Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Biomater Sci ; 12(13): 3411-3422, 2024 Jun 25.
Artículo en Inglés | MEDLINE | ID: mdl-38809118

RESUMEN

Bacteria have evolved survival mechanisms that enable them to live within host cells, triggering persistent intracellular infections that present significant clinical challenges due to the inability for conventional antibiotics to permeate cell membranes. In recent years, antibiotic nanocarriers or 'nanoantibiotics' have presented a promising strategy for overcoming intracellular infections by facilitating cellular uptake of antibiotics, thus improving targeting to the bacteria. However, prior to reaching host cells, nanocarriers experience interactions with proteins that form a corona and alter their physiological response. The influence of this protein corona on the cellular uptake, drug release and efficacy of nanoantibiotics for intracellular infections is poorly understood and commonly overlooked in preclinical studies. In this study, protein corona influence on cellular uptake was investigated for two nanoparticles; liposomes and cubosomes in macrophage and epithelial cells that are commonly infected with pathogens. Studies were conducted in presence of fetal bovine serum (FBS) to form a biologically relevant protein corona in an in vitro setting. Protein corona impact on cellular uptake was shown to be nanoparticle-dependent, where reduced internalization was observed for liposomes, the opposite was observed for cubosomes. Subsequently, vancomycin-loaded cubosomes were explored for their drug delivery performance against intracellular small colony variants of Staphylococcus aureus. We demonstrated improved bacterial killing in macrophages, with greater reduction in bacterial viability upon internalization of cubosomes mediated by the protein corona. However, no differences in efficacy were observed in epithelial cells. Thus, this study provides insights and evidence to the role of protein corona in modulating the performance of nanoparticles in a dynamic manner; these findings will facilitate improved understanding and translation of future investigations from in vitro to in vivo.


Asunto(s)
Antibacterianos , Liposomas , Nanopartículas , Corona de Proteínas , Staphylococcus aureus , Antibacterianos/farmacología , Antibacterianos/química , Antibacterianos/administración & dosificación , Corona de Proteínas/química , Corona de Proteínas/metabolismo , Staphylococcus aureus/efectos de los fármacos , Animales , Humanos , Liposomas/química , Nanopartículas/química , Vancomicina/farmacología , Vancomicina/química , Vancomicina/administración & dosificación , Ratones , Macrófagos/metabolismo , Macrófagos/efectos de los fármacos , Células RAW 264.7 , Pruebas de Sensibilidad Microbiana , Lípidos/química , Portadores de Fármacos/química
2.
Pharmaceutics ; 16(3)2024 Mar 14.
Artículo en Inglés | MEDLINE | ID: mdl-38543290

RESUMEN

The opportunistic bacteria growing in biofilms play a decisive role in the pathogenesis of chronic infectious diseases. Biofilm-dwelling bacteria behave differently than planktonic bacteria and are likely to increase resistance and tolerance to antimicrobial therapeutics. Antimicrobial adjuvants have emerged as a promising strategy to combat antimicrobial resistance (AMR) and restore the efficacy of existing antibiotics. A combination of antibiotics and potential antimicrobial adjuvants, (e.g., extracellular polymeric substance (EPS)-degrading enzymes and quorum sensing inhibitors (QSI) can improve the effects of antibiotics and potentially reduce bacterial resistance). In addition, encapsulation of antimicrobials within nanoparticulate systems can improve their stability and their delivery into biofilms. Lipid nanocarriers (LNCs) have been established as having the potential to improve the efficacy of existing antibiotics in combination with antimicrobial adjuvants. Among them, liquid crystal nanoparticles (LCNPs), liposomes, solid lipid nanoparticles (SLNs), and nanostructured lipid carriers (NLCs) are promising due to their superior properties compared to traditional formulations, including their greater biocompatibility, higher drug loading capacity, drug protection from chemical or enzymatic degradation, controlled drug release, targeted delivery, ease of preparation, and scale-up feasibility. This article reviews the recent advances in developing various LNCs to co-deliver some well-studied antimicrobial adjuvants combined with antibiotics from different classes. The efficacy of various combination treatments is compared against bacterial biofilms, and synergistic therapeutics that deserve further investigation are also highlighted. This review identifies promising LNCs for the delivery of combination therapies that are in recent development. It discusses how LNC-enabled co-delivery of antibiotics and adjuvants can advance current clinical antimicrobial treatments, leading to innovative products, enabling the reuse of antibiotics, and providing opportunities for saving millions of lives from bacterial infections.

3.
ACS Infect Dis ; 10(2): 337-349, 2024 02 09.
Artículo en Inglés | MEDLINE | ID: mdl-38295053

RESUMEN

Bacterial pathogens are constantly evolving to outsmart the host immune system and antibiotics developed to eradicate them. One key strategy involves the ability of bacteria to survive and replicate within host cells, thereby causing intracellular infections. To address this unmet clinical need, researchers are adopting new approaches, such as the development of novel molecules that can penetrate host cells, thus exerting their antimicrobial activity intracellularly, or repurposing existing antibiotics using nanocarriers (i.e., nanoantibiotics) for site-specific delivery. However, inconsistency in information reported across published studies makes it challenging for scientific comparison and judgment of experiments for future direction by researchers. Together with the lack of reproducibility of experiments, these inconsistencies limit the translation of experimental results beyond pre-clinical evaluation. Minimum information guidelines have been instrumental in addressing such challenges in other fields of biomedical research. Guidelines and recommendations provided herein have been designed for researchers as essential parameters to be disclosed when publishing their methodology and results, divided into four main categories: (i) experimental design, (ii) establishing an in vitro model, (iii) assessment of efficacy of novel therapeutics, and (iv) statistical assessment. These guidelines have been designed with the intention to improve the reproducibility and rigor of future studies while enabling quantitative comparisons of published studies, ultimately facilitating translation of emerging antimicrobial technologies into clinically viable therapies that safely and effectively treat intracellular infections.


Asunto(s)
Antiinfecciosos , Proyectos de Investigación , Reproducibilidad de los Resultados , Antibacterianos/farmacología , Antibacterianos/uso terapéutico , Bacterias
4.
Int J Pharm ; 648: 123614, 2023 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-37979632

RESUMEN

Self-emulsifying drug delivery systems (i.e. SEDDS, SMEDDS and SNEDDS) are widely employed as solubility and bioavailability enhancing formulation strategies for poorly water-soluble drugs. Despite the capacity for SEDDS to effectively facilitate oral drug absorption, tolerability concerns exist due to the capacity for high concentrations of surfactants (typically present within SEDDS) to induce gastrointestinal toxicity and mucosal irritation. With new knowledge surrounding the role of the gut microbiota in modulating intestinal inflammation and mucosal injury, there is a clear need to determine the impact of SEDDS on the gut microbiota. The current study is the first of its kind to demonstrate the detrimental impact of SEDDS on the gut microbiota of Sprague-Dawley rats, following daily oral administration (100 mg/kg) for 21 days. SEDDS comprising a lipid phase (i.e. Type I, II and III formulations according to the Lipid Formulation Classification Scheme) induced significant changes to the composition and diversity of the gut microbiota, evidenced through a reduction in operational taxonomic units (OTUs) and alpha diversity (Shannon's index), along with statistically significant shifts in beta diversity (according to PERMANOVA of multi-dimensional Bray-Curtis plots). Key signatures of gut microbiota dysbiosis correlated with the increased expression of pro-inflammatory cytokines within the jejunum, while mucosal injury was characterised by significant reductions in plasma citrulline levels, a validated biomarker of enterocyte mass and mucosal barrier integrity. These findings have potential clinical ramifications for chronically administered drugs that are formulated with SEDDS and stresses the need for further studies that investigate dose-dependent effects of SEDDS on the gastrointestinal microenvironment in a clinical setting.


Asunto(s)
Microbioma Gastrointestinal , Ratas , Animales , Ratas Sprague-Dawley , Química Farmacéutica/métodos , Sistemas de Liberación de Medicamentos/métodos , Preparaciones Farmacéuticas , Administración Oral , Disponibilidad Biológica , Solubilidad , Lípidos , Emulsiones
5.
Expert Opin Drug Deliv ; 20(10): 1297-1314, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37307224

RESUMEN

INTRODUCTION: Increasing attention is being afforded to understanding the bidirectional relationships that exist between oral medications and the gut microbiota, in an attempt to optimize pharmacokinetic performance and mitigate unwanted side effects. While a wealth of research has investigated the direct impact of active pharmaceutical ingredients (APIs) on the gut microbiota, the interactions between inactive pharmaceutical ingredients (i.e. excipients) and the gut microbiota are commonly overlooked, despite excipients typically representing over 90% of the final dosage form. AREAS COVERED: Known excipient-gut microbiota interactions for various classes of inactive pharmaceutical ingredients, including solubilizing agents, binders, fillers, sweeteners, and color additives, are reviewed in detail. EXPERT OPINION: Clear evidence indicates that orally administered pharmaceutical excipients directly interact with gut microbes and can either positively or negatively impact gut microbiota diversity and composition. However, these relationships and mechanisms are commonly overlooked during drug formulation, despite the potential for excipient-microbiota interactions to alter drug pharmacokinetics and interfere with host metabolic health. The insights derived from this review will inform pharmaceutical scientists with the necessary design considerations for mitigating potential adverse pharmacomicrobiomic interactions when formulating oral dosage forms, ultimately providing clear avenues for improving therapeutic safety and efficacy.


Asunto(s)
Efectos Colaterales y Reacciones Adversas Relacionados con Medicamentos , Microbioma Gastrointestinal , Humanos , Excipientes , Química Farmacéutica , Composición de Medicamentos
6.
Int J Pharm ; 639: 122927, 2023 May 25.
Artículo en Inglés | MEDLINE | ID: mdl-37059243

RESUMEN

The intracellular survival of bacteria is a significant challenge in the fight against antimicrobial resistance. Currently available antibiotics suffer from limited penetration across host cell membranes, resulting in suboptimal treatment against the internalised bacteria. Liquid crystalline nanoparticles (LCNP) are gaining significant research interest in promoting the cellular uptake of therapeutics due to their fusogenic properties; however, they have not been reported for targeting intracellular bacteria. Herein, the cellular internalisation of LCNPs in RAW 264.7 macrophages and A549 epithelial cells was investigated and optimized through the incorporation of a cationic lipid, dimethyldioctadecylammonium bromide (DDAB). LCNPs displayed a honeycomb-like structure, while the inclusion of DDAB resulted into an onion-like organisation with larger internal pores. Cationic LCNPs enhanced the cellular uptake in both cells, reaching up to âˆ¼ 90% uptake in cells. Further, LCNPs were encapsulated with tobramycin or vancomycin to improve their activity against intracellular gram-negative, Pseudomonas aeruginosa (P. aeruginosa) and gram-positive, Staphylococcus aureus (S. aureus) bacteria. The enhanced cellular uptake of cationic LCNP resulted in significant reduction of intracellular bacterial load (up to 90% reduction), compared to antibiotic dosed in its free form; with reduced performance observed for epithelial cells infected with S. aureus. Specifically engineered LCNP can re-sensitise antibiotics against both intracellular Gram positive and negative bacteria in diverse cell lines.


Asunto(s)
Nanopartículas , Infecciones Estafilocócicas , Humanos , Vancomicina , Tobramicina/farmacología , Staphylococcus aureus , Pseudomonas aeruginosa/metabolismo , Antibacterianos/farmacología , Antibacterianos/metabolismo , Nanopartículas/química , Bacterias , Pruebas de Sensibilidad Microbiana
7.
J Colloid Interface Sci ; 641: 36-47, 2023 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-36924544

RESUMEN

The inhalable administration of lipid nanoparticles is an effective strategy for localised delivery of therapeutics against various lung diseases. Of this, improved intracellular delivery of pharmaceuticals for infectious disease and cancer management is of high significance. However, the influence of lipid nanoparticle composition and structure on uptake in pulmonary cell lines, especially in the presence of biologically relevant media is poorly understood. Here, the uptake of lamellar (liposomes) versus non-lamellar (cubosomes) lipid nanoparticles in macrophages and lung epithelial cells was quantified and the influence of bronchoalveolar lavage fluid (BALF), containing native pulmonary protein and surfactant molecules is determined. Cubosome uptake in both macrophages and epithelial cells was strongly mediated by a high percentage of molecular function regulatory and binding proteins present within the protein corona. In contrast, the protein corona did not influence the uptake of liposomes in epithelial cells. In macrophages, the proteins mediated a rapid internalisation, followed by exocytosis of liposomes after 6 h incubation. These findings on the influence of biological fluid in regulating lipid nanoparticle uptake mechanisms may guide future development of optimal intracellular delivery systems for therapeutics via the pulmonary route.


Asunto(s)
Nanopartículas , Corona de Proteínas , Liposomas/química , Corona de Proteínas/química , Adsorción , Pulmón/metabolismo , Proteínas/química , Nanopartículas/química , Lípidos/química
8.
Adv Drug Deliv Rev ; 177: 113948, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34464665

RESUMEN

Bacteria have developed a wealth of strategies to avoid and resist the action of antibiotics, one of which involves pathogens invading and forming reservoirs within host cells. Due to the poor cell membrane permeability, stability and retention of conventional antibiotics, this renders current treatments largely ineffective, since achieving a therapeutically relevant antibiotic concentration at the site of intracellular infection is not possible. To overcome such challenges, current antibiotics are 'repurposed' via reformulation using micro- or nano-carrier systems that effectively encapsulate and deliver therapeutics across cellular membranes of infected cells. Bioinspired materials that imitate the uptake of biological particulates and release antibiotics in response to natural stimuli are recently explored to improve the targeting and specificity of this 'nanoantibiotic' approach. In this review, the mechanisms of internalization and survival of intracellular bacteria are elucidated, effectively accentuating the current treatment challenges for intracellular infections and the implications for repurposing conventional antibiotics. Key case studies of nanoantibiotics that have drawn inspiration from natural biological particles and cellular uptake pathways to effectively eradicate intracellular pathogens are detailed, clearly highlighting the rational for harnessing bioinspired drug delivery strategies.


Asunto(s)
Antibacterianos/administración & dosificación , Infecciones Bacterianas/tratamiento farmacológico , Sistemas de Liberación de Medicamentos , Reposicionamiento de Medicamentos , Animales , Humanos , Evasión Inmune
9.
Nanomaterials (Basel) ; 10(4)2020 Apr 24.
Artículo en Inglés | MEDLINE | ID: mdl-32344619

RESUMEN

An urgent demand exists for the development of novel delivery systems that efficiently transport antibacterial agents across cellular membranes for the eradication of intracellular pathogens. In this study, the clinically relevant poorly water-soluble antibiotic, rifampicin, was confined within mesoporous silica nanoparticles (MSN) to investigate their ability to serve as an efficacious nanocarrier system against small colony variants of Staphylococcus aureus (SCV S. aureus) hosted within Caco-2 cells. The surface chemistry and particle size of MSN were varied through modifications during synthesis, where 40 nm particles with high silanol group densities promoted enhanced cellular uptake. Extensive biophysical analysis was performed, using quartz crystal microbalance with dissipation (QCM-D) and total internal reflection fluorescence (TIRF) microscopy, to elucidate the mechanism of MSN adsorption onto semi-native supported lipid bilayers (snSLB) and, thus, uncover potential cellular uptake mechanisms of MSN into Caco-2 cells. Such studies revealed that MSN with reduced silanol group densities were prone to greater particle aggregation on snSLB, which was expected to restrict endocytosis. MSN adsorption and uptake into Caco-2 cells correlated well with antibacterial efficacy against SCV S. aureus, with 40 nm hydrophilic particles triggering a ~2.5-log greater reduction in colony forming units, compared to the pure rifampicin. Thus, this study provides evidence for the potential to design silica nanocarrier systems with controlled surface chemistries that can be used to re-sensitise intracellular bacteria to antibiotics by delivering them to the site of infection.

10.
Antibiotics (Basel) ; 8(2)2019 Apr 11.
Artículo en Inglés | MEDLINE | ID: mdl-30979069

RESUMEN

Infectious diseases remain a major burden in today's world, causing high mortality rates and significant economic losses, with >9 million deaths per year predicted by 2030. Invasion of host cells by intracellular bacteria poses treatment challenges due to the poor permeation of antimicrobials into the infected cells. To overcome these limitations, mesoporous silica nanoparticles (MSNP) loaded with the antibiotic rifampicin were investigated as a nanocarrier system for the treatment of intracellular bacterial infection with specific interest in the influence of particle size on treatment efficiency. An intracellular infection model was established using small colony variants (SCV) of S. aureus in macrophages to systemically evaluate the efficacy of rifampicin-loaded MSNP against the pathogen as compared to a rifampicin solution. As hypothesized, the superior uptake of MSNP by macrophages resulted in an enhanced treatment efficacy of the encapsulated rifampicin as compared to free antibiotic. This study provides a potential platform to improve the performance of currently available antibiotics against intracellular infections.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...