Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 15.460
Filtrar
1.
Int J Biol Macromol ; 267(Pt 1): 131328, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38574901

RESUMEN

The management of invasive fungal infections in humans poses significant challenges due to the intricate nature of the treatment, which is both arduous and costly, necessitating routine diagnostic procedures. Consequently, this investigation aimed to formulate a chitosan-based nanoemulsion (CS NEMs) incorporating the antifungal agent undecanoic acid (UDA), characterizing these NEMs and assessing their antifungal efficacy against both filamentous and non-filamentous fungal pathogens. The CS-based UDA NEMs were synthesized by introducing the surfactant Triton X-100 and the stabilizer glycerol. Nanoparticle tracking analysis (NTA) and SEM demonstrated the CS-UDA NEMs with an average size of 145 nm and 164.5 ± 24 nm, respectively. The successful formation of CS-UDA NEMs was verified through FTIR and XRD. CS-UDA NEMs exhibited exceptional inhibition against Aspergillus flavus, Aspergillus fumigatus, Aspergillus niger, and Candida albicans with MFC of 500, 500, 250 and 250 µg/mL, respectively. Additionally, CS-UDA NEMs displayed comparatively lower antioxidant activity as determined by DPPH and ABTS radical scavenging assays. Importantly, CS-UDA NEMs demonstrated no cytotoxic effects on NIH3T3 cells even at higher concentration (1000 µg/mL), as confirmed by cell viability and fluorescent staining assays. In conclusion, this study suggests that the developed CS-UDA NEMs hold promise as potent antifungal agents with diverse potential applications.


Asunto(s)
Antifúngicos , Quitosano , Emulsiones , Ácidos Grasos , Quitosano/química , Quitosano/farmacología , Antifúngicos/farmacología , Antifúngicos/química , Emulsiones/química , Ácidos Grasos/química , Ratones , Animales , Materiales Biocompatibles/química , Materiales Biocompatibles/farmacología , Pruebas de Sensibilidad Microbiana , Nanopartículas/química , Células 3T3 NIH , Antioxidantes/farmacología , Antioxidantes/química , Candida albicans/efectos de los fármacos
2.
Int J Biol Macromol ; 267(Pt 2): 131375, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38604424

RESUMEN

Compostable zein-polycaprolactone (PZ) electrospun nanofiber integrated with different concentrations of Aster yomena extract loaded halloysite nanotubes (A. yomena-HNT) as bioactive nanofibrous food packaging is reported. SEM micrographs reveal heterogeneous nanofibers. A. yomena extract used in the study showed weak antioxidant activity with AAI and TEAC values of 0.229 and 0.346. In vitro, release profile over 7 days of A. yomena indicates a controlled, sustained, and prolonged release. The prepared nanofibers were effective against both gram-positive and gram-negative bacteria. The prepared composite nanofibers were rendered biocompatible and nontoxic when subjected to WST-1 and LDH assay after incubating with NIH 3T3 mouse fibroblast cell line. PZ-15 nanofiber packaging showed the best postharvest quality preservation in Black mulberry fruits after 4 days of storage at 25 °C and 85 % Rh. Moreover, the in vitro decomposition test reveals that the fabricated nanofibers decompose in the soil and do not pose as a threat to the environment.


Asunto(s)
Arcilla , Embalaje de Alimentos , Nanofibras , Nanotubos , Extractos Vegetales , Poliésteres , Zeína , Nanofibras/química , Poliésteres/química , Zeína/química , Ratones , Animales , Arcilla/química , Embalaje de Alimentos/métodos , Nanotubos/química , Células 3T3 NIH , Extractos Vegetales/química , Extractos Vegetales/farmacología , Antioxidantes/farmacología , Antioxidantes/química , Antibacterianos/farmacología , Antibacterianos/química
3.
Biomed Mater ; 19(3)2024 Apr 26.
Artículo en Inglés | MEDLINE | ID: mdl-38574669

RESUMEN

Recently,in vitromodels of intestinal mucosa have become important tools for drug screening and studying the physiology and pathology of the intestine. These models enable the examination of cellular behavior in diseased states or in reaction to alterations in the microenvironment, potentially serving as alternatives to animal models. One of the major challenges in constructing physiologically relevantin vitromodels of intestinal mucosa is the creation of three-dimensional microstructures that accurately mimic the integration of intestinal epithelium and vascularized stroma. Here, core-shell alginate (Alg) microspheres were generated to create the compartmentalized extracellular matrix microenvironment needed to simulate the epithelial and vascularized stromal compartments of the intestinal mucosa. We demonstrated that NIH-3T3 and human umbilical vein endothelial cells embedded in the core of the microspheres can proliferate and develop a vascular network, while human colorectal adenocarcinoma cells (Caco-2) can form an epithelial monolayer in the shell. Compared to Caco-2 monolayer encapsulated within the shell, the presence of the vascularized stroma enhances their proliferation and functionality. As such, our core-shell Alg microspheres provide a valuable method for generatingin vitromodels of vascularized intestinal mucosa with epithelial and vascularized stroma arranged in a spatially relevant manner and demonstrating near-physiological functionality.


Asunto(s)
Alginatos , Proliferación Celular , Células Endoteliales de la Vena Umbilical Humana , Mucosa Intestinal , Microesferas , Ingeniería de Tejidos , Alginatos/química , Humanos , Mucosa Intestinal/metabolismo , Animales , Ratones , Células CACO-2 , Ingeniería de Tejidos/métodos , Células 3T3 NIH , Matriz Extracelular/metabolismo , Andamios del Tejido/química , Ácidos Hexurónicos/química
4.
ACS Appl Mater Interfaces ; 16(15): 18327-18343, 2024 Apr 17.
Artículo en Inglés | MEDLINE | ID: mdl-38588343

RESUMEN

58S bioactive glass (BG) has effective biocompatibility and bioresorbable properties for bone tissue engineering; however, it has limitations regarding antibacterial, antioxidant, and mechanical properties. Therefore, we have developed BGAC biocomposites by reinforcing 58S BG with silver and ceria nanoparticles, which showed effective bactericidal properties by forming inhibited zones of 2.13 mm (against Escherichia coli) and 1.96 mm (against Staphylococcus aureus; evidenced by disc diffusion assay) and an increment in the antioxidant properties by 39.9%. Moreover, the elastic modulus, hardness, and fracture toughness were observed to be increased by ∼84.7% (∼51.9 GPa), ∼54.5% (∼3.4 GPa), and ∼160% (∼1.3 MPam1/2), whereas the specific wear rate was decreased by ∼55.2% (∼1.9 × 10-11 m3/Nm). X-ray diffraction, high-resolution transmission electron microscopy, and field emission scanning electron microscopy confirmed the fabrication of biocomposites and the uniform distribution of the nanomaterials in the BG matrix. The addition of silver nanoparticles in the 58S BG matrix (in BGA) increased mechanical properties by composite strengthening and bactericidal properties by damaging the cytoplasmic membrane of bacterial cells. The addition of nanoceria in 58S BG (BGC) increased the antioxidant properties by 44.5% (as evidenced by the 2,2-diphenyl-1-picrylhydrazyl assay). The resazurin reduction assay and MTT assay confirmed the effective cytocompatibility for BGAC biocomposites against mouse embryonic fibroblast cells (NIH3T3) and mouse bone marrow stromal cells. Overall, BGAC resulted in mechanical properties comparable to those of cancellous bone, and its effective antibacterial and cytocompatibility properties make it a good candidate for bone healing.


Asunto(s)
Cerio , Nanopartículas del Metal , Plata , Animales , Ratones , Antioxidantes , Células 3T3 NIH , Fibroblastos , Antibacterianos/farmacología , Vidrio
5.
Int J Mol Sci ; 25(8)2024 Apr 13.
Artículo en Inglés | MEDLINE | ID: mdl-38673904

RESUMEN

Chagas disease is one of the world's neglected tropical diseases, caused by the human pathogenic protozoan parasite Trypanosoma cruzi. There is currently a lack of effective and tolerable clinically available therapeutics to treat this life-threatening illness and the discovery of modern alternative options is an urgent matter. T. cruzi glucokinase (TcGlcK) is a potential drug target because its product, d-glucose-6-phosphate, serves as a key metabolite in the pentose phosphate pathway, glycolysis, and gluconeogenesis. In 2019, we identified a novel cluster of TcGlcK inhibitors that also exhibited anti-T. cruzi efficacy called the 3-nitro-2-phenyl-2H-chromene analogues. This was achieved by performing a target-based high-throughput screening (HTS) campaign of 13,040 compounds. The selection criteria were based on first determining which compounds strongly inhibited TcGlcK in a primary screen, followed by establishing on-target confirmed hits from a confirmatory assay. Compounds that exhibited notable in vitro trypanocidal activity over the T. cruzi infective form (trypomastigotes and intracellular amastigotes) co-cultured in NIH-3T3 mammalian host cells, as well as having revealed low NIH-3T3 cytotoxicity, were further considered. Compounds GLK2-003 and GLK2-004 were determined to inhibit TcGlcK quite well with IC50 values of 6.1 µM and 4.8 µM, respectively. Illuminated by these findings, we herein screened a small compound library consisting of thirteen commercially available 3-nitro-2-phenyl-2H-chromene analogues, two of which were GLK2-003 and GLK2-004 (compounds 1 and 9, respectively). Twelve of these compounds had a one-point change from the chemical structure of GLK2-003. The analogues were run through a similar primary screening and confirmatory assay protocol to our previous HTS campaign. Subsequently, three in vitro biological assays were performed where compounds were screened against (a) T. cruzi (Tulahuen strain) infective form co-cultured within NIH-3T3 cells, (b) T. brucei brucei (427 strain) bloodstream form, and (c) NIH-3T3 host cells alone. We report on the TcGlcK inhibitor constant determinations, mode of enzyme inhibition, in vitro antitrypanosomal IC50 determinations, and an assessment of structure-activity relationships. Our results reveal that the 3-nitro-2-phenyl-2H-chromene scaffold holds promise and can be further optimized for both Chagas disease and human African trypanosomiasis early-stage drug discovery research.


Asunto(s)
Benzopiranos , Glucoquinasa , Tripanocidas , Trypanosoma cruzi , Trypanosoma cruzi/efectos de los fármacos , Trypanosoma cruzi/enzimología , Tripanocidas/farmacología , Tripanocidas/química , Animales , Ratones , Benzopiranos/farmacología , Benzopiranos/química , Glucoquinasa/metabolismo , Glucoquinasa/antagonistas & inhibidores , Ensayos Analíticos de Alto Rendimiento , Enfermedad de Chagas/tratamiento farmacológico , Enfermedad de Chagas/parasitología , Inhibidores Enzimáticos/farmacología , Inhibidores Enzimáticos/química , Células 3T3 NIH , Descubrimiento de Drogas/métodos , Humanos , Simulación del Acoplamiento Molecular , Relación Estructura-Actividad
6.
Cells ; 13(8)2024 Apr 17.
Artículo en Inglés | MEDLINE | ID: mdl-38667312

RESUMEN

The assessment of nanoparticle cytotoxicity is challenging due to the lack of customized and standardized guidelines for nanoparticle testing. Nanoparticles, with their unique properties, can interfere with biochemical test methods, so multiple tests are required to fully assess their cellular effects. For a more reliable and comprehensive assessment, it is therefore imperative to include methods in nanoparticle testing routines that are not affected by particles and allow for the efficient integration of additional molecular techniques into the workflow. Digital holographic microscopy (DHM), an interferometric variant of quantitative phase imaging (QPI), has been demonstrated as a promising method for the label-free assessment of the cytotoxic potential of nanoparticles. Due to minimal interactions with the sample, DHM allows for further downstream analyses. In this study, we investigated the capabilities of DHM in a multimodal approach to assess cytotoxicity by directly comparing DHM-detected effects on the same cell population with two downstream biochemical assays. Therefore, the dry mass increase in RAW 264.7 macrophages and NIH-3T3 fibroblast populations measured by quantitative DHM phase contrast after incubation with poly(alkyl cyanoacrylate) nanoparticles for 24 h was compared to the cytotoxic control digitonin, and cell culture medium control. Viability was then determined using a metabolic activity assay (WST-8). Moreover, to determine cell death, supernatants were analyzed for the release of the enzyme lactate dehydrogenase (LDH assay). In a comparative analysis, in which the average half-maximal effective concentration (EC50) of the nanocarriers on the cells was determined, DHM was more sensitive to the effect of the nanoparticles on the used cell lines compared to the biochemical assays.


Asunto(s)
Nanopartículas , Animales , Ratones , Células 3T3 NIH , Nanopartículas/toxicidad , Nanopartículas/química , Células RAW 264.7 , Supervivencia Celular/efectos de los fármacos , Holografía/métodos , Imágenes de Fase Cuantitativa
7.
Biomed Phys Eng Express ; 10(3)2024 Apr 02.
Artículo en Inglés | MEDLINE | ID: mdl-38564254

RESUMEN

The high demand for bone grafts has motivated the development of implants with excellent osteogenic activity, whereas the risk of implant-associated infection, particularly given the rise of antimicrobial resistance, has compelled the development of implants with innovative antimicrobial strategies in which a small amount of bactericidal agent can effectively kill a wide range of bacteria. To induce antibacterial property, the surface of Grade-5 bone plate titanium implants used in clinical applications was modified using direct current (DC) sputter coating followed by thermal annealing. The 15 nm silver film-coated implants were thermally annealed in the furnace for 15 min at 750 °C. The modified implant surface's antibacterial efficacy againstEscherichia coli(E. coli),Staphylococcus aureus(S. aureus),Salmonella typhi, andMethicillin-resistant staphylococcus aureusbacteria has been assessed using a colony-forming assay. On the modified implant surface, the growth ofE. coliandS. aureusbacteria is reduced by 99.72%, while highly drug-resistant bacteria are inhibited by 96.59%. The MTT assay was used to assess the cytotoxicity of the modified bone-implant surface against NIH3T3 mouse fibroblast cells. The modified bone-implant surface promoted fibroblast growth and demonstrated good cytocompatibility. Furthermore, the mechanical properties of the implant were not harmed by this novel surface modification method. This method is simple and provides new insight into surface modification of commercial metallic implants to have effective antibacterial properties against various classes of bacteria.


Asunto(s)
Aleaciones , Staphylococcus aureus Resistente a Meticilina , Plata , Animales , Ratones , Titanio , Placas Óseas , Escherichia coli , Células 3T3 NIH , Staphylococcus aureus , Antibacterianos/farmacología
8.
Int J Biol Macromol ; 267(Pt 2): 131389, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38582461

RESUMEN

This work developed Acer tegmentosum extract-mediated silver nanoparticles (AgNPs) loaded chitosan (CS)/alginic acid (AL) scaffolds (CS/AL-AgNPs) to enhance the healing of E. coli-infected wounds. The SEM-EDS and XRD results revealed the successful formation of the CS/AL-AgNPs. FTIR analysis evidenced that the anionic group of AL (-COO-) and cationic amine groups of CS (-NH3+) were ionically crosslinked to form scaffold (CS/AL). The CS/AL-AgNPs exhibited significant antimicrobial activity against both Gram-positive (G+) and Gram-negative (G-) bacterial pathogens, while being non-toxic to red blood cells (RBCs), the hen's egg chorioallantoic membrane (HET-CAM), and a non-cancerous cell line (NIH3T3). Treatment with CS/AL-AgNPs significantly accelerated the healing of E. coli-infected wounds by regulating the collagen deposition and blood parameters as evidenced by in vivo experiments. Overall, these findings suggest that CS/AL-AgNPs are promising for the treatment of infected wounds.


Asunto(s)
Acer , Alginatos , Antibacterianos , Quitosano , Escherichia coli , Nanopartículas del Metal , Extractos Vegetales , Plata , Cicatrización de Heridas , Quitosano/química , Quitosano/farmacología , Nanopartículas del Metal/química , Plata/química , Plata/farmacología , Animales , Cicatrización de Heridas/efectos de los fármacos , Escherichia coli/efectos de los fármacos , Ratones , Acer/química , Extractos Vegetales/química , Extractos Vegetales/farmacología , Células 3T3 NIH , Antibacterianos/farmacología , Antibacterianos/química , Alginatos/química , Alginatos/farmacología , Infecciones por Escherichia coli/tratamiento farmacológico , Andamios del Tejido/química
9.
J Vis Exp ; (204)2024 Feb 16.
Artículo en Inglés | MEDLINE | ID: mdl-38436407

RESUMEN

Most living organisms possess circadian rhythms, which are biological processes that occur within a period of approximately 24 h and regulate a diverse repertoire of cellular and physiological processes ranging from sleep-wake cycles to metabolism. This clock mechanism entrains the organism based on environmental changes and coordinates the temporal regulation of molecular and physiological events. Previously, it was demonstrated that autonomous circadian rhythms are maintained even at the single-cell level using cell lines such as NIH3T3 fibroblasts, which were instrumental in uncovering the mechanisms of circadian rhythms. However, these cell lines are homogeneous cultures lacking multicellularity and robust intercellular communications. In the past decade, extensive work has been performed on the development, characterization, and application of 3D organoids, which are in vitro multicellular systems that resemble in vivo morphological structures and functions. This paper describes a protocol for detecting circadian rhythms using a bioluminescent reporter in human intestinal enteroids, which enables the investigation of circadian rhythms in multicellular systems in vitro.


Asunto(s)
Comunicación Celular , Organoides , Humanos , Ratones , Animales , Células 3T3 NIH , Ritmo Circadiano , Fibroblastos
10.
Int J Pharm ; 654: 123994, 2024 Apr 10.
Artículo en Inglés | MEDLINE | ID: mdl-38484859

RESUMEN

Small interfering RNA (siRNA) holds great potential to treat many difficult-to-treat diseases, but its delivery remains the central challenge. This study aimed at investigating the suitability of polymer-lipid hybrid nanomedicines (HNMeds) as novel siRNA delivery platforms for locoregional therapy of glioblastoma. Two HNMed formulations were developed from poly(lactic-co-glycolic acid) polymer and a cationic lipid: 1,2-dioleoyl-3-trimethylammonium-propane (DOTAP) or 3ß-[N-(N',N'-dimethylaminoethane)-carbamoyl]cholesterol (DC-Chol). After characterization of the HNMeds, a model siRNA was complexed onto their surface to form HNMed/siRNA complexes. The physicochemical properties and siRNA binding ability of complexes were assessed over a range of nitrogen-to-phosphate (N/P) ratios to optimize the formulations. At the optimal N/P ratio of 10, complexes effectively bound siRNA and improved its protection from enzymatic degradation. Using the NIH3T3 mouse fibroblast cell line, DOTAP-based HNMeds were shown to possess higher cytocompatibility in vitro over the DC-Chol-based ones. As proof-of-concept, uptake and bioefficacy of formulations were also assessed in vitro on U87MG human glioblastoma cell line expressing luciferase gene. Complexes were able to deliver anti-luciferase siRNA and induce a remarkable suppression of gene expression. Noteworthy, the effect of DOTAP-based formulation was not only about three-times higher than DC-Chol-based one, but also comparable to lipofectamine model transfection reagent. These findings set the basis to exploit this nanosystem for silencing relevant GB-related genes in further in vitro and in vivo studies.


Asunto(s)
Ácidos Grasos Monoinsaturados , Glioblastoma , Liposomas , Compuestos de Amonio Cuaternario , Ratones , Animales , Humanos , Liposomas/química , Polímeros/química , ARN Interferente Pequeño , Glioblastoma/genética , Glioblastoma/terapia , Células 3T3 NIH , Nanomedicina , Lípidos/química
11.
Cell Biol Int ; 48(5): 726-736, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38439187

RESUMEN

Cellular senescence is an irreversible cell-cycle arrest in response to a variety of cellular stresses, which contribute to the pathogenesis of a variety of age-related degenerative diseases. However, effective antisenescence strategies are still lacking. Drugs that selectively target senescent cells represent an intriguing therapeutic strategy to delay aging and age-related diseases. Thus, we thought to investigate the effects of dihydroartemisinin (DHA) on senescent cells and elucidated its mechanisms underlying aging. Stress-induced premature senescence (SIPS) model was built in NIH3T3 cells using H2O2 and evaluated by ß-galactosidase staining. Cells were exposed to DHA and subjected to cellular activity assays including viability, ferroptosis, and autophagy. The number of microtubule-associated protein light-chain 3 puncta was detected by immunofluorescence staining. The iron content was assessed by spectrophotometer and intracellular reactive oxygen species (ROS) was measured by fluorescent probe dichlorodihydrofluorescein diacetate. We found that DHA triggered senescent cell death via ferroptosis. DHA accelerated ferritin degradation via promoting autophagy, increasing the iron contents, promoting ROS accumulation, thus leading to ferroptotic cell death in SIPS cells. In addition, autophagy inhibitor BafA1 preconditioning inhibited ferroptosis induced by DHA. Moreover, Atg5 silencing and autophagy inhibitor BafA1 preconditioning inhibited ferroptosis induced by DHA. We also revealed that the expression of p-AMP-activated protein kinase (AMPK) and p-mammalian target of rapamycin (mTOR) in senescent cells was downregulated. These results suggested that DHA may be a promising drug candidate for clearing senescent cells by inducing autophagy-dependent ferroptosis via AMPK/mTOR signaling pathway.


Asunto(s)
Proteínas Quinasas Activadas por AMP , Artemisininas , Ferroptosis , Animales , Ratones , Proteínas Quinasas Activadas por AMP/metabolismo , Autofagia , Senescencia Celular , Peróxido de Hidrógeno/farmacología , Hierro , Células 3T3 NIH , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal , Serina-Treonina Quinasas TOR/metabolismo
12.
Int J Biol Macromol ; 266(Pt 1): 131257, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38554908

RESUMEN

The infected wounds pose one of the major threats to human health today. To address this issue, it is necessary to develop innovative wound dressings with superior antibacterial activity and other properties. Due to its potent antibacterial, antioxidant, and immune-boosting properties, epigallocatechin gallate (EGCG) has been widely utilized. In this study, a multifunctional curdlan hydrogel loading EGCG (Cur-EGCGH3) was designed. Cur-EGCGH3 exhibited excellent physicochemical properties, good biocompatibility, hemostatic, antibacterial, and antioxidant activities. Also, ELISA data showed that Cur-EGCGH3 stimulated macrophages to secrete pro-inflammatory and pro-regenerative cytokines. Cell scratch results indicated that Cur-EGCGH3 promoted the migration of NIH3T3 and HUVECs. In vivo experiments confirmed that Cur-EGCGH3 could inhibit bacterial infection of the infected wounds, accelerate hemostasis, and promote epithelial regeneration and collagen deposition. These results demonstrated that Cur-EGCGH3 holds promise for promoting healing of the infected wounds.


Asunto(s)
Antibacterianos , Catequina , Catequina/análogos & derivados , Hemostáticos , Hidrogeles , Cicatrización de Heridas , beta-Glucanos , Catequina/farmacología , Catequina/química , Animales , Cicatrización de Heridas/efectos de los fármacos , Ratones , beta-Glucanos/química , beta-Glucanos/farmacología , Hidrogeles/química , Hidrogeles/farmacología , Antibacterianos/farmacología , Antibacterianos/química , Humanos , Células 3T3 NIH , Hemostáticos/farmacología , Hemostáticos/química , Infección de Heridas/tratamiento farmacológico , Infección de Heridas/microbiología , Antioxidantes/farmacología , Antioxidantes/química , Células Endoteliales de la Vena Umbilical Humana/efectos de los fármacos
13.
Int J Biol Macromol ; 262(Pt 2): 130140, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38365152

RESUMEN

The current research work focuses on preparing the polycaprolactone (PCL) based nanocomposite films embedded with surface modified Halloysite Nanotube (HNT). The avenue of the study is to unravel the applicability of polymer nanocomposites for wound healing. The flexible property of HNT was taken as the major force to accomplish the addition of biopolymer pectin onto its surface. Functionalization of HNT with pectin has certainly enhanced its binding nature with the polymer. The PCL nanocomposite films were characterized by several promising techniques such as FTIR, XRD, DSC-TGA, FESEM, TEM, AFM, and mechanical properties were too examined along. When compared to the plane PCL film, the nanocomposite films manifested favorable results in terms of mechanical and chemical properties. Additionally, biometric studies such as in-vitro swelling, enzymatic degradation, and hemolysis performed on the films gave extremely good results. The haemolytic percentage recorded for the films exhibited a steady decrease with increasing amount of nanofillers. The MTT assay showed cell proliferation and its increase as the embedded HNT is more in the matrix. Wound closure study performed on NIH3T3 cell line with 1, 3 and 5wt% of films has given a strong proof for the involvement of polymer and HNT in the healing procedure.


Asunto(s)
Nanocompuestos , Nanotubos , Poliésteres , Ratones , Animales , Arcilla/química , Pectinas/farmacología , Pectinas/química , Células 3T3 NIH , Cicatrización de Heridas , Polímeros , Nanotubos/química , Nanocompuestos/química
14.
Biomed Mater ; 19(3)2024 Mar 20.
Artículo en Inglés | MEDLINE | ID: mdl-38417147

RESUMEN

The present work focuses on developing 5% w/v oxidized alginate (alginate di aldehyde, ADA)-7.5% w/v gelatin (GEL) hydrogels incorporating 0.25% w/v silk fibroin (SF) and loaded with 0.3% w/v Cu-Ag doped mesoporous bioactive glass nanoparticles (Cu-Ag MBGNs). The microstructural, mechanical, and biological properties of the composite hydrogels were characterized in detail. The porous microstructure of the developed ADA-GEL based hydrogels was confirmed by scanning electron microscopy, while the presence of Cu-Ag MBGNs in the synthesized hydrogels was determined using energy dispersive x-ray spectroscopy. The incorporation of 0.3% w/v Cu-Ag MBGNs reduced the mechanical properties of the synthesized hydrogels, as investigated using micro-tensile testing. The synthesized ADA-GEL loaded with 0.25% w/v SF and 0.3% w/v Cu-Ag MBGNs showed a potent antibacterial effect againstEscherichia coliandStaphylococcus aureus. Cellular studies using the NIH3T3-E1 fibroblast cell line confirmed that ADA-GEL films incorporated with 0.3% w/v Cu-Ag MBGNs exhibited promising cellular viability as compared to pure ADA-GEL (determined by WST-8 assay). The addition of SF improved the biocompatibility, degradation rate, moisturizing effects, and stretchability of the developed hydrogels, as determinedin vitro. Such multimaterial hydrogels can stimulate angiogenesis and exhibit desirable antibacterial properties. Therefore further (in vivo) tests are justified to assess the hydrogels' potential for wound dressing and skin tissue healing applications.


Asunto(s)
Fibroínas , Nanopartículas , Ratones , Animales , Hidrogeles/química , Fibroínas/química , Células 3T3 NIH , Alginatos/química , Gelatina/química , Antibacterianos , Nanopartículas/química
15.
Hum Exp Toxicol ; 43: 9603271241231947, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38324556

RESUMEN

Objectives: Doxorubicin (DOX) is a highly effective chemotherapeutic used to treat many adult and pediatric cancers, such as solid tumors, leukemia, lymphomas and breast cancer. It can also cause injuries to multiple organs, including the heart, liver, and brain or kidney, although cardiotoxicity is the most prominent side effect of DOX. In this study, we examined the potential effects of DOX on autophagy activity in two different mouse fibroblasts.Methods: Mouse embryonic fibroblasts (NIH3T3) and mouse primary cardiac fibroblasts (CFs) were treated with DOX to assess changes in the expression of two commonly used autophagy protein markers, LC3II and p62. We also examined the effects of DOX the on expression of key genes that encode components of the molecular machinery and regulators modulating autophagy in response to both extracellular and intracellular signals.Results: We observed that LC3II levels increased and p62 levels decreased following the DOX treatment in NIH3T3 cells. However, similar effects were not observed in primary cardiac fibroblasts. In addition, DOX treatment induced the upregulation of a significant number of genes involved in autophagy in NIH3T3 cells, but not in primary cardiac fibroblasts.Conclusions: Taken together, these results indicate that DOX upregulates autophagy in fibroblasts in a cell-specific manner.


Asunto(s)
Estrés Oxidativo , Transducción de Señal , Humanos , Niño , Animales , Ratones , Células 3T3 NIH , Fibroblastos , Doxorrubicina/toxicidad , Autofagia , Cardiotoxicidad/metabolismo , Miocitos Cardíacos/metabolismo , Apoptosis
16.
Int J Nanomedicine ; 19: 1539-1556, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38406603

RESUMEN

Purpose: Persistent Infections and inflammation are associated with impaired wound healing in diabetic patients. There is a pressing demand for innovative antimicrobial strategies to address infections arising from antibiotic-resistant bacteria. Polymer-modified gold nanoparticles (AuNPs) show broad-spectrum antibacterial properties and significant biocompatibility. This study investigated the antibacterial and wound healing efficacy of hydrogel dressings conjugated with chitosan-AuNPs in diabetic model rats. Methods: Chitosan (CS)-functionalized gold nanoparticles (CS-AuNPs) were incorporated into hydrogel dressings (Gel/CS-AuNPs), which were formulated through the chemical cross-linking of gelatin with sodium alginate (SA). The basic characteristics of Gel/CS-AuNPs were analyzed by TEM, SEM, XRD, and UV-visible spectra. Rheological, swelling, degradation, and adhesive properties of Gel/CS-AuNPs were also determined. In vitro anti-bactericidal effects of the Gel/CS-AuNPs were analyzed with E. coli, S. aureus, and MRSA. In vitro biocompatibility of the Gel/CS-AuNPs was evaluated using NIH3T3 cells. The in vivo antibacterial and wound healing efficacy of the Gel/CS-AuNPs was analyzed in the diabetic wound model rats. Histological and immunofluorescence staining were performed to determine the status of angiogenesis, epithelization, inflammation response, and collagen deposition. Results: Gel/CS-AuNPs demonstrated significant high biodegradability, water absorption bactericidal, and biocompatibility, and slight adhesiveness. Gel/CS-AuNPs exhibited pronounced antibacterial efficacy against gram-negative, gram-positive, and MRSA in a CS-AuNPs-dose-dependent manner. In the diabetic wound model rats, Gel/CS-AuNPs effectively killed MRSA, reduced inflammation, and promoted angiogenesis and collagen deposition and remodeling at the wound site. As a result, Gel/CS-AuNPs expedited the recovery process for infected diabetic wounds. Among the hydrogels with different CS-AuNPs concentrations, Gel/CS-Au25 with 25% CS-AuNPs showed the best bactericidal and wound healing performance. Conclusion: Gel/CS-AuNPs significantly improve the healing of MRSA-infected diabetic wounds in the rat model. Therefore, Gel/CS-AuNPs show great promise for the treatment of diabetic infection wound healing.


Asunto(s)
Quitosano , Diabetes Mellitus , Nanopartículas del Metal , Humanos , Ratones , Ratas , Animales , Antibacterianos/farmacología , Antibacterianos/química , Quitosano/química , Oro/química , Staphylococcus aureus/metabolismo , Hidrogeles/química , Escherichia coli , Células 3T3 NIH , Nanopartículas del Metal/química , Cicatrización de Heridas , Colágeno/metabolismo , Bacterias/metabolismo , Inflamación
17.
Int J Nanomedicine ; 19: 1749-1766, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38414527

RESUMEN

Purpose: Triple-negative breast cancer (TNBC) is the most lethal subtype of breast cancer. However, the effect of current treatment strategies by inducing tumor cell apoptosis alone is not satisfactory. The growth, metastasis and treatment sensitivity of tumors can be strongly influenced by cancer-associated fibroblasts (CAFs) in the microenvironment. Effective cancer therapies may need to target not only the tumor cells directly but also the CAFs that protect them. Methods: Celastrol and small-sized micelles containing betulinic acid were co-encapsulated into liposomes using the thin-film hydration method (CL@BM). Folic acid was further introduced to modify liposomes as the targeting moiety (F/CL@BM). We established a novel NIH3T3+4T1 co-culture model to mimic the tumor microenvironment and assessed the nanocarrier's inhibitory effects on CAFs-induced drug resistance and migration in the co-culture model. The in vivo biological distribution, fluorescence imaging, biological safety evaluation, and combined therapeutic effect evaluation of the nanocarrier were carried out based on a triple-negative breast cancer model. Results: In the present study, a novel multifunctional nano-formulation was designed by combining the advantages of sequential release, co-loading of tretinoin and betulinic acid, and folic acid-mediated active targeting. As expected, the nano-formulation exhibited enhanced cytotoxicity in different cellular models and effectively increased drug accumulation at the tumor site by disrupting the cellular barrier composed of CAFs by tretinoin. Notably, the co-loaded nano-formulations proved to be more potent in inhibiting tumor growth in mice and also showed better anti-metastatic effects in lung metastasis models compared to the formulations with either drug alone. This novel drug delivery system has the potential to be used to develop more effective cancer therapies. Conclusion: Targeting CAFs with celastrol sensitizes tumor cells to chemotherapy, increasing the efficacy of betulinic acid. The combination of drugs targeting tumor cells and CAFs may lead to more effective therapies against various cancers.


Asunto(s)
Fibroblastos Asociados al Cáncer , Triterpenos Pentacíclicos , Neoplasias de la Mama Triple Negativas , Humanos , Animales , Ratones , Liposomas/farmacología , Neoplasias de la Mama Triple Negativas/tratamiento farmacológico , Neoplasias de la Mama Triple Negativas/patología , Células 3T3 NIH , Ácido Betulínico , Tretinoina/farmacología , Ácido Fólico/farmacología , Línea Celular Tumoral , Microambiente Tumoral
18.
Biomolecules ; 14(2)2024 Feb 16.
Artículo en Inglés | MEDLINE | ID: mdl-38397463

RESUMEN

Antimicrobial resistance (AMR) poses a significant global health risk as a consequence of misuse of antibiotics. Owing to the increasing antimicrobial resistance, it became imperative to develop novel molecules and materials with antimicrobial properties. Porphyrins and metalloporphyrins are compounds which present antimicrobial properties especially after irradiation. As a consequence, porphyrinoids have recently been utilized as antimicrobial agents in antimicrobial photodynamic inactivation in bacteria and other microorganisms. Herein, we report the encapsulation of porphyrins into peptide hydrogels which serve as delivery vehicles. We selected the self-assembling Fmoc-Phe-Phe dipeptide, a potent gelator, as a scaffold due to its previously reported biocompatibility and three different water-soluble porphyrins as photosensitizers. We evaluated the structural, mechanical and in vitro degradation properties of these hydrogels, their interaction with NIH3T3 mouse skin fibroblasts, and we assessed their antimicrobial efficacy against Gram-positive Staphylococcus aureus (S. aureus) and Gram-negative Escherichia coli (E. coli) bacteria. We found out that the hydrogels are cytocompatible and display antimicrobial efficiency against both strains with the zinc porphyrins being more efficient. Therefore, these hydrogels present a promising alternative for combating bacterial infections in the face of growing AMR concerns.


Asunto(s)
Antiinfecciosos , Fluorenos , Porfirinas , Animales , Ratones , Porfirinas/farmacología , Porfirinas/química , Antibacterianos/farmacología , Antibacterianos/química , Escherichia coli , Staphylococcus aureus , Hidrogeles/farmacología , Células 3T3 NIH , Farmacorresistencia Bacteriana , Antiinfecciosos/farmacología , Antiinfecciosos/química , Dipéptidos/farmacología
19.
Genes Chromosomes Cancer ; 63(2): e23226, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-38380774

RESUMEN

Epithelioid hemangioendothelioma (EHE) is a rare endothelial sarcoma associated with a high incidence of metastases and for which there are no standard treatment options. Based on disease-defining mutations, most EHEs are classified into two subtypes: WWTR1::CAMTA1-fused EHE or YAP1::TFE3-fused EHE. However, rare non-canonical fusions have been identified in clinical samples of EHE cases and are challenging to classify. In this study, we report the identification of a novel WWTR1::TFE3 fusion variant in an EHE patient using targeted RNA sequencing. Histologically, the tumor exhibited hybrid morphological characteristics between WWTR1::CAMTA1-fused EHE and YAP1::TFE3-fused EHE. In addition to the driver fusion, there were six additional secondary mutations identified, including a loss-of-function FANCA mutation. Furthermore, in vitro studies were conducted to investigate the tumorigenic function of the WWTR1::TFE3 fusion protein in NIH3T3 cells and demonstrated that WWTR1::TFE3 promotes colony formation in soft agar. Finally, as the wild-type WWTR1 protein relies on binding the TEAD family of transcription factors to affect gene transcription, mutation of the WWTR1 domain of the fusion protein to inhibit such binding abrogates the transformative effect of WWTR1::TFE3. Overall, we describe a novel gene fusion in EHE with a hybrid histological appearance between the two major genetic subtypes of EHE. Further cases of this very rare subtype of EHE will need to be identified to fully elucidate the clinical and pathological characteristics of this unusual subtype of EHE.


Asunto(s)
Hemangioendotelioma Epitelioide , Transactivadores , Humanos , Ratones , Animales , Transactivadores/genética , Hemangioendotelioma Epitelioide/genética , Hemangioendotelioma Epitelioide/patología , Células 3T3 NIH , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Péptidos y Proteínas de Señalización Intracelular/genética , Fusión Génica , Factores de Transcripción Básicos con Cremalleras de Leucinas y Motivos Hélice-Asa-Hélice/genética , Factores de Transcripción Básicos con Cremalleras de Leucinas y Motivos Hélice-Asa-Hélice/metabolismo , Proteínas Coactivadoras Transcripcionales con Motivo de Unión a PDZ
20.
Oncol Res ; 32(3): 477-487, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38361760

RESUMEN

Intracellular communications between breast cancer and fibroblast cells were reported to be involved in cancer proliferation, growth, and therapy resistance. The hallmarks of cancer-fibroblast interactions, consisting of caveolin 1 (Cav1) and mono-carboxylate transporter 4 (MCT4) (metabolic coupling markers), along with IL-6, TGFß, and lactate secretion, are considered robust biomarkers predicting recurrence and metastasis. In order to promote a novel phenotype in normal fibroblasts, we predicted that breast cancer cells could be able to cause loss of Cav1 and increase of MCT4, as well as elevate IL-6 and TGFß in nearby normal fibroblasts. We created a co-culture model using breast cancer (4T1) and normal fibroblast (NIH3T3) cell lines cultured under specific experimental conditions in order to directly test our theory. Moreover, we show that long-term co-culture of breast cancer cells and normal fibroblasts promotes loss of Cav1 and gain of MCT4 in adjacent fibroblasts and increase lactate secretion. These results were validated using the monoculture of each group separately as a control. In this system, we show that metformin inhibits IL-6 and TGFß secretion and re-expresses Cav1 in both cells. However, MCT4 and lactate stayed high after treatment with metformin. In conclusion, our work shows that co-culture with breast cancer cells may cause significant alterations in the phenotype and secretion of normal fibroblasts. Metformin, however, may change this state and affect fibroblasts' acquired phenotypes. Moreover, mitochondrial inhibition by metformin after 8 days of treatment, significantly hinders tumor growth in mouse model of breast cancer.


Asunto(s)
Neoplasias de la Mama , Metformina , Animales , Ratones , Humanos , Femenino , Metformina/farmacología , Metformina/metabolismo , Técnicas de Cocultivo , Interleucina-6/metabolismo , Interleucina-6/farmacología , Células 3T3 NIH , Estrés Oxidativo , Neoplasias de la Mama/patología , Fibroblastos/metabolismo , Fenotipo , Ácido Láctico/metabolismo , Ácido Láctico/farmacología , Factor de Crecimiento Transformador beta/metabolismo , Línea Celular Tumoral
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...