1
|
Jiao M, Kong W, Liu W, Dong Z, Yang J, Wei Z, Lu X, Wei Y, Zhuang J. Boosting the antibacterial potency of natural products through nanotechnologies. Int J Pharm 2025; 674:125437. [PMID: 40057213 DOI: 10.1016/j.ijpharm.2025.125437] [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: 01/11/2025] [Revised: 03/01/2025] [Accepted: 03/06/2025] [Indexed: 03/17/2025]
Abstract
The advent of bacterial resistance has led to a notable challenge in effectively treating bacterial infections. This highlights the urgent need for the development of novel and effective drugs to combat bacterial infections. Medicinal plants, with their rich and diverse natural compounds, represent a valuable source for the discovery of novel antibacterial agents. Many of these natural compounds exhibit strong antibacterial functions, offering a promising direction for the development of antibacterial drugs. Furthermore, the application of nanotechnology in the development of antibacterial natural products has become a topic of considerable interest due to the advantages it offers, including the potential to enhance drug solubility. The efficacy of natural antibacterial agents is significantly enhanced through nanotechnology. This review offers a comprehensive overview of recent advances in the delivery of natural antibacterial compounds using a range of nanoformulation strategies.
Collapse
Affiliation(s)
- Min Jiao
- Graduate School, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; School of Pharmacy, Shanghai University of Medicine and Health Sciences, Shanghai 201318, China
| | - Weiwen Kong
- School of Pharmacy, Fudan University, Key Laboratory of Smart Drug Delivery, Ministry of Education, Shanghai 201203, China
| | - Wenjuan Liu
- School of Pharmacy, Fudan University, Key Laboratory of Smart Drug Delivery, Ministry of Education, Shanghai 201203, China
| | - Zirong Dong
- School of Pharmacy, Fudan University, Key Laboratory of Smart Drug Delivery, Ministry of Education, Shanghai 201203, China
| | - Jinlong Yang
- School of Pharmacy, Fudan University, Key Laboratory of Smart Drug Delivery, Ministry of Education, Shanghai 201203, China; Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai 200443, China
| | - Zibo Wei
- Graduate School, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; School of Pharmacy, Shanghai University of Medicine and Health Sciences, Shanghai 201318, China
| | - Xinrui Lu
- School of Pharmacy, Fudan University, Key Laboratory of Smart Drug Delivery, Ministry of Education, Shanghai 201203, China
| | - Yuning Wei
- School of Pharmacy, Fudan University, Key Laboratory of Smart Drug Delivery, Ministry of Education, Shanghai 201203, China
| | - Jie Zhuang
- Graduate School, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; School of Pharmacy, Shanghai University of Medicine and Health Sciences, Shanghai 201318, China.
| |
Collapse
|
2
|
Yuan P, Zhang M, Wang S, Li L, Zuo R, Qu S. Bacteria-Responsive Drug Delivery System Utilizing Carboxymethyl Cellulose-Functionalized Metal-Organic Framework for Enhanced Antibacterial Efficacy. ACS Biomater Sci Eng 2025; 11:2216-2225. [PMID: 40131182 DOI: 10.1021/acsbiomaterials.5c00084] [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: 03/26/2025]
Abstract
Bacterial infections pose a significant threat to human health and economic stability. The overuse of antibiotics has exacerbated bacterial resistance, highlighting the urgent need for innovative strategies to combat this issue. Bacteria-responsive drug delivery systems present a promising solution to overcoming bacterial resistance. Metal-organic frameworks (MOFs), versatile porous materials created by linking metal clusters with organic ligands, are ideal candidates for such applications. Here, we employed the zeolite imidazole framework 8 (ZIF-8) as a carrier for ceftiofur (EFT), enhanced with carboxymethyl cellulose to develop a smart drug delivery system (CMC-EFT@ZIF-8) responsive to pH and cellulase. In vitro tests demonstrated that this system released a higher quantity of EFT under acidic conditions and in the presence of cellulase, leading to more effective disruption of bacterial membranes and subsequent bacterial death. The CMC-EFT@ZIF-8 system achieved a 99% clearance of Pseudomonas aeruginosa within 6 h and showed superior efficacy in a mouse skin wound model. These findings underscore the potential of our smart drug delivery system to significantly improve treatment outcomes for bacterial infections, representing a significant advancement in the fight against antibiotic resistance.
Collapse
Affiliation(s)
- Pingping Yuan
- Animal-Derived Food Safety Innovation Team, College of Veterinary Medicine, Anhui Agricultural University, Hefei 230036, China
- Joint Research Center for Food Nutrition and Health of IHM, Anhui Agricultural University, Hefei 230036, China
| | - Mengying Zhang
- Animal-Derived Food Safety Innovation Team, College of Veterinary Medicine, Anhui Agricultural University, Hefei 230036, China
| | - Sheng Wang
- Animal-Derived Food Safety Innovation Team, College of Veterinary Medicine, Anhui Agricultural University, Hefei 230036, China
| | - Lin Li
- Animal-Derived Food Safety Innovation Team, College of Veterinary Medicine, Anhui Agricultural University, Hefei 230036, China
| | - Runan Zuo
- Animal-Derived Food Safety Innovation Team, College of Veterinary Medicine, Anhui Agricultural University, Hefei 230036, China
| | - Shaoqi Qu
- Animal-Derived Food Safety Innovation Team, College of Veterinary Medicine, Anhui Agricultural University, Hefei 230036, China
- Joint Research Center for Food Nutrition and Health of IHM, Anhui Agricultural University, Hefei 230036, China
| |
Collapse
|
3
|
Niculescu AG, Mitache MM, Grumezescu AM, Chifiriuc MC, Mihai MM, Tantu MM, Tantu AC, Popa LG, Grigore GA, Cristian RE, Popa MI, Vrancianu CO. From Microbial Ecology to Clinical Challenges: The Respiratory Microbiome's Role in Antibiotic Resistance. Pathogens 2025; 14:355. [PMID: 40333133 PMCID: PMC12030467 DOI: 10.3390/pathogens14040355] [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: 03/01/2025] [Revised: 04/01/2025] [Accepted: 04/03/2025] [Indexed: 05/09/2025] Open
Abstract
Antibiotic resistance represents a growing public health threat, with airborne drug-resistant strains being especially alarming due to their ease of transmission and association with severe respiratory infections. The respiratory microbiome plays a pivotal role in maintaining respiratory health, influencing the dynamics of antibiotic resistance among airborne pathogenic microorganisms. In this context, this review proposes the exploration of the complex interplay between the respiratory microbiota and antimicrobial resistance, highlighting the implications of microbiome diversity in health and disease. Moreover, strategies to mitigate antibiotic resistance, including stewardship programs, alternatives to traditional antibiotics, probiotics, microbiota restoration techniques, and nanotechnology-based therapeutic interventions, are critically presented, setting an updated framework of current management options. Therefore, through a better understanding of respiratory microbiome roles in antibiotic resistance, alongside emerging therapeutic strategies, this paper aims to shed light on how the global health challenges posed by multi-drug-resistant pathogens can be addressed.
Collapse
Affiliation(s)
- Adelina-Gabriela Niculescu
- Department of Science and Engineering of Oxide Materials and Nanomaterials, National University of Science and Technology POLITEHNICA Bucharest, 011061 Bucharest, Romania; (A.-G.N.); (A.M.G.)
- Research Institute of the University of Bucharest—ICUB, University of Bucharest, 050663 Bucharest, Romania; (G.A.G.)
| | - Mihaela Magdalena Mitache
- Department of Preclinical Disciplines, Faculty of Medicine, Titu Maiorescu University, 031593 Bucharest, Romania;
| | - Alexandru Mihai Grumezescu
- Department of Science and Engineering of Oxide Materials and Nanomaterials, National University of Science and Technology POLITEHNICA Bucharest, 011061 Bucharest, Romania; (A.-G.N.); (A.M.G.)
- Research Institute of the University of Bucharest—ICUB, University of Bucharest, 050663 Bucharest, Romania; (G.A.G.)
| | - Mariana Carmen Chifiriuc
- Research Institute of the University of Bucharest—ICUB, University of Bucharest, 050663 Bucharest, Romania; (G.A.G.)
- Microbiology-Immunology Department, Faculty of Biology, University of Bucharest, 050095 Bucharest, Romania
- Biological Sciences Division, Romanian Academy, Calea Victoriei 125, Sector 1, 010071 Bucharest, Romania
| | - Mara Madalina Mihai
- Faculty of Medicine, “Carol Davila” University of Medicine and Pharmacy, 020021 Bucharest, Romania;
- Department of Oncologic Dermatology, “Elias” University Emergency Hospital, 010024 Bucharest, Romania
| | - Monica Marilena Tantu
- Department of Medical Assistance and Physical Therapy, Pitesti University Center, Târgu din Vale 1, 110040 Pitești, Romania;
- Faculty of Science, Physical Education and Informatics, National University of Science and Technology, Politehnica, Splaiul Independenței 313, District 6, 060042 Bucharest, Romania
| | - Ana Catalina Tantu
- Doctoral School, University of Medicine and Pharmacy of Craiova, Petru Rareș 2, 200349 Craiova, Romania;
- Emergency Clinical County Hospital of Craiova, Tabaci 1, 200642 Craiova, Romania
| | - Loredana Gabriela Popa
- Microbiology Discipline II, Faculty of Medicine, Carol Davila University of Medicine and Pharmacy, 020021 Bucharest, Romania; (L.G.P.); (M.I.P.)
| | - Georgiana Alexandra Grigore
- Research Institute of the University of Bucharest—ICUB, University of Bucharest, 050663 Bucharest, Romania; (G.A.G.)
- Microbiology-Immunology Department, Faculty of Biology, University of Bucharest, 050095 Bucharest, Romania
- National Institute of Research and Development for Biological Sciences, 296 Splaiul Independentei, District 6, 060031 Bucharest, Romania
| | - Roxana-Elena Cristian
- Research Institute of the University of Bucharest—ICUB, University of Bucharest, 050663 Bucharest, Romania; (G.A.G.)
- National Institute of Research and Development for Biological Sciences, 296 Splaiul Independentei, District 6, 060031 Bucharest, Romania
| | - Mircea Ioan Popa
- Microbiology Discipline II, Faculty of Medicine, Carol Davila University of Medicine and Pharmacy, 020021 Bucharest, Romania; (L.G.P.); (M.I.P.)
- Preclinical Testing Unit, Cantacuzino National Military Medical Institute for Research and Development, 050096 Bucharest, Romania
| | - Corneliu Ovidiu Vrancianu
- Research Institute of the University of Bucharest—ICUB, University of Bucharest, 050663 Bucharest, Romania; (G.A.G.)
- National Institute of Research and Development for Biological Sciences, 296 Splaiul Independentei, District 6, 060031 Bucharest, Romania
- Doctoral School, Carol Davila University of Medicine and Pharmacy, Eroii Sanitari 8, District 5, 050474 Bucharest, Romania
| |
Collapse
|
4
|
Ghouri I, Demir M, Khan SA, Mansoor MA, Iqbal M. Unveiling the Potential of Protein-Based Sustainable Antibacterial Materials. Probiotics Antimicrob Proteins 2025; 17:737-762. [PMID: 39422822 DOI: 10.1007/s12602-024-10381-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/05/2024] [Indexed: 10/19/2024]
Abstract
The surge in bacterial growth and the escalating resistance against a multitude of antibiotic drugs have burgeoned into an alarming global threat, necessitating urgent and innovative interventions. In response to this peril, scientists have embarked on the development of advanced biocompatible antibacterial materials, aiming to counteract not only bacterial infections but also the pervasive issue of food spoilage resulting from microbial proliferation. Protein-based biopolymers and their meticulously engineered composites are at the forefront of this endeavor. Their potential in combating this severe global concern presents an approach that intersects the domains of biomedicine and environmental science. The present review article delves into the intricate extraction processes employed to derive various proteins from their natural sources, unraveling the complex biochemical pathways that underpin their antibacterial properties. Expanding on the foundational knowledge, the review also provides a comprehensive synthesis of functionalized proteins modified to enhance their antibacterial efficacy, unveiling a realm of possibilities for tailoring solutions to specific biomedical and environmental applications. The present review navigates through their antibacterial applications; from wound dressings to packaging materials with inherent antibacterial properties, the potential applications underscore the versatility and adaptability of these materials. Moreover, this comprehensive review serves as a valuable roadmap, guiding future research endeavors in reshaping the landscape of natural antibacterial materials on a global scale.
Collapse
Affiliation(s)
- Iqra Ghouri
- Department of Chemistry, School of Natural Sciences (SNS), National University of Sciences & Technology (NUST), H-12, Islamabad, 44000, Pakistan
| | - Muslum Demir
- Department of Chemical Engineering, Bogazici University, 34342, Istanbul, Turkey
- Materials Institute, TUBITAK Marmara Research Center, 41470, Gebze, Turkey
| | - Shahid Ali Khan
- Department of Chemistry, School of Natural Sciences (SNS), National University of Sciences & Technology (NUST), H-12, Islamabad, 44000, Pakistan
| | - Muhammad Adil Mansoor
- Department of Chemistry, School of Natural Sciences (SNS), National University of Sciences & Technology (NUST), H-12, Islamabad, 44000, Pakistan
| | - Mudassir Iqbal
- Department of Chemistry, School of Natural Sciences (SNS), National University of Sciences & Technology (NUST), H-12, Islamabad, 44000, Pakistan.
| |
Collapse
|
5
|
Madineh H, Mansourinia F, Zarrintaj P, Poostchi M, Gnatowski P, Kucinska-Lipka J, Ghaffari M, Hasanin MS, Chapi S, Yazdi MK, Ashrafizadeh M, Bączek T, Saeb MR, Wang G. Stimuli-responsive delivery systems using carbohydrate polymers: A review. Int J Biol Macromol 2025; 310:142648. [PMID: 40174846 DOI: 10.1016/j.ijbiomac.2025.142648] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2024] [Revised: 03/08/2025] [Accepted: 03/27/2025] [Indexed: 04/04/2025]
Abstract
Carbohydrate polymers, including Chitosan, Cellulose, Starch, Dextran, Pectin, Alginate, and Hyaluronic Acid, have been considered as stimuli-responsive biopolymers demonstrating significant potential for drug delivery approaches. Relying on the specific design and fabrication, such biopolymers are able to respond to fluctuations in pH, temperature, or enzymatic activity. This review investigates stimuli-responsive biopolymers, known as carbohydrate polymers, mainly chitosan, cellulose, and alginate, utilized as drug delivery approaches, emphasizing that these stimuli-responsive biopolymers accelerate controlled drug release. The pH-responsive delivery systems selectively target acidic tumor microenvironments, while temperature-responsive materials provide precise control for drug release produced by hyperthermia. Light-responsive biopolymers provide spatial and temporal control, providing appropriate for targeted therapy. Redox-responsive structures are especially efficient in responding to elevated glutathione (GSH) in tumor microenvironment, facilitating targeted drug release. Electro- and magnetic-responsive systems provide remote control functionalities, improving the accuracy of drug administration. The incorporation of multi-stimuli-responsive mechanisms implies a remarkable progression in drug delivery, providing a more versatile and adaptable framework for therapeutic applications. Accordingly, the future research on carbohydrate polymer-based stimuli-responsive delivery systems should focus on improving the responsiveness and targeting efficacy through complicated optimization of features and performance of carbohydrate polymers, where the integration of multifunctional moieties facilitates transformation of targeted drugs for broader biological functions.
Collapse
Affiliation(s)
- Hossein Madineh
- Polymer Engineering Department, Chemical Engineering Faculty, Tarbiat Modares University, Tehran, Iran
| | - Fatemeh Mansourinia
- Polymer Engineering Department, Chemical Engineering Faculty, Tarbiat Modares University, Tehran, Iran
| | | | - Maryam Poostchi
- Department of Polymer Technology, Faculty of Chemistry, Gdańsk University of Technology, Gabriela Narutowicza 11/12, 80-233 Gdańsk, Poland
| | - Przemysław Gnatowski
- Department of Polymer Technology, Faculty of Chemistry, Gdańsk University of Technology, Gabriela Narutowicza 11/12, 80-233 Gdańsk, Poland; Department of Environmental Toxicology, Faculty of Health Sciences with the Institute of Maritime and Tropical Medicine, Medical University of Gdańsk, Dębowa 23A, Gdańsk 80-204, Poland.
| | - Justyna Kucinska-Lipka
- Department of Polymer Technology, Faculty of Chemistry, Gdańsk University of Technology, Gabriela Narutowicza 11/12, 80-233 Gdańsk, Poland
| | - Mehdi Ghaffari
- Polymer Group, Faculty of Technical and Engineering, Golestan University, P. O. Box 155, Gorgan, Golestan, Iran
| | - Mohamed S Hasanin
- Cellulose and Paper Department, National Research Centre, 33 El Bohouth St., Cairo 12622, Egypt; Department of Polymer and Biomaterials Science, West Pomeranian University of Technology in Szczecin, Al. Piastow 45, 70-311 Szczecin, Poland
| | - Sharanappa Chapi
- Department of Physics, B.M.S. College of Engineering, Basavanagudi - 560019, Bengaluru, Karnataka, India
| | - Mohsen Khodadadi Yazdi
- Division of Electrochemistry and Surface Physical Chemistry, Faculty of Applied Physics and Mathematics, Gdańsk University of Technology, Narutowicza 11/12, 80-233 Gdańsk, Poland; Advanced Materials Center, Gdańsk University of Technology, Narutowicza 11/12, 80-233 Gdańsk, Poland
| | - Milad Ashrafizadeh
- Department of Radiation Oncology and Shandong Provincial Key Laboratory of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong 250117, China
| | - Tomasz Bączek
- Department of Pharmaceutical Chemistry, Medical University of Gdańsk, J. Hallera 107, 80-416 Gdańsk, Poland
| | - Mohammad Reza Saeb
- Department of Pharmaceutical Chemistry, Medical University of Gdańsk, J. Hallera 107, 80-416 Gdańsk, Poland.
| | - Guizhen Wang
- Department of Emergency, Shanghai Tenth People's Hospital, School of Medicine Tongji University, Shanghai 200072, China.
| |
Collapse
|
6
|
Sonbol H, Ahmed EZ, Mohamed ET, Abdelmonem AF, El-Sayed H. Exploring the multifunctionality of myco-synthesized selenium nanoparticles: biological, docking study and plant growth-promoting perspectives. Front Microbiol 2025; 16:1565907. [PMID: 40160268 PMCID: PMC11952769 DOI: 10.3389/fmicb.2025.1565907] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2025] [Accepted: 02/26/2025] [Indexed: 04/02/2025] Open
Abstract
Selenium is a vital trace mineral that regulates essential physiological functions, and the development of sustainable methods for synthesizing selenium nanoparticles (SeNPs) is an active area of research. This study reported the mycosynthesis of SeNPs using the aqueous extract of the edible mushroom Pleurotus ostreatus. The synthesized SeNPs were characterized using various analytical techniques, including UV-visible spectroscopy, dynamic light scattering, transmission electron microscopy (TEM), and Fourier-transform infrared spectroscopy (FTIR). The results revealed that the SeNPs exhibited spherical morphology with a diameter range of 72-148 nm, moderate stability with a zeta potential of -10.5 mV, and a polydispersity index of 0.27. The myco-synthesized SeNPs demonstrated potent antioxidant activity with a DPPH radical scavenging IC50 value of 662.1 ± 1.05 μg/mL, comparable to the standard antioxidant Trolox (IC50 = 24.42 ± 0.87 μM). Furthermore, SeNPs exhibited considerable antimicrobial efficacy against Staphylococcus aureus (17 ± 0.02 mm inhibition zone), followed by Escherichia coli (16 ± 1.04 mm) and Candida albicans (12 ± 0.3 mm). Additionally, SeNPs displayed moderate antiviral activity against a low-pathogenic coronavirus (229E) strain, with a selectivity index (SI) of 5. In agriculture, the application of SeNPs at 10 μM significantly increased primary metabolite production in wheat (Triticum aestivum), with total soluble sugars reaching 54.32 mg/g and soluble proteins increasing to 139.66 mg/g, promoting both shoot and root growth. The comprehensive characterization and evaluation of SeNPs provide valuable insights into their multifunctionality, paving the way for further exploration in medicine, agriculture, and environmental applications.
Collapse
Affiliation(s)
- Hana Sonbol
- Department of Biology, College of Sciences, Princess Nourah Bint Abdulrahman University, Riyadh, Saudi Arabia
| | - Eman Zakaria Ahmed
- Botany and Microbiology Department, Faculty of Science, Helwan University, Helwan, Egypt
| | - Eslam T. Mohamed
- Botany and Microbiology Department, Faculty of Science, Helwan University, Helwan, Egypt
| | - Asmaa F. Abdelmonem
- Botany and Microbiology Department, Faculty of Science, Helwan University, Helwan, Egypt
| | - Heba El-Sayed
- Botany and Microbiology Department, Faculty of Science, Helwan University, Helwan, Egypt
| |
Collapse
|
7
|
Ramadhan SA, Ali DS. Innovations in Core-Shell Nanoparticles: Advancing Drug Delivery Solutions and Precision Medicine. OMICS : A JOURNAL OF INTEGRATIVE BIOLOGY 2025; 29:73-86. [PMID: 39981668 DOI: 10.1089/omi.2024.0182] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/22/2025]
Abstract
Drug delivery innovation is an important pillar of systems pharmacology wherein nanotechnology offers significant prospects. This expert review examines and unpacks how core-shell nanoparticles (NPs) could revolutionize drug delivery systems and play a key role in advancing personalized and precision medicine. Core-shell NPs have gained attention as flexible tools for drug delivery due to their distinct structure, which features a core material enclosed by a protective shell. This setup offers multiple benefits, such as effective drug encapsulation, shielding the drug from degradation, and allowing for controlled release. Accordingly, the core serves as a safe storage area for the drug while the shell manages the release speed, providing added stability and supporting sustained delivery. By enabling targeted drug release, this controlled mechanism can help improve treatment outcomes and reduce side effects. Various materials, including polymers, lipids, and inorganic substances create these NPs. Biodegradable polymers, such as poly(lactic-co-glycolic acid) and poly(lactic acid), are popular choices because they offer adjustable degradation rates, which further control how the drug is released. These materials can be tailored for better drug loading, compatibility with the host organism, and specific chemical properties to suit different therapeutic needs. Research into core-shell NPs has been advancing in many therapeutic areas, highlighting their potential for drug delivery innovations. The potential of core-shell NPs to revolutionize drug delivery is not just a possibility but a promising reality that could significantly advance the field of personalized/precision medicine.
Collapse
Affiliation(s)
- Suren A Ramadhan
- Department of Pharmacy, College of Pharmacy, Knowledge University, Erbil, Iraq
| | - Diyar S Ali
- Department of Pharmacy, College of Pharmacy, Knowledge University, Erbil, Iraq
- Department of Chemistry, College of Science, Salahaddin University-Erbil, Erbil, Iraq
| |
Collapse
|
8
|
Alberts A, Tudorache DI, Niculescu AG, Grumezescu AM. Advancements in Wound Dressing Materials: Highlighting Recent Progress in Hydrogels, Foams, and Antimicrobial Dressings. Gels 2025; 11:123. [PMID: 39996666 PMCID: PMC11854827 DOI: 10.3390/gels11020123] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2024] [Revised: 01/27/2025] [Accepted: 02/05/2025] [Indexed: 02/26/2025] Open
Abstract
Recent advancements in wound dressing materials have significantly improved acute and chronic wound management by addressing challenges such as infection control, moisture balance, and enhanced healing. Important progress has been made, especially with hydrogels, foams, and antimicrobial materials for creating optimized dressings. Hydrogels are known for maintaining optimal moisture levels, while foam dressings are excellent exudate absorbents. Meanwhile, antimicrobial dressing incorporates various antimicrobial agents to reduce infection risks. These dressing options reduce wound healing time while focusing on customized patient needs. Therefore, this review highlights the newest research materials and prototypes for wound healing applications, emphasizing their particular benefits and clinical importance. Innovations such as stimuli-responsive hydrogels and hybrid bioengineered composites are discussed in relation to their enhanced properties, including responsiveness to pH, temperature, glucose, or enzymes and drug delivery precision. Moreover, ongoing clinical trials have been included, demonstrating the potential of emerging solutions to be soon translated from the laboratory to clinical settings. By discussing interdisciplinary approaches that integrate advanced materials, nanotechnology, and biological insights, this work provides a contemporary framework for patient-centric, efficient wound care strategies.
Collapse
Affiliation(s)
- Adina Alberts
- Carol Davila University of Medicine and Pharmacy, 050474 Bucharest, Romania;
| | - Dana-Ionela Tudorache
- National University of Science and Technology Politehnica Bucharest, 011061 Bucharest, Romania; (D.-I.T.); (A.M.G.)
| | - Adelina-Gabriela Niculescu
- National University of Science and Technology Politehnica Bucharest, 011061 Bucharest, Romania; (D.-I.T.); (A.M.G.)
- Research Institute of the University of Bucharest—ICUB, University of Bucharest, 050657 Bucharest, Romania
| | - Alexandru Mihai Grumezescu
- National University of Science and Technology Politehnica Bucharest, 011061 Bucharest, Romania; (D.-I.T.); (A.M.G.)
- Research Institute of the University of Bucharest—ICUB, University of Bucharest, 050657 Bucharest, Romania
| |
Collapse
|
9
|
Puspadewi R, Milanda T, Muhaimin M, Chaerunisaa AY. Nanoparticle-Encapsulated Plant Polyphenols and Flavonoids as an Enhanced Delivery System for Anti-Acne Therapy. Pharmaceuticals (Basel) 2025; 18:209. [PMID: 40006023 PMCID: PMC11858878 DOI: 10.3390/ph18020209] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2024] [Revised: 01/29/2025] [Accepted: 01/29/2025] [Indexed: 02/27/2025] Open
Abstract
This study conducted a literature review by searching for articles related to the treatment of skin infections/wrinkles using nano-delivery systems containing natural compounds. The search was conducted in various databases for articles published in the last 10 years, with strict inclusion and exclusion criteria. Of the 490 articles found, 40 were considered relevant. Acne vulgaris is a common dermatological disorder characterised by inflammation of the sebaceous glands, often resulting in the development of pimples, cysts, and scarring. Conventional treatments, including antibiotics and topical retinoids, frequently demonstrate limitations such as side effects, resistance, and insufficient skin absorption. Recent advancements in nanotechnology have enabled the creation of innovative drug-delivery systems that enhance the effectiveness and reduce the adverse effects of anti-acne medications. Polyphenols and flavonoids, natural bioactive compounds with notable anti-inflammatory, antioxidant, and antibacterial properties, are recognised for their therapeutic effectiveness in acne treatment. However, their practical application is hindered by insufficient solubility, stability, and bioavailability. The incorporation of these compounds into nanoparticle-based delivery systems has shown promise in resolving these challenges. Various nanoparticle platforms, including lipid-based nanoparticles, polymeric nanoparticles, and solid lipid nanoparticles, are evaluated for their ability to improve the stability, controlled release, and targeted delivery of polyphenols and flavonoids to the skin. The advent of polyphenol and flavonoid-loaded nanoparticles marks a new acne therapy era.
Collapse
Affiliation(s)
- Ririn Puspadewi
- Doctoral Program of Pharmacy, Faculty of Pharmacy, Padjadjaran University, Sumedang 45363, Indonesia;
- Faculty of Pharmacy, Jenderal Achmad Yani University, Cimahi 40531, Indonesia
| | - Tiana Milanda
- Department of Pharmaceutical Biology, Faculty of Pharmacy, Padjadjaran University, Sumedang 45363, Indonesia
| | - Muhaimin Muhaimin
- Department of Pharmaceutical Biology, Faculty of Pharmacy, Padjadjaran University, Sumedang 45363, Indonesia
- Center of Herbal Studies, Padjadjaran University, Sumedang 45363, Indonesia
| | - Anis Yohana Chaerunisaa
- Department of Pharmaceutics and Pharmaceutical Technology, Faculty of Pharmacy, Padjadjaran University, Sumedang 45363, Indonesia
| |
Collapse
|
10
|
Yazdan M, Naghib SM. Smart Ultrasound-responsive Polymers for Drug Delivery: An Overview on Advanced Stimuli-sensitive Materials and Techniques. Curr Drug Deliv 2025; 22:283-309. [PMID: 38288800 DOI: 10.2174/0115672018283792240115053302] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2023] [Revised: 12/11/2023] [Accepted: 12/28/2023] [Indexed: 04/11/2025]
Abstract
In recent years, a notable advancement has occurred in the domain of drug delivery systems via the integration of intelligent polymers that respond to ultrasound. The implementation of this groundbreaking methodology has significantly revolutionised the controlled and precise delivery of therapeutic interventions. An in-depth investigation is conducted into the most recent developments in ultrasonic stimulus-responsive materials and techniques for the purpose of accomplishing precise medication administration. The investigation begins with an exhaustive synopsis of the foundational principles underlying drug delivery systems that react to ultrasonic stimuli, focusing specifically on the complex interplay between polymers and ultrasound waves. Significant attention is devoted to the development of polymers that demonstrate tailored responsiveness to ultrasound, thereby exemplifying their versatility in generating controlled drug release patterns. Numerous classifications of intelligent polymers are examined in the discussion, including those that react to variations in temperature, pH, and enzymes. When coupled with ultrasonic stimuli, these polymers offer a sophisticated framework for the precise manipulation of drug release in terms of both temporal and spatial dimensions. The present study aims to examine the synergistic effects of responsive polymers and ultrasound in overcoming biological barriers such as the blood-brain barrier and the gastrointestinal tract. By doing so, it seeks to shed light on the potential applications of these materials in intricate clinical scenarios. The issues and future prospects of intelligent ultrasound-responsive polymers in the context of drug delivery are critically analysed in this article. The objective of this study is to offer valuable perspectives on the challenges that must be overcome to enable the effective implementation of these technologies. The primary objective of this comprehensive review is to furnish researchers, clinicians, and pharmaceutical scientists with a wealth of information that will serve as a guide for forthcoming developments in the development and enhancement of intelligent drug delivery systems that employ ultrasound-responsive polymers to attain superior therapeutic outcomes.
Collapse
Affiliation(s)
- Mostafa Yazdan
- Department of Nanotechnology, School of Advanced Technologies, Iran University of Science and Technology (IUST), Tehran, 1684613114, Iran
| | - Seyed Morteza Naghib
- Department of Nanotechnology, School of Advanced Technologies, Iran University of Science and Technology (IUST), Tehran, 1684613114, Iran
| |
Collapse
|
11
|
Singhal M, Seaton CC, Surtees A, Katsikogianni MG. Formulation, Characterisation, and Biocompatibility Assessment of Rifampicin-Loaded Poly(d,l-lactide-co-glycolide) Composites for Local Treatment of Orthopaedic and Wound Infections. Pharmaceutics 2024; 16:1467. [PMID: 39598590 PMCID: PMC11597898 DOI: 10.3390/pharmaceutics16111467] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2024] [Revised: 11/10/2024] [Accepted: 11/12/2024] [Indexed: 11/29/2024] Open
Abstract
Background/Objectives: The escalating challenge of antimicrobial resistance (AMR) necessitates the development of targeted antibiotic delivery platforms, minimising systemic administration. Polymer-based drug delivery emerges as a promising solution, ensuring sustained release and prolonged efficacy of bioactive compounds, ensuring long-term efficacy. Methods: This study focuses on encapsulating rifampicin (RIF), a key antibiotic for orthopaedic and wound-related infections, within Poly(d,l-lactide-co-glycolide) (PLGA), a biodegradable polymer, through solvent casting, to formulate a PLGA-RIF composite membrane. Comprehensive characterisation, employing Fourier-transformed infrared spectroscopy (FT-IR), scanning electron microscopy (SEM), thermal analysis and X-ray Diffraction (XRD), confirmed the integrity of both the starting and produced materials. UV-Vis spectroscopy revealed a controlled drug release profile over 21 days in various media, with the chosen media influencing the drug release, notably the tryptic soya broth (TSB) caused the highest release. The quantitative assessment of the antimicrobial efficacy of the developed PLGA-RIF composite was conducted by measuring the size of the inhibition zones against both Gram-negative and Gram-positive bacteria. Results: The results confirmed the composite's potential as a robust antibacterial biomaterial, demonstrating a rapid and effective antibacterial response. Cytocompatibility tests incorporated human fibroblast and osteoblast-like cell lines and demonstrated that the RIF:PLGA (1:8) formulation maintained eukaryotic cell viability, indicating the composite's potential for targeted medical applications in combating bacterial infections with minimal systemic impact. Conclusions: This study presents the significance of investigating drug release within appropriate and relevant physiological media. A key novelty of this work therefore lies in the exploration of drug release dynamics across different media, allowing for a comprehensive understanding of how varying physiological conditions may influence drug release and its effect on biological responses.
Collapse
Affiliation(s)
- Mitali Singhal
- School of Pharmacy and Medical Science, Institute of Cancer Therapeutics, University of Bradford, Bradford BD7 1DP, UK;
| | - Colin C. Seaton
- School of Chemistry and Biosciences, University of Bradford, Bradford BD7 1DP, UK;
| | - Alexander Surtees
- School of Archaeological and Forensic Sciences, University of Bradford, Bradford BD7 1DP, UK;
| | | |
Collapse
|
12
|
Zhang Q. Bacteria carrying mobile colistin resistance genes and their control measures, an updated review. Arch Microbiol 2024; 206:462. [PMID: 39516398 DOI: 10.1007/s00203-024-04188-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2024] [Revised: 10/23/2024] [Accepted: 10/28/2024] [Indexed: 11/16/2024]
Abstract
The plasmid encoded mobile colistin resistance (MCRs) enzyme poses a significant challenge to the clinical efficacy of colistin, which is frequently employed as a last resort antibiotic for treating infections caused by multidrug resistant bacteria. This transferase catalyzes the addition of positively charged phosphoethanolamine to lipid A of the outer membrane of gram-negative bacteria, thereby facilitating the acquired colistin resistance. This review aims to summarize and critically discuss recent advancements in the distribution and pathogenesis of mcr-positive bacteria, as well as the various control measures available for treating these infections. In addition, the ecology of mcr genes, colistin-resistance mechanism, co-existence with other antibiotic resistant genes, and their impact on clinical treatment are also analyzed to address the colistin resistance crisis. These insights provide a comprehensive perspective on MCRs and serve as a valuable reference for future therapeutic approaches to effectively combat mcr-positive bacterial infections.
Collapse
Affiliation(s)
- Qi Zhang
- Centre for Eye and Vision Research, Hong Kong Science Park, Hong Kong, China.
| |
Collapse
|
13
|
Chen X, Wu D, Chen Z. Biomedical applications of stimuli-responsive nanomaterials. MedComm (Beijing) 2024; 5:e643. [PMID: 39036340 PMCID: PMC11260173 DOI: 10.1002/mco2.643] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2023] [Revised: 06/12/2024] [Accepted: 06/13/2024] [Indexed: 07/23/2024] Open
Abstract
Nanomaterials have aroused great interests in drug delivery due to their nanoscale structure, facile modifiability, and multifunctional physicochemical properties. Currently, stimuli-responsive nanomaterials that can respond to endogenous or exogenous stimulus display strong potentials in biomedical applications. In comparison with conventional nanomaterials, stimuli-responsive nanomaterials can improve therapeutic efficiency and reduce the toxicity of drugs toward normal tissues through specific targeting and on-demand drug release at pathological sites. In this review, we summarize the responsive mechanism of a variety of stimulus, including pH, redox, and enzymes within pathological microenvironment, as well as exogenous stimulus such as thermal effect, magnetic field, light, and ultrasound. After that, biomedical applications (e.g., drug delivery, imaging, and theranostics) of stimuli-responsive nanomaterials in a diverse array of common diseases, including cardiovascular diseases, cancer, neurological disorders, inflammation, and bacterial infection, are presented and discussed. Finally, the remaining challenges and outlooks of future research directions for the biomedical applications of stimuli-responsive nanomaterials are also discussed. We hope that this review can provide valuable guidance for developing stimuli-responsive nanomaterials and accelerate their biomedical applications in diseases diagnosis and treatment.
Collapse
Affiliation(s)
- Xiaojie Chen
- Key Laboratory of Neuropharmacology and Translational Medicine of Zhejiang ProvinceSchool of Pharmaceutical SciencesDepartment of NeurologyThe First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine)HangzhouChina
| | - Di Wu
- Key Laboratory of Neuropharmacology and Translational Medicine of Zhejiang ProvinceSchool of Pharmaceutical SciencesDepartment of NeurologyThe First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine)HangzhouChina
| | - Zhong Chen
- Key Laboratory of Neuropharmacology and Translational Medicine of Zhejiang ProvinceSchool of Pharmaceutical SciencesDepartment of NeurologyThe First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine)HangzhouChina
| |
Collapse
|
14
|
Tageldin A, Omolo CA, Nyandoro VO, Elhassan E, Kassam SZF, Peters XQ, Govender T. Engineering dynamic covalent bond-based nanosystems for delivery of antimicrobials against bacterial infections. J Control Release 2024; 371:237-257. [PMID: 38815705 DOI: 10.1016/j.jconrel.2024.05.047] [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: 01/27/2024] [Revised: 05/22/2024] [Accepted: 05/27/2024] [Indexed: 06/01/2024]
Abstract
Nanodrug delivery systems (NDDS) continue to be explored as novel strategies enhance therapy outcomes and combat microbial resistance. The need for the formulation of smart drug delivery systems for targeting infection sites calls for the engineering of responsive chemical designs such as dynamic covalent bonds (DCBs). Stimuli response due to DCBs incorporated into nanosystems are emerging as an alternative way to target infection sites, thus enhancing the delivery of antibacterial agents. This leads to the eradication of bacterial infections and the reduction of antimicrobial resistance. Incorporating DCBs on the backbone of the nanoparticles endows the systems with several properties, including self-healing, controlled disassembly, and stimuli responsiveness, which are beneficial in the delivery and release of the antimicrobial at the infection site. This review provides a comprehensive and current overview of conventional DCBs-based nanosystems, stimuli-responsive DCBs-based nanosystems, and targeted DCBs-based nanosystems that have been reported in the literature for antibacterial delivery. The review emphasizes the DCBs used in their design, the nanomaterials constructed, the drug release-triggering stimuli, and the antibacterial efficacy of the reported DCBs-based nanosystems. Additionally, the review underlines future strategies that can be used to improve the potential of DCBs-based nanosystems to treat bacterial infections and overcome antibacterial resistance.
Collapse
Affiliation(s)
- Abdelrahman Tageldin
- Discipline of Pharmaceutical Sciences, College of Health Sciences, University of KwaZulu-Natal, Private Bag X54001, Durban, South Africa
| | - Calvin A Omolo
- Discipline of Pharmaceutical Sciences, College of Health Sciences, University of KwaZulu-Natal, Private Bag X54001, Durban, South Africa; Department of Pharmaceutics and Pharmacy Practice, School of Pharmacy and Health Sciences, United States International University-Africa, P. O. Box 14634-00800, Nairobi, Kenya.
| | - Vincent O Nyandoro
- Discipline of Pharmaceutical Sciences, College of Health Sciences, University of KwaZulu-Natal, Private Bag X54001, Durban, South Africa
| | - Eman Elhassan
- Discipline of Pharmaceutical Sciences, College of Health Sciences, University of KwaZulu-Natal, Private Bag X54001, Durban, South Africa
| | - Sania Z F Kassam
- Discipline of Pharmaceutical Sciences, College of Health Sciences, University of KwaZulu-Natal, Private Bag X54001, Durban, South Africa
| | - Xylia Q Peters
- Discipline of Pharmaceutical Sciences, College of Health Sciences, University of KwaZulu-Natal, Private Bag X54001, Durban, South Africa
| | - Thirumala Govender
- Discipline of Pharmaceutical Sciences, College of Health Sciences, University of KwaZulu-Natal, Private Bag X54001, Durban, South Africa.
| |
Collapse
|
15
|
Saadh MJ, Mustafa MA, Kumar A, Alamir HTA, Kumar A, Khudair SA, Faisal A, Alubiady MHS, Jalal SS, Shafik SS, Ahmad I, Khry FAF, Abosaoda MK. Stealth Nanocarriers in Cancer Therapy: a Comprehensive Review of Design, Functionality, and Clinical Applications. AAPS PharmSciTech 2024; 25:140. [PMID: 38890191 DOI: 10.1208/s12249-024-02843-5] [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: 02/17/2024] [Accepted: 05/13/2024] [Indexed: 06/20/2024] Open
Abstract
Nanotechnology has significantly transformed cancer treatment by introducing innovative methods for delivering drugs effectively. This literature review provided an in-depth analysis of the role of nanocarriers in cancer therapy, with a particular focus on the critical concept of the 'stealth effect.' The stealth effect refers to the ability of nanocarriers to evade the immune system and overcome physiological barriers. The review investigated the design and composition of various nanocarriers, such as liposomes, micelles, and inorganic nanoparticles, highlighting the importance of surface modifications and functionalization. The complex interaction between the immune system, opsonization, phagocytosis, and the protein corona was examined to understand the stealth effect. The review carefully evaluated strategies to enhance the stealth effect, including surface coating with polymers, biomimetic camouflage, and targeting ligands. The in vivo behavior of stealth nanocarriers and their impact on pharmacokinetics, biodistribution, and toxicity were also systematically examined. Additionally, the review presented clinical applications, case studies of approved nanocarrier-based cancer therapies, and emerging formulations in clinical trials. Future directions and obstacles in the field, such as advancements in nanocarrier engineering, personalized nanomedicine, regulatory considerations, and ethical implications, were discussed in detail. The review concluded by summarizing key findings and emphasizing the transformative potential of stealth nanocarriers in revolutionizing cancer therapy. This review enhanced the comprehension of nanocarrier-based cancer therapies and their potential impact by providing insights into advanced studies, clinical applications, and regulatory considerations.
Collapse
Affiliation(s)
- Mohamed J Saadh
- Faculty of Pharmacy, Middle East University, Amman, 11831, Jordan.
| | - Mohammed Ahmed Mustafa
- Department of Medical Laboratory Technology, University of Imam Jaafar AL-Sadiq, Baghdad, Iraq
| | - Ashwani Kumar
- Department of Life Sciences, School of Sciences, Jain (Deemed-to-be) University, Bengaluru, Karnataka, India
- Department of Pharmacy, Vivekananda Global University, Jaipur, Rajasthan, India
| | | | - Abhishek Kumar
- School of Pharmacy-Adarsh Vijendra Institute of Pharmaceutical Sciences, Shobhit University, Gangoh, 247341, Uttar Pradesh, India
- Department of Pharmacy, Arka Jain University, Jamshedpur, Jharkhand, 831001, India
| | | | - Ahmed Faisal
- Department of Pharmacy, Al-Noor University College, Nineveh, Iraq
| | | | - Sarah Salah Jalal
- College of Pharmacy, National University of Science and Technology, Nasiriyah, Dhi Qar, Iraq
| | - Shafik Shaker Shafik
- Experimental Nuclear Radiation Group, Scientific Research Center, Al-Ayen University, Thi-Qar, Iraq
| | - Irfan Ahmad
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Khalid University, Abha, Saudi Arabia
| | - Faeza A F Khry
- Faculty of pharmacy, department of pharmaceutics, Al-Esraa University, Baghdad, Iraq
| | - Munther Kadhim Abosaoda
- College of Technical Engineering, The Islamic University, Najaf, Iraq
- College of Technical Engineering, The Islamic University of Al Diwaniyah, Qadisiyyah, Iraq
- College of Technical Engineering, The Islamic University of Babylon, Babylon, Iraq
| |
Collapse
|
16
|
Naser IH, Zaid M, Ali E, Jabar HI, Mustafa AN, Alubiady MHS, Ramadan MF, Muzammil K, Khalaf RM, Jalal SS, Alawadi AH, Alsalamy A. Unveiling innovative therapeutic strategies and future trajectories on stimuli-responsive drug delivery systems for targeted treatment of breast carcinoma. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024; 397:3747-3770. [PMID: 38095649 DOI: 10.1007/s00210-023-02885-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/10/2023] [Accepted: 12/02/2023] [Indexed: 05/23/2024]
Abstract
This comprehensive review delineates the latest advancements in stimuli-responsive drug delivery systems engineered for the targeted treatment of breast carcinoma. The manuscript commences by introducing mammary carcinoma and the current therapeutic methodologies, underscoring the urgency for innovative therapeutic strategies. Subsequently, it elucidates the logic behind the employment of stimuli-responsive drug delivery systems, which promise targeted drug administration and the minimization of adverse reactions. The review proffers an in-depth analysis of diverse types of stimuli-responsive systems, including thermoresponsive, pH-responsive, and enzyme-responsive nanocarriers. The paramount importance of material choice, biocompatibility, and drug loading strategies in the design of these systems is accentuated. The review explores characterization methodologies for stimuli-responsive nanocarriers and probes preclinical evaluations of their efficacy, toxicity, pharmacokinetics, and biodistribution in mammary carcinoma models. Clinical applications of stimuli-responsive systems, ongoing clinical trials, the potential of combination therapies, and the utility of multifunctional nanocarriers for the co-delivery of assorted drugs and therapies are also discussed. The manuscript addresses the persistent challenge of drug resistance in mammary carcinoma and the potential of stimuli-responsive systems in surmounting it. Regulatory and safety considerations, including FDA guidelines and biocompatibility assessments, are outlined. The review concludes by spotlighting future trajectories and emergent technologies in stimuli-responsive drug delivery, focusing on pioneering approaches, advancements in nanotechnology, and personalized medicine considerations. This review aims to serve as a valuable compendium for researchers and clinicians interested in the development of efficacious and safe stimuli-responsive drug delivery systems for the treatment of breast carcinoma.
Collapse
Affiliation(s)
- Israa Habeeb Naser
- Medical Laboratories Techniques Department, AL-Mustaqbal University, Hillah, Babil, Iraq
| | - Muhaned Zaid
- Department of Pharmacy, Al-Manara College for Medical Sciences, Maysan, Amarah, Iraq
| | - Eyhab Ali
- Al-Zahraa University for Women, Karbala, Iraq
| | - Hayder Imad Jabar
- Department of Pharmaceutics, College of Pharmacy, University of Al-Ameed, Karbala, Iraq
| | | | | | | | - Khursheed Muzammil
- Department of Public Health, College of Applied Medical Sciences, Khamis Mushait Campus, King Khalid University, Abha, Saudi Arabia
| | | | - Sarah Salah Jalal
- College of Pharmacy, National University of Science and Technology, Dhi Qar, Iraq
| | - Ahmed Hussien Alawadi
- College of Technical Engineering, the Islamic University, Najaf, Iraq
- College of Technical Engineering, the Islamic University of Al Diwaniyah, Al Diwaniyah, Iraq
- College of Technical Engineering, the Islamic University of Babylon, Babylon, Iraq
| | - Ali Alsalamy
- College of Technical Engineering, Imam Ja'afar Al-Sadiq University, Al-Muthanna, Iraq.
| |
Collapse
|
17
|
Singh G, Rana A, Smriti. Decoding antimicrobial resistance: unraveling molecular mechanisms and targeted strategies. Arch Microbiol 2024; 206:280. [PMID: 38805035 DOI: 10.1007/s00203-024-03998-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2024] [Revised: 05/07/2024] [Accepted: 05/08/2024] [Indexed: 05/29/2024]
Abstract
Antimicrobial resistance poses a significant global health threat, necessitating innovative approaches for combatting it. This review explores various mechanisms of antimicrobial resistance observed in various strains of bacteria. We examine various strategies, including antimicrobial peptides (AMPs), novel antimicrobial materials, drug delivery systems, vaccines, antibody therapies, and non-traditional antibiotic treatments. Through a comprehensive literature review, the efficacy and challenges of these strategies are evaluated. Findings reveal the potential of AMPs in combating resistance due to their unique mechanisms and lower propensity for resistance development. Additionally, novel drug delivery systems, such as nanoparticles, show promise in enhancing antibiotic efficacy and overcoming resistance mechanisms. Vaccines and antibody therapies offer preventive measures, although challenges exist in their development. Non-traditional antibiotic treatments, including CRISPR-Cas systems, present alternative approaches to combat resistance. Overall, this review underscores the importance of multifaceted strategies and coordinated global efforts to address antimicrobial resistance effectively.
Collapse
Affiliation(s)
- Gagandeep Singh
- Department of Biosciences (UIBT), Chandigarh University, Punjab, 140413, India
| | - Anita Rana
- Department of Biosciences (UIBT), Chandigarh University, Punjab, 140413, India.
| | - Smriti
- Department of Biosciences (UIBT), Chandigarh University, Punjab, 140413, India
| |
Collapse
|
18
|
Yu YM, Lu YP, Zhang T, Zheng YF, Liu YS, Xia DD. Biomaterials science and surface engineering strategies for dental peri-implantitis management. Mil Med Res 2024; 11:29. [PMID: 38741175 DOI: 10.1186/s40779-024-00532-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/16/2023] [Accepted: 04/29/2024] [Indexed: 05/16/2024] Open
Abstract
Peri-implantitis is a bacterial infection that causes soft tissue inflammatory lesions and alveolar bone resorption, ultimately resulting in implant failure. Dental implants for clinical use barely have antibacterial properties, and bacterial colonization and biofilm formation on the dental implants are major causes of peri-implantitis. Treatment strategies such as mechanical debridement and antibiotic therapy have been used to remove dental plaque. However, it is particularly important to prevent the occurrence of peri-implantitis rather than treatment. Therefore, the current research spot has focused on improving the antibacterial properties of dental implants, such as the construction of specific micro-nano surface texture, the introduction of diverse functional coatings, or the application of materials with intrinsic antibacterial properties. The aforementioned antibacterial surfaces can be incorporated with bioactive molecules, metallic nanoparticles, or other functional components to further enhance the osteogenic properties and accelerate the healing process. In this review, we summarize the recent developments in biomaterial science and the modification strategies applied to dental implants to inhibit biofilm formation and facilitate bone-implant integration. Furthermore, we summarized the obstacles existing in the process of laboratory research to reach the clinic products, and propose corresponding directions for future developments and research perspectives, so that to provide insights into the rational design and construction of dental implants with the aim to balance antibacterial efficacy, biological safety, and osteogenic property.
Collapse
Affiliation(s)
- Ya-Meng Yu
- Department of Dental Materials, Peking University School and Hospital of Stomatology, Beijing, 100081, China
- National Center for Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, Beijing Key Laboratory of Digital Stomatology & NHC Key Laboratory of Digital Stomatology & NMPA Key Laboratory for Dental Materials, Beijing, 100081, China
| | - Yu-Pu Lu
- Department of Dental Materials, Peking University School and Hospital of Stomatology, Beijing, 100081, China
- National Center for Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, Beijing Key Laboratory of Digital Stomatology & NHC Key Laboratory of Digital Stomatology & NMPA Key Laboratory for Dental Materials, Beijing, 100081, China
| | - Ting Zhang
- School of Materials Science and Engineering, Peking University, Beijing, 100871, China
| | - Yu-Feng Zheng
- School of Materials Science and Engineering, Peking University, Beijing, 100871, China.
| | - Yun-Song Liu
- National Center for Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, Beijing Key Laboratory of Digital Stomatology & NHC Key Laboratory of Digital Stomatology & NMPA Key Laboratory for Dental Materials, Beijing, 100081, China.
- Department of Prosthodontics, Peking University School and Hospital of Stomatology, Beijing, 100081, China.
| | - Dan-Dan Xia
- Department of Dental Materials, Peking University School and Hospital of Stomatology, Beijing, 100081, China.
- National Center for Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, Beijing Key Laboratory of Digital Stomatology & NHC Key Laboratory of Digital Stomatology & NMPA Key Laboratory for Dental Materials, Beijing, 100081, China.
| |
Collapse
|
19
|
Ahmad N, Bukhari SNA, Hussain MA, Ejaz H, Munir MU, Amjad MW. Nanoparticles incorporated hydrogels for delivery of antimicrobial agents: developments and trends. RSC Adv 2024; 14:13535-13564. [PMID: 38665493 PMCID: PMC11043667 DOI: 10.1039/d4ra00631c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Accepted: 04/01/2024] [Indexed: 04/28/2024] Open
Abstract
The prevention and treatment of microbial infections is an imminent global public health concern due to the poor antimicrobial performance of the existing antimicrobial regime and rapidly emerging antibiotic resistance in pathogenic microbes. In order to overcome these problems and effectively control bacterial infections, various new treatment modalities have been identified. To attempt this, various micro- and macro-molecular antimicrobial agents that function by microbial membrane disruption have been developed with improved antimicrobial activity and lesser resistance. Antimicrobial nanoparticle-hydrogels systems comprising antimicrobial agents (antibiotics, biological extracts, and antimicrobial peptides) loaded nanoparticles or antimicrobial nanoparticles (metal or metal oxide) constitute an important class of biomaterials for the prevention and treatment of infections. Hydrogels that incorporate nanoparticles can offer an effective strategy for delivering antimicrobial agents (or nanoparticles) in a controlled, sustained, and targeted manner. In this review, we have described an overview of recent advancements in nanoparticle-hydrogel hybrid systems for antimicrobial agent delivery. Firstly, we have provided an overview of the nanoparticle hydrogel system and discussed various advantages of these systems in biomedical and pharmaceutical applications. Thereafter, different hybrid hydrogel systems encapsulating antibacterial metal/metal oxide nanoparticles, polymeric nanoparticles, antibiotics, biological extracts, and antimicrobial peptides for controlling infections have been reviewed in detail. Finally, the challenges and future prospects of nanoparticle-hydrogel systems have been discussed.
Collapse
Affiliation(s)
- Naveed Ahmad
- Department of Pharmaceutics, College of Pharmacy, Jouf University Sakaka 72388 Aljouf Saudi Arabia
| | - Syed Nasir Abbas Bukhari
- Department of Pharmaceutical Chemistry, College of Pharmacy, Jouf University Sakaka 72388 Aljouf Saudi Arabia
| | - Muhammad Ajaz Hussain
- Centre for Organic Chemistry, School of Chemistry, University of the Punjab Lahore 54590 Pakistan
| | - Hasan Ejaz
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Jouf University Sakaka 72388 Aljouf Saudi Arabia
| | - Muhammad Usman Munir
- Australian Institute for Bioengineering & Nanotechnology, The University of Queensland Brisbane Queens-land 4072 Australia
| | - Muhammad Wahab Amjad
- 6 Center for Ultrasound Molecular Imaging and Therapeutics, School of Medicine, University of Pittsburgh 15213 Pittsburgh Pennsylvania USA
| |
Collapse
|
20
|
Singh A, Sharma JJ, Mohanta B, Sood A, Han SS, Sharma A. Synthetic and biopolymers-based antimicrobial hybrid hydrogels: a focused review. JOURNAL OF BIOMATERIALS SCIENCE. POLYMER EDITION 2024; 35:675-716. [PMID: 37943320 DOI: 10.1080/09205063.2023.2278814] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/15/2023] [Accepted: 10/29/2023] [Indexed: 11/10/2023]
Abstract
The constantly accelerating occurrence of microbial infections and their antibiotic resistance has spurred advancement in the field of material sciences and has guided the development of novel materials with anti-bacterial properties. To address the clinical exigencies, the material of choice should be biodegradable, biocompatible, and able to offer prolonged antibacterial effects. As an attractive option, hydrogels have been explored globally as a potent biomaterial platform that can furnish essential antibacterial attributes owing to its three-dimensional (3D) hydrophilic polymeric network, adequate biocompatibility, and cellular adhesion. The current review focuses on the utilization of different antimicrobial hydrogels based on their sources (natural and synthetic). Further, the review also highlights the strategies for the generation of hydrogels with their advantages and disadvantages and their applications in different biomedical fields. Finally, the prospects in the development of hydrogels-based antimicrobial biomaterials are discussed along with some key challenges encountered during their development and clinical translation.
Collapse
Affiliation(s)
- Anand Singh
- University Institute of Biotechnology, Chandigarh University, Mohali, Punjab, India
| | - Janmay Jai Sharma
- University Institute of Biotechnology, Chandigarh University, Mohali, Punjab, India
| | - Billeswar Mohanta
- University Institute of Biotechnology, Chandigarh University, Mohali, Punjab, India
| | - Ankur Sood
- School of Chemical Engineering, Yeungnam University, Gyeongsan, South Korea
| | - Sung Soo Han
- School of Chemical Engineering, Yeungnam University, Gyeongsan, South Korea
| | - Anirudh Sharma
- Department of Biotechnology, Jaypee Institute of Information Technology, Noida, India
| |
Collapse
|
21
|
Osman N, Omolo CA, Gafar MA, Devnarain N, Rambharose S, Ibrahim UH, Fasiku VO, Govender T. Niosomes modified with a novel pH-responsive coating (mPEG-OA) enhance the antibacterial and anti-biofilm activity of vancomycin against methicillin-resistant Staphylococcus aureus. NANO EXPRESS 2024; 5:015008. [DOI: 10.1088/2632-959x/ad1d02] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
Abstract
Abstract
Surface functionalization of nanoparticles has shown potential in enhancing the efficacy of antibiotic-loaded nanosystems against drug-resistant bacteria. The objective of this study was to synthesize and characterize an acid-cleavable pH-responsive polymer from methoxy polyethylene glycol and oleylamine (mPEG-OA) to surface modify vancomycin (VCM)-loaded niosomes and to evaluate their antibacterial and anti-biofilm effectiveness against methicillin-resistant Staphylococcus aureus (MRSA). The novel mPEG-OA-coated niosomes were biocompatible, hemocompatible with size, polydispersity index, and zeta potential of 169.2 ± 1.6 nm, 0.21 ± 0.01 and −0.82 ± 0.22 mV, respectively. Under acidic conditions, mPEG-OA-coated niosomes exhibited a pH-responsive and sustained VCM release profile and in vitro antibacterial activity than non-coated niosomes and bare VCM. mPEG-OA-coated niosomes showed a significant reduction in biofilm formation at pH 6 compared to pH 7.4 (p = 0,0119). The in vivo efficacy of mPEG-OA-coated niosomes in the BALB/c mice skin infection model showed a 9.9-fold reduction in MRSA load compared to bare VCM. Histomorphologically, the mPEG-OA-coated niosomes group displayed the lowest bacterial load, tissue swelling, and inflammation. The results of this study demonstrate the potential of novel pH-responsive mPEG-OA-derived polymer coating to enhance bacterial killing kinetics, and antibacterial and anti-biofilm efficacies over conventional antibiotic and non-functionalized nano delivery systems.
Collapse
|
22
|
Ijaz M, Aslam B, Hasan I, Ullah Z, Roy S, Guo B. Cell membrane-coated biomimetic nanomedicines: productive cancer theranostic tools. Biomater Sci 2024; 12:863-895. [PMID: 38230669 DOI: 10.1039/d3bm01552a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2024]
Abstract
As the second-leading cause of human death, cancer has drawn attention in the area of biomedical research and therapy from all around the world. Certainly, the development of nanotechnology has made it possible for nanoparticles (NPs) to be used as a carrier for delivery systems in the treatment of tumors. This is a biomimetic approach established to craft remedial strategies comprising NPs cloaked with membrane obtained from various natural cells like blood cells, bacterial cells, cancer cells, etc. Here we conduct an in-depth exploration of cell membrane-coated NPs (CMNPs) and their extensive array of applications including drug delivery, vaccination, phototherapy, immunotherapy, MRI imaging, PET imaging, multimodal imaging, gene therapy and a combination of photothermal and chemotherapy. This review article provides a thorough summary of the most recent developments in the use of CMNPs for the diagnosis and treatment of cancer. It critically assesses the state of research while recognizing significant accomplishments and innovations. Additionally, it indicates ongoing problems in clinical translation and associated queries that warrant deeper research. By doing so, this study encourages creative thinking for future projects in the field of tumor therapy using CMNPs while also educating academics on the present status of CMNP research.
Collapse
Affiliation(s)
- Muhammad Ijaz
- School of Science, Shenzhen Key Laboratory of Flexible Printed Electronics Technology, Shenzhen Key Laboratory of Advanced Functional Carbon Materials Research and Comprehensive Application, Harbin Institute of Technology, Shenzhen-518055, China.
- Institute of Microbiology, Government College University Faisalabad Pakistan, Pakistan
| | - Bilal Aslam
- Institute of Microbiology, Government College University Faisalabad Pakistan, Pakistan
| | - Ikram Hasan
- School of Biomedical Engineering, Medical School, Shenzhen University, Shenzhen, Guangdong, 518060, China
| | - Zia Ullah
- School of Science, Shenzhen Key Laboratory of Flexible Printed Electronics Technology, Shenzhen Key Laboratory of Advanced Functional Carbon Materials Research and Comprehensive Application, Harbin Institute of Technology, Shenzhen-518055, China.
| | - Shubham Roy
- School of Science, Shenzhen Key Laboratory of Flexible Printed Electronics Technology, Shenzhen Key Laboratory of Advanced Functional Carbon Materials Research and Comprehensive Application, Harbin Institute of Technology, Shenzhen-518055, China.
| | - Bing Guo
- School of Science, Shenzhen Key Laboratory of Flexible Printed Electronics Technology, Shenzhen Key Laboratory of Advanced Functional Carbon Materials Research and Comprehensive Application, Harbin Institute of Technology, Shenzhen-518055, China.
| |
Collapse
|
23
|
Weaver E, Macartney RA, Irwin R, Uddin S, Hooker A, Burke GA, Wylie MP, Lamprou DA. Liposomal encapsulation of amoxicillin via microfluidics with subsequent investigation of the significance of PEGylated therapeutics. Int J Pharm 2024; 650:123710. [PMID: 38097147 DOI: 10.1016/j.ijpharm.2023.123710] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2023] [Revised: 12/11/2023] [Accepted: 12/12/2023] [Indexed: 12/19/2023]
Abstract
With an increasing concern of global antimicrobial resistance, the efforts to improve the formulation of a narrowing library of therapeutic antibiotics must be confronted. The liposomal encapsulation of antibiotics using a novel and sustainable microfluidic method has been employed in this study to address this pressing issue, via a targeted, lower-dose medical approach. The study focusses upon microfluidic parameter optimisation, formulation stability, cytotoxicity, and future applications. Particle sizes of circa. 130 nm, with viable short-term (28-day) physical stability were obtained, using two different non-cytotoxic liposomal formulations, both of which displayed suitable antibacterial efficacy. The microfluidic method allowed for high encapsulation efficiencies (≈77 %) and the subsequent in vitro release profile suggested high limits of antibiotic dissociation from the nanovessels, achieving 90% release within 72 h. In addition to the experimental data, the growing use of poly(ethylene) glycol (PEG) within lipid-based formulations is discussed in relation to anti-PEG antibodies, highlighting the key pharmacological differences between PEGylated and non-PEGylated formulations and their respective advantages and drawbacks. It's surmised that in the case of the formulations used in this study, the addition of PEG upon the liposomal membrane would still be a beneficial feature to possess owing to beneficial features such as stability, antibiotic efficacy and the capacity to further modify the liposomal membrane.
Collapse
Affiliation(s)
- Edward Weaver
- School of Pharmacy, Queen's University Belfast, 97 Lisburn Road, Belfast BT9 7BL, UK
| | - Robyn A Macartney
- School of Pharmacy, Queen's University Belfast, 97 Lisburn Road, Belfast BT9 7BL, UK; Nanotechnology & Integrated Bioengineering Centre (NIBEC), School of Engineering, Ulster University, York Street, Belfast BT15 1ED, UK
| | - Robyn Irwin
- School of Pharmacy, Queen's University Belfast, 97 Lisburn Road, Belfast BT9 7BL, UK
| | - Shahid Uddin
- Immunocore Ltd, 92 Park Dr, Milton, Abingdon OX14 4RY, UK
| | - Andrew Hooker
- Immunocore Ltd, 92 Park Dr, Milton, Abingdon OX14 4RY, UK
| | - George A Burke
- Nanotechnology & Integrated Bioengineering Centre (NIBEC), School of Engineering, Ulster University, York Street, Belfast BT15 1ED, UK
| | - Matthew P Wylie
- School of Pharmacy, Queen's University Belfast, 97 Lisburn Road, Belfast BT9 7BL, UK
| | - Dimitrios A Lamprou
- School of Pharmacy, Queen's University Belfast, 97 Lisburn Road, Belfast BT9 7BL, UK.
| |
Collapse
|
24
|
Jiang H, Li L, Li Z, Chu X. Metal-based nanoparticles in antibacterial application in biomedical field: Current development and potential mechanisms. Biomed Microdevices 2024; 26:12. [PMID: 38261085 PMCID: PMC10806003 DOI: 10.1007/s10544-023-00686-8] [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] [Accepted: 11/27/2023] [Indexed: 01/24/2024]
Abstract
The rise in drug resistance in pathogenic bacteria greatly endangers public health in the post-antibiotic era, and drug-resistant bacteria currently pose a great challenge not only to the community but also to clinical procedures, including surgery, stent implantation, organ transplantation, and other medical procedures involving any open wound and compromised human immunity. Biofilm-associated drug failure, as well as rapid resistance to last-resort antibiotics, necessitates the search for novel treatments against bacterial infection. In recent years, the flourishing development of nanotechnology has provided new insights for exploiting promising alternative therapeutics for drug-resistant bacteria. Metallic agents have been applied in antibacterial usage for several centuries, and the functional modification of metal-based biomaterials using nanotechnology has now attracted great interest in the antibacterial field, not only for their intrinsic antibacterial nature but also for their ready on-demand functionalization and enhanced interaction with bacteria, rendering them with good potential in further translation. However, the possible toxicity of MNPs to the host cells and tissue still hinders its application, and current knowledge on their interaction with cellular pathways is not enough. This review will focus on recent advances in developing metallic nanoparticles (MNPs), including silver, gold, copper, and other metallic nanoparticles, for antibacterial applications, and their potential mechanisms of interaction with pathogenic bacteria as well as hosts.
Collapse
Affiliation(s)
- Hao Jiang
- Sichuan Provincial Laboratory of Orthopaedic Engineering, Department of Orthopaedics, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, Sichuan, China
| | - Lingzhi Li
- Sichuan Provincial Laboratory of Orthopaedic Engineering, Department of Orthopaedics, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, Sichuan, China
| | - Zhong Li
- Sichuan Provincial Laboratory of Orthopaedic Engineering, Department of Orthopaedics, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, Sichuan, China.
| | - Xiang Chu
- State Key Laboratory of Trauma, Burn and Combined Injury, Department of Emergency, Daping Hospital, Army Medical University, Chongqing, 400042, China
| |
Collapse
|
25
|
Patel M, Andoy NMO, Tran SM, Jeon K, Sullan RMA. Different drug loading methods and antibiotic structure modulate the efficacy of polydopamine nanoparticles as drug nanocarriers. J Mater Chem B 2023; 11:11335-11343. [PMID: 37990852 DOI: 10.1039/d3tb01490h] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2023]
Abstract
The inefficient delivery of antimicrobials to their target is a significant factor contributing to antibiotic resistance. As such, smart nanomaterials that respond to external stimuli are extensively explored for precise drug delivery. Here, we investigate how drug loading methods and the structure of antibiotics impact the effectiveness of photothermally active polydopamine nanoparticles (PDNPs) as a laser-responsive drug delivery system. We examine two loading methods: in-synthesis and post-synthesis, and evaluate how laser irradiation affects drug release. Density functional theory calculations are also performed to gain deeper insights into the drug-PDNP interactions. Our findings point to the critical role of antibiotic structure and drug loading method in the laser-responsive capabilities of PDNPs as drug nanocarriers. Our study offers valuable insights for optimizing the design and efficiency of PDNP-based drug delivery systems.
Collapse
Affiliation(s)
- Meera Patel
- Department of Physical and Environmental Sciences, University of Toronto Scarborough, 1265 Military Trail, Toronto, Ontario, Canada, M1C 1A4
- Department of Chemistry, University of Toronto, 80 St. George St., Toronto, Ontario, Canada, M5S 3H6.
| | - Nesha May O Andoy
- Department of Physical and Environmental Sciences, University of Toronto Scarborough, 1265 Military Trail, Toronto, Ontario, Canada, M1C 1A4
| | - Susannah Megan Tran
- Department of Physical and Environmental Sciences, University of Toronto Scarborough, 1265 Military Trail, Toronto, Ontario, Canada, M1C 1A4
| | - Keuna Jeon
- Department of Physical and Environmental Sciences, University of Toronto Scarborough, 1265 Military Trail, Toronto, Ontario, Canada, M1C 1A4
- Department of Chemistry, University of Toronto, 80 St. George St., Toronto, Ontario, Canada, M5S 3H6.
| | - Ruby May A Sullan
- Department of Physical and Environmental Sciences, University of Toronto Scarborough, 1265 Military Trail, Toronto, Ontario, Canada, M1C 1A4
- Department of Chemistry, University of Toronto, 80 St. George St., Toronto, Ontario, Canada, M5S 3H6.
| |
Collapse
|
26
|
Wu Z, Nie R, Wang Y, Wang Q, Li X, Liu Y. Precise antibacterial therapeutics based on stimuli-responsive nanomaterials. Front Bioeng Biotechnol 2023; 11:1289323. [PMID: 37920242 PMCID: PMC10619694 DOI: 10.3389/fbioe.2023.1289323] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Accepted: 10/09/2023] [Indexed: 11/04/2023] Open
Abstract
Bacterial infection refers to the process in which bacteria invade, grow, reproduce, and interact with the body, ultimately causing a series of pathological changes. Nowadays, bacterial infection remains a significant public health issue, posing a huge threat to human health and a serious financial burden. In the post-antibiotic era, traditional antibiotics are prone to inducing bacterial resistance and difficulty in removing bacterial biofilm. In recent years, antibacterial therapy based on nanomaterials has developed rapidly. Compared with traditional antibiotics, nanomaterials effectively remove bacterial biofilms and rarely result in bacterial resistance. However, due to nanomaterials' strong permeability and effectiveness, they will easily cause cytotoxicity when they are not controlled. In addition, the antibacterial effect of non-responsive nanomaterials cannot be perfectly exerted since the drug release property or other antibacterial effects of these nano-materials are not be positively correlated with the intensity of bacterial infection. Stimuli-responsive antibacterial nanomaterials are a more advanced and intelligent class of nano drugs, which are controlled by exogenous stimuli and microenvironmental stimuli to change the dosage and intensity of treatment. The excellent spatiotemporal controllability enables stimuli-responsive nanomaterials to treat bacterial infections precisely. In this review, we first elaborate on the design principles of various stimuli-responsive antibacterial nanomaterials. Then, we analyze and summarizes the antibacterial properties, advantages and shortcomings of different applied anti-bacterial strategies based on stimuli-responsive nanomaterials. Finally, we propose the challenges of employing stimuli-responsive nanomaterials and corresponding potential solutions.
Collapse
Affiliation(s)
| | | | | | | | | | - Yuguang Liu
- Department of Stomatology, The First Hospital of Jilin University, Changchun, China
| |
Collapse
|
27
|
Mkangara M. Prevention and Control of Human Salmonella enterica Infections: An Implication in Food Safety. INTERNATIONAL JOURNAL OF FOOD SCIENCE 2023; 2023:8899596. [PMID: 37727836 PMCID: PMC10506869 DOI: 10.1155/2023/8899596] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/10/2022] [Revised: 08/19/2023] [Accepted: 08/26/2023] [Indexed: 09/21/2023]
Abstract
Salmonella is a foodborne zoonotic pathogen causing diarrhoeal disease to humans after consuming contaminated water, animal, and plant products. The bacterium is the third leading cause of human death among diarrhoeal diseases worldwide. Therefore, human salmonellosis is of public health concern demanding integrated interventions against the causative agent, Salmonella enterica. The prevention of salmonellosis in humans is intricate due to several factors, including an immune-stable individual infected with S. enterica continuing to shed live bacteria without showing any clinical signs. Similarly, the asymptomatic Salmonella animals are the source of salmonellosis in humans after consuming contaminated food products. Furthermore, the contaminated products of plant and animal origin are a menace in food industries due to Salmonella biofilms, which enhance colonization, persistence, and survival of bacteria on equipment. The contaminated food products resulting from bacteria on equipment offset the economic competition of food industries and partner institutions in international business. The most worldwide prevalent broad-range Salmonella serovars affecting humans are Salmonella Typhimurium and Salmonella Enteritidis, and poultry products, among others, are the primary source of infection. The broader range of Salmonella serovars creates concern over multiple strategies for preventing and controlling Salmonella contamination in foods to enhance food safety for humans. Among the strategies for preventing and controlling Salmonella spread in animal and plant products include biosecurity measures, isolation and quarantine, epidemiological surveillance, farming systems, herbs and spices, and vaccination. Other measures are the application of phages, probiotics, prebiotics, and nanoparticles reduced and capped with antimicrobial agents. Therefore, Salmonella-free products, such as beef, pork, poultry meat, eggs, milk, and plant foods, such as vegetables and fruits, will prevent humans from Salmonella infection. This review explains Salmonella infection in humans caused by consuming contaminated foods and the interventions against Salmonella contamination in foods to enhance food safety and quality for humans.
Collapse
Affiliation(s)
- Mwanaisha Mkangara
- Department of Science and Laboratory Technology, Dar es Salaam Institute of Technology, P.O. Box 2958, Dar es Salaam, Tanzania
| |
Collapse
|
28
|
El-Sayed NS, Hashem AH, Khattab TA, Kamel S. New antibacterial hydrogels based on sodium alginate. Int J Biol Macromol 2023; 248:125872. [PMID: 37482158 DOI: 10.1016/j.ijbiomac.2023.125872] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2022] [Revised: 07/12/2023] [Accepted: 07/15/2023] [Indexed: 07/25/2023]
Abstract
Nowadays, the combined knowledge and experience in biomedical research and material sciences results in the innovation of smart materials that could efficiently overcome the problems of microbial contaminations. Herein, a new drug delivery platform prepared by grafting sodium alginate with β-carboxyethyl acrylate and acrylamide was described and characterized. 9-Aminoacridine (9-AA), and kanamycin sulfate (KS) were separately loaded into the hydrogel in situ during graft polymerization. The grafting efficiency for the resulting hydrogels was 70.01-78.08 %. The chemical structure of the hydrogels, thermogravimetric analysis, and morphological features were investigated. The swelling study revealed that the hydrogel without drugs achieved a superior swelling rate compared to drug-loaded hydrogels. The hydrogel tuned the drug-release rate in a pH-dependent manner. Furthermore, the antibacterial study suggested that the hydrogels encapsulating 9-AA (88.6 %) or KS (89.3 %) exhibited comparable antibacterial activity against Gram-positive and Gram-negative bacterial strains. Finally, the cytocompatibility study conducted on normal lung cell line (Vero cells) demonstrated neglectable to tolerable toxicity for the drug-loaded hydrogel. More interestingly, the cell viability for the blank hydrogel was 92.5 %, implying its suitability for biomedical applications.
Collapse
Affiliation(s)
| | - Amr H Hashem
- Botany and Microbiology Department, Faculty of Science, Al-Azhar University, Cairo 11884, Egypt
| | - Tawfik A Khattab
- Dyeing, Printing and Auxiliaries Department, National Research Centre, Cairo, P.O. 12622, Egypt
| | - Samir Kamel
- Cellulose and Paper Department, National Research Centre, Cairo, P.O. 12622, Egypt
| |
Collapse
|
29
|
Kauser A, Parisini E, Suarato G, Castagna R. Light-Based Anti-Biofilm and Antibacterial Strategies. Pharmaceutics 2023; 15:2106. [PMID: 37631320 PMCID: PMC10457815 DOI: 10.3390/pharmaceutics15082106] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 07/29/2023] [Accepted: 08/02/2023] [Indexed: 08/27/2023] Open
Abstract
Biofilm formation and antimicrobial resistance pose significant challenges not only in clinical settings (i.e., implant-associated infections, endocarditis, and urinary tract infections) but also in industrial settings and in the environment, where the spreading of antibiotic-resistant bacteria is on the rise. Indeed, developing effective strategies to prevent biofilm formation and treat infections will be one of the major global challenges in the next few years. As traditional pharmacological treatments are becoming inadequate to curb this problem, a constant commitment to the exploration of novel therapeutic strategies is necessary. Light-triggered therapies have emerged as promising alternatives to traditional approaches due to their non-invasive nature, precise spatial and temporal control, and potential multifunctional properties. Here, we provide a comprehensive overview of the different biofilm formation stages and the molecular mechanism of biofilm disruption, with a major focus on the quorum sensing machinery. Moreover, we highlight the principal guidelines for the development of light-responsive materials and photosensitive compounds. The synergistic effects of combining light-triggered therapies with conventional treatments are also discussed. Through elegant molecular and material design solutions, remarkable results have been achieved in the fight against biofilm formation and antibacterial resistance. However, further research and development in this field are essential to optimize therapeutic strategies and translate them into clinical and industrial applications, ultimately addressing the global challenges posed by biofilm and antimicrobial resistance.
Collapse
Affiliation(s)
- Ambreen Kauser
- Department of Biotechnology, Latvian Institute of Organic Synthesis, Aizkraukles 21, LV-1006 Riga, Latvia; (A.K.); (E.P.)
- Faculty of Materials Science and Applied Chemistry, Riga Technical University, Paula Valdena 3, LV-1048 Riga, Latvia
| | - Emilio Parisini
- Department of Biotechnology, Latvian Institute of Organic Synthesis, Aizkraukles 21, LV-1006 Riga, Latvia; (A.K.); (E.P.)
- Department of Chemistry “G. Ciamician”, University of Bologna, Via Selmi 2, 40126 Bologna, Italy
| | - Giulia Suarato
- Istituto di Elettronica e di Ingegneria dell’Informazione e delle Telecomunicazioni, Consiglio Nazionale delle Ricerche, CNR-IEIIT, Piazza Leonardo da Vinci 32, 20133 Milano, Italy
| | - Rossella Castagna
- Department of Biotechnology, Latvian Institute of Organic Synthesis, Aizkraukles 21, LV-1006 Riga, Latvia; (A.K.); (E.P.)
- Dipartimento di Chimica, Materiali e Ingegneria Chimica “G. Natta”, Politecnico di Milano, Piazza Leonardo da Vinci 32, 20133 Milano, Italy
| |
Collapse
|
30
|
Santonoceta GDG, Sgarlata C. pH-Responsive Cobalt(II)-Coordinated Assembly Containing Quercetin for Antimicrobial Applications. Molecules 2023; 28:5581. [PMID: 37513453 PMCID: PMC10386366 DOI: 10.3390/molecules28145581] [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: 06/28/2023] [Revised: 07/17/2023] [Accepted: 07/18/2023] [Indexed: 07/30/2023] Open
Abstract
The development of novel drug delivery systems (DDSs) with promising antibacterial properties is essential for facing the emergency of increasing resistance to antimicrobial agents. The antibacterial features of quercetin and its metal complexes have been broadly investigated. However, several drawbacks affect their activity and effectiveness. In this work, we propose a DDS based on a pH-responsive cobalt(II)-coordinated assembly containing quercetin and polyacrylic acid. This system is suggested to trigger the release of the model drug in a pH-dependent mode by exploiting the localized acidic environment at the bacterial infection sites under anaerobic conditions. The delivery system has been designed by accurately examining the species and the multiple equilibria occurring in solution among the assembly components. The formation of cobalt(II) complexes with quercetin in the absence or presence of the pH-responsive polyacrylic acid was investigated in buffered aqueous solution at pH 7.4 using spectrophotometric (UV-Vis) and calorimetric (ITC) techniques. The determined binding affinities and thermodynamic parameters that resulted are essential for the development of a DDS with improved binding and release capabilities. Furthermore, the affinity of the polymer-cobalt(II) complex toward the model antimicrobial flavonoid was explored at the solid-liquid interface by quartz crystal microbalance (QCM-D) experiments, which provided marked evidence for drug loading and release under pH control.
Collapse
Affiliation(s)
| | - Carmelo Sgarlata
- Department of Chemical Sciences, University of Catania, Viale Andrea Doria 6, 95125 Catania, Italy
| |
Collapse
|
31
|
Serpico L, Dello Iacono S, Cammarano A, De Stefano L. Recent Advances in Stimuli-Responsive Hydrogel-Based Wound Dressing. Gels 2023; 9:451. [PMID: 37367122 DOI: 10.3390/gels9060451] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Revised: 05/24/2023] [Accepted: 05/28/2023] [Indexed: 06/28/2023] Open
Abstract
Polymeric materials have found increasing use in biomedical applications in the last decades. Among them, hydrogels represent the chosen class of materials to use in this field, in particular as wound dressings. They are generally non-toxic, biocompatible, and biodegradable, and they can absorb large amounts of exudates. Moreover, hydrogels actively contribute to skin repair promoting fibroblast proliferation and keratinocyte migration, allowing oxygen to permeate, and protecting wounds from microbial invasion. As wound dressing, stimuli-responsive systems are particularly advantageous since they can be active only in response to specific environmental stimuli (such as pH, light, ROS concentration, temperature, and glucose level). In this review, we briefly resume the human skin's structure and functions, as well as the wound healing phases; then, we present recent advances in stimuli-responsive hydrogels-based wound dressings. Lastly, we provide a bibliometric analysis of knowledge produced in the field.
Collapse
Affiliation(s)
- Luigia Serpico
- Institute of Applied Sciences and Intelligent Systems (ISASI), National Research Council, Via P. Castellino 111, 80131 Naples, Italy
- Materias Srl, Corso N. Protopisani 50, 80146 Naples, Italy
| | - Stefania Dello Iacono
- Institute of Polymers, Composites and Biomaterials (IPCB), National Research Council, P.le E. Fermi 1, 80055 Portici, Italy
| | | | - Luca De Stefano
- Institute of Applied Sciences and Intelligent Systems (ISASI), National Research Council, Via P. Castellino 111, 80131 Naples, Italy
| |
Collapse
|
32
|
Shao M, Bigham A, Yousefiasl S, Yiu CKY, Girish YR, Ghomi M, Sharifi E, Sezen S, Nazarzadeh Zare E, Zarrabi A, Rabiee N, Paiva-Santos AC, Del Turco S, Guo B, Wang X, Mattoli V, Wu A. Recapitulating Antioxidant and Antibacterial Compounds into a Package for Tissue Regeneration: Dual Function Materials with Synergistic Effect. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2023; 19:e2207057. [PMID: 36775954 DOI: 10.1002/smll.202207057] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/13/2022] [Revised: 01/18/2023] [Indexed: 05/11/2023]
Abstract
Oxidative damage and infection can prevent or delay tissue repair. Moreover, infection reinforces reactive oxygen species (ROS) formation, which makes the wound's condition even worse. Therefore, the need for antioxidant and antibacterial agents is felt for tissue regeneration. There are emerging up-and-coming biomaterials that recapitulate both properties into a package, offering an effective solution to turn the wound back into a healing state. In this article, the principles of antioxidant and antibacterial activity are summarized. The review starts with biological aspects, getting the readers to familiarize themselves with tissue barriers against infection. This is followed by the chemistry and mechanism of action of antioxidant and antibacterial materials (dual function). Eventually, the outlook and challenges are underlined to provide where the dual-function biomaterials are and where they are going in the future. It is expected that the present article inspires the designing of dual-function biomaterials to more advanced levels by providing the fundamentals and comparative points of view and paving the clinical way for these materials.
Collapse
Affiliation(s)
- Minmin Shao
- Department of Otorhinolaryngology, The Second Affiliated Hospital of Shanghai University, Wenzhou Central Hospital, Wenzhou, 325000, P. R. China
| | - Ashkan Bigham
- Institute of Polymers, Composites and Biomaterials, National Research Council (IPCB-CNR), 80125, Naples, Italy
| | - Satar Yousefiasl
- School of Dentistry, Hamadan University of Medical Sciences, Hamadan, 6517838736, Iran
| | - Cynthia K Y Yiu
- Paediatric Dentistry and Orthodontics, Faculty of Dentistry, The University of Hong Kong, Prince Philip Dental Hospital, Hong Kong, 999077, P. R. China
| | - Yarabahally R Girish
- Centre for Research and Innovations, School of Natural Sciences, BGSIT, Adichunchanagiri University, B.G. Nagara, Mandya District, Mandya, Karnataka, 571448, India
| | - Matineh Ghomi
- School of Chemistry, Damghan University, Damghan, 36716-45667, Iran
| | - Esmaeel Sharifi
- Department of Tissue Engineering and Biomaterials, School of Advanced Medical Sciences and Technologies, Hamadan University of Medical Sciences, Hamadan, 6517838736, Iran
| | - Serap Sezen
- Faculty of Engineering and Natural Sciences, Sabanci University, Orta Mahalle, Üniversite Caddesi No. 27, Orhanlı, Tuzla, Istanbul, 34956, Turkey
| | | | - Ali Zarrabi
- Department of Biomedical Engineering, Faculty of Engineering and Natural Sciences, Istinye University, Sariyer, Istanbul, 34396, Turkey
| | - Navid Rabiee
- Centre for Molecular Medicine and Innovative Therapeutics, Murdoch University, Perth, Western Australia, 6150, Australia
- School of Engineering, Macquarie University, Sydney, New South Wales, 2109, Australia
| | - Ana Cláudia Paiva-Santos
- Department of Pharmaceutical Technology, Faculty of Pharmacy of the University of Coimbra, University of Coimbra, 3000-548, Coimbra, Portugal
- REQUIMTE/LAQV, Group of Pharmaceutical Technology, Faculty of Pharmacy of the University of Coimbra, University of Coimbra, 3000-548, Coimbra, Portugal
| | - Serena Del Turco
- National Research Council, Institute of Clinical Physiology, 56124, Pisa, Italy
- Istituto Italiano di Tecnologia, Centre for Materials Interfaces, 56025, Pontedera, Pisa, Italy
| | - Baolin Guo
- State Key Laboratory for Mechanical Behavior of Materials, and Frontier Institute of Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, P. R. China
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University, Xi'an, 710049, P. R. China
| | - Xiangdong Wang
- Department of Pulmonary and Critical Care Medicine, Zhongshan Hospital, Fudan University Shanghai Medical College, Shanghai, 200032, P. R. China
| | - Virgilio Mattoli
- Istituto Italiano di Tecnologia, Centre for Materials Interfaces, 56025, Pontedera, Pisa, Italy
| | - Aimin Wu
- Department of Orthopaedics, Key Laboratory of Structural Malformations in Children of Zhejiang Province, Key Laboratory of Orthopaedics of Zhejiang Province, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, 325000, P. R. China
| |
Collapse
|
33
|
Abbasian M, Khayyatalimohammadi M. Ultrasound-assisted synthesis of MIL-88(Fe) conjugated starch-Fe 3O 4 nanocomposite: A safe antibacterial carrier for controlled release of tetracycline. Int J Biol Macromol 2023; 234:123665. [PMID: 36791936 DOI: 10.1016/j.ijbiomac.2023.123665] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Revised: 01/30/2023] [Accepted: 02/09/2023] [Indexed: 02/15/2023]
Abstract
A constructing antibiotic carrier with a sustained release profile is a promising method to stop long-term bacterial infection, which is of ideal interest in different biomedical fields. To end this, the present study aims to design a novel carrier based on the modification of biopolymeric starch for the rising possible interaction between carrier and antibiotic agent. We established an in-situ ultrasound-assisted method was applied to grow and create MIL-88(Fe) framework in the structure of magnetic polysaccharide (i.e., St/Fe3O4) synthesized by precipitation method resulting in St/Fe3O4/MIL-88(Fe) nanocomposite. It was loaded with a high amount of Tetracycline (TC) through its immersion into the TC aqueous solution. The release profile of TC-loaded St/Fe3O4/MIL-88(Fe) displays a lower initial burst release (about 26 % after 12 h) and followed by a controlled and sustained release (about 73 % up to 168 h) in the simulated physiological environment at pH 7.4. The in vitro cytotoxicity showed good cytocompatibility against Human skin fibroblast (HFF-1) cells. TC-loaded St/Fe3O4/MIL-88(Fe) showed higher antibacterial activity against both S. aureus and E. coli with the MIC value of 64 and 128 μg·mL-1, respectively.
Collapse
Affiliation(s)
- Mojtaba Abbasian
- Department of Chemical Engineering, Faculty of Engineering, University of Bonab, Bonab, Iran.
| | | |
Collapse
|
34
|
Ameh T, Zarzosa K, Dickinson J, Braswell WE, Sayes CM. Nanoparticle surface stabilizing agents influence antibacterial action. Front Microbiol 2023; 14:1119550. [PMID: 36846763 PMCID: PMC9947285 DOI: 10.3389/fmicb.2023.1119550] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Accepted: 01/24/2023] [Indexed: 02/11/2023] Open
Abstract
The antibacterial properties of nanoparticles are of particular interest because of their potential to serve as an alternative therapy to combat antimicrobial resistance. Metal nanoparticles such as silver and copper nanoparticles have been investigated for their antibacterial properties. Silver and copper nanoparticles were synthesized with the surface stabilizing agents cetyltrimethylammonium bromide (CTAB, to confer a positive surface charge) and polyvinyl pyrrolidone (PVP, to confer a neutral surface charge). Minimum inhibitory concentration (MIC), minimum bactericidal concentration (MBC), and viable plate count assays were used to determine effective doses of silver and copper nanoparticles treatment against Escherichia coli, Staphylococcus aureus and Sphingobacterium multivorum. Results show that CTAB stabilized silver and copper nanoparticles were more effective antibacterial agents than PVP stabilized metal nanoparticles, with MIC values in a range of 0.003 μM to 0.25 μM for CTAB stabilized metal nanoparticles and 0.25 μM to 2 μM for PVP stabilized metal nanoparticles. The recorded MIC and MBC values of the surface stabilized metal nanoparticles show that they can serve as effective antibacterial agents at low doses.
Collapse
Affiliation(s)
- Thelma Ameh
- Department of Environmental Science, Baylor University, Waco, TX, United States
| | - Kusy Zarzosa
- Department of Environmental Science, Baylor University, Waco, TX, United States,United States Department of Agriculture, Animal and Plant Health Inspection Services, Plant Protection and Quarantine, Science and Technology, Insect Management and Molecular Diagnostics Laboratory, Edinburg, TX, United States
| | - Jake Dickinson
- Department of Environmental Science, Baylor University, Waco, TX, United States
| | - W. Evan Braswell
- United States Department of Agriculture, Animal and Plant Health Inspection Services, Plant Protection and Quarantine, Science and Technology, Insect Management and Molecular Diagnostics Laboratory, Edinburg, TX, United States
| | - Christie M. Sayes
- Department of Environmental Science, Baylor University, Waco, TX, United States,*Correspondence: Christie M. Sayes, ✉
| |
Collapse
|
35
|
Yousefi M, Andishmand H, Assadpour E, Barzegar A, Kharazmi MS, Jafari SM. Nanoliposomal delivery systems of natural antibacterial compounds; properties, applications, and recent advances. Crit Rev Food Sci Nutr 2023; 64:6498-6511. [PMID: 36728840 DOI: 10.1080/10408398.2023.2170318] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Todays, nanoliposomes (NLPs) are considered as one of the most efficient nanocarriers to deal with bacteria, practically in food products. These nanodelivery systems are able to be loaded with different bioactive compounds. The main aim of this review is investigating recent approaches (mostly from the years of 2018 to 2022) regarding development of nanoliposomal natural antibacterial compounds. In this regard, NLPs alone, combined with films, coatings, or fibers, and in coated forms are reviewed as advanced delivery systems of antibacterial substances. Moreover, a robust and comprehensive coverage of the morphological and physical properties of formulated NLPs as well as their interactions with antibacterial substances are discussed. The importance of NLPs to encapsulate antibacterial ingredients, advantages and drawbacks, antibacterial pathways of formulated NLPs, and comparison of them with pure antibacterial bioactive compounds are also explained.
Collapse
Affiliation(s)
- Mohammad Yousefi
- Food and Beverage Safety Research Center, Urmia University of Medical Sciences, Urmia, Iran
| | - Hashem Andishmand
- Department of Food Science and Technology, Faculty of Nutrition and Food Science, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Elham Assadpour
- Food Industry Research Co, Gorgan, Iran
- Food and Bio-Nanotech International Research Center (Fabiano), Gorgan University of Agricultural Sciences and Natural Resources, Gorgan, Iran
| | - Ali Barzegar
- Department of Community Nutrition, Faculty of Nutrition and Food Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Seid Mahdi Jafari
- Department of Food Materials and Process Design Engineering, Gorgan University of Agricultural Sciences and Natural Resources, Gorgan, Iran
- Department of Analytical Chemistry and Food Science, Faculty of Science, Universidade De Vigo, Nutrition and Bromatology Group, Ourense, Spain
- College Of Food Science and Technology, Hebei Agricultural University, Baoding, China
| |
Collapse
|
36
|
Geng Z, Cao Z, Liu J. Recent advances in targeted antibacterial therapy basing on nanomaterials. EXPLORATION (BEIJING, CHINA) 2023; 3:20210117. [PMID: 37323620 PMCID: PMC10191045 DOI: 10.1002/exp.20210117] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Accepted: 05/19/2022] [Indexed: 06/17/2023]
Abstract
Bacterial infection has become one of the leading causes of death worldwide, particularly in low-income countries. Despite the fact that antibiotics have provided successful management in bacterial infections, the long-term overconsumption and abuse of antibiotics has contributed to the emergence of multidrug resistant bacteria. To address this challenge, nanomaterials with intrinsic antibacterial properties or that serve as drug carriers have been substantially developed as an alternative to fight against bacterial infection. Systematically and deeply understanding the antibacterial mechanisms of nanomaterials is extremely important for designing new therapeutics. Recently, nanomaterials-mediated targeted bacteria depletion in either a passive or active manner is one of the most promising approaches for antibacterial treatment by increasing local concentration around bacterial cells to enhance inhibitory activity and reduce side effects. Passive targeting approach is widely explored by searching nanomaterial-based alternatives to antibiotics, while active targeting strategy relies on biomimetic or biomolecular surface feature that can selectively recognize targeted bacteria. In this review article, we summarize the recent developments in the field of targeted antibacterial therapy based on nanomaterials, which will promote more innovative thinking focusing on the treatment of multidrug-resistant bacteria.
Collapse
Affiliation(s)
- Zhongmin Geng
- Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine, Institute of Molecular Medicine, State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, School of MedicineShanghai Jiao Tong UniversityShanghaiChina
- The Affiliated Hospital of Qingdao UniversityQingdao UniversityQingdaoChina
- Qingdao Cancer InstituteQingdao UniversityQingdaoChina
| | - Zhenping Cao
- Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine, Institute of Molecular Medicine, State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, School of MedicineShanghai Jiao Tong UniversityShanghaiChina
| | - Jinyao Liu
- Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine, Institute of Molecular Medicine, State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, School of MedicineShanghai Jiao Tong UniversityShanghaiChina
| |
Collapse
|
37
|
Sousa A, Phung AN, Škalko-Basnet N, Obuobi S. Smart delivery systems for microbial biofilm therapy: Dissecting design, drug release and toxicological features. J Control Release 2023; 354:394-416. [PMID: 36638844 DOI: 10.1016/j.jconrel.2023.01.003] [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: 09/01/2022] [Revised: 11/14/2022] [Accepted: 01/02/2023] [Indexed: 01/15/2023]
Abstract
Bacterial biofilms are highly protected surface attached communities of bacteria that typically cause chronic infections. To address their recalcitrance to antibiotics and minimise side effects of current therapies, smart drug carriers are being explored as promising platforms for antimicrobials. Herein, we briefly summarize recent efforts and considerations that have been applied in the design of these smart carriers. We guide readers on a journey on how they can leverage the inherent biofilm microenvironment, external stimuli, or combine both types of stimuli in a predictable manner. The specific carrier features that are responsible for their 'on-demand' properties are detailed and their impact on antibiofilm property are further discussed. Moreover, an analysis on the impact of such features on drug release profiles is provided. Since nanotechnology represents a significant slice of the drug delivery pie, some insights on the potential toxicity are also depicted. We hope that this review inspires researchers to use their knowledge and creativity to design responsive systems that can eradicate biofilm infections.
Collapse
Affiliation(s)
- A Sousa
- Drug Transport and Delivery Research Group, Department of Pharmacy, UIT The Arctic University of Norway, Tromsø, Norway
| | - A Ngoc Phung
- Drug Transport and Delivery Research Group, Department of Pharmacy, UIT The Arctic University of Norway, Tromsø, Norway
| | - N Škalko-Basnet
- Drug Transport and Delivery Research Group, Department of Pharmacy, UIT The Arctic University of Norway, Tromsø, Norway
| | - S Obuobi
- Drug Transport and Delivery Research Group, Department of Pharmacy, UIT The Arctic University of Norway, Tromsø, Norway.
| |
Collapse
|
38
|
van Gent ME, van Baaren T, Kłodzińska SN, Ali M, Dolezal N, van Doodewaerd BR, Bos E, de Waal AM, Koning RI, Drijfhout JW, Nielsen HM, Nibbering PH. Encapsulation of SAAP-148 in Octenyl Succinic Anhydride-Modified Hyaluronic Acid Nanogels for Treatment of Skin Wound Infections. Pharmaceutics 2023; 15:pharmaceutics15020429. [PMID: 36839751 PMCID: PMC9967827 DOI: 10.3390/pharmaceutics15020429] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2022] [Revised: 01/23/2023] [Accepted: 01/26/2023] [Indexed: 01/31/2023] Open
Abstract
Chronic wound infections colonized by bacteria are becoming more difficult to treat with current antibiotics due to the development of antimicrobial resistance (AMR) as well as biofilm and persister cell formation. Synthetic antibacterial and antibiofilm peptide (SAAP)-148 is an excellent alternative for treatment of such infections but suffers from limitations related to its cationic peptidic nature and thus instability and possible cytotoxicity, resulting in a narrow therapeutic window. Here, we evaluated SAAP-148 encapsulation in nanogels composed of octenyl succinic anhydride (OSA)-modified hyaluronic acid (HA) to circumvent these limitations. SAAP-148 was efficiently (>98%) encapsulated with high drug loading (23%), resulting in monodispersed anionic OSA-HA nanogels with sizes ranging 204-253 nm. Nanogel lyophilization in presence of polyvinyl alcohol maintained their sizes and morphology. SAAP-148 was sustainedly released from lyophilized nanogels (37-41% in 72 h) upon reconstitution. Lyophilized SAAP-148-loaded nanogels showed similar antimicrobial activity as SAAP-148 against planktonic and biofilm-residing AMR Staphylococcus aureus and Acinetobacter baumannii. Importantly, formulated SAAP-148 showed reduced cytotoxicity against human erythrocytes, primary human skin fibroblasts and human keratinocytes. Additionally, lyophilized SAAP-148-loaded nanogels eradicated AMR S. aureus and A. baumannii colonizing a 3D human epidermal model, without inducing any cytotoxicity in contrast to SAAP-148. These findings indicate that OSA-HA nanogels increase SAAP-148's therapeutic potential for treatment of skin wound infections.
Collapse
Affiliation(s)
- Miriam E. van Gent
- Department of Infectious Diseases, Leiden University Medical Center, 2300 RC Leiden, The Netherlands
- Correspondence:
| | - Tom van Baaren
- Department of Infectious Diseases, Leiden University Medical Center, 2300 RC Leiden, The Netherlands
| | - Sylvia N. Kłodzińska
- Center for Biopharmaceuticals and Biobarriers in Drug Delivery (BioDelivery), Department of Pharmacy, Faculty of Health and Medical Sciences, University of Copenhagen, DK-2100 Copenhagen, Denmark
| | - Muhanad Ali
- Department of Infectious Diseases, Leiden University Medical Center, 2300 RC Leiden, The Netherlands
| | - Natasja Dolezal
- Department of Immunology, Leiden University Medical Center, 2300 RC Leiden, The Netherlands
| | - Bjorn R. van Doodewaerd
- Department of Cell and Chemical Biology, Leiden University Medical Center, 2300 RC Leiden, The Netherlands
| | - Erik Bos
- Electron Microscopy Facility, Department of Cell and Chemical Biology, Leiden University Medical Center, 2300 RC Leiden, The Netherlands
| | - Amy M. de Waal
- Department of Infectious Diseases, Leiden University Medical Center, 2300 RC Leiden, The Netherlands
| | - Roman I. Koning
- Electron Microscopy Facility, Department of Cell and Chemical Biology, Leiden University Medical Center, 2300 RC Leiden, The Netherlands
| | - Jan Wouter Drijfhout
- Department of Immunology, Leiden University Medical Center, 2300 RC Leiden, The Netherlands
| | - Hanne Mørck Nielsen
- Center for Biopharmaceuticals and Biobarriers in Drug Delivery (BioDelivery), Department of Pharmacy, Faculty of Health and Medical Sciences, University of Copenhagen, DK-2100 Copenhagen, Denmark
| | - Peter H. Nibbering
- Department of Infectious Diseases, Leiden University Medical Center, 2300 RC Leiden, The Netherlands
| |
Collapse
|
39
|
Song Y, Zheng X, Hu J, Ma S, Li K, Chen J, Xu X, Lu X, Wang X. Recent advances of cell membrane-coated nanoparticles for therapy of bacterial infection. Front Microbiol 2023; 14:1083007. [PMID: 36876074 PMCID: PMC9981803 DOI: 10.3389/fmicb.2023.1083007] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Accepted: 02/01/2023] [Indexed: 02/19/2023] Open
Abstract
The rapid evolution of antibiotic resistance and the complicated bacterial infection microenvironments are serious obstacles to traditional antibiotic therapy. Developing novel antibacterial agents or strategy to prevent the occurrence of antibiotic resistance and enhance antibacterial efficiency is of the utmost importance. Cell membrane-coated nanoparticles (CM-NPs) combine the characteristics of the naturally occurring membranes with those of the synthetic core materials. CM-NPs have shown considerable promise in neutralizing toxins, evading clearance by the immune system, targeting specific bacteria, delivering antibiotics, achieving responsive antibiotic released to the microenvironments, and eradicating biofilms. Additionally, CM-NPs can be utilized in conjunction with photodynamic, sonodynamic, and photothermal therapies. In this review, the process for preparing CM-NPs is briefly described. We focus on the functions and the recent advances in applications of several types of CM-NPs in bacterial infection, including CM-NPs derived from red blood cells, white blood cells, platelet, bacteria. CM-NPs derived from other cells, such as dendritic cells, genetically engineered cells, gastric epithelial cells and plant-derived extracellular vesicles are introduced as well. Finally, we place a novel perspective on CM-NPs' applications in bacterial infection, and list the challenges encountered in this field from the preparation and application standpoint. We believe that advances in this technology will reduce threats posed by bacteria resistance and save lives from infectious diseases in the future.
Collapse
Affiliation(s)
- Yue Song
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Hangzhou, China
| | - Xia Zheng
- The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Juan Hu
- The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Subo Ma
- The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Kun Li
- The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Junyao Chen
- Shulan International Medical College, Zhejiang Shuren University, Hangzhou, China
| | - Xiaoling Xu
- Shulan International Medical College, Zhejiang Shuren University, Hangzhou, China
| | - Xiaoyang Lu
- The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Xiaojuan Wang
- The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| |
Collapse
|
40
|
Wang C, Xu P, Li X, Zheng Y, Song Z. Research progress of stimulus-responsive antibacterial materials for bone infection. Front Bioeng Biotechnol 2022; 10:1069932. [PMID: 36636700 PMCID: PMC9831006 DOI: 10.3389/fbioe.2022.1069932] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Accepted: 12/09/2022] [Indexed: 12/24/2022] Open
Abstract
Infection is one of the most serious complications harmful to human health, which brings a huge burden to human health. Bone infection is one of the most common and serious complications of fracture and orthopaedic surgery. Antibacterial treatment is the premise of bone defect healing. Among all the antibacterial strategies, irritant antibacterial materials have unique advantages and the ability of targeted therapy. In this review, we focus on the research progress of irritating materials, the development of antibacterial materials and their advantages and disadvantages potential applications in bone infection.
Collapse
Affiliation(s)
| | | | | | - Yuhao Zheng
- Department of Sports Medicine, Orthopaedic Center, The First Hospital of Jilin University, Changchun, China
| | - Zhiming Song
- Department of Sports Medicine, Orthopaedic Center, The First Hospital of Jilin University, Changchun, China
| |
Collapse
|
41
|
Aliakbar Ahovan Z, Esmaeili Z, Eftekhari BS, Khosravimelal S, Alehosseini M, Orive G, Dolatshahi-Pirouz A, Pal Singh Chauhan N, Janmey PA, Hashemi A, Kundu SC, Gholipourmalekabadi M. Antibacterial smart hydrogels: New hope for infectious wound management. Mater Today Bio 2022; 17:100499. [PMID: 36466959 PMCID: PMC9709163 DOI: 10.1016/j.mtbio.2022.100499] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Revised: 10/31/2022] [Accepted: 11/18/2022] [Indexed: 11/21/2022] Open
Abstract
Millions of people die annually due to uncured wound infections. Healthcare systems incur high costs to treat wound infections. Tt is predicted to become more challenging due to the rise of multidrug-resistant conditions. During the last decades, smart antibacterial hydrogels could attract attention as a promising solution, especially for skin wound infections. These antibacterial hydrogels are termed 'smart' due to their response to specific physical and chemical environmental stimuli. To deliver different drugs to particular sites in a controlled manner, various types of crosslinking strategies are used in the manufacturing process. Smart hydrogels are designed to provide antimicrobial agents to the infected sites or are built from polymers with inherent disinfectant properties. This paper aims to critically review recent pre-clinical and clinical advances in using smart hydrogels against skin wound infections and propose the next best thing for future trends. For this purpose, an introduction to skin wound healing and disease is presented and intelligent hydrogels responding to different stimuli are introduced. Finally, the most promising investigations are discussed in their related sections. These studies can pave the way for producing new biomaterials with clinical applications.
Collapse
Affiliation(s)
- Zahra Aliakbar Ahovan
- Department of Microbiology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Zahra Esmaeili
- Cellular and Molecular Research Centre, Iran University of Medical Sciences, Tehran, Iran
- Department of Medical Biotechnology, Faculty of Allied Medicine, Iran University of Medical Sciences, Tehran, Iran
| | | | - Sadjad Khosravimelal
- Cellular and Molecular Research Centre, Iran University of Medical Sciences, Tehran, Iran
- Department of Medical Biotechnology, Faculty of Allied Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Morteza Alehosseini
- Department of Materials Engineering, Isfahan University of Technology, Isfahan 84156-83111, Iran
| | - Gorka Orive
- NanoBioCel Research Group, School of Pharmacy, University of the Basque Country (UPV/EHU), Vitoria-Gasteiz, Spain
- Biomedical Research Networking Centre in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN). Vitoria-Gasteiz, Spain
- University Institute for Regenerative Medicine and Oral Implantology - UIRMI (UPV/EHU-Fundación Eduardo Anitua). Vitoria-Gasteiz, Spain
- Bioaraba, NanoBioCel Research Group, Vitoria-Gasteiz, Spain
- Singapore Eye Research Institute, The Academia, 20 College Road, Discovery Tower, Singapore
| | | | | | - Paul A. Janmey
- Bioengineering Department, University of Pennsylvania, Philadelphia, USA
| | - Ali Hashemi
- Department of Microbiology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Subhas C. Kundu
- 3Bs Research Group, I3Bs - Research Institute on Biomaterials, Biodegradable and Biomimetics, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, University of Minho, AvePark, Guimaraes, Portugal
| | - Mazaher Gholipourmalekabadi
- Cellular and Molecular Research Centre, Iran University of Medical Sciences, Tehran, Iran
- Department of Medical Biotechnology, Faculty of Allied Medicine, Iran University of Medical Sciences, Tehran, Iran
- Department of Tissue Engineering & Regenerative Medicine, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
42
|
Zhou Q, Si Z, Wang K, Li K, Hong W, Zhang Y, Li P. Enzyme-triggered smart antimicrobial drug release systems against bacterial infections. J Control Release 2022; 352:507-526. [PMID: 36341932 DOI: 10.1016/j.jconrel.2022.10.038] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2022] [Revised: 10/17/2022] [Accepted: 10/22/2022] [Indexed: 11/06/2022]
Abstract
The rapid emergence and spread of drug-resistant bacteria, as one of the most pressing public health threats, are declining our arsenal of available antimicrobial drugs. Advanced antimicrobial drug delivery systems that can achieve precise and controlled release of antimicrobial agents in the microenvironment of bacterial infections will retard the development of antimicrobial resistance. A variety of extracellular enzymes are secreted by bacteria to destroy physical integrity of tissue during their invasion of host body, which can be utilized as stimuli to trigger "on-demand" release of antimicrobials. In the past decade, such bacterial enzyme responsive drug release systems have been intensively studied but few review has been released. Herein, we systematically summarize the recent progress of smart antimicrobial drug delivery systems triggered by bacteria secreted enzymes such as lipase, hyaluronidase, protease and antibiotic degrading enzymes. The perspectives and existing key issues of this field will also be discussed to fuel the innovative research and translational application in the future.
Collapse
Affiliation(s)
- Qian Zhou
- Frontiers Science Center for Flexible Electronics, (FSCFE), Xi'an Institute of Flexible Electronics (IFE) and Xi'an Institute of Biomedical Materials & Engineering (IBME), Northwestern Polytechnical University, 127 West Youyi Road, Xi'an 710072, China
| | - Zhangyong Si
- School of Chemical and Biomedical Engineering, Nanyang Technological University, Singapore 637459, Singapore
| | - Kun Wang
- Frontiers Science Center for Flexible Electronics, (FSCFE), Xi'an Institute of Flexible Electronics (IFE) and Xi'an Institute of Biomedical Materials & Engineering (IBME), Northwestern Polytechnical University, 127 West Youyi Road, Xi'an 710072, China
| | - Kunpeng Li
- Frontiers Science Center for Flexible Electronics, (FSCFE), Xi'an Institute of Flexible Electronics (IFE) and Xi'an Institute of Biomedical Materials & Engineering (IBME), Northwestern Polytechnical University, 127 West Youyi Road, Xi'an 710072, China
| | - Weilin Hong
- Frontiers Science Center for Flexible Electronics, (FSCFE), Xi'an Institute of Flexible Electronics (IFE) and Xi'an Institute of Biomedical Materials & Engineering (IBME), Northwestern Polytechnical University, 127 West Youyi Road, Xi'an 710072, China
| | - Yuezhou Zhang
- Frontiers Science Center for Flexible Electronics, (FSCFE), Xi'an Institute of Flexible Electronics (IFE) and Xi'an Institute of Biomedical Materials & Engineering (IBME), Northwestern Polytechnical University, 127 West Youyi Road, Xi'an 710072, China.
| | - Peng Li
- Frontiers Science Center for Flexible Electronics, (FSCFE), Xi'an Institute of Flexible Electronics (IFE) and Xi'an Institute of Biomedical Materials & Engineering (IBME), Northwestern Polytechnical University, 127 West Youyi Road, Xi'an 710072, China.
| |
Collapse
|
43
|
The New Strategy for Studying Drug-Delivery Systems with Prolonged Release: Seven-Day In Vitro Antibacterial Action. MOLECULES (BASEL, SWITZERLAND) 2022; 27:molecules27228026. [PMID: 36432127 PMCID: PMC9695913 DOI: 10.3390/molecules27228026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Revised: 11/14/2022] [Accepted: 11/15/2022] [Indexed: 11/22/2022]
Abstract
The new method of antibacterial-drug-activity investigation in vitro is proposed as a powerful strategy for understanding how carriers affect drug action during long periods (7 days). In this paper, we observed fluoroquinolone moxifloxacin (MF) antibacterial-efficiency in non-covalent complexes, with the sulfobutyl ether derivative of β-cyclodextrin (SCD) and its polymer (SCDpol). We conducted in vitro studies on two Escherichia coli strains that differed in surface morphology. It was found that MF loses its antibacterial action after 3-4 days in liquid media, whereas the inclusion of the drug in SCD led to the increase of MF antibacterial activity by up to 1.4 times within 1-5 days of the experiment. In the case of MF-SCDpol, we observed a 12-fold increase in the MF action, and a tendency to prolonged antibacterial activity. We visualized this phenomenon (the state of bacteria, cell membrane, and surface morphology) during MF and MF-carrier exposure by TEM. SCD and SCDpol did not change the drug's mechanism of action. Particle adsorption on cells was the crucial factor for determining the observed effects. The proteinaceous fimbriae on the bacteria surface gave a 2-fold increase of the drug carrier adsorption, hence the strains with fimbriae are more preferable for the proposed treatment. Furthermore, the approach to visualize the CD polymer adsorption on bacteria via TEM is suggested. We hope that the proposed comprehensive method will be useful for the studies of drug-delivery systems to uncover long-term antibacterial action.
Collapse
|
44
|
Nanoparticles for Antimicrobial Agents Delivery-An Up-to-Date Review. Int J Mol Sci 2022; 23:ijms232213862. [PMID: 36430343 PMCID: PMC9696780 DOI: 10.3390/ijms232213862] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Revised: 11/06/2022] [Accepted: 11/08/2022] [Indexed: 11/12/2022] Open
Abstract
Infectious diseases constitute an increasing threat to public health and medical systems worldwide. Particularly, the emergence of multidrug-resistant pathogens has left the pharmaceutical arsenal unarmed to fight against such severe microbial infections. Thus, the context has called for a paradigm shift in managing bacterial, fungal, viral, and parasitic infections, leading to the collision of medicine with nanotechnology. As a result, renewed research interest has been noted in utilizing various nanoparticles as drug delivery vehicles, aiming to overcome the limitations of current treatment options. In more detail, numerous studies have loaded natural and synthetic antimicrobial agents into different inorganic, lipid, and polymeric-based nanomaterials and tested them against clinically relevant pathogens. In this respect, this paper reviews the most recently reported successfully fabricated nanoformulations that demonstrated a great potential against bacteria, fungi, viruses, and parasites of interest for human medicine.
Collapse
|
45
|
Pseudomonas aeruginosa Clusters Toxic Nickel Nanoparticles to Enhance Survival. Microorganisms 2022; 10:microorganisms10112220. [PMID: 36363812 PMCID: PMC9694399 DOI: 10.3390/microorganisms10112220] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Revised: 11/02/2022] [Accepted: 11/07/2022] [Indexed: 11/11/2022] Open
Abstract
Microorganisms forming a biofilm might become multidrug-resistant by information exchange. Multi-resistant, biofilm-producing microorganisms are responsible for a major portion of hospital-acquired infections. Additionally, these microorganisms cause considerable damage in the industrial sector. Here, we screened several nanoparticles of transition metals for their antibacterial properties. The nanoparticles sizes of nickel (<300 nm) and nickel oxide (<50 nm) were analyzed with transmission electron microscopy. We could show that the antibacterial efficacy of nickel and nickel oxide nanoparticles on Pseudomonas aeruginosa isolated from household appliances and Staphylococcus aureus was the highest. Interestingly, only P. aeruginosa was able to survive at high concentrations (up to 50 mM) due to clustering toxic nanoparticles out of the medium by biofilm formation. This clustering served to make the medium nearly free of nanoparticles, allowing the bacteria to continue living without contact to the stressor. We observed these clusters by CLSM, SEM, and light microscopy. Moreover, we calculated the volume of NiO particles in the bacterial biofilms based on an estimated thickness of 5 nm from the TEM images as an average volume of 3.5 × 10−6 µm3. These results give us a new perspective on bacterial defense mechanisms and might be useful in industries such as water purification.
Collapse
|
46
|
Duan W, Liu X, Zhao J, Zheng Y, Wu J. Porous Silicon Carrier Endowed with Photothermal and Therapeutic Effects for Synergistic Wound Disinfection. ACS APPLIED MATERIALS & INTERFACES 2022; 14:48368-48383. [PMID: 36278256 DOI: 10.1021/acsami.2c12012] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
Drug carriers endowed with photothermal effects will allow the drug delivery system to release drugs in a thermal-stimuli manner. In addition, the photothermal therapy (PTT) will also interplay with therapeutic drugs loaded in the carrier to exhibit synergistic bioactivity for various disease treatment. However, endowing the drug carrier with photothermal and synergistic therapeutic effects still has challenge. Herein, we demonstrate that surface modification of porous silicon (PSi) with polydopamine (PDA) could endow the classical drug carrier with a significant photothermal effect for advanced antibacterial therapy and wound disinfection. Specifically, the PSi surface interacts with a Cu2+/PDA complex via a simple and fast surface reduction-induced deposition method, forming the unique CuPDA coated PSi microcarrier (CuPPSi) without blocking the mesoporous structure. The CuPPSi carrier generates a higher near-infrared (NIR) photothermal efficiency and improved drug loading capacity owing to the abundant functional groups of PDA. Stimuli-responsive release of antibacterial Cu2+ and loaded curcumin (Cur) from CuPPSi can be realized under multiple stimuli including pH, reactive oxygen species and NIR laser irradition. Benefited from the carrier's intrinsic multimodal therapy, the CuPPSi-Cur platform exhibits amplified, broad-spectrum, and synergistic antibacterial effect, killing more than 98% for both Staphylococcus aureus and Escherichia coli at a mild PTT temperature (∼45 °C). Notably, the combined therapy promotes migration of fibroblasts with no significant cytotoxicity as revealed through cell experiments in vitro. In bacteria-infected mice model, efficient bacterial ablation and wound healing are further demonstrated with negligible side effects in vivo. Overall, the rational design of a drug carrier with photothermal and therapeutic effects provides a novel intervention for amplifing wound disinfection clinically.
Collapse
Affiliation(s)
- Wei Duan
- Lab of Nanomedicine and Omic-based Diagnostics, Institute of Analytical Chemistry, Department of Chemistry, Zhejiang University, Hangzhou310058, China
| | - Xingyue Liu
- Lab of Nanomedicine and Omic-based Diagnostics, Institute of Analytical Chemistry, Department of Chemistry, Zhejiang University, Hangzhou310058, China
| | - Jingwen Zhao
- Lab of Nanomedicine and Omic-based Diagnostics, Institute of Analytical Chemistry, Department of Chemistry, Zhejiang University, Hangzhou310058, China
| | - Yongke Zheng
- Department of Intensive Care Unit, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou310006, China
| | - Jianmin Wu
- Lab of Nanomedicine and Omic-based Diagnostics, Institute of Analytical Chemistry, Department of Chemistry, Zhejiang University, Hangzhou310058, China
| |
Collapse
|
47
|
Synthesis of pH-responsive dimethylglycine surface-modified branched lipids for targeted delivery of antibiotics. Chem Phys Lipids 2022; 249:105241. [PMID: 36152880 DOI: 10.1016/j.chemphyslip.2022.105241] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2022] [Revised: 09/10/2022] [Accepted: 09/19/2022] [Indexed: 01/25/2023]
Abstract
The rampant antimicrobial resistance crisis calls for efficient and targeted drug delivery of antibiotics at the infectious site. Hence, this study aimed to synthesize a pH-responsive dimethylglycine surface-modified branched lipid (DMGSAD-lipid). The structure of the synthesized lipid was fully confirmed. The lipid polymer hybrid nanoparticles (LPHNPs) were formulated using the solvent evaporation method and characterised. Two LPHNPs (VCM_HS15_LPHNPs and VCM_RH40_LPHNPs) were formulated and characterised for size, polydispersity index (PDI), and zeta potential (ZP). Atomistic molecular dynamics simulations revealed that both the systems self-assembled to form energetically stable aggregates. The ZP of RH40_VCM_LPHNPs changed from 0.55 ± 0.14-9.44 ± 0.33 Vm, whereas for SH15_VCM_LPHNPs, ZP changed from - 1.55 ± 0.184 Vm to 9.83 ± 0.52 Vm at pH 7.4 and 6.0, respectively. The encapsulation efficiencies of VCM were above 40% while the drug release was faster at acidic pH when compared to pH 7.4. The antibacterial activity of LPHNPs against MRSA was eight-fold better in MICs at pH 6.0, compared to 7.4, when compared to bare VCM-treated specimens. The study confirms that pH-responsive LPHNPs have the potential for enhancing the treatment of bacterial infections and other diseases characterised by acidic conditions at the target site.
Collapse
|
48
|
A comparison study about antibacterial activity of zeolitic imidazolate frameworks (ZIFs) prepared with various metal ions. Inorganica Chim Acta 2022. [DOI: 10.1016/j.ica.2022.121110] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
49
|
Nazli A, He DL, Liao D, Khan MZI, Huang C, He Y. Strategies and progresses for enhancing targeted antibiotic delivery. Adv Drug Deliv Rev 2022; 189:114502. [PMID: 35998828 DOI: 10.1016/j.addr.2022.114502] [Citation(s) in RCA: 42] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Revised: 08/10/2022] [Accepted: 08/16/2022] [Indexed: 01/24/2023]
Abstract
Antibiotic resistance is a global health issue and a potential risk for society. Antibiotics administered through conventional formulations are devoid of targeting effect and often spread to various undesired body sites, leading to sub-lethal concentrations at the site of action and thus resulting in emergence of resistance, as well as side effects. Moreover, we have a very slim antibiotic pipeline. Drug-delivery systems have been designed to control the rate, time, and site of drug release, and innovative approaches for antibiotic delivery provide a glint of hope for addressing these issues. This review elaborates different delivery strategies and approaches employed to overcome the limitations of conventional antibiotic therapy. These include antibiotic conjugates, prodrugs, and nanocarriers for local and targeted antibiotic release. In addition, a wide range of stimuli-responsive nanocarriers and biological carriers for targeted antibiotic delivery are discussed. The potential advantages and limitations of targeted antibiotic delivery strategies are described along with possible solutions to avoid these limitations. A number of antibiotics successfully delivered through these approaches with attained outcomes and potentials are reviewed.
Collapse
Affiliation(s)
- Adila Nazli
- Chongqing Key Laboratory of Natural Product Synthesis and Drug Research, School of Pharmaceutical Sciences, Chongqing University, Chongqing 401331, PR China
| | - David L He
- College of Chemistry, University of California, Berkeley, CA 94720, United States
| | - Dandan Liao
- Chongqing Key Laboratory of Natural Product Synthesis and Drug Research, School of Pharmaceutical Sciences, Chongqing University, Chongqing 401331, PR China
| | | | - Chao Huang
- Chongqing Key Laboratory of Natural Product Synthesis and Drug Research, School of Pharmaceutical Sciences, Chongqing University, Chongqing 401331, PR China.
| | - Yun He
- Chongqing Key Laboratory of Natural Product Synthesis and Drug Research, School of Pharmaceutical Sciences, Chongqing University, Chongqing 401331, PR China.
| |
Collapse
|
50
|
Bhattacharjee R, Dey T, Kumar L, Kar S, Sarkar R, Ghorai M, Malik S, Jha NK, Vellingiri B, Kesari KK, Pérez de la Lastra JM, Dey A. Cellular landscaping of cisplatin resistance in cervical cancer. Biomed Pharmacother 2022; 153:113345. [PMID: 35810692 DOI: 10.1016/j.biopha.2022.113345] [Citation(s) in RCA: 45] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Revised: 06/22/2022] [Accepted: 06/24/2022] [Indexed: 12/11/2022] Open
Abstract
Cervical cancer (CC) caused by human papillomavirus (HPV) is one of the largest causes of malignancies in women worldwide. Cisplatin is one of the widely used drugs for the treatment of CC is rendered ineffective owing to drug resistance. This review highlights the cause of resistance and the mechanism of cisplatin resistance cells in CC to develop therapeutic ventures and strategies that could be utilized to overcome the aforementioned issue. These strategies would include the application of nanocarries, miRNA, CRIPSR/Cas system, and chemotherapeutics in synergy with cisplatin to not only overcome the issues of drug resistance but also enhance its anti-cancer efficiency. Moreover, we have also discussed the signaling network of cisplatin resistance cells in CC that would provide insights to develop therapeutic target sites and inhibitors. Furthermore, we have discussed the role of CC metabolism on cisplatin resistance cells and the physical and biological factors affecting the tumor microenvironments.
Collapse
Affiliation(s)
- Rahul Bhattacharjee
- KIIT School of Biotechnology, Kalinga Institute of Industrial Technology (KIIT-DU), Bhubaneswar 751024, Odisha, India
| | - Tanima Dey
- KIIT School of Biotechnology, Kalinga Institute of Industrial Technology (KIIT-DU), Bhubaneswar 751024, Odisha, India
| | - Lamha Kumar
- School of Biology, Indian Institute of Science Education and Research, Thiruvananthapuram 695551, Kerala, India
| | - Sulagna Kar
- KIIT School of Biotechnology, Kalinga Institute of Industrial Technology (KIIT-DU), Bhubaneswar 751024, Odisha, India
| | - Ritayan Sarkar
- KIIT School of Biotechnology, Kalinga Institute of Industrial Technology (KIIT-DU), Bhubaneswar 751024, Odisha, India
| | - Mimosa Ghorai
- Department of Life Sciences, Presidency University, 86/1 College Street, Kolkata 700073, West Bengal, India
| | - Sumira Malik
- Amity Institute of Biotechnology, Amity University Jharkhand, Ranchi, Jharkhand 834001, India
| | - Niraj Kumar Jha
- Department of Biotechnology, School of Engineering and Technology (SET), Sharda University, Greater Noida, Uttar Pradesh 201310, India; Department of Biotechnology, School of Applied & Life Sciences (SALS), Uttaranchal University, Dehradun 248007, India; Department of Biotechnology Engineering and Food Technology, Chandigarh University, Mohali 140413, India.
| | - Balachandar Vellingiri
- Human Molecular Cytogenetics and Stem Cell Laboratory, Department of Human Genetics and Molecular Biology, Bharathiar University, Coimbatore 641-046, India
| | - Kavindra Kumar Kesari
- Department of Applied Physics, School of Science, Aalto University, Espoo 00076, Finland; Department of Bio-products and Bio-systems, School of Chemical Engineering, Aalto University, Espoo 00076, Finland
| | - José M Pérez de la Lastra
- Biotechnology of Macromolecules, Instituto de Productos Naturales y Agrobiología, IPNA (CSIC), Avda. Astrofísico Francisco Sánchez, 3, 38206 San Cristóbal de la Laguna (Santa Cruz de Tenerife), Spain.
| | - Abhijit Dey
- Department of Life Sciences, Presidency University, 86/1 College Street, Kolkata 700073, West Bengal, India.
| |
Collapse
|