Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 36
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Microbes Infect ; 26(5-6): 105365, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38777105

RESUMEN

Aureocin A53 is a peptide bacteriocin produced by an opportunistic pathogen Staphylococcus aureus strain A53. The spatial structure of aureocin, unlike its amino acid sequence, is similar to the bacteriocin BacSp222, which was recently found to have the ability to induce the inflammatory response in the host cells. The presented research aimed to verify such properties also for aureocin A53. We demonstrated that the synthetic aureocin has slight cytotoxic activity towards murine monocytic-macrophage cells. This molecule was also able to activate murine P388.D1 and RAW 264.7 cells to IFN-γ-dependent production of nitric oxide and to activate production of the pro-inflammatory cytokine - TNF. We also proved that the observed pro-inflammatory activity of the studied bacteriocin is related to the stimulation of the TLR2/TLR6 heterodimer and, consequently, activation of the NF-κB transcription factor. To sum up, A53 is the second bacteriocin described in the literature, showing the pro-inflammatory activity against murine macrophage-like cells.


Asunto(s)
Bacteriocinas , Macrófagos , Óxido Nítrico , Staphylococcus aureus , Ratones , Animales , Bacteriocinas/farmacología , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Macrófagos/inmunología , Staphylococcus aureus/efectos de los fármacos , Células RAW 264.7 , Óxido Nítrico/metabolismo , FN-kappa B/metabolismo , Receptor Toll-Like 2/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo , Línea Celular , Citocinas/metabolismo , Interferón gamma/metabolismo , Inflamación/metabolismo
2.
Int J Biol Macromol ; 262(Pt 2): 130142, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38365151

RESUMEN

Injectable hydrogel-based materials have emerged as promising alendronate (ALN) delivery systems for the treatment of osteoporosis. However, their intrinsic permeability limits the sustained delivery of small-molecule drugs. In response to this challenge, we present the multifunctional hybrids composed of mesoporous silica particles decorated with hydroxyapatite and loaded with alendronate (MSP-NH2-HAp-ALN), which are immobilized in collagen/chitosan/hyaluronic acid-based hydrogel. We have mainly focused on the biological in vitro/ex vivo evaluation of developed composites. It was found that the extracts released from tested systems do not exhibit hemolytic properties and are safe for blood elements and the human liver cell model. The resulting materials create an environment conducive to differentiating human bone marrow mesenchymal stem cells and reduce the viability of osteoclast precursors (RAW 264.7). Importantly, even the system with the lowest concentration of ALN caused a substantial cytotoxic effect on RAW 264.7 cells; their viability decreased to 20 % and 10 % of control on 3 and 7 day of culture. Additionally, prolonged ALN release (up to 20 days) with minimized burst release was observed, while material features (wettability, swellability, degradation, mechanical properties) depended on MSP-NH2-HAp-ALN content. The obtained data indicate that developed composites establish a high-potential formulation for safe and effective osteoporosis therapy.


Asunto(s)
Quitosano , Osteoporosis , Humanos , Alendronato/farmacología , Ácido Hialurónico , Hidrogeles , Colágeno/farmacología , Osteoporosis/tratamiento farmacológico
3.
Inflamm Res ; 72(5): 915-928, 2023 May.
Artículo en Inglés | MEDLINE | ID: mdl-36964784

RESUMEN

OBJECTIVE AND DESIGN: BacSp222 bacteriocin is a bactericidal and proinflammatory peptide stimulating immune cells to produce selected cytokines and NO in NF-ĸB dependent manner. This study aims to identify the receptor which mediates this activity. METHODS: We applied fluorescently labeled BacSp222 and a confocal microscopy imaging to analyze the direct interaction of the bacteriocin with the cells. Reporter HEK-Blue cells overexpressing human toll-like receptors (TLR2, TLR4, TLR5 or TLR2/TLR1 and TLR2/TLR6 heterodimers) were stimulated with BacSp222, and then the activity of NF-ĸB-dependent secreted embryonic alkaline phosphatase (SEAP) was measured. In turn, formylated peptide receptor (FPR) or TLR2 antagonists were used to verify bacteriocin-stimulated TNF production by murine monocyte-macrophage cell lines. RESULTS: BacSp222 undergoes internalization into cells without disturbing the cell membrane. FPR antagonists do not affect TNF produced by BacSp222-stimulated murine macrophage-like cells. In contrast, BacSp222 stimulates NF-ĸB activation in HEK-Blue overexpressing TLR2 or TLR2/TLR6 heterodimer, but not TLR2/TLR1, TLR4 or TLR5 receptors. Moreover, TLR2-specific antagonists inhibit NF-ĸB signaling in BacSp222-stimulated HEK-Blue TLR2/TLR6 cells and reduce TNF release by BacSp222-treated RAW 264.7 and P388.D1. CONCLUSIONS: BacSp222 is a novel ligand for TLR2/TLR6 heterodimer. By binding TLR complex the bacteriocin undergoes internalization, inducing proinflammatory signaling that employs MyD88 and NF-ĸB pathways.


Asunto(s)
Bacteriocinas , Receptor Toll-Like 6 , Humanos , Animales , Ratones , Ligandos , Receptor Toll-Like 6/metabolismo , FN-kappa B/metabolismo , Receptor Toll-Like 1 , Receptor Toll-Like 2/metabolismo , Receptor Toll-Like 5 , Receptor Toll-Like 4 , Bacteriocinas/farmacología
4.
J Inflamm Res ; 15: 4601-4621, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35982757

RESUMEN

Purpose: The zoonotic opportunistic pathogen Staphylococcus pseudintermedius 222 produces BacSp222 - an atypical peptide exhibiting the features of a bacteriocin, a virulence factor, and a molecule modulating the host inflammatory reaction. The peptide is secreted in an unmodified form and, additionally, two forms modified posttranslationally by succinylation. This study is a comprehensive report focusing on the proinflammatory properties of such molecules. Methods: The study was performed on mouse monocyte/macrophage-like and endothelial cell lines as well as human neutrophils. The following peptides were studied: BacSp222, its succinylated forms, the form deprived of formylated methionine, and a reference bacteriocin - nisin. The measurements of the nitric oxide (NO) level, induced NO synthase (iNOS) expression, the profile of secreted cytokines, NF-kappa-B activation, reactive oxygen species (ROS) biosynthesis, and the formation of extracellular traps were conducted to evaluate the proinflammatory activity of the studied peptides. Results: BacSp222 and its succinylated forms effectively induced NO production and iNOS expression when combined with IFN-gamma in macrophage-like cells. All natural BacSp222 forms used alone or with IFN-gamma stimulated the production of TNF-alpha, MCP-1, and IL-1-alpha, while the co-stimulation with IFN-gamma increased IL-10 and IL-27. Upregulated TNF-alpha secretion observed after BacSp222 exposition resulted from increased expression but not from membrane TNF-alpha proteolysis. In neutrophils, all forms of bacteriocin upregulated IL-8, but did not induce ROS production or NETs formation. In all experiments, the activities of deformylated bacteriocin were lower or unequivocal in comparison to other forms of the peptide. Conclusion: All naturally secreted forms of BacSp222 exhibit proinflammatory activity against monocyte-macrophage cells and neutrophils, confirming that the biological role of BacSp222 goes beyond bactericidal and cytotoxic effects. The atypical posttranslational modification (succinylation) does not diminish its immunomodulatory activity in contrast to the lower antibacterial potential or cytotoxicity of such modified form established in previous studies.

5.
Int J Nanomedicine ; 17: 577-588, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35173431

RESUMEN

INTRODUCTION: Biodistribution of nanocarriers with a structure consisting of core and shell is most often analyzed using methods based on labeling subsequent compartments of nanocarriers. This approach may have serious limitations due to the instability of such complex systems under in vivo conditions. METHODS: The core-shell polyelectrolyte nanocarriers were intravenously administered to healthy BALB/c mice with breast cancer. Next, biodistribution profiles and elimination routes were determined post mortem based on fluorescence measurements performed for isolated blood, tissue homogenates, collected urine, and feces. RESULTS: Despite the surface PEGylation with PLL-g-PEG, multilayer polyelectrolyte nanocarriers undergo rapid degradation after intravenous administration. This process releases the shell components but not free Rhodamine B. Elements of polyelectrolyte shells are removed by hepatobiliary and renal clearance. CONCLUSION: Multilayer polyelectrolyte nanocarriers are prone to rapid degradation after intravenous administration. Fluorophore localization determines the obtained results of biodistribution and elimination routes of core-shell nanomaterials. Therefore, precise and reliable analysis of in vivo stability and biodistribution of nanomaterials composed of several compartments requires nanomaterials labeled within each compartment.


Asunto(s)
Nanopartículas , Nanoestructuras , Animales , Portadores de Fármacos/química , Colorantes Fluorescentes , Ratones , Nanopartículas/química , Nanoestructuras/química , Polielectrolitos/química , Polietilenglicoles/química , Distribución Tisular
6.
Materials (Basel) ; 14(24)2021 Dec 11.
Artículo en Inglés | MEDLINE | ID: mdl-34947226

RESUMEN

Control of nonspecific/specific protein adsorption is the main goal in the design of novel biomaterials, implants, drug delivery systems, and sensors. The specific functionalization of biomaterials can be achieved by proper surface modification. One of the important strategies is covering the materials with functional coatings. Therefore, our work aimed to functionalize multilayer coating to control nonspecific/specific protein adsorption. The polyelectrolyte coating was formed using a layer-by-layer technique (LbL) with biocompatible polyelectrolytes poly-L-lysine hydrobromide (PLL) and poly-L-glutamic acid (PGA). Nonspecific protein adsorption was minimized/eliminated by pegylation of multilayer films, which was achieved by adsorption of pegylated polycations (PLL-g-PEG). The influence of poly (ethylene glycol) chain length on eliminating nonspecific protein adsorption was confirmed. Moreover, to achieve specific protein adsorption, the multilayer film was also functionalized by immobilization of antibodies via a streptavidin bridge. The functional coatings were tested, and the adsorption of the following proteins confirmed the ability to control nonspecific/specific adsorption: human serum albumin (HSA), fibrinogen (FIB), fetal bovine serum (FBS), carcinoembryonic antigen human (CEA) monitored by quartz crystal microbalance with dissipation (QCM-D). AFM imaging of unmodified and modified multilayer surfaces was also performed. Functional multilayer films are believed to have the potential as a novel platform for biotechnological applications, such as biosensors and nanocarriers for drug delivery systems.

7.
Int J Mol Sci ; 22(23)2021 Nov 25.
Artículo en Inglés | MEDLINE | ID: mdl-34884566

RESUMEN

Cancer is one of the most important health problems of our population, and one of the common anticancer treatments is chemotherapy. The disadvantages of chemotherapy are related to the drug's toxic effects, which act on cancer cells and the healthy part of the body. The solution of the problem is drug encapsulation and drug targeting. The present study aimed to develop a novel method of preparing multifunctional 5-Fluorouracil (5-FU) nanocarriers and their in vitro characterization. 5-FU polyaminoacid-based core@shell nanocarriers were formed by encapsulation drug-loaded nanocores with polyaminoacids multilayer shell via layer-by-layer method. The size of prepared nanocarriers ranged between 80-200 nm. Biocompatibility of our nanocarriers as well as activity of the encapsulated drug were confirmed by MTT tests. Moreover, the ability to the real-time observation of developed nanocarriers and drug accumulation inside the target was confirmed by fluorine magnetic resonance imaging (19F-MRI).


Asunto(s)
Aminoácidos/química , Portadores de Fármacos/administración & dosificación , Portadores de Fármacos/síntesis química , Fluorouracilo/farmacología , Neoplasias Mamarias Experimentales/tratamiento farmacológico , Nanopartículas/administración & dosificación , Animales , Antimetabolitos Antineoplásicos/química , Antimetabolitos Antineoplásicos/farmacología , Femenino , Fluorouracilo/química , Neoplasias Mamarias Experimentales/patología , Nanopartículas/química , Células Tumorales Cultivadas
8.
ACS Appl Mater Interfaces ; 13(42): 49762-49779, 2021 Oct 27.
Artículo en Inglés | MEDLINE | ID: mdl-34643364

RESUMEN

Novel multifunctional biomimetic injectable hybrid systems were synthesized. The physicochemical as well as biological in vitro and in vivo tests demonstrated that they are promising candidates for bone tissue regeneration. The hybrids are composed of a biopolymeric collagen/chitosan/hyaluronic acid matrix and amine group-functionalized silica particles decorated with apatite to which the alendronate molecules were coordinated. The components of these systems were integrated and stabilized by cross-linking with genipin, a compound of natural origin. They can be precisely injected into the diseased tissue in the form of a viscous sol or a partially cross-linked hydrogel, where they can serve as scaffolds for locally controlled bone tissue regeneration/remodeling by supporting the osteoblast formation/proliferation and maintaining the optimal osteoclast level. These materials lack systemic toxicity. They can be particularly useful for the repair of small osteoporotic bone defects.


Asunto(s)
Materiales Biocompatibles/farmacología , Osteoporosis/tratamiento farmacológico , Ingeniería de Tejidos , Andamios del Tejido/química , Aminas/administración & dosificación , Aminas/química , Aminas/farmacología , Animales , Materiales Biocompatibles/administración & dosificación , Materiales Biocompatibles/química , Regeneración Ósea/efectos de los fármacos , Línea Celular , Quitosano/administración & dosificación , Quitosano/química , Quitosano/farmacología , Colágeno/administración & dosificación , Colágeno/química , Colágeno/farmacología , Liberación de Fármacos , Humanos , Ácido Hialurónico/administración & dosificación , Ácido Hialurónico/química , Ácido Hialurónico/farmacología , Inyecciones Subcutáneas , Ensayo de Materiales , Ratones , Ratones Endogámicos C57BL , Osteoporosis/patología , Tamaño de la Partícula , Dióxido de Silicio/administración & dosificación , Dióxido de Silicio/química , Dióxido de Silicio/farmacología
9.
J Mater Chem B ; 9(36): 7482-7491, 2021 09 22.
Artículo en Inglés | MEDLINE | ID: mdl-34551060

RESUMEN

The introduction of ibuprofen into mesopores of SBA-15 has been accomplished using the melting method. Samples exhibit from 9 to 33% of the hydrophobic drug. They are not toxic to mouse monocyte-macrophage cells and do not stimulate a pro-inflammatory response. The sample with 25% of the drug showed no crystalline ibuprofen and almost filled the mesopores, while the sample with 33% showed a total filling of the mesopores with some crystalline ibuprofen present. By means of 1D (1H, 13C HPDEC, 13C CP MAS) and 2D (1H-1H NOESY) MAS NMR spectroscopy, it has been shown that water coexists with ibuprofen in mesopores and has an impact on the mobility of ibuprofen molecules and their location within the sample (outside or inside mesopores). Studies in the dehydrated state show for the first time that the high mobility of ibuprofen in mesopores is directly connected to the presence of water. Dehydrated samples show slightly slower release rates in comparison to their hydrated counterparts.


Asunto(s)
Ibuprofeno/química , Dióxido de Silicio/química , Agua/química , Animales , Línea Celular , Supervivencia Celular/efectos de los fármacos , Ibuprofeno/metabolismo , Ibuprofeno/farmacología , Espectroscopía de Resonancia Magnética , Ratones , Dióxido de Silicio/farmacología
10.
Int J Mol Sci ; 22(12)2021 Jun 10.
Artículo en Inglés | MEDLINE | ID: mdl-34200765

RESUMEN

BacSp222 is a multifunctional peptide produced by Staphylococcus pseudintermedius 222. This 50-amino acid long peptide belongs to subclass IId of bacteriocins and forms a four-helix bundle molecule. In addition to bactericidal functions, BacSp222 possesses also features of a virulence factor, manifested in immunomodulatory and cytotoxic activities toward eukaryotic cells. In the present study, we demonstrate that BacSp222 is produced in several post-translationally modified forms, succinylated at the ε-amino group of lysine residues. Such modifications have not been previously described for any bacteriocins. NMR and circular dichroism spectroscopy studies have shown that the modifications do not alter the spatial structure of the peptide. At the same time, succinylation significantly diminishes its bactericidal and cytotoxic potential. We demonstrate that the modification of the bacteriocin is an effect of non-enzymatic reaction with a highly reactive intracellular metabolite, i.e., succinyl-coenzyme A. The production of succinylated forms of the bacteriocin depends on environmental factors and on the access of bacteria to nutrients. Our study indicates that the production of succinylated forms of bacteriocin occurs in response to the changing environment, protects producer cells against the autotoxicity of the excreted peptide, and limits the pathogenicity of the strain.


Asunto(s)
Bacteriocinas/química , Bacteriocinas/farmacología , Macrófagos/efectos de los fármacos , Neutrófilos/efectos de los fármacos , Staphylococcus/fisiología , Acilcoenzima A/metabolismo , Animales , Antibacterianos/farmacología , Humanos , Lisina/química , Lisina/metabolismo , Macrófagos/patología , Ratones , Neutrófilos/patología , Fragmentos de Péptidos/química , Fragmentos de Péptidos/farmacología , Procesamiento Proteico-Postraduccional
11.
Sci Rep ; 11(1): 10295, 2021 05 13.
Artículo en Inglés | MEDLINE | ID: mdl-33986441

RESUMEN

The binding of mouse IgG3 to Fcγ receptors (FcγR) and the existence of a mouse IgG3-specific receptor have been discussed for 40 years. Recently, integrin beta-1 (ITGB1) was proposed to be a part of an IgG3 receptor involved in the phagocytosis of IgG3-coated pathogens. We investigated the interaction of mouse IgG3 with macrophage-like J774A.1 and P388D1 cells. The existence of an IgG3-specific receptor was verified using flow cytometry and a rosetting assay, in which erythrocytes clustered around the macrophage-like cells coated with an erythrocyte-specific IgG3. Our findings confirmed that receptors binding antigen-free IgG3 are present on J774A.1 and P388D1 cells. We demonstrated for the first time that the removal of N-glycans from IgG3 completely abolished its binding to the cells. Moreover, we discovered that the cells treated with Accutase did not bind IgG3, indicating that IgG3-specific receptors are substrates of this enzyme. The results of antibody-mediated blocking of putative IgG3 receptors suggested that apart from previously proposed ITGB1, FcγRII, FcγRIII, also additional, still unknown, receptor is involved in IgG3 binding. These findings indicate that there is a complex network of glycan-dependent interactions between mouse IgG3 and the surface of effector immune cells.


Asunto(s)
Colagenasas/farmacología , Inmunoglobulina G/inmunología , Macrófagos/efectos de los fármacos , Péptido Hidrolasas/farmacología , Animales , Citometría de Flujo , Glicosilación , Macrófagos/inmunología , Ratones , Unión Proteica
12.
mBio ; 12(1)2021 02 23.
Artículo en Inglés | MEDLINE | ID: mdl-33622730

RESUMEN

Cargo proteins of the type IX secretion system (T9SS) in human pathogens from the Bacteroidetes phylum invariably possess a conserved C-terminal domain (CTD) that functions as a signal for outer membrane (OM) translocation. In Porphyromonas gingivalis, the CTD of cargos is cleaved off after translocation, and anionic lipopolysaccharide (A-LPS) is attached. This transpeptidase reaction anchors secreted proteins to the OM. PorZ, a cell surface-associated protein, is an essential component of the T9SS whose function was previously unknown. We recently solved the crystal structure of PorZ and found that it consists of two ß-propeller moieties, followed by a CTD. In this study, we performed structure-based modeling, suggesting that PorZ is a carbohydrate-binding protein. Indeed, we found that recombinant PorZ specifically binds A-LPS in vitro Binding was blocked by monoclonal antibodies that specifically react with a phosphorylated branched mannan in the anionic polysaccharide (A-PS) component of A-LPS, but not with the core oligosaccharide or the lipid A endotoxin. Examination of A-LPS derived from a cohort of mutants producing various truncations of A-PS confirmed that the phosphorylated branched mannan is indeed the PorZ ligand. Moreover, purified recombinant PorZ interacted with the PorU sortase in an A-LPS-dependent manner. This interaction on the cell surface is crucial for the function of the "attachment complex" composed of PorU, PorZ, and the integral OM ß-barrel proteins PorV and PorQ, which is involved in posttranslational modification and retention of T9SS cargos on the bacterial surface.IMPORTANCE Bacteria have evolved multiple systems to transport effector proteins to their surface or into the surrounding milieu. These proteins have a wide range of functions, including attachment, motility, nutrient acquisition, and toxicity in the host. Porphyromonas gingivalis, the human pathogen responsible for severe gum diseases (periodontitis), uses a recently characterized type IX secretion system (T9SS) to translocate and anchor secreted virulence effectors to the cell surface. Anchorage is facilitated by sortase, an enzyme that covalently attaches T9SS cargo proteins to a unique anionic lipopolysaccharide (A-LPS) moiety of P. gingivalis Here, we show that the T9SS component PorZ interacts with sortase and specifically binds A-LPS. Binding is mediated by a phosphorylated branched mannan repeat in A-LPS polysaccharide. A-LPS-bound PorZ interacts with sortase with significantly higher affinity, facilitating modification of cargo proteins by the cell surface attachment complex of the T9SS.


Asunto(s)
Aminoaciltransferasas/metabolismo , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Sistemas de Secreción Bacterianos/metabolismo , Cisteína Endopeptidasas/metabolismo , Lipopolisacáridos/metabolismo , Peptidil Transferasas/metabolismo , Porphyromonas gingivalis/genética , Sistemas de Secreción Bacterianos/genética , Peptidil Transferasas/genética , Porphyromonas gingivalis/enzimología , Porphyromonas gingivalis/metabolismo , Unión Proteica , Procesamiento Proteico-Postraduccional , Transporte de Proteínas
13.
Int J Nanomedicine ; 15: 8673-8696, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33192061

RESUMEN

Polymeric nanomaterials have become a prominent area of research in the field of drug delivery. Their application in nanomedicine can improve bioavailability, pharmacokinetics, and, therefore, the effectiveness of various therapeutics or contrast agents. There are many studies for developing new polymeric nanocarriers; however, their clinical application is somewhat limited. In this review, we present new complex and multifunctional polymeric nanocarriers as promising and innovative diagnostic or therapeutic systems. Their multifunctionality, resulting from the unique chemical and biological properties of the polymers used, ensures better delivery, and a controlled, sequential release of many different therapeutics to the diseased tissue. We present a brief introduction of the classical formulation techniques and describe examples of multifunctional nanocarriers, whose biological assessment has been carried out at least in vitro. Most of them, however, also underwent evaluation in vivo on animal models. Selected polymeric nanocarriers were grouped depending on their medical application: anti-cancer drug nanocarriers, nanomaterials delivering compounds for cancer immunotherapy or regenerative medicine, components of vaccines nanomaterials used for topical application, and lifestyle diseases, ie, diabetes.


Asunto(s)
Portadores de Fármacos/administración & dosificación , Portadores de Fármacos/química , Sistemas de Liberación de Medicamentos/métodos , Nanoestructuras/química , Administración Tópica , Animales , Antineoplásicos/administración & dosificación , Humanos , Nanomedicina/métodos , Nanoestructuras/administración & dosificación , Polímeros/química , Medicina Regenerativa/métodos
14.
Colloids Surf B Biointerfaces ; 195: 111272, 2020 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-32791473

RESUMEN

The liposomes are among the most promising types of drug delivery systems but low stability significantly limits their application. Some approaches proposed to overcome this drawback may affect the liposomes toxicity profile. It is assumed that developed by us and presented here stabilization method involving formation of silicone network within the liposomal bilayer will improve elastomechanical properties of vesicles while not deteriorating their biocompatibility. The silicone-stabilized liposomes were prepared by base-catalyzed polycondensation process of the 1,3,5,7-tetramethylcyclotetrasiloxane (D4H) within the liposomal bilayer. The systematic biological in vitro studies of vesicles obtained were carried out. Moreover, the elastomechanical features investigation employing atomic force microscopy (AFM) measurements was performed. These properties of the liposome membrane are of great importance since they define the nanocarriers' stability as well as play a significant role in their cellular uptake via endocytosis. Applying the Derjaguin-Muller-Toporov (DMT) model, the elastic modulus of the silicone-stabilized liposomes was determined and compared to that characteristic for the pristine liposomes. The in vitro biological evaluation of silicone-stabilized liposomes demonstrated that these vesicles are not toxic for blood cells isolated from healthy donors and they do not induce oxidative stress in HepG2 cells. AFM results confirmed the stabilizing effect of silicone and revealed that the silicone network improves the elastomechanical properties of the resulted liposomes. This is the first report demonstrating that the silicone-stabilized liposomes retain biocompatibility of pristine liposomes' while acquire significantly better elastomechanical features.


Asunto(s)
Liposomas , Siliconas , Sistemas de Liberación de Medicamentos , Módulo de Elasticidad , Microscopía de Fuerza Atómica
15.
Int J Nanomedicine ; 14: 9587-9602, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31824153

RESUMEN

BACKGROUND: The functionalization of a nanoparticle surface with PEG (polyethylene glycol) is an approach most often used for extending nanomaterial circulation time, enhancing its delivery and retention in the target tissues, and decreasing systemic toxicity of nanocarriers and their cargos. However, because PEGylated nanomedicines were reported to induce immune response including production of anti-PEG antibodies, activation of the complement system as well as hypersensitivity reactions, hydrophilic polymers other than PEG are gaining interest as its replacement in nanomaterial functionalization. Here, we present the results of in vivo evaluation of polyelectrolyte nanocapsules with biodegradable, polyelectrolyte multilayer shells consisting of poly-l-lysine (PLL) and poly-l-glutamic (PGA) acid as a potential drug delivery system. We compared the effects of nanocapsules functionalized with two different "stealth" polymers as the external layer of tested nanocapsules was composed of PGA (PGA-terminated nanocapsules, NC-PGA) or the copolymer of poly-l-lysine and polyethylene glycol (PEG-terminated nanocapsules, NC-PEG). METHODS: Nanocapsules pharmacokinetics, biodistribution and routes of eliminations were analysed postmortem by fluorescence intensity measurement. Toxicity of intravenously injected nanocapsules was evaluated with analyses of blood morphology and biochemistry and by histological tissue analysis. DNA integrity was determined by comet assay, cytokine profiling was performed using flow cytometer and detection of antibodies specific to PEG was performed by ELISA assay. RESULTS: We found that NC-PGA and NC-PEG had similar pharmacokinetic and biodistribution profiles and both were eliminated by hepatobiliary and renal clearance. Biochemical and histopathological evaluation of long-term toxicity performed after a single as well as repeated intravenous injections of nanomaterials demonstrated that neither NC-PGA nor NC-PEG had any acute or chronic hemato-, hepato- or nephrotoxic effects. In contrast to NC-PGA, repeated administration of NC-PEG resulted in prolonged increased serum levels of a number of cytokines. CONCLUSION: Our results indicate that NC-PEG may cause undesirable activation of the immune system. Therefore, PGA compares favorably with PEG in equipping nanomaterials with stealth properties. Our research points to the importance of a thorough assessment of the potential influence of nanomaterials on the immune system.


Asunto(s)
Nanocápsulas/toxicidad , Polielectrolitos/farmacocinética , Polielectrolitos/toxicidad , Polietilenglicoles/farmacocinética , Polietilenglicoles/toxicidad , Ácido Poliglutámico/farmacocinética , Ácido Poliglutámico/toxicidad , Animales , Citocinas/sangre , Sistemas de Liberación de Medicamentos , Femenino , Fluorescencia , Ratones Endogámicos BALB C , Nanocápsulas/química , Especificidad de Órganos/efectos de los fármacos , Polielectrolitos/química , Polietilenglicoles/química , Ácido Poliglutámico/química , Rodaminas/química , Distribución Tisular , Regulación hacia Arriba/efectos de los fármacos
16.
Int J Nanomedicine ; 14: 7249-7262, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31564877

RESUMEN

BACKGROUND: Curcumin is a natural polyphenol with anti-inflammatory, chemopreventive and anticancer activity. However, its high hydrophobicity and poor bioavailability limit its medical application. The development of nanocarriers for curcumin delivery is an attractive approach to overcome its low bioavailability and fast metabolism in the liver. We synthesized a blood compatible alginate-curcumin conjugate, AA-Cur, which formed colloidally stable micelles of approximately 200 nm and, as previously shown, exerted strong cytotoxicity against mouse cancer cell lines. Here we analyze in vivo toxicity and antitumor activity of AA-Cur in two different mouse tumor models. METHOD: Potential toxicity of intravenously injected AA-Cur was evaluated by: i) analyses of blood parameters (morphology and biochemistry), ii) histology, iii) DNA integrity (comet assay), and iv) cytokine profiling (flow cytometry). Antitumor activity of AA-Cur was evaluated by measuring the growth of subcutaneously inoculated colon MC38-CEA- or orthotopically injected breast 4T1 tumor cells in control mice vs mice treated with AA-Cur. RESULTS: Injections of four doses of AA-Cur did not reveal any toxicity of the conjugate, thus indicating the safety of its use. AA-Cur elicited moderate anti-tumor activity toward colon MC38-CEA or breast 4T1 carcinomas. CONCLUSION: The tested conjugate of alginate and curcumin, AA-Cur, is non-toxic and safe, but exhibits limited anticancer activity.


Asunto(s)
Alginatos/farmacología , Alginatos/toxicidad , Curcumina/farmacología , Curcumina/toxicidad , Micelas , Pruebas de Toxicidad , Alginatos/uso terapéutico , Animales , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Antineoplásicos/toxicidad , Materiales Biocompatibles/química , Células de la Médula Ósea/metabolismo , Línea Celular Tumoral , Curcumina/administración & dosificación , Citocinas/sangre , Femenino , Humanos , Hidrodinámica , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Especificidad de Órganos
17.
Colloids Surf B Biointerfaces ; 183: 110396, 2019 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-31394424

RESUMEN

Here, we designed a novel Gadolinium (Gd) labeled drug-loaded polyelectrolyte nanocarriers for theranostics. The nanocarriers were formed via layer-by-layer technique with biodegradable polyelectrolytes: PLL (Poly-L-lysine), PLL-Gd (Gadolinium-labeled Poly-L-lysine) and PGA (Poly-L-glutamic acid). Anticancer drug (Paclitaxel) was encapsulated in the formed nanocarriers. The average size of synthesized nanocarriers was around 150 nm. The empty gadolinium labeled nanocarriers did not show any deleterious effects on tested cells (CT26-CEA, B16F10, 4T1 and PBMC), whereas encapsulated paclitaxel retained its cytotoxic/cytostatic activity. Using T2 and T1 NMR relaxation measurements with 9.4 T preclinical MRI scanner, we demonstrated that gadolinium labeled nanocarriers can be detected due to a locally altered contrast in the MR image. Thus, they may become a promising platform for future theranostic applications.


Asunto(s)
Antineoplásicos Fitogénicos/farmacología , Portadores de Fármacos , Gadolinio/química , Nanocápsulas/química , Paclitaxel/farmacología , Polilisina/química , Animales , Antineoplásicos Fitogénicos/química , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Composición de Medicamentos/métodos , Humanos , Leucocitos Mononucleares/citología , Leucocitos Mononucleares/efectos de los fármacos , Imagen por Resonancia Magnética , Ratones , Nanocápsulas/ultraestructura , Paclitaxel/química , Polielectrolitos/química , Cultivo Primario de Células , Coloración y Etiquetado/métodos , Nanomedicina Teranóstica/métodos
18.
Viruses ; 11(4)2019 04 05.
Artículo en Inglés | MEDLINE | ID: mdl-30959796

RESUMEN

Despite high similarity of canine respiratory coronavirus (CRCoV), bovine coronavirus, (BCoV) and human coronavirus OC43 (HCoV-OC43), these viruses differ in species specificity. For years it was believed that they share receptor specificity, utilizing sialic acids for cell surface attachment, internalization, and entry. Interestingly, careful literature analysis shows that viruses indeed bind to the cell surface via sialic acids, but there is no solid data that these moieties mediate virus entry. In our study, using a number of techniques, we showed that all three viruses are indeed able to bind to sialic acids to a different extent, but these molecules render the cells permissive only for the clinical strain of HCoV-OC43, while for others they serve only as attachment receptors. CRCoV and BCoV appear to employ human leukocyte antigen class I (HLA-1) as the entry receptor. Furthermore, we identified heparan sulfate as an alternative attachment factor, but this may be related to the cell culture adaptation, as in ex vivo conditions, it does not seem to play a significant role. Summarizing, we delineated early events during CRCoV, BCoV, and HCoV-OC43 entry and systematically studied the attachment and entry receptor utilized by these viruses.


Asunto(s)
Coronavirus Humano OC43/fisiología , Coronavirus Bovino/fisiología , Coronavirus Canino/fisiología , Receptores Virales/análisis , Acoplamiento Viral , Células Cultivadas , Heparitina Sulfato/metabolismo , Antígenos de Histocompatibilidad Clase I/metabolismo , Humanos , Ácidos Siálicos/metabolismo
19.
Biotechnol J ; 14(2): e1700722, 2019 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-29781256

RESUMEN

Mammalian cell perfusion cultures represent a promising alternative to the current fed-batch technology for the production of various biopharmaceuticals. Long-term operation at a fixed viable cell density (VCD) requires a viable culture and a constant removal of excessive cells. Product loss in the cell removing bleed stream deteriorates the process yield. In this study, the authors investigate the use of chemical and environmental growth inhibition on culture performance by either adding valeric acid (VA) to the production media or by reducing the culture temperature (33.0 °C) with respect to control conditions (36.5 °C, no VA). Low temperature significantly reduces cellular growth, thus, resulting in lower bleed rates accompanied by a reduced product loss of 11% compared to 26% under control conditions. Additionally, the cell specific productivity of the target protein improves and maintained stable leading to media savings per mass of product. VA shows initially an inhibitory effect on cellular growth. However, cells seemed to adapt to the presence of the inhibitor resulting in a recovery of the cellular growth. Cell cycle and Western blot analyses support the observed results. This work underlines the role of temperature as a key operating variable for the optimization of perfusion cultures.


Asunto(s)
Técnicas de Cultivo Celular por Lotes/métodos , Reactores Biológicos , Inhibidores de Crecimiento/farmacología , Animales , Células CHO , Proliferación Celular/efectos de los fármacos , Frío , Cricetulus , Ácidos Pentanoicos/farmacología
20.
Int J Nanomedicine ; 13: 5159-5172, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30233178

RESUMEN

BACKGROUND: Toxicity of nanomaterials is one of the most important factors limiting their medical application. Evaluation of in vitro nanotoxicity allows for the identification and elimination of most of the toxic materials prior to animal testing. The current knowledge of the possible side effects of biodegradable nanomaterials, such as liposomes and polymeric organic nanoparticles, is limited. Previously, we developed a potential drug delivery system in the form of nanocapsules with polyelectrolyte, biodegradable shells consisting of poly-l-lysine and poly-l-glutamic acid (PGA), formed by the layer-by-layer adsorption technique. METHODS: Hemolysis assay, viability tests, flow cytometry analysis of vascular cell adhesion molecule-1 expression on endothelium, analysis of nitric oxide production, measurement of intracellular reactive oxygen species levels, detection of antioxidant enzyme activity, and analysis of DNA damage with comet assay were performed to study the in vitro toxicity of nanocapsules. RESULTS: In this work, we present the results of an in vitro analysis of toxicity of five-layer positively charged poly-l-lysine-terminated nanocapsules (NC5), six-layer negatively charged PGA-terminated nanocapsules (NC6) and five-layer PEGylated nanocapsules (NC5-PEG). PGA and polyethylene glycol (PEG) were used as two different "stealth" polymers. Of all the polyelectrolyte nanocapsules tested for blood compatibility, only cationic NC5 showed acute toxicity toward blood cells, expressed as hemolysis and aggregation. Neither NC6 nor NC5-PEG had proinflammatory activity evaluated through changes in the expression of NF-κB-dependent genes, iNOS and vascular cell adhesion molecule-1, induced oxidative stress, or promoted DNA damage in various cells. CONCLUSION: Our studies clearly indicate that PGA-coated (negatively charged) and PEGylated polyelectrolyte nanocapsules do not show in vitro toxicity, and their potential as a drug delivery system may be safely studied in vivo.


Asunto(s)
Nanocápsulas/toxicidad , Polielectrolitos/toxicidad , Pruebas de Toxicidad , Animales , Muerte Celular/efectos de los fármacos , Daño del ADN , Eritrocitos/efectos de los fármacos , Eritrocitos/metabolismo , Hemólisis/efectos de los fármacos , Células Hep G2 , Humanos , Inflamación/patología , Leucocitos Mononucleares/efectos de los fármacos , Leucocitos Mononucleares/metabolismo , Ratones , Mutágenos/toxicidad , Estrés Oxidativo/efectos de los fármacos , Tamaño de la Partícula , Polielectrolitos/síntesis química , Polietilenglicoles/química
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...