Medical University, Yantai, People’s Republic of China
| | - Wenting Li
- School of Pharmacy, Shandong New Drug Loading & Release Technology and Preparation Engineering Laboratory, Binzhou Medical University, Yantai, People’s Republic of China
| | - Xueling Li
- School of Pharmacy, Shandong New Drug Loading & Release Technology and Preparation Engineering Laboratory, Binzhou Medical University, Yantai, People’s Republic of China
| | - Keke Li
- School of Pharmacy, Shandong New Drug Loading & Release Technology and Preparation Engineering Laboratory, Binzhou Medical University, Yantai, People’s Republic of China
| | - Guilong Zhang
- School of Pharmacy, Shandong New Drug Loading & Release Technology and Preparation Engineering Laboratory, Binzhou Medical University, Yantai, People’s Republic of China
| | - Wei Hong
- School of Pharmacy, Shandong New Drug Loading & Release Technology and Preparation Engineering Laboratory, Binzhou Medical University, Yantai, People’s Republic of China
| |
Collapse
|
15
|
Zhang Y, Wong CYJ, Gholizadeh H, Aluigi A, Tiboni M, Casettari L, Young P, Traini D, Li M, Cheng S, Ong HX. Microfluidics assembly of inhalable liposomal ciprofloxacin characterised by an innovative in vitro pulmonary model. Int J Pharm 2023; 635:122667. [PMID: 36738806 DOI: 10.1016/j.ijpharm.2023.122667] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Revised: 01/19/2023] [Accepted: 01/28/2023] [Indexed: 02/05/2023]
Abstract
Respiratory tract infections (RTIs) are reported to be the leading cause of death worldwide. Delivery of liposomal antibiotic nano-systems via the inhalation route has drawn significant interest in RTIs treatment as it can directly target the site of infection and reduces the risk of systemic exposure and side effects. Moreover, this formulation system can improve pharmacokinetics and biodistribution and enhance the activity against intracellular pathogens. Microfluidics is an innovative manufacturing technology that can produce nanomedicines in a homogenous and scalable way. The objective of this study was to evaluate the antibiofilm efficacy of two liposomal ciprofloxacin formulations with different vesicle sizes manufactured by using a 3D-printed microfluidic chip. Each formulation was characterised in terms of size, polydispersity index, charge and encapsulation. Moreover, the aerosolisation characteristics of the liposomal formulations were investigated and compared with free ciprofloxacin solution using laser diffraction and cascade impaction methods. The in vitro drug release was tested using the dialysis bag method. Furthermore, the drug transport and drug release studies were conducted using the alveolar epithelial H441 cell line integrated next-generation impactor in vitro model. Finally, the biofilm eradication efficacy was evaluated using a dual-chamber microfluidic in vitro model. Results showed that both liposomal-loaded ciprofloxacin formulations and free ciprofloxacin solution had comparable aerosolisation characteristics and biofilm-killing efficacy. The liposomal ciprofloxacin formulation of smaller vesicle size showed significantly slower drug release in the dialysis bag technique compared to the free ciprofloxacin solution. Interestingly, liposomal ciprofloxacin formulations successfully controlled the release of the drug in the epithelial cell model and showed different drug transport profiles on H441 cell lines compared to the free ciprofloxacin solution, supporting the potential for inhaled liposomal ciprofloxacin to provide a promising treatment for respiratory infections.
Collapse
Affiliation(s)
- Ye Zhang
- School of Engineering, Faculty of Science and Engineering, Macquarie University, Sydney, NSW, Australia; Woolcock Institute of Medical Research, Sydney, NSW, Australia
| | | | - Hanieh Gholizadeh
- Woolcock Institute of Medical Research, Sydney, NSW, Australia; Macquarie Medical School, Faculty of Medicine, Health and Human Sciences, Macquarie University, Sydney, NSW, Australia
| | - Annalisa Aluigi
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, Piazza del Rinascimento, 6, 61029 Urbino, PU, Italy
| | - Mattia Tiboni
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, Piazza del Rinascimento, 6, 61029 Urbino, PU, Italy
| | - Luca Casettari
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, Piazza del Rinascimento, 6, 61029 Urbino, PU, Italy
| | - Paul Young
- Woolcock Institute of Medical Research, Sydney, NSW, Australia; Department of Marketing, Macquarie Business School, Macquarie University, Sydney, NSW, Australia
| | - Daniela Traini
- Woolcock Institute of Medical Research, Sydney, NSW, Australia; Macquarie Medical School, Faculty of Medicine, Health and Human Sciences, Macquarie University, Sydney, NSW, Australia
| | - Ming Li
- School of Engineering, Faculty of Science and Engineering, Macquarie University, Sydney, NSW, Australia
| | - Shaokoon Cheng
- School of Engineering, Faculty of Science and Engineering, Macquarie University, Sydney, NSW, Australia.
| | - Hui Xin Ong
- Woolcock Institute of Medical Research, Sydney, NSW, Australia; Macquarie Medical School, Faculty of Medicine, Health and Human Sciences, Macquarie University, Sydney, NSW, Australia.
| |
Collapse
|
16
|
Crintea A, Carpa R, Mitre AO, Petho RI, Chelaru VF, Nădășan SM, Neamti L, Dutu AG. Nanotechnology Involved in Treating Urinary Tract Infections: An Overview. NANOMATERIALS (BASEL, SWITZERLAND) 2023; 13:555. [PMID: 36770516 PMCID: PMC9919202 DOI: 10.3390/nano13030555] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Revised: 01/22/2023] [Accepted: 01/27/2023] [Indexed: 06/18/2023]
Abstract
Considered as the most frequent contaminations that do not require hospitalization, urinary tract infections (UTIs) are largely known to cause significant personal burdens on patients. Although UTIs overall are highly preventable health issues, the recourse to antibiotics as drug treatments for these infections is a worryingly spread approach that should be addressed and gradually overcome in a contemporary, modernized healthcare system. With a virtually alarming global rise of antibiotic resistance overall, nanotechnologies may prove to be the much-needed 'lifebuoy' that will eventually suppress this prejudicial phenomenon. This review aims to present the most promising, currently known nano-solutions, with glimpses on clinical and epidemiological aspects of the UTIs, prospective diagnostic instruments, and non-antibiotic treatments, all of these engulfed in a comprehensive overview.
Collapse
Affiliation(s)
- Andreea Crintea
- Department of Medical Biochemistry, Faculty of Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, 400349 Cluj-Napoca, Romania
| | - Rahela Carpa
- Department of Molecular Biology and Biotechnology, Faculty of Biology and Geology, Babes-Bolyai University, 400084 Cluj-Napoca, Romania
| | - Andrei-Otto Mitre
- Department of Pathophysiology, Faculty of Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, 400349 Cluj-Napoca, Romania
| | - Robert Istvan Petho
- Faculty of Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, 400349 Cluj-Napoca, Romania
| | - Vlad-Florin Chelaru
- Faculty of Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, 400349 Cluj-Napoca, Romania
| | - Sebastian-Mihail Nădășan
- Faculty of Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, 400349 Cluj-Napoca, Romania
| | - Lidia Neamti
- Department of Medical Biochemistry, Faculty of Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, 400349 Cluj-Napoca, Romania
| | - Alina Gabriela Dutu
- Department of Medical Biochemistry, Faculty of Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, 400349 Cluj-Napoca, Romania
| |
Collapse
|
17
|
Ye J, Chen X. Current Promising Strategies against Antibiotic-Resistant Bacterial Infections. Antibiotics (Basel) 2022; 12:antibiotics12010067. [PMID: 36671268 PMCID: PMC9854991 DOI: 10.3390/antibiotics12010067] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2022] [Revised: 12/27/2022] [Accepted: 12/28/2022] [Indexed: 12/31/2022] Open
Abstract
Infections caused by antibiotic-resistant bacteria (ARB) are one of the major global health challenges of our time. In addition to developing new antibiotics to combat ARB, sensitizing ARB, or pursuing alternatives to existing antibiotics are promising options to counter antibiotic resistance. This review compiles the most promising anti-ARB strategies currently under development. These strategies include the following: (i) discovery of novel antibiotics by modification of existing antibiotics, screening of small-molecule libraries, or exploration of peculiar places; (ii) improvement in the efficacy of existing antibiotics through metabolic stimulation or by loading a novel, more efficient delivery systems; (iii) development of alternatives to conventional antibiotics such as bacteriophages and their encoded endolysins, anti-biofilm drugs, probiotics, nanomaterials, vaccines, and antibody therapies. Clinical or preclinical studies show that these treatments possess great potential against ARB. Some anti-ARB products are expected to become commercially available in the near future.
Collapse
|
18
|
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: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar 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
|
19
|
Braťka P, Fenclová T, Hlinková J, Uherková L, Šebová E, Hefka Blahnová V, Hedvičáková V, Žižková R, Litvinec A, Trč T, Rosina J, Filová E. The Preparation and Biological Testing of Novel Wound Dressings with an Encapsulated Antibacterial and Antioxidant Substance. NANOMATERIALS (BASEL, SWITZERLAND) 2022; 12:3824. [PMID: 36364600 PMCID: PMC9656126 DOI: 10.3390/nano12213824] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Revised: 10/25/2022] [Accepted: 10/26/2022] [Indexed: 06/16/2023]
Abstract
Chronic wounds represent a significant socio-economic problem, and the improvement of their healing is therefore an essential issue. This paper describes the preparation and biological properties of a novel functionalized nanofiber wound dressing consisting of a polycaprolactone nanofiber carrier modified by a drug delivery system, based on the lipid particles formed by 1-tetradecanol and encapsulated gentamicin and tocopherol acetate. The cytotoxicity of extracts was tested using a metabolic activity assay, and the antibacterial properties of the extracts were tested in vitro on the bacterial strains Staphylococcus aureus and Pseudomonas aeruginosa. The effect of the wound dressing on chronic wound healing was subsequently tested using a mouse model. Fourteen days after surgery, the groups treated by the examined wound cover showed a lower granulation, reepithelization, and inflammation score compared to both the uninfected groups, a lower dermis organization compared to the control, a higher scar thickness compared to the other groups, and a higher thickness of hypodermis and bacteria score compared to both the uninfected groups. This work demonstrates the basic parameters of the safety (biocompatibility) and performance (effect on healing) of the dressing as a medical device and indicates the feasibility of the concept of its preparation in outpatient conditions using a suitable functionalization device.
Collapse
Affiliation(s)
- Petr Braťka
- Faculty of Biomedical Engineering, Czech Technical University, Náměstí Sítná 3105, 27201 Kladno, Czech Republic
- Grade Medical s.r.o., Náměstí Sítná 3105, 27201 Kladno, Czech Republic
| | - Taťána Fenclová
- Grade Medical s.r.o., Náměstí Sítná 3105, 27201 Kladno, Czech Republic
| | - Jana Hlinková
- Department of Tissue Engineering, Institute of Experimental Medicine of the Czech Academy of Sciences, Vídeňská 1083, 14220 Prague, Czech Republic
| | - Lenka Uherková
- Department of Tissue Engineering, Institute of Experimental Medicine of the Czech Academy of Sciences, Vídeňská 1083, 14220 Prague, Czech Republic
| | - Eva Šebová
- Department of Tissue Engineering, Institute of Experimental Medicine of the Czech Academy of Sciences, Vídeňská 1083, 14220 Prague, Czech Republic
| | - Veronika Hefka Blahnová
- Department of Tissue Engineering, Institute of Experimental Medicine of the Czech Academy of Sciences, Vídeňská 1083, 14220 Prague, Czech Republic
| | - Věra Hedvičáková
- Department of Tissue Engineering, Institute of Experimental Medicine of the Czech Academy of Sciences, Vídeňská 1083, 14220 Prague, Czech Republic
| | - Radmila Žižková
- Department of Tissue Engineering, Institute of Experimental Medicine of the Czech Academy of Sciences, Vídeňská 1083, 14220 Prague, Czech Republic
| | - Andrej Litvinec
- Department of Tissue Engineering, Institute of Experimental Medicine of the Czech Academy of Sciences, Vídeňská 1083, 14220 Prague, Czech Republic
| | - Tomáš Trč
- Grade Medical s.r.o., Náměstí Sítná 3105, 27201 Kladno, Czech Republic
| | - Jozef Rosina
- Faculty of Biomedical Engineering, Czech Technical University, Náměstí Sítná 3105, 27201 Kladno, Czech Republic
| | - Eva Filová
- Department of Tissue Engineering, Institute of Experimental Medicine of the Czech Academy of Sciences, Vídeňská 1083, 14220 Prague, Czech Republic
| |
Collapse
|
20
|
Maxwell A, Chaudhari BB, Chaudhari P, Ananthamurthy K, Aranjani J, Moorkoth S, Ghate V, Lewis S. In vitro antibacterial activity and in vivo pharmacokinetics of intravenously administered Amikacin-loaded Liposomes for the management of bacterial septicaemia. Colloids Surf B Biointerfaces 2022; 220:112892. [DOI: 10.1016/j.colsurfb.2022.112892] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Revised: 09/10/2022] [Accepted: 09/28/2022] [Indexed: 10/14/2022]
|
21
|
Monteiro KLC, Silva ON, Dos Santos Nascimento IJ, Mendonça Júnior FJB, Aquino PGV, da Silva-Júnior EF, de Aquino TM. Medicinal Chemistry of Inhibitors Targeting Resistant Bacteria. Curr Top Med Chem 2022; 22:1983-2028. [PMID: 35319372 DOI: 10.2174/1568026622666220321124452] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2021] [Revised: 02/01/2022] [Accepted: 02/13/2022] [Indexed: 12/15/2022]
Abstract
The discovery of antibiotics was a revolutionary feat that provided countless health benefits. The identification of penicillin by Alexander Fleming initiated the era of antibiotics, represented by constant discoveries that enabled effective treatments for the different classes of diseases caused by bacteria. However, the indiscriminate use of these drugs allowed the emergence of resistance mechanisms of these microorganisms against the available drugs. In addition, the constant discoveries in the 20th century generated a shortage of new molecules, worrying health agencies and professionals about the appearance of multidrug-resistant strains against available drugs. In this context, the advances of recent years in molecular biology and microbiology have allowed new perspectives in drug design and development, using the findings related to the mechanisms of bacterial resistance to generate new drugs that are not affected by such mechanisms and supply new molecules to be used to treat resistant bacterial infections. Besides, a promising strategy against bacterial resistance is the combination of drugs through adjuvants, providing new expectations in designing new antibiotics and new antimicrobial therapies. Thus, this manuscript will address the main mechanisms of bacterial resistance under the understanding of medicinal chemistry, showing the main active compounds against efflux mechanisms, and also the application of the use of drug delivery systems, and finally, the main potential natural products as adjuvants or with promising activity against resistant strains.
Collapse
Affiliation(s)
- Kadja Luana Chagas Monteiro
- Research Group on Therapeutic Strategies - GPET, Laboratory of Synthesis and Research in Medicinal Chemistry - LSPMED, Institute of Chemistry and Biotechnology, Federal University of Alagoas, 57072-970, Maceió, Alagoas, Brazil
| | - Osmar Nascimento Silva
- Faculty of Pharmacy, University Center of Anápolis, Unievangélica, 75083-515, Anápolis, Goiás, Brazil
| | - Igor José Dos Santos Nascimento
- Research Group on Therapeutic Strategies - GPET, Laboratory of Synthesis and Research in Medicinal Chemistry - LSPMED, Institute of Chemistry and Biotechnology, Federal University of Alagoas, 57072-970, Maceió, Alagoas, Brazil
| | | | | | - Edeildo Ferreira da Silva-Júnior
- Laboratory of Medicinal Chemistry, Institute of Pharmaceutical Sciences, Federal University of Alagoas, 57072-970, Maceió, Alagoas, Brazil
| | - Thiago Mendonça de Aquino
- Research Group on Therapeutic Strategies - GPET, Laboratory of Synthesis and Research in Medicinal Chemistry - LSPMED, Institute of Chemistry and Biotechnology, Federal University of Alagoas, 57072-970, Maceió, Alagoas, Brazil
| |
Collapse
|
22
|
Silver nanoparticle effect on Salmonella enterica isolated from Northern West Egypt food, poultry, and calves. Appl Microbiol Biotechnol 2022; 106:5701-5713. [PMID: 35945362 PMCID: PMC9418292 DOI: 10.1007/s00253-022-12102-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2022] [Revised: 06/11/2022] [Accepted: 07/23/2022] [Indexed: 11/03/2022]
Abstract
A total no. of 65 Salmonella enterica isolates recovered from food samples, feces of diarrheic calves, poultry, and hospital patient in large five cities at Northern West Egypt were obtained from the Department of Microbiology, Faculty of Veterinary Medicine, Alexandria University, Alexandria, Egypt. The 65 Salmonella enterica isolates had the invA gene were grouped into 11 Salmonella enterica serovars with dominance of S. Enteritidis and S. Kentucky serovars. Their resistance pattern were characterized by using 18 antibiotics from different classes. Approximately 80% of the isolates were multidrug resistant (MDR). Enterobacterial repetitive intergenic consequences polymerase chain reaction (ERIC-PCR) typing of 7 strains of S. Enteritidis showed 5 clusters with dissimilarity 25%. S. Enteritidis clusters in 2 main groups A and B. Group A have 2 human strain (HE2 and HE3) and one food origin (FE7) with a similarity 99%. Group B divided into B1 (FE2) and B2 (FE3) with a similarity ratio ≥ 93%, while ERIC-PCR analysis of 5 strains of S. Kentucky revealed 4 ERIC types, clustered in 2 main groups A and B with similarity 75%. We studied the effect of silver nanoparticles (Ag-NPs) on 10 antibiotic resistant strains of S. Enteritidis and S. Kentucky. The broth microdilution minimum inhibitory concentration (MIC) and minimum bactericidal concentration (MBC) were detected. Evaluation of the affection using scanning electron microscopy (SEM) and transmission electron microscopy (TEM) showed different ratios of Ag-NPs and microorganism as well as at different contact time ended finally with morphological alteration of the bacteria. We submitted new method in vivo to explore the activity of nanosilver in chicken. KEY POINTS: • Importance of ERIC-PCR to determine the relatedness between Salmonella isolates. • Effect of silver nanoparticles to confront the antibacterial resistance. • Studying the effect of silver nanoparticles in vivo on infected chicken with Salmonella.
Collapse
|
23
|
Tagrida M, Benjakul S. Liposomes loaded with betel leaf (Piper betle L.) extract: Antibacterial activity and preservative effect in combination with hurdle technologies on tilapia slices. Food Control 2022. [DOI: 10.1016/j.foodcont.2022.108999] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
24
|
Fangary S, Abdel-Halim M, Fathalla RK, Hassan R, Farag N, Engel M, Mansour S, Tammam SN. Nanoparticle Fraught Liposomes: A Platform for Increased Antibiotic Selectivity in Multidrug Resistant Bacteria. Mol Pharm 2022; 19:3163-3177. [PMID: 35876358 DOI: 10.1021/acs.molpharmaceut.2c00258] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Increasing antibiotic concentrations within bacterial cells while reducing them in mammalian ones would ultimately result in an enhancement of antibacterial actions, overcoming multidrug resistance, all while minimizing toxicity. Nanoparticles (NPs) have been used in numerous occasions to overcome antibiotic resistance, poor drug solubility, and stability. However, the concomitant increase in antibiotic concentration in mammalian cells and the resultant toxicity are usually overlooked. Without compromising bacterial cell fusion, large liposomes (Lip) have been reported to show reduced uptake in mammalian cells. Therefore, in this work, small NP fraught liposomes (NP-Lip) were formulated with the aim of increasing NP uptake and antibiotic delivery in bacterial cells but not in mammalian ones. Small polylactic-co-glycolic acid NPs were therefore loaded with erythromycin (Er), an antibiotic with low membrane permeability that is susceptible to drug efflux, and 3c, a 5-cyanothiazolyl urea derivative with low solubility and stability. In vitro experiments demonstrated that the incorporation of small NPs into large Lip resulted in a reduction in NP uptake by HEK293 cells while increasing it in Gram-negative bacteria (Escherichia coli DH5α, E. coli K12, and Pseudomonas aeruginosa), consequently resulting in an enhancement of antibiotic selectivity by fourfold toward E. coli (both strains) and eightfold toward P. aeruginosa. Ocular administration of NP-Lip in a P. aeruginosa keratitis mouse model demonstrated the ability of Er/3c-loaded NP-Lip to result in a complete recovery. More importantly, in comparison to NPs, the ocular administration of NP-Lip showed a reduction in TNF-alpha and IL-6 levels, implying reduced interaction with mammalian cells in vivo. This work therefore clearly demonstrated how tailoring the nano-bio interaction could result in selective drug delivery and a reduction in toxicity.
Collapse
Affiliation(s)
- Suzan Fangary
- Department of Pharmaceutical Technology, German University in Cairo (GUC), New Cairo 11835, Egypt
| | - Mohammad Abdel-Halim
- Department of Pharmaceutical Chemistry, German University in Cairo (GUC), New Cairo 24681, Egypt
| | - Reem K Fathalla
- Pharmaceutical and Medicinal Chemistry, Saarland University, D-66123 Saarbrücken, Germany
| | - Raghda Hassan
- Department of Pharmaceutical Technology, German University in Cairo (GUC), New Cairo 11835, Egypt
| | - Noha Farag
- Department of Microbiology and Immunology, German University in Cairo (GUC), New Cairo 11835, Egypt
| | - Matthias Engel
- Pharmaceutical and Medicinal Chemistry, Saarland University, D-66123 Saarbrücken, Germany
| | - Samar Mansour
- Department of Pharmaceutical Technology, German University in Cairo (GUC), New Cairo 11835, Egypt.,Department of Pharmaceutics and Industrial Pharmacy-Faculty of Pharmacy, Ain Shams University, Cairo 11566, Egypt
| | - Salma N Tammam
- Department of Pharmaceutical Technology, German University in Cairo (GUC), New Cairo 11835, Egypt
| |
Collapse
|
25
|
Zhang Y, Lin S, Fu J, Zhang W, Shu G, Lin J, Li H, Xu F, Tang H, Peng G, Zhao L, Chen S, Fu H. Nanocarriers for combating biofilms: advantages and challenges. J Appl Microbiol 2022; 133:1273-1287. [PMID: 35621701 DOI: 10.1111/jam.15640] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Revised: 03/08/2022] [Accepted: 05/19/2022] [Indexed: 11/27/2022]
Abstract
Bacterial biofilms are highly resistant to antibiotics and pose a great threat to human and animal health. The control and removal of bacterial biofilms have become an important topic in the field of bacterial infectious diseases. Nanocarriers show great anti-biofilm potential because of their small particle size and strong permeability. In this review, the advantages of nanocarriers for combating biofilms are analyzed. Nanocarriers can act on all stages of bacterial biofilm formation and diffusion. They can improve the scavenging effect of biofilm by targeting biofilm, destroying extracellular polymeric substances, and enhancing the biofilm permeability of antimicrobial substances. Nanocarriers can also improve the antibacterial ability of antimicrobial drugs against bacteria in biofilm by protecting the loaded drugs and controlling the release of antimicrobial substances. Additionally, we emphasize the challenges faced in using nanocarrier formulations and translating them from a preclinical level to the clinical setting.
Collapse
Affiliation(s)
- Yuning Zhang
- Innovative Engineering Research Center of Veterinary Pharmaceutics, Department of Pharmacy, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, 611130, China
| | - Shiyu Lin
- Innovative Engineering Research Center of Veterinary Pharmaceutics, Department of Pharmacy, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, 611130, China
| | - Jingyuan Fu
- Innovative Engineering Research Center of Veterinary Pharmaceutics, Department of Pharmacy, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, 611130, China
| | - Wei Zhang
- Innovative Engineering Research Center of Veterinary Pharmaceutics, Department of Pharmacy, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, 611130, China
| | - Gang Shu
- Innovative Engineering Research Center of Veterinary Pharmaceutics, Department of Pharmacy, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, 611130, China
| | - Juchun Lin
- Innovative Engineering Research Center of Veterinary Pharmaceutics, Department of Pharmacy, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, 611130, China
| | - Haohuan Li
- Innovative Engineering Research Center of Veterinary Pharmaceutics, Department of Pharmacy, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, 611130, China
| | - Funeng Xu
- Innovative Engineering Research Center of Veterinary Pharmaceutics, Department of Pharmacy, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, 611130, China
| | - Huaqiao Tang
- Innovative Engineering Research Center of Veterinary Pharmaceutics, Department of Pharmacy, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, 611130, China
| | - Guangneng Peng
- Innovative Engineering Research Center of Veterinary Pharmaceutics, Department of Pharmacy, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, 611130, China
| | - Ling Zhao
- Innovative Engineering Research Center of Veterinary Pharmaceutics, Department of Pharmacy, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, 611130, China
| | - Shiqi Chen
- Innovative Engineering Research Center of Veterinary Pharmaceutics, Department of Pharmacy, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, 611130, China
| | - Hualin Fu
- Innovative Engineering Research Center of Veterinary Pharmaceutics, Department of Pharmacy, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, 611130, China
| |
Collapse
|
26
|
Bera S, Mondal D. Antibacterial Efficacies of Nanostructured Aminoglycosides. ACS OMEGA 2022; 7:4724-4734. [PMID: 35187293 PMCID: PMC8851436 DOI: 10.1021/acsomega.1c04399] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/14/2021] [Accepted: 01/18/2022] [Indexed: 06/14/2023]
Abstract
The widespread use of broad-spectrum aminoglycoside antibiotics is restricted from various clinical applications due to the emergence of bacterial resistance and the adverse effects such as ototoxicity and nephrotoxicity. The intensive applicability of nanoparticles in modern medicinal chemistry has gained the interest of researchers for modification of aminoglycosides as nanoconjugates either via covalent conjugation or physical interactions to alleviate their undesirable effects and bacterial resistance. In this context, various carbohydrates, polymers, lipids, silver, gold, and silica-attached aminoglycoside nanoparticles have been reported with improvements in physicochemical properties, bioavailability, and biocompatibility in physiological medium. Overall, this review encompassed the synthesis of nanostructured aminoglycosides and their applications in the development of new antibacterial therapeutics.
Collapse
|
27
|
Alarfaj RE, Alkhulaifi MM, Al-Fahad AJ, Aljihani S, Yassin AEB, Alghoribi MF, Halwani MA. Antibacterial Efficacy of Liposomal Formulations Containing Tobramycin and N-Acetylcysteine against Tobramycin-Resistant Escherichia coli, Klebsiella pneumoniae, and Acinetobacter baumannii. Pharmaceutics 2022; 14:130. [PMID: 35057026 PMCID: PMC8778299 DOI: 10.3390/pharmaceutics14010130] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Revised: 12/23/2021] [Accepted: 12/28/2021] [Indexed: 12/13/2022] Open
Abstract
The antibacterial activity and biofilm reduction capability of liposome formulations encapsulating tobramycin (TL), and Tobramycin-N-acetylcysteine (TNL) were tested against tobramycin-resistant strains of E. coli, K. pneumoniae and A. baumannii in the presence of several resistant genes. All antibacterial activity were assessed against tobramycin-resistant bacterial clinical isolate strains, which were fully characterized by whole-genome sequencing (WGS). All isolates acquired one or more of AMEs genes, efflux pump genes, OMP genes, and biofilm formation genes. TL formulation inhibited the growth of EC_089 and KP_002 isolates from 64 mg/L and 1024 mg/L to 8 mg/L. TNL formulation reduced the MIC of the same isolates to 16 mg/L. TNL formulation was the only effective formulation against all A. baumannii strains compared with TL and conventional tobramycin (in the plektonic environment). Biofilm reduction was significantly observed when TL and TNL formulations were used against E. coli and K. pneumoniae strains. TNL formulation reduced biofilm formation at a low concentration of 16 mg/L compared with TL and conventional tobramycin. In conclusion, TL and TNL formulations particularly need to be tested on animal models, where they may pave the way to considering drug delivery for the treatment of serious infectious diseases.
Collapse
Affiliation(s)
- Reem E. Alarfaj
- Department of Botany and Microbiology, College of Science, King Saud University, Riyadh 11451, Saudi Arabia; (R.E.A.); (M.M.A.)
- Infectious Diseases Research Department, King Abdullah International Medical Research Center, King Saud bin Abdulaziz University for Health Sciences, National Guard Health Affairs, Riyadh 11481, Saudi Arabia
| | - Manal M. Alkhulaifi
- Department of Botany and Microbiology, College of Science, King Saud University, Riyadh 11451, Saudi Arabia; (R.E.A.); (M.M.A.)
| | - Ahmed J. Al-Fahad
- National Center for Biotechnology, Life Science & Environment Research Institute, King Abdulaziz City for Science and Technology (KACST), Riyadh 12354, Saudi Arabia;
| | - Shokran Aljihani
- Nanomedicine Department, King Abdullah International Medical Research Center, King Saud bin Abdulaziz University for Health Sciences, National Guard Health Affairs, Riyadh 11481, Saudi Arabia;
| | - Alaa Eldeen B. Yassin
- Pharmaceutical Sciences Department, College of Pharmacy, King Saud bin Abdulaziz University for Health Sciences, National Guard Health Affairs, Riyadh 11481, Saudi Arabia;
| | - Majed F. Alghoribi
- Infectious Diseases Research Department, King Abdullah International Medical Research Center, King Saud bin Abdulaziz University for Health Sciences, National Guard Health Affairs, Riyadh 11481, Saudi Arabia
| | - Majed A. Halwani
- Nanomedicine Department, King Abdullah International Medical Research Center, King Saud bin Abdulaziz University for Health Sciences, National Guard Health Affairs, Riyadh 11481, Saudi Arabia;
| |
Collapse
|
28
|
Cyclodextrin nanosponges as potential anticancer drug delivery systems to be introduced into the market, compared with liposomes. J Drug Deliv Sci Technol 2022. [DOI: 10.1016/j.jddst.2021.102931] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
29
|
Sangboonruang S, Semakul N, Obeid MA, Ruano M, Kitidee K, Anukool U, Pringproa K, Chantawannakul P, Ferro VA, Tragoolpua Y, Tragoolpua K. Potentiality of Melittin-Loaded Niosomal Vesicles Against Vancomycin-Intermediate Staphylococcus aureus and Staphylococcal Skin Infection. Int J Nanomedicine 2021; 16:7639-7661. [PMID: 34819727 PMCID: PMC8606986 DOI: 10.2147/ijn.s325901] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Accepted: 10/28/2021] [Indexed: 11/23/2022] Open
Abstract
Background Staphylococcus aureus is an important human pathogen, especially causing skin and soft tissue infections (SSTIs). Over the decades, the infections caused by antibiotic-resistant strains have often become life-threatening. Consequently, exploration and development of competent approaches to combat these serious circumstances are urgently required. Methods The antibacterial activity of melittin (Mel) on S. aureus, methicillin-resistant S. aureus (MRSA) and clinical isolates of vancomycin-intermediate S. aureus (VISA) was investigated by minimum inhibitory concentration (MIC) and time-killing assays. The localization of Mel on the bacterial cell was visualized by confocal laser scanning microscopy and its effect on the membrane was indicated based on propidium iodide uptake. The non-ionic surfactant vesicle (NISV) or niosome nanocarrier was established for Mel loading (Mel-loaded NISV) by the thin-film hydration method. Physicochemical and in vitro biological properties of Mel-loaded NISVs were characterized. The cellular uptake of Mel-loaded NISVs was evaluated by holotomography analysis. In addition, an ex vivo study was conducted on a porcine ear skin model to assess the permeation ability of Mel-loaded NISVs and their potential to inhibit bacterial skin infection. Results The effective inhibitory activity of Mel on skin pathogens was demonstrated. Among the tested strains, VISA was most susceptible to Mel. Regarding to its function, Mel targeted the bacterial cell envelope and disrupted cell membrane integrity. Mel-loaded NISVs were successfully fabricated with a nano-size of 120-200 nm and entrapment efficiency of greater than 90%. Moreover, Mel-loaded NISVs were taken up and accumulated in the intracellular space. Meanwhile, Mel was released and distributed throughout the cytosol and nucleus. Mel-loaded NISVs efficiently inhibited the growth of bacteria, particularly MRSA and VISA. Importantly, they not only penetrated epidermal and dermal skin layers, but also reduced the bacterial growth in infected skin. Conclusion Mel-loaded NISVs have a great potential to exhibit antibacterial activity. Therapeutic application of Mel-loaded NISVs could be further developed as an alternative platform for the treatment of skin infection via dermal and transdermal delivery.
Collapse
Affiliation(s)
| | - Natthawat Semakul
- Department of Chemistry, Faculty of Science, Chiang Mai University, Chiang Mai, Thailand
| | - Mohammad A Obeid
- Department of Pharmaceutics and Pharmaceutical Technology, Faculty of Pharmacy, Yarmouk University, Irbid, Jordan
| | - Marta Ruano
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, UK
| | - Kuntida Kitidee
- Center for Research and Innovation, Faculty of Medical Technology, Mahidol University, Salaya, Nakhon Pathom, Thailand
| | - Usanee Anukool
- Division of Clinical Microbiology, Department of Medical Technology, Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai, Thailand.,Infectious Diseases Research Unit (IDRU), Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai, Thailand
| | - Kidsadagon Pringproa
- Department of Veterinary Biosciences and Veterinary Public Health, Faculty of Veterinary Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Panuwan Chantawannakul
- Division of Microbiology, Department of Biology, Faculty of Sciences, Chiang Mai University, Chiang Mai, Thailand
| | - Valerie A Ferro
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, UK
| | - Yingmanee Tragoolpua
- Division of Microbiology, Department of Biology, Faculty of Sciences, Chiang Mai University, Chiang Mai, Thailand
| | - Khajornsak Tragoolpua
- Division of Clinical Microbiology, Department of Medical Technology, Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai, Thailand.,Infectious Diseases Research Unit (IDRU), Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai, Thailand
| |
Collapse
|
30
|
Sudagidan M, Yildiz G, Onen S, Al R, Temiz ŞN, Yurt MNZ, Tasbasi BB, Acar EE, Coban A, Aydin A, Dursun AD, Ozalp VC. Targeted mesoporous silica nanoparticles for improved inhibition of disinfectant resistant Listeria monocytogenes and lower environmental pollution. JOURNAL OF HAZARDOUS MATERIALS 2021; 418:126364. [PMID: 34329020 DOI: 10.1016/j.jhazmat.2021.126364] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Revised: 05/25/2021] [Accepted: 06/07/2021] [Indexed: 06/13/2023]
Abstract
Benzalkonium chloride (BAC) is a common ingredient of disinfectants used for industrial, medical, food safety and domestic applications. It is a common pollutant detected in surface and wastewaters to induce adverse effects on Human health as well as aquatic and terrestrial life forms. Since disinfectant use is essential in combatting against microorganisms, the best approach to reduce ecotoxicity level is to restrict BAC use. We report here that encapsulation of BAC in mesoporous silica nanoparticles can provide an efficient strategy for inhibition of microbial activity with lower than usual concentrations of disinfectants. As a proof-of-concept, Listeria monocytogenes was evaluated for minimum inhibitory concentration (MIC) of nanomaterial encapsulated BAC. Aptamer molecular gate structures provided a specific targeting of the disinfectant to Listeria cells, leading to high BAC concentrations around bacterial cells, but significantly reduced amounts in total. This strategy allowed to inhibition of BAC resistant Listeria strains with 8 times less the usual disinfectant dose. BAC encapsulated and aptamer functionalized silica nanoparticles (AptBACNP) effectively killed only target bacteria L. monocytogenes, but not the non-target cells, Staphylococcus aureus or Escherichia coli. AptBACNP was not cytotoxic to Human cells as determined by in vitro viability assays.
Collapse
Affiliation(s)
- Mert Sudagidan
- Kit-ARGEM R&D Center, Konya Food and Agriculture University, Konya 42080, Turkey.
| | - Gulsah Yildiz
- Kit-ARGEM R&D Center, Konya Food and Agriculture University, Konya 42080, Turkey.
| | - Selin Onen
- Department of Biology, Medical School, Atilim University, 06830 Ankara, Turkey; Department of Stem Cell Sciences, Graduate School of Health Sciences, Hacettepe University, Ankara 06100, Turkey.
| | - Rabia Al
- Department of Molecular Biology and Genetics, Muğla Sıtkı Koçman University, 48000 Muğla, Turkey.
| | | | | | - Behiye Busra Tasbasi
- Kit-ARGEM R&D Center, Konya Food and Agriculture University, Konya 42080, Turkey.
| | - Elif Esma Acar
- Kit-ARGEM R&D Center, Konya Food and Agriculture University, Konya 42080, Turkey.
| | - Aysen Coban
- Department of Gastronomy and Culinary Arts, Istanbul Gedik University, Kartal, 34876 Istanbul, Turkey.
| | - Ali Aydin
- Department of Food Hygiene and Technology, Faculty of Veterinary Medicine, Istanbul University-Cerrahpaşa, Avcilar, Istanbul, Turkey.
| | - Ali D Dursun
- Department of Physiology, Medical School, Atilim University, 06830 Ankara, Turkey.
| | - Veli C Ozalp
- Department of Biology, Medical School, Atilim University, 06830 Ankara, Turkey.
| |
Collapse
|
31
|
Hong Q, Huo S, Tang H, Qu X, Yue B. Smart Nanomaterials for Treatment of Biofilm in Orthopedic Implants. Front Bioeng Biotechnol 2021; 9:694635. [PMID: 34589470 PMCID: PMC8473796 DOI: 10.3389/fbioe.2021.694635] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2021] [Accepted: 07/23/2021] [Indexed: 01/02/2023] Open
Abstract
Biofilms refer to complex bacterial communities that are attached to the surface of animate or inanimate objects, which highly resist the antibiotics or the host immune defense mechanisms. Pathogenic biofilms in medicine are general, chronic, and even costly, especially on medical devices and orthopedic implants. Bacteria within biofilms are the cause of many persistent infections, which are almost impossible to eradicate. Though some progress has been made in comprehending the mechanisms of biofilm formation and persistence, novel alternative compounds or strategies and effective anti-biofilm antibiotics are still lacking. Smart materials of nano size which are able to respond to an external stimulus or internal environment have a great range of applications in clinic. Recently, smart nanomaterials with or without carriage of antibiotics, targeting specific bacteria and biofilm under some stimuli, have shown great potential for pathogenic biofilm and resident bacteria eradication. First, this review briefly summarizes and describes the significance of biofilms and the process of biofilm formation. Then, we focus on some of the latest research studies involving biofilm elimination, which probably could be applied in orthopedic implants. Finally, some outstanding challenges and limitations that need to be settled urgently in order to make smart nanomaterials effectively target and treat implant biofilms are also discussed. It is hoped that there will be more novel anti-biofilm strategies for biofilm infection in the prospective future.
Collapse
Affiliation(s)
| | | | | | - Xinhua Qu
- Department of Bone and Joint Surgery, Department of Orthopaedics, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Bing Yue
- Department of Bone and Joint Surgery, Department of Orthopaedics, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
32
|
Maxwell A, Ghate V, Aranjani J, Lewis S. Breaking the barriers for the delivery of amikacin: Challenges, strategies, and opportunities. Life Sci 2021; 284:119883. [PMID: 34390724 DOI: 10.1016/j.lfs.2021.119883] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Revised: 04/30/2021] [Accepted: 07/30/2021] [Indexed: 12/14/2022]
Abstract
Hypodermic delivery of amikacin is a widely adopted treatment modality for severe infections, including bacterial septicemia, meningitis, intra-abdominal infections, burns, postoperative complications, and urinary tract infections in both paediatric and adult populations. In most instances, the course of treatment requires repeated bolus doses of amikacin, prolonged hospitalization, and the presence of a skilled healthcare worker for administration and continuous therapeutic monitoring to manage the severe adverse effects. Amikacin is hydrophilic and exhibits a short half-life, which further challenges the delivery of sufficient systemic concentrations when administered by the oral or transdermal route. In this purview, the exploitation of novel controlled and sustained release drug delivery platforms is warranted. Furthermore, it has been shown that novel delivery systems are capable of increasing the antibacterial activity of amikacin at lower doses when compared to the conventional formulations and also aid in overcoming the development of drug-resistance, which currently is a significant threat to the healthcare system worldwide. The current review presents a comprehensive overview of the developmental history of amikacin, the mechanism of action in virulent strains as well as the occurrence of resistance, and various emerging drug delivery solutions developed both by the academia and the industry. The examples outlined within the review provides significant pieces of evidence on novel amikacin formulations in the field of antimicrobial research paving the path for future therapeutic interventions that will result in improved clinical outcome.
Collapse
Affiliation(s)
- Amala Maxwell
- Department of Pharmaceutics, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education (MAHE), Manipal 576104, Karnataka, India
| | - Vivek Ghate
- Mechatronics Lab, Department of Electronic System Engineering, Indian Institute of Science, Bengaluru 560012, Karnataka, India
| | - Jesil Aranjani
- Department of Pharmaceutical Biotechnology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education (MAHE), Manipal 576104, Karnataka, India
| | - Shaila Lewis
- Department of Pharmaceutics, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education (MAHE), Manipal 576104, Karnataka, India.
| |
Collapse
|
33
|
Responsive Polymeric Nanoparticles for Biofilm-infection Control. CHINESE JOURNAL OF POLYMER SCIENCE 2021. [DOI: 10.1007/s10118-021-2610-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
|
34
|
Pereira S, Santos RS, Moreira L, Guimarães N, Gomes M, Zhang H, Remaut K, Braeckmans K, De Smedt S, Azevedo NF. Lipoplexes to Deliver Oligonucleotides in Gram-Positive and Gram-Negative Bacteria: Towards Treatment of Blood Infections. Pharmaceutics 2021; 13:pharmaceutics13070989. [PMID: 34210111 PMCID: PMC8309032 DOI: 10.3390/pharmaceutics13070989] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Revised: 06/25/2021] [Accepted: 06/26/2021] [Indexed: 11/16/2022] Open
Abstract
Bacterial resistance to antibiotics threatens the ability to treat life-threatening bloodstream infections. Oligonucleotides (ONs) composed of nucleic acid mimics (NAMs) able to inhibit essential genes can become an alternative to traditional antibiotics, as long as they are safely transported in human serum upon intravenous administration and they are carried across the multilayered bacterial envelopes, impermeable to ONs. In this study, fusogenic liposomes were considered to transport the ONs and promote their internalization in clinically relevant bacteria. Locked nucleic acids and 2′-OMethyl RNA were evaluated as model NAMs and formulated into DOTAP–DOPE liposomes followed by post-PEGylation. Our data showed a complexation stability between the post-PEGylated liposomes and the ONs of over 82%, during 24 h in native human serum, as determined by fluorescence correlation spectroscopy. Quantification by a lipid-mixing assay showed that liposomes, with and without post-PEGylation, fused with all bacteria tested. Such fusion promoted the delivery of a fraction of the ONs into the bacterial cytosol, as observed by fluorescence in situ hybridization and bacterial fractionation. In short, we demonstrated for the first time that liposomes can safely transport ONs in human serum and intracellularly deliver them in both Gram-negative and -positive bacteria, which holds promise towards the treatment of bloodstream infections.
Collapse
Affiliation(s)
- Sara Pereira
- Laboratory for Process Engineering, Environment, Biotechnology and Energy (LEPABE), Faculty of Engineering, University of Porto, 4200-465 Porto, Portugal; (S.P.); (L.M.); (N.G.); (M.G.); (N.F.A.)
| | - Rita Sobral Santos
- Laboratory for Process Engineering, Environment, Biotechnology and Energy (LEPABE), Faculty of Engineering, University of Porto, 4200-465 Porto, Portugal; (S.P.); (L.M.); (N.G.); (M.G.); (N.F.A.)
- Correspondence: ; Tel.: +351-225-08-48-71
| | - Luís Moreira
- Laboratory for Process Engineering, Environment, Biotechnology and Energy (LEPABE), Faculty of Engineering, University of Porto, 4200-465 Porto, Portugal; (S.P.); (L.M.); (N.G.); (M.G.); (N.F.A.)
| | - Nuno Guimarães
- Laboratory for Process Engineering, Environment, Biotechnology and Energy (LEPABE), Faculty of Engineering, University of Porto, 4200-465 Porto, Portugal; (S.P.); (L.M.); (N.G.); (M.G.); (N.F.A.)
| | - Mariana Gomes
- Laboratory for Process Engineering, Environment, Biotechnology and Energy (LEPABE), Faculty of Engineering, University of Porto, 4200-465 Porto, Portugal; (S.P.); (L.M.); (N.G.); (M.G.); (N.F.A.)
| | - Heyang Zhang
- Ghent Research Group on Nanomedicine, Laboratory of General Biochemistry and Physical Pharmacy, Faculty of Pharmaceutical Sciences, Ghent University, 9000 Ghent, Belgium; (H.Z.); (K.R.); (K.B.); (S.D.S.)
| | - Katrien Remaut
- Ghent Research Group on Nanomedicine, Laboratory of General Biochemistry and Physical Pharmacy, Faculty of Pharmaceutical Sciences, Ghent University, 9000 Ghent, Belgium; (H.Z.); (K.R.); (K.B.); (S.D.S.)
| | - Kevin Braeckmans
- Ghent Research Group on Nanomedicine, Laboratory of General Biochemistry and Physical Pharmacy, Faculty of Pharmaceutical Sciences, Ghent University, 9000 Ghent, Belgium; (H.Z.); (K.R.); (K.B.); (S.D.S.)
- Centre for Advanced Light Microscopy, Ghent University, 9000 Ghent, Belgium
| | - Stefaan De Smedt
- Ghent Research Group on Nanomedicine, Laboratory of General Biochemistry and Physical Pharmacy, Faculty of Pharmaceutical Sciences, Ghent University, 9000 Ghent, Belgium; (H.Z.); (K.R.); (K.B.); (S.D.S.)
- Centre for Advanced Light Microscopy, Ghent University, 9000 Ghent, Belgium
| | - Nuno Filipe Azevedo
- Laboratory for Process Engineering, Environment, Biotechnology and Energy (LEPABE), Faculty of Engineering, University of Porto, 4200-465 Porto, Portugal; (S.P.); (L.M.); (N.G.); (M.G.); (N.F.A.)
| |
Collapse
|
35
|
Malathi S, Balashanmugam P, Devasena T, Kalkura SN. Enhanced antibacterial activity and wound healing by a novel collagen blended ZnO nanoparticles embedded niosome nanocomposites. J Drug Deliv Sci Technol 2021. [DOI: 10.1016/j.jddst.2021.102498] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
|
36
|
Gbian DL, Omri A. The Impact of an Efflux Pump Inhibitor on the Activity of Free and Liposomal Antibiotics against Pseudomonas aeruginosa. Pharmaceutics 2021; 13:pharmaceutics13040577. [PMID: 33919624 PMCID: PMC8072581 DOI: 10.3390/pharmaceutics13040577] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2021] [Revised: 04/14/2021] [Accepted: 04/15/2021] [Indexed: 11/21/2022] Open
Abstract
The eradication of Pseudomonas aeruginosa in cystic fibrosis patients has become continuously difficult due to its increased resistance to treatments. This study assessed the efficacy of free and liposomal gentamicin and erythromycin, combined with Phenylalanine arginine beta-naphthylamide (PABN), a broad-spectrum efflux pump inhibitor, against P. aeruginosa isolates. Liposomes were prepared and characterized for their sizes and encapsulation efficiencies. The antimicrobial activities of formulations were determined by the microbroth dilution method. Their activity on P. aeruginosa biofilms was assessed, and the effect of sub-inhibitory concentrations on bacterial virulence factors, quorum sensing (QS) signals and bacterial motility was also evaluated. The average diameters of liposomes were 562.67 ± 33.74 nm for gentamicin and 3086.35 ± 553.95 nm for erythromycin, with encapsulation efficiencies of 13.89 ± 1.54% and 51.58 ± 2.84%, respectively. Liposomes and PABN combinations potentiated antibiotics by reducing minimum inhibitory and bactericidal concentrations by 4–32 fold overall. The formulations significantly inhibited biofilm formation and differentially attenuated virulence factor production as well as motility. Unexpectedly, QS signal production was not affected by treatments. Taken together, the results indicate that PABN shows potential as an adjuvant of liposomal macrolides and aminoglycosides in the management of lung infections in cystic fibrosis patients.
Collapse
Affiliation(s)
| | - Abdelwahab Omri
- Correspondence: ; Tel.: +1-705-675-1151-2190; Fax: +1-705-675-4844
| |
Collapse
|
37
|
Ferreira M, Ogren M, Dias JNR, Silva M, Gil S, Tavares L, Aires-da-Silva F, Gaspar MM, Aguiar SI. Liposomes as Antibiotic Delivery Systems: A Promising Nanotechnological Strategy against Antimicrobial Resistance. Molecules 2021; 26:2047. [PMID: 33918529 PMCID: PMC8038399 DOI: 10.3390/molecules26072047] [Citation(s) in RCA: 95] [Impact Index Per Article: 23.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2021] [Revised: 03/30/2021] [Accepted: 03/31/2021] [Indexed: 12/17/2022] Open
Abstract
Antimicrobial drugs are key tools to prevent and treat bacterial infections. Despite the early success of antibiotics, the current treatment of bacterial infections faces serious challenges due to the emergence and spread of resistant bacteria. Moreover, the decline of research and private investment in new antibiotics further aggravates this antibiotic crisis era. Overcoming the complexity of antimicrobial resistance must go beyond the search of new classes of antibiotics and include the development of alternative solutions. The evolution of nanomedicine has allowed the design of new drug delivery systems with improved therapeutic index for the incorporated compounds. One of the most promising strategies is their association to lipid-based delivery (nano)systems. A drug's encapsulation in liposomes has been demonstrated to increase its accumulation at the infection site, minimizing drug toxicity and protecting the antibiotic from peripheral degradation. In addition, liposomes may be designed to fuse with bacterial cells, holding the potential to overcome antimicrobial resistance and biofilm formation and constituting a promising solution for the treatment of potential fatal multidrug-resistant bacterial infections, such as methicillin resistant Staphylococcus aureus. In this review, we aim to address the applicability of antibiotic encapsulated liposomes as an effective therapeutic strategy for bacterial infections.
Collapse
Affiliation(s)
- Magda Ferreira
- Centro de Investigação Interdisciplinar em Sanidade Animal, Faculdade de Medicina Veterinária, Universidade de Lisboa, Avenida da Universidade Técnica, 1300-477 Lisbon, Portugal; (M.F.); (M.O.); (J.N.R.D.); (M.S.); (S.G.); (L.T.); (F.A.-d.-S.)
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003 Lisbon, Portugal
| | - Maria Ogren
- Centro de Investigação Interdisciplinar em Sanidade Animal, Faculdade de Medicina Veterinária, Universidade de Lisboa, Avenida da Universidade Técnica, 1300-477 Lisbon, Portugal; (M.F.); (M.O.); (J.N.R.D.); (M.S.); (S.G.); (L.T.); (F.A.-d.-S.)
| | - Joana N. R. Dias
- Centro de Investigação Interdisciplinar em Sanidade Animal, Faculdade de Medicina Veterinária, Universidade de Lisboa, Avenida da Universidade Técnica, 1300-477 Lisbon, Portugal; (M.F.); (M.O.); (J.N.R.D.); (M.S.); (S.G.); (L.T.); (F.A.-d.-S.)
| | - Marta Silva
- Centro de Investigação Interdisciplinar em Sanidade Animal, Faculdade de Medicina Veterinária, Universidade de Lisboa, Avenida da Universidade Técnica, 1300-477 Lisbon, Portugal; (M.F.); (M.O.); (J.N.R.D.); (M.S.); (S.G.); (L.T.); (F.A.-d.-S.)
| | - Solange Gil
- Centro de Investigação Interdisciplinar em Sanidade Animal, Faculdade de Medicina Veterinária, Universidade de Lisboa, Avenida da Universidade Técnica, 1300-477 Lisbon, Portugal; (M.F.); (M.O.); (J.N.R.D.); (M.S.); (S.G.); (L.T.); (F.A.-d.-S.)
| | - Luís Tavares
- Centro de Investigação Interdisciplinar em Sanidade Animal, Faculdade de Medicina Veterinária, Universidade de Lisboa, Avenida da Universidade Técnica, 1300-477 Lisbon, Portugal; (M.F.); (M.O.); (J.N.R.D.); (M.S.); (S.G.); (L.T.); (F.A.-d.-S.)
| | - Frederico Aires-da-Silva
- Centro de Investigação Interdisciplinar em Sanidade Animal, Faculdade de Medicina Veterinária, Universidade de Lisboa, Avenida da Universidade Técnica, 1300-477 Lisbon, Portugal; (M.F.); (M.O.); (J.N.R.D.); (M.S.); (S.G.); (L.T.); (F.A.-d.-S.)
| | - Maria Manuela Gaspar
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003 Lisbon, Portugal
| | - Sandra Isabel Aguiar
- Centro de Investigação Interdisciplinar em Sanidade Animal, Faculdade de Medicina Veterinária, Universidade de Lisboa, Avenida da Universidade Técnica, 1300-477 Lisbon, Portugal; (M.F.); (M.O.); (J.N.R.D.); (M.S.); (S.G.); (L.T.); (F.A.-d.-S.)
| |
Collapse
|
38
|
Liu Y, Li Y, Shi L. Controlled drug delivery systems in eradicating bacterial biofilm-associated infections. J Control Release 2021; 329:1102-1116. [DOI: 10.1016/j.jconrel.2020.10.038] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2020] [Revised: 10/15/2020] [Accepted: 10/18/2020] [Indexed: 12/14/2022]
|
39
|
Aljihani SA, Alehaideb Z, Alarfaj RE, Alghoribi MF, Akiel MA, Alenazi TH, Al-Fahad AJ, Al Tamimi SM, Albakr TM, Alshehri A, Alyahya SM, Yassin AEB, Halwani MA. Enhancing azithromycin antibacterial activity by encapsulation in liposomes/liposomal-N-acetylcysteine formulations against resistant clinical strains of Escherichia coli. Saudi J Biol Sci 2020; 27:3065-3071. [PMID: 33100866 PMCID: PMC7569117 DOI: 10.1016/j.sjbs.2020.09.012] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Revised: 08/10/2020] [Accepted: 09/06/2020] [Indexed: 12/25/2022] Open
Abstract
E. coli is an Enterobacteriaceae that could develop resistance to various antibiotics and become a multi-drug resistant (MDR) bacterium. Options for treating MDR E. coli are limited and the pipeline is somewhat dry when it comes to antibiotics for MDR bacteria, so we aimed to explore more options to help in treating MDR E. coli. The purpose of this study is to examine the synergistic effect of a liposomal formulations of co-encapsulated azithromycin and N-acetylcysteine against E. coli. Liposomal azithromycin (LA) and liposomal azithromycin/N-acetylcysteine (LAN) were compared to free azithromycin. A broth dilution was used to measure the MIC and MBC of both formulations. The biofilm reduction activity, thermal stability measurements, stability studies, and cell toxicity analysis were performed. LA and LAN effectively reduced the MIC of E. coli SA10 strain, to 3 μg/ml and 2.5 μg/ml respectively. LAN at 1 × MIC recorded a 93.22% effectiveness in reducing an E. coli SA10 biofilm. The LA and LAN formulations were also structurally stable to 212 ± 2 °C and 198 ± 3 °C, respectively. In biological conditions, the formulations were largely stable in PBS conditions; however, they illustrated limited stability in sputum and plasma. We conclude that the formulation presented could be a promising therapy for E. coli resistance circumstances, providing the stability conditions have been enhanced.
Collapse
Affiliation(s)
- Shokran A. Aljihani
- Nanomedicine Department, King Abdullah International Medical Research Center, King Saud bin Abdulaziz University for Health Sciences, Riyadh, Saudi Arabia
| | - Zeyad Alehaideb
- Department of Medical Genomics, King Abdullah International Medical Research Center, King Saud bin Abdulaziz University for Health Sciences, Riyadh, Saudi Arabia
| | - Reem E. Alarfaj
- Infectious Diseases Research Department, King Abdullah International Medical Research Center, King Saud bin Abdulaziz University for Health Sciences, Riyadh, Saudi Arabia
| | - Majed F. Alghoribi
- Infectious Diseases Research Department, King Abdullah International Medical Research Center, King Saud bin Abdulaziz University for Health Sciences, Riyadh, Saudi Arabia
| | - Maaged A. Akiel
- Departmentof Clinical Laboratory Sciences, College of Applied Medical Sciences, King Saud Bin Abdulaziz University for Health Sciences (KSAU-HS), King Abdullah International Medical Research Center (KAIMRC), Riyadh, Saudi Arabia
- Department of Human and Molecular Genetics, Virginia Commonwealth University, Richmond, VA, United States
| | - Thamer H. Alenazi
- College of Medicine, King Saud Bin Abdulaziz University for Health Sciences, King Abdullah International Medical Research Center, Infectious Disease Division, King Abdulaziz Medical City, Riyadh, Saudi Arabia
| | - Ahmed J. Al-Fahad
- National Center for Biotechnology, Life Science & Environment Research Institute, King Abdulaziz City for Science and Technology (KACST), Riyadh, Saudi Arabia
| | - Saad M. Al Tamimi
- College of Pharmacy, King Saud bin Abdulaziz University for Health Sciences, King Abdullah International Medical Research Center (KAIMRC), Ministry of the National Guard - Health Affair, Riyadh, Saudi Arabia
| | - Turki M. Albakr
- College of Pharmacy, King Saud bin Abdulaziz University for Health Sciences, King Abdullah International Medical Research Center (KAIMRC), Ministry of the National Guard - Health Affair, Riyadh, Saudi Arabia
| | - Abdulrahman Alshehri
- College of Pharmacy, King Saud bin Abdulaziz University for Health Sciences, King Abdullah International Medical Research Center (KAIMRC), Ministry of the National Guard - Health Affair, Riyadh, Saudi Arabia
| | - Saad M. Alyahya
- College of Pharmacy, King Saud bin Abdulaziz University for Health Sciences, King Abdullah International Medical Research Center (KAIMRC), Ministry of the National Guard - Health Affair, Riyadh, Saudi Arabia
| | - Alaa Eldeen B. Yassin
- College of Pharmacy, King Saud bin Abdulaziz University for Health Sciences, King Abdullah International Medical Research Center (KAIMRC), Ministry of the National Guard - Health Affair, Riyadh, Saudi Arabia
- Corresponding authors.
| | - Majed A. Halwani
- Nanomedicine Department, King Abdullah International Medical Research Center, King Saud bin Abdulaziz University for Health Sciences, Riyadh, Saudi Arabia
- Corresponding authors.
| |
Collapse
|
40
|
Ghodake V, Vishwakarma J, Vavilala SL, Patravale V. Cefoperazone sodium liposomal formulation to mitigate P. aeruginosa biofilm in Cystic fibrosis infection: A QbD approach. Int J Pharm 2020; 587:119696. [DOI: 10.1016/j.ijpharm.2020.119696] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2020] [Revised: 07/21/2020] [Accepted: 07/23/2020] [Indexed: 12/19/2022]
|
41
|
|
42
|
Guo R, Li K, Qin J, Niu S, Hong W. Development of polycationic micelles as an efficient delivery system of antibiotics for overcoming the biological barriers to reverse multidrug resistance in Escherichia coli. NANOSCALE 2020; 12:11251-11266. [PMID: 32412567 DOI: 10.1039/d0nr01366h] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/20/2023]
Abstract
Highly pathogenic Gram-negative bacteria (G-) are tenacious and pose a serious threat to public health, mainly because of three biological barriers: cell envelope blockages, biofilm protection, and macrophages shelter. One strategy to bypass the biological barriers and consequently achieve a satisfying G- bactericidal effect is to utilize polymeric micelles with superior bacterial recognition and binding capabilities. In the current study, we explored the biological barriers penetration ability of a traditional polycationic micellar system (PP-PEI) based on a copolymer of polylactide-poly (ethylene glycol)-polyethylenimine (PLA5K-PEG2K-PEI2K). Subsequently, tetracycline (TC) with good fluorescence property was encapsulated into the PLA core of the micelle (PP-PEI/TC) through hydrophobic interaction. The combination of a PEI shell and loaded antibiotic drug endowed the polycationic micelles with a greater capacity for killing drug-resistant bacteria, destructing biofilms, and eradicating intracellular bacteria, compared with free TC and micelles without the inoculation of a PEI moiety. Confocal laser scanning microscopy (CLSM) and flow cytometry illustrated that PP-PEI/TC could completely penetrate and accumulate in drug-resistant E. coli, biofilms, and infected macrophages. The efficient biological barrier penetration was elucidated as due to the strong electrostatic interactions between the polycationic PEI block and the anionic composition of the bacterial outer membrane (e.g., LPS), macrophage cell membrane (e.g., phospholipid), and extracellular polymeric substances (e.g., eDNA), which was confirmed by biolayer interferometry (BLI). Once the micellar system was bound to a negatively-charged surface, bacterial and cellular enzymes could degrade the PP-PEI core to release its antibacterial content and finally kill planktonic bacteria, bacteria over the depth of a biofilm, and/or intracellular bacteria. In vivo imaging indicated that fluorescent polycationic micelles accumulated in bacterial infection sites with strong fluorescence. In vivo antibacterial experiments showed that PP-PEI/TC could dramatically reduce the number of drug-resistant E. coli EB1-1 in the peritoneal cavity of acute peritonitis BALB/c mice compared with its counterparts. In conclusion, our study demonstrated that polycationic micelles with a PEI shell could penetrate into drug-resistant bacteria, the biofilm matrix, and infected macrophages and lead to the spatiotemporal release of antibacterial agents for the comprehensive treatment of drug-resistant relevant infections.
Collapse
Affiliation(s)
- Rong Guo
- School of Pharmacy, the Key Laboratory of Prescription Effect and Clinical Evaluation of State Administration of Traditional Chinese Medicine of China, Binzhou Medical University, Guanhai Road 346, Yantai, 264003, P. R. China.
| | - Keke Li
- School of Pharmacy, the Key Laboratory of Prescription Effect and Clinical Evaluation of State Administration of Traditional Chinese Medicine of China, Binzhou Medical University, Guanhai Road 346, Yantai, 264003, P. R. China.
| | - Jing Qin
- Department of Pharmaceutics, School of Pharmacy, Institute of Integrative Medicine, Fudan University, Key Laboratory of Smart Drug Delivery, Ministry of Education, Zhangheng Road 826, Shanghai, 200433, P. R. China
| | - Shengli Niu
- Key laboratory of Zoonosis of Liaoning Province, College of Animal Science and Veterinary Medicine, Shenyang Agricultural University, Dongling Road 120, Shenyang, 110866, P.R. China
| | - Wei Hong
- School of Pharmacy, the Key Laboratory of Prescription Effect and Clinical Evaluation of State Administration of Traditional Chinese Medicine of China, Binzhou Medical University, Guanhai Road 346, Yantai, 264003, P. R. China.
| |
Collapse
|
43
|
Abstract
When antibiotics are administered, orally or intravenously, they pass through different organs and layers of tissue on their way to the site of infection; this can cause dilution and/or intoxication. To overcome these problems, drug delivery vehicles have been used to encapsulate and deliver antibiotics, improving their therapeutic index while minimizing their adverse effects. Liposomes are self-assembled lipid vesicles made from at least one bilayer of phospholipids with an inner aqueous compartment. Liposomes are attractive vehicles to deliver antibiotics because they can encapsulate both hydrophobic and hydrophilic antibiotics, they have low toxicity, and they can change the biodistribution of the drug. Furthermore, liposomes have been approved by regulatory agencies. However, most developmental and mechanistic research in the field has been focused on encapsulation and delivery of anticancer drugs, a class of molecules that differ significantly in chemistry from antibiotics. In this critical Review, we discuss the state of knowledge regarding the design of liposomes for encapsulation and delivery of antibiotics and offer insight into the challenges and promises of using liposomes for antibiotic delivery.
Collapse
Affiliation(s)
- Azucena Gonzalez Gomez
- Department of Chemical Engineering, McMaster University, Hamilton, Ontario L9S 8L7, Canada
| | - Zeinab Hosseinidoust
- Department of Chemical Engineering, McMaster University, Hamilton, Ontario L9S 8L7, Canada
- Michael DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, Ontario L98 4LS, Canada
| |
Collapse
|
44
|
Kelly SA, Rodgers AM, O'Brien SC, Donnelly RF, Gilmore BF. Gut Check Time: Antibiotic Delivery Strategies to Reduce Antimicrobial Resistance. Trends Biotechnol 2020; 38:447-462. [PMID: 31757410 DOI: 10.1016/j.tibtech.2019.10.008] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2019] [Revised: 10/18/2019] [Accepted: 10/21/2019] [Indexed: 02/08/2023]
Abstract
Antimicrobial resistance (AMR) has developed into a huge threat to global health, and reducing it is an urgent priority for public health authorities. The importance of a healthy and balanced gut microbiome has been identified as a key protective factor against AMR development, but this can be significantly affected by antibiotic therapy, resulting in dysbiosis and reduction of taxonomic richness. The way in which antibiotics are administered could form an important part of future antimicrobial stewardship strategies, where drug delivery is ideally placed to play a key role in the fight against AMR. This review focuses on drug delivery strategies for antibiotic administration, including avoidance of the gut microbiome and targeted delivery approaches, which may reduce AMR.
Collapse
Affiliation(s)
- Stephen A Kelly
- School of Pharmacy, Queen's University Belfast, Belfast, Northern Ireland
| | - Aoife M Rodgers
- Centre for Experimental Medicine, Queen's University Belfast, Belfast, Northern Ireland; Department of Biology, Maynooth University, Maynooth, Kildare, Ireland
| | - Séamus C O'Brien
- School of Pharmacy, Queen's University Belfast, Belfast, Northern Ireland
| | - Ryan F Donnelly
- School of Pharmacy, Queen's University Belfast, Belfast, Northern Ireland
| | - Brendan F Gilmore
- School of Pharmacy, Queen's University Belfast, Belfast, Northern Ireland.
| |
Collapse
|
45
|
Hashemzadeh H, Javadi H, Darvishi MH. Study of Structural stability and formation mechanisms in DSPC and DPSM liposomes: A coarse-grained molecular dynamics simulation. Sci Rep 2020; 10:1837. [PMID: 32020000 PMCID: PMC7000798 DOI: 10.1038/s41598-020-58730-z] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2019] [Accepted: 01/19/2020] [Indexed: 01/09/2023] Open
Abstract
Liposomes or biological vesicles can be created from cholesterol, phospholipid, and water. Their stability is affected by their phospholipid composition which can influence disease treatment and drug delivery efficacy. In this study, the effect of phospholipid type on the formation and stability of liposomes using coarse-grained molecular dynamics simulations is investigated. For this purpose, the simulation study of the DSPC (1,2-distearoyl-sn-glycero-3-phosphocholine) and DPSM (Egg sphingomyelin) lipids were considered. All simulations were carried out using the Gromacs software and Martini force field 2.2. Energy minimization (3000 steps) model, equilibrium at constant volume to adjust the temperature at 400 Kelvin and equilibrium at constant pressure to adjust the pressure, at atmospheric pressure (1 bar) have been validated. Microsecond simulations, as well as formation analysis including density, radial distribution function, and solvent accessible surface area, demonstrated spherical nanodisc structures for the DPSM and DSPC liposomes. The results revealed that due to the cylindrical geometric structure and small-size head group, the DSPC lipid maintained its perfectly spherical structure. However, the DPSM lipid showed a conical geometric structure with larger head group than other lipids, which allows the liposome to form a micelle structure. Although the DSPC and DPSM lipids used in the laboratory tests exhibit liposome and micelle behaviors, the simulation results revealed their nanodisc structures. Energy analysis including overall energy, Van der Waals interaction energy, and electrostatic interaction energy showed that DPSM liposome is more stable than DSPC liposome.
Collapse
Affiliation(s)
- H Hashemzadeh
- Nanobiotechnology Department, Faculty of Bioscience, Tarbiat Modares University, Tehran, Iran
| | - H Javadi
- Nanobiotechnology Research Center, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - M H Darvishi
- Nanobiotechnology Research Center, Baqiyatallah University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
46
|
Alternative strategies for the application of aminoglycoside antibiotics against the biofilm-forming human pathogenic bacteria. Appl Microbiol Biotechnol 2020; 104:1955-1976. [DOI: 10.1007/s00253-020-10360-1] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2019] [Revised: 12/29/2019] [Accepted: 01/05/2020] [Indexed: 12/17/2022]
|
47
|
Wang DY, van der Mei HC, Ren Y, Busscher HJ, Shi L. Lipid-Based Antimicrobial Delivery-Systems for the Treatment of Bacterial Infections. Front Chem 2020; 7:872. [PMID: 31998680 PMCID: PMC6965326 DOI: 10.3389/fchem.2019.00872] [Citation(s) in RCA: 107] [Impact Index Per Article: 21.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2019] [Accepted: 12/03/2019] [Indexed: 02/06/2023] Open
Abstract
Many nanotechnology-based antimicrobials and antimicrobial-delivery-systems have been developed over the past decades with the aim to provide alternatives to antibiotic treatment of infectious-biofilms across the human body. Antimicrobials can be loaded into nanocarriers to protect them against de-activation, and to reduce their toxicity and potential, harmful side-effects. Moreover, antimicrobial nanocarriers such as micelles, can be equipped with stealth and pH-responsive features that allow self-targeting and accumulation in infectious-biofilms at high concentrations. Micellar and liposomal nanocarriers differ in hydrophilicity of their outer-surface and inner-core. Micelles are self-assembled, spherical core-shell structures composed of single layers of surfactants, with hydrophilic head-groups and hydrophobic tail-groups pointing to the micellar core. Liposomes are composed of lipids, self-assembled into bilayers. The hydrophilic head of the lipids determines the surface properties of liposomes, while the hydrophobic tail, internal to the bilayer, determines the fluidity of liposomal-membranes. Therefore, whereas micelles can only be loaded with hydrophobic antimicrobials, hydrophilic antimicrobials can be encapsulated in the hydrophilic, aqueous core of liposomes and hydrophobic or amphiphilic antimicrobials can be inserted in the phospholipid bilayer. Nanotechnology-derived liposomes can be prepared with diameters <100-200 nm, required to prevent reticulo-endothelial rejection and allow penetration into infectious-biofilms. However, surface-functionalization of liposomes is considerably more difficult than of micelles, which explains while self-targeting, pH-responsive liposomes that find their way through the blood circulation toward infectious-biofilms are still challenging to prepare. Equally, development of liposomes that penetrate over the entire thickness of biofilms to provide deep killing of biofilm inhabitants still provides a challenge. The liposomal phospholipid bilayer easily fuses with bacterial cell membranes to release high antimicrobial-doses directly inside bacteria. Arguably, protection against de-activation of antibiotics in liposomal nanocarriers and their fusogenicity constitute the biggest advantage of liposomal antimicrobial carriers over antimicrobials free in solution. Many Gram-negative and Gram-positive bacterial strains, resistant to specific antibiotics, have been demonstrated to be susceptible to these antibiotics when encapsulated in liposomal nanocarriers. Recently, also progress has been made concerning large-scale production and long-term storage of liposomes. Therewith, the remaining challenges to develop self-targeting liposomes that penetrate, accumulate and kill deeply in infectious-biofilms remain worthwhile to pursue.
Collapse
Affiliation(s)
- Da-Yuan Wang
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Functional Polymer Materials, Ministry of Education, Institute of Polymer Chemistry, College of Chemistry, Nankai University, Tianjin, China
- Department of Biomedical Engineering, University of Groningen and University Medical Center Groningen, Groningen, Netherlands
| | - Henny C. van der Mei
- Department of Biomedical Engineering, University of Groningen and University Medical Center Groningen, Groningen, Netherlands
| | - Yijin Ren
- Department of Orthodontics, University of Groningen and University Medical Center Groningen, Groningen, Netherlands
| | - Henk J. Busscher
- Department of Biomedical Engineering, University of Groningen and University Medical Center Groningen, Groningen, Netherlands
| | - Linqi Shi
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Functional Polymer Materials, Ministry of Education, Institute of Polymer Chemistry, College of Chemistry, Nankai University, Tianjin, China
| |
Collapse
|
48
|
Khan S, Akhtar MU, Khan S, Javed F, Khan AA. Nanoniosome-encapsulated levoflaxicin as an antibacterial agent against Brucella. J Basic Microbiol 2019; 60:281-290. [PMID: 31856360 DOI: 10.1002/jobm.201900454] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2019] [Revised: 10/17/2019] [Accepted: 10/30/2019] [Indexed: 01/09/2023]
Abstract
A study was conducted to examine the prevalence of brucellosis (in animal farms) in the vicinity of Islamabad and Rawalpindi. A total of 170 milk samples were collected randomly from several farmhouses. The collected milk samples were initially screened by a Brucella selective medium. The bacterial isolates grown on the selective medium were subjected to biochemical identification for further confirmation of Brucella species. Among the tested samples, 28 (16.4%) were found positive for selective medium and 14 (8.2%) were found positive after biochemical confirmation. The antimicrobial susceptibility of several antibiotics performed by the disc-diffusion method did not yield any significant findings. Encapsulating antimicrobial drugs in unilamellar niosomes is an effective approach to treat the endemic infection. In this study, the antimicrobial activity of niosome-encapsulated levofloxacin is compared with free drug. The drug-encapsulating and empty niosomes were synthesized by using two surfactants Tween 80 and Span 40. Niosomal characterization included electron microscopy, dynamic light scattering, and zeta potential. The encapsulation efficiency was found to be 78% and 74% for Span 40 and Tween 80 niosomes, respectively. The antibacterial activity of niosomal levofloxacin was evaluated against the identified Brucella species and the antimicrobial activity of the free drug was increased many folds after encapsulation. In this study, levofloxacin niosomes were successfully synthesized against Brucellosis.
Collapse
Affiliation(s)
- Sana Khan
- Department of Biosciences, COMASTS University Islamabad, Islamabad, Pakistan
| | | | - Suhaib Khan
- Department of Biosciences, COMASTS University Islamabad, Islamabad, Pakistan
| | - Farakh Javed
- Department of Microbiology, University of Haripur, Haripur, KP, Pakistan
| | - Abid Ali Khan
- Department of Biosciences, COMASTS University Islamabad, Islamabad, Pakistan.,Institute of Microbiology and Epizootics, Free University of Berlin, Berlin, Germany
| |
Collapse
|
49
|
Velino C, Carella F, Adamiano A, Sanguinetti M, Vitali A, Catalucci D, Bugli F, Iafisco M. Nanomedicine Approaches for the Pulmonary Treatment of Cystic Fibrosis. Front Bioeng Biotechnol 2019; 7:406. [PMID: 31921811 PMCID: PMC6927921 DOI: 10.3389/fbioe.2019.00406] [Citation(s) in RCA: 57] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2019] [Accepted: 11/27/2019] [Indexed: 12/24/2022] Open
Abstract
Cystic fibrosis (CF) is a genetic disease affecting today nearly 70,000 patients worldwide and characterized by a hypersecretion of thick mucus difficult to clear arising from the defective CFTR protein. The over-production of the mucus secreted in the lungs, along with its altered composition and consistency, results in airway obstruction that makes the lungs susceptible to recurrent and persistent bacterial infections and endobronchial chronic inflammation, which are considered the primary cause of bronchiectasis, respiratory failure, and consequent death of patients. Despite the difficulty of treating the continuous infections caused by pathogens in CF patients, various strategies focused on the symptomatic therapy have been developed during the last few decades, showing significant positive impact on prognosis. Moreover, nowadays, the discovery of CFTR modulators as well as the development of gene therapy have provided new opportunity to treat CF. However, the lack of effective methods for delivery and especially targeted delivery of therapeutics specifically to lung tissues and cells limits the efficiency of the treatments. Nanomedicine represents an extraordinary opportunity for the improvement of current therapies and for the development of innovative treatment options for CF previously considered hard or impossible to treat. Due to the peculiar environment in which the therapies have to operate characterized by several biological barriers (pulmonary tract, mucus, epithelia, bacterial biofilm) the use of nanotechnologies to improve and enhance drug delivery or gene therapies is an extremely promising way to be pursued. The aim of this review is to revise the currently used treatments and to outline the most recent progresses about the use of nanotechnology for the management of CF.
Collapse
Affiliation(s)
- Cecilia Velino
- Institute of Science and Technology for Ceramics (ISTEC), National Research Council (CNR), Faenza, Italy
| | - Francesca Carella
- Institute of Science and Technology for Ceramics (ISTEC), National Research Council (CNR), Faenza, Italy
| | - Alessio Adamiano
- Institute of Science and Technology for Ceramics (ISTEC), National Research Council (CNR), Faenza, Italy
| | - Maurizio Sanguinetti
- Fondazione Policlinico Universitario “A. Gemelli” IRCCS, Dipartimento di Scienze di Laboratorio e Infettivologiche, Rome, Italy
- Istituto di Microbiologia, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Alberto Vitali
- Institute for the Chemistry of Molecular Recognition (ICRM), National Research Council (CNR), c/o Institute of Biochemistry and Clinical Biochemistry, Catholic University, Rome, Italy
| | - Daniele Catalucci
- Humanitas Clinical and Research Center, Rozzano, Italy
- Institute of Genetic and Biomedical Research (IRGB) - UOS Milan, National Research Council (CNR), Milan, Italy
| | - Francesca Bugli
- Fondazione Policlinico Universitario “A. Gemelli” IRCCS, Dipartimento di Scienze di Laboratorio e Infettivologiche, Rome, Italy
- Istituto di Microbiologia, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Michele Iafisco
- Institute of Science and Technology for Ceramics (ISTEC), National Research Council (CNR), Faenza, Italy
| |
Collapse
|
50
|
Olatunde OO, Benjakul S, Vongkamjan K, Amnuaikit T. Liposomal Encapsulated Ethanolic Coconut Husk Extract: Antioxidant and Antibacterial Properties. J Food Sci 2019; 84:3664-3673. [PMID: 31724746 DOI: 10.1111/1750-3841.14853] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2019] [Revised: 09/04/2019] [Accepted: 09/30/2019] [Indexed: 11/26/2022]
Abstract
Characteristics of liposomal encapsulated ethanolic coconut husk extract (LE-ECHE) prepared using two levels of lipid phase (LP) containing soybean phosphatidylcholine/cholesterol mixture of 4:1 mol ratio (60 and 80 µmol/mL) and two ECHE concentrations (1% and 2%) were investigated. Poly-dispersity index, zeta-potential, and particle size of LE-ECHE samples were 0.22% to 0.28%, -70.4 to -53.63 mV, and 232 to 697.65 nm, respectively. Encapsulation efficiency of all samples was 75.25% to 90.11%. LE-ECHE prepared with LP content of 60 µmol/mL and 1% ECHE (LP60-EC1) was milky, whereas UN-EC1 (un-encapsulated ECHE) was brownish in color. ECHE retained its antioxidant activity even after entrapment in liposome, although higher activity was recorded for UN-EC1. Encapsulation of ECHE in liposome enhanced antibacterial properties of ECHE. Hence, LP60-EC1 showed promising potential as a delivery based system for lowering dark color, a drawback associated with ECHE as well as improving the antibacterial properties of ECHE. PRACTICAL APPLICATION: Ethanolic coconut husk extract (ECHE) contains polyphenols with diverse biological activities such as antimicrobial and antioxidant properties. However, there are limited applications of ECHE in food industries, mainly because of its distinctive dark brown color. A homogeneous and stable liposomal system was demonstrated to be an efficient delivery based system for ECHE. Remarkably, antimicrobial property of ECHE was enhanced with liposomal encapsulation, whereas antioxidant activities of ECHE were retained. Also, liposomal encapsulation was shown as the potential technique to mask the undesirable dark brown, a drawback associated with ECHE for wider application.
Collapse
Affiliation(s)
- Oladipupo Odunayo Olatunde
- Dept. of Food Technology, Faculty of Agro-Industry, Prince of Songkla Univ., Hat Yai, Songkhla, 90112, Thailand
| | - Soottawat Benjakul
- Dept. of Food Technology, Faculty of Agro-Industry, Prince of Songkla Univ., Hat Yai, Songkhla, 90112, Thailand
| | - Kitiya Vongkamjan
- Dept. of Food Technology, Faculty of Agro-Industry, Prince of Songkla Univ., Hat Yai, Songkhla, 90112, Thailand
| | - Thanaporn Amnuaikit
- Dept. of Pharmaceutical Technology, Faculty of Pharmaceutical Sciences, Prince of Songkla Univ., Hat Yai, Songkhla, 90112, Thailand
| |
Collapse
|