Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Arch Toxicol ; 96(5): 1455-1471, 2022 05.
Artículo en Inglés | MEDLINE | ID: mdl-35226136

RESUMEN

The micronucleus (MN) assay is widely used as part of a battery of tests applied to evaluate the genotoxic potential of chemicals, including new food additives and novel food ingredients. Micronucleus assays typically utilise homogenous in vitro cell lines which poorly recapitulate the physiology, biochemistry and genomic events in the gut, the site of first contact for ingested materials. Here we have adapted and validated the MN endpoint assay protocol for use with complex 3D reconstructed intestinal microtissues; we have named this new protocol the reconstructed intestine micronucleus cytome (RICyt) assay. Our data suggest the commercial 3D microtissues replicate the physiological, biochemical and genomic responses of native human small intestine to exogenous compounds. Tissues were shown to maintain log-phase proliferation throughout the period of exposure and expressed low background MN. Analysis using the RICyt assay protocol revealed the presence of diverse cell types and nuclear anomalies (cytome) in addition to MN, indicating evidence for comprehensive DNA damage and mode(s) of cell death reported by the assay. The assay correctly identified and discriminated direct-acting clastogen, aneugen and clastogen requiring exogenous metabolic activation, and a non-genotoxic chemical. We are confident that the genotoxic response in the 3D microtissues more closely resembles the native tissues due to the inherent tissue architecture, surface area, barrier effects and tissue matrix interactions. This proof-of-concept study highlights the RICyt MN cytome assay in 3D reconstructed intestinal microtissues is a promising tool for applications in predictive toxicology.


Asunto(s)
Daño del ADN , Micronúcleos con Defecto Cromosómico , Aneugénicos , Humanos , Pruebas de Micronúcleos/métodos , Mutágenos/toxicidad
2.
Nanotechnology ; 32(50)2021 Oct 07.
Artículo en Inglés | MEDLINE | ID: mdl-34536952

RESUMEN

Liposomes are potential drug carriers for atherosclerosis therapy due to low immunogenicity and ease of surface modifications that allow them to have prolonged circulation half-life and specifically target atherosclerotic sites to increase uptake efficiency. However, the effects of their size, charge, and lipid compositions on macrophage and foam cell behaviour are not fully understood. In this study, liposomes of different sizes (60 nm, 100 nm and 180 nm), charges (-40 mV, -20 mV, neutral, +15 mV and +30 mV) and lipid compositions (1,2-dipalmitoyl-sn-glycero-3-phosphatidylcholine, 1,2-dipalmitoyl-sn-glycero-3-phosphocholine, L-a-phosphatidylcholine, and egg sphingomyelin) were synthesized, characterized and exposed to macrophages and foam cells. Compared to 100 nm neutral 1,2-dipalmitoyl-sn-glycero-3-phosphatidylcholine (DPPC) liposomes, flow cytometry and confocal imaging indicated that cationic liposomes and 1,2-dipalmitoyl-sn-glycero-3-phosphocholine (DSPC) liposomes were internalized more by both macrophages and foam cells. Through endocytosis inhibition, phagocytosis and clathrin-mediated endocytosis were identified as the dominant mechanisms of uptake. Anionic and DSPC liposomes induced more cholesterol efflux capacity in foam cells. These results provide a guide for the optimal size, charge, and lipid composition of liposomes as drug carriers for atherosclerosis treatment.


Asunto(s)
Endocitosis/efectos de los fármacos , Liposomas/farmacología , Fagocitosis/efectos de los fármacos , 1,2-Dipalmitoilfosfatidilcolina/análogos & derivados , 1,2-Dipalmitoilfosfatidilcolina/química , Aterosclerosis/tratamiento farmacológico , Línea Celular , Supervivencia Celular/efectos de los fármacos , Colesterol/metabolismo , Células Espumosas/citología , Células Espumosas/metabolismo , Humanos , Liposomas/química , Liposomas/uso terapéutico , Macrófagos/citología , Macrófagos/metabolismo , Tamaño de la Partícula , Propiedades de Superficie
3.
Int J Mol Sci ; 22(12)2021 Jun 16.
Artículo en Inglés | MEDLINE | ID: mdl-34208563

RESUMEN

Bone exhibits piezoelectric properties. Thus, electrical stimulations such as pulsed electromagnetic fields (PEMFs) and stimuli-responsive piezoelectric properties of scaffolds have been investigated separately to evaluate their efficacy in supporting osteogenesis. However, current understanding of cells responding under the combined influence of PEMF and piezoelectric properties in scaffolds is still lacking. Therefore, in this study, we fabricated piezoelectric scaffolds by functionalization of polycaprolactone-tricalcium phosphate (PCL-TCP) films with a polyvinylidene fluoride (PVDF) coating that is self-polarized by a modified breath-figure technique. The osteoinductive properties of these PVDF-coated PCL-TCP films on MC3T3-E1 cells were studied under the stimulation of PEMF. Piezoelectric and ferroelectric characterization demonstrated that scaffolds with piezoelectric coefficient d33 = -1.2 pC/N were obtained at a powder dissolution temperature of 100 °C and coating relative humidity (RH) of 56%. DNA quantification showed that cell proliferation was significantly enhanced by PEMF as low as 0.6 mT and 50 Hz. Hydroxyapatite staining showed that cell mineralization was significantly enhanced by incorporation of PVDF coating. Gene expression study showed that the combination of PEMF and PVDF coating promoted late osteogenic gene expression marker most significantly. Collectively, our results suggest that the synergistic effects of PEMF and piezoelectric scaffolds on osteogenesis provide a promising alternative strategy for electrically augmented osteoinduction. The piezoelectric response of PVDF by PEMF, which could provide mechanical strain, is particularly interesting as it could deliver local mechanical stimulation to osteogenic cells using PEMF.


Asunto(s)
Fosfatos de Calcio , Materiales Biocompatibles Revestidos , Campos Electromagnéticos , Osteogénesis , Poliésteres , Polivinilos , Andamios del Tejido , Regeneración Ósea , Diferenciación Celular , Proliferación Celular , Células Cultivadas , Materiales Biocompatibles Revestidos/química , Expresión Génica , Osteogénesis/efectos de los fármacos , Osteogénesis/genética , Osteogénesis/efectos de la radiación , Poliésteres/química , Poliésteres/farmacología , Polivinilos/química , Solventes , Ingeniería de Tejidos , Difracción de Rayos X
4.
Nanomedicine ; 37: 102434, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34214684

RESUMEN

Atherosclerosis is a multifactorial disease triggered and sustained by risk factors such as high cholesterol, high blood pressure and unhealthy lifestyle. Inflammation plays a pivotal role in atherosclerosis pathogenesis. In this study, we developed a simvastatin (STAT) loaded nanoliposomal formulation (LIPOSTAT) which can deliver the drug into atherosclerotic plaque, when administered intravenously. This formulation is easily prepared, stable, and biocompatible with minimal burst release for effective drug delivery. 2D and 3D in vitro models were examined towards anti-inflammatory effects of STAT, both free and in combination with liposomes. LIPOSTAT induced greater cholesterol efflux in the 2D foam cells and significantly reduced inflammation in both 2D and 3D models. LIPOSTAT alleviated inflammation by reducing the secretion of early and late phase pro-inflammatory cytokines, monocyte adherence marker, and lipid accumulation cytokines. Additionally, the 3D foam cell spheroid model is a convenient and practical approach in testing various anti-atherosclerotic drugs without the need for human tissue.


Asunto(s)
Aterosclerosis/tratamiento farmacológico , Inflamación/tratamiento farmacológico , Liposomas/farmacología , Nanopartículas/química , Simvastatina/farmacología , Aterosclerosis/genética , Aterosclerosis/patología , Línea Celular , Sistemas de Liberación de Medicamentos/métodos , Células Espumosas/efectos de los fármacos , Células Espumosas/patología , Humanos , Inflamación/genética , Inflamación/patología , Liposomas/química , Placa Aterosclerótica/tratamiento farmacológico , Placa Aterosclerótica/patología , Simvastatina/química , Esferoides Celulares/química , Esferoides Celulares/efectos de los fármacos
5.
ACS Appl Mater Interfaces ; 13(21): 24422-24430, 2021 Jun 02.
Artículo en Inglés | MEDLINE | ID: mdl-34019376

RESUMEN

For site-specific diseases such as atherosclerosis, it is desirable to noninvasively and locally deliver therapeutics for extended periods of time. High-intensity focused ultrasound (HIFU) provides targeted drug delivery, yet remains unable to sustain delivery beyond the HIFU treatment time. Furthermore, methods to validate HIFU-enhanced drug delivery remain limited. In this study, we report on HIFU-targeted implantation of degradable drug-loaded sound-sensitive multicavity PLGA microparticles (mcPLGA MPs) as a theranostic agent for the treatment of arterial lesions. Once implanted into the targeted tissue, mcPLGA MPs eluted dexamethasone for several days, thereby reducing inflammatory markers linked to oxidized lipid uptake in a foam cell spheroid model. Furthermore, implanted mcPLGA MPs created hyperechoic regions on diagnostic ultrasound images, and thus noninvasively verified that the target region was treated with the theranostic agents. This novel and innovative multifunctional theranostic platform may serve as a promising candidate for noninvasive imaging and treatment for site-specific diseases such as atherosclerosis.


Asunto(s)
Arteritis/diagnóstico por imagen , Medicina de Precisión , Ondas Ultrasónicas , Arteritis/terapia , Humanos
6.
Bioorg Med Chem ; 37: 116112, 2021 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-33751939

RESUMEN

Natural compounds isolated from different medicinal plants remain one of the major resources of anticancer drugs due to their enormous chemical diversity. Studies suggested therapeutic potential for various tanshinones, key bioactive lipophilic compounds from the root extracts of Salvia miltiorrhiza Bunge, against multiple cancers including breast carcinoma. We designed, synthesized and evaluated anti-cancer properties of a series of condensed and doubly condensed furophenanthraquinones of tanshinone derivatives on two breast cancer lines - MCF7 and MDA-MB-231. We identified two thiophene analogues - compounds 48 and 52 with greater anti-proliferative efficiency (~4 fold) as compared to the natural tanshinones. Mechanistically, we showed that both compounds induced autophagy mediated cell death and partial but significant restoration of cell death in the presence of autophagy inhibitor further supported this notion. Both compounds transcriptionally activated several autophagy genes responsible for autophagosome formation along with two death regulators - GADD34 and CHOP for inducing cell death. Altogether, our studies provide strong evidence to support compounds 48 and 52 as promising leads for further development as anticancer agents through modulating autophagy mechanism.


Asunto(s)
Antineoplásicos/farmacología , Autofagia/efectos de los fármacos , Neoplasias de la Mama/tratamiento farmacológico , Tiofenos/farmacología , Antineoplásicos/síntesis química , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Ensayos de Selección de Medicamentos Antitumorales , Humanos , Tiofenos/síntesis química , Respuesta de Proteína Desplegada/efectos de los fármacos
7.
ACS Appl Bio Mater ; 4(9): 7044-7058, 2021 09 20.
Artículo en Inglés | MEDLINE | ID: mdl-35006937

RESUMEN

There is an urgent clinical need for wound dressings to treat skin injuries, particularly full-thickness wounds caused by acute and chronic wounds. Marine collagen has emerged as an attractive and safer alternative due to its biocompatibility, diversity, and sustainability. It has minimum risk of zoonotic diseases and less religious constraints as compared to mammalian collagen. In this study, we reported the development of a self-assembled nanofibrous barramundi (Lates calcarifer) collagen matrix (Nano-BCM), which showed good biocompatibility for full-thickness wound-healing applications. The collagen was extracted and purified from barramundi scales and skin. Thereafter, the physicochemical properties of collagen were systematically evaluated. The process to extract barramundi skin collagen (BC) gave an excellent 45% yield and superior purity (∼100%). More importantly, BC demonstrated structural integrity, native triple helix structure, and good thermal stability. BC demonstrated its efficacy in promoting human primary dermal fibroblast (HDF) and immortalized human keratinocytes (HaCaT) proliferation and migration. Nano-BCM has been prepared via self-assembly of collagen molecules in physiological conditions, which resembled the native extracellular matrix (ECM). The clinical therapeutic efficacy of the Nano-BCM was further evaluated in a full-thickness splinted skin wound mice model. In comparison to a clinically used wound dressing (DuoDerm), the Nano-BCM demonstrated significantly accelerated wound closure and re-epithelization. Moreover, Nano-BCM nanofibrous architecture and its ability to facilitate early inflammatory response significantly promoted angiogenesis and differentiated myofibroblast, leading to enhanced wound healing. Consequently, Nano-BCM demonstrates great potential as an economical and effective nonmammalian substitute to achieve skin regeneration.


Asunto(s)
Nanofibras , Animales , Colágeno/farmacología , Matriz Extracelular , Mamíferos , Ratones , Nanofibras/uso terapéutico , Piel , Cicatrización de Heridas
8.
Int J Nanomedicine ; 15: 4943-4956, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32764927

RESUMEN

BACKGROUND: Hydroxyapatite (HA) [Ca5(PO4)3(OH)] is a naturally occurring calcium phosphate which makes up 60-70% of the dry weight of human bones. Nano-scale HA particles are increasingly being used as carriers for controlled and targeted delivery of bioactive agents like drugs, proteins, and nucleic acids due to their high porosity, negative charge, and biodegradability. PURPOSE: Although much effort has been devoted to understanding the delivery kinetics and effects of the payloads in such carriers, a thorough understanding of the influence of the carriers themselves is lacking. METHODS: HA particles (300 µg/mL) were administered to primary human dermal fibroblasts (HDFs). The uptake and intracellular localization of the particles were determined by flow cytometry, confocal imaging, and transmission electron microscopy (TEM). Immunological assays and PCR were performed to determine the levels of pro-inflammatory cytokines and collagens in cell lysates and media supernatant. RESULTS: The current study explores the effects of poly-dispersed HA particles on primary HDFs as a model system. The majority of the particles were determined to range between 150 and 200 nm in diameter. Upon exposure to HA suspensions, primary HDFs internalized the particles by endocytosis within 6 hours of exposure, showing maximum uptake at 72 hours following which the particles were exocytosed by 168 hours. This correlated to reduced secretion of various pro-inflammatory and pro-collagenic cytokines. Biochemical analysis further revealed a reduction in Type I collagen expression and secretion. CONCLUSION: HA particles have an immune-modulatory effect on dermal fibroblasts and reduce collagen production, which may impact the integrity of the extracellular matrix (ECM). This study demonstrates the need to consider the secondary effects of particulate carriers like HA, beyond basic cytotoxicity, in the specific tissue environment where the intended function is to be realized.


Asunto(s)
Colágeno/metabolismo , Durapatita/farmacología , Fibroblastos/efectos de los fármacos , Fibroblastos/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Piel/citología , Colágeno Tipo I/metabolismo , Citocinas/metabolismo , Durapatita/química , Matriz Extracelular/efectos de los fármacos , Matriz Extracelular/metabolismo , Fibroblastos/citología , Humanos
9.
Artículo en Inglés | MEDLINE | ID: mdl-32656197

RESUMEN

3-dimensional (3D) in vitro models were developed in order to mimic the complexity of real organ/tissue in a dish. They offer new possibilities to model biological processes in more physiologically relevant ways which can be applied to a myriad of applications including drug development, toxicity screening and regenerative medicine. Hydrogels are the most relevant tissue-like matrices to support the development of 3D in vitro models since they are in many ways akin to the native extracellular matrix (ECM). For the purpose of further improving matrix relevance or to impart specific functionalities, composite hydrogels have attracted increasing attention. These could incorporate drugs to control cell fates, additional ECM elements to improve mechanical properties, biomolecules to improve biological activities or any combinations of the above. In this Review, recent developments in using composite hydrogels laden with cells as biomimetic tissue- or organ-like constructs, and as matrices for multi-cell type organoid cultures are highlighted. The latest composite hydrogel systems that contain nanomaterials, biological factors, and combinations of biopolymers (e.g., proteins and polysaccharide), such as Interpenetrating Networks (IPNs) and Soft Network Composites (SNCs) are also presented. While promising, challenges remain. These will be discussed in light of future perspectives toward encompassing diverse composite hydrogel platforms for an improved organ environment in vitro.

10.
Nanotoxicology ; 12(10): 1215-1229, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-30428752

RESUMEN

Zinc oxide nanoparticles (Nano-ZnO) is currently one of the most extensively used inorganic particles in a wide range of skin care and consumable products. Therefore, examining the biological effects of Nano-ZnO, especially in the non-cytotoxic levels, thus holds important contemporary practical implications. Herein, our study demonstrates that long-term conditioning of human keratinocytes (HaCaTs) to non-cytoxic dose of Nano-ZnO (∼100 nm) can induce an adaptive response, leading to an enhancement of the cells tolerance against cytotoxic level of Nano-ZnO. It was found that the Nano-ZnO induced adaptive alteration is mediated by a strong synergism between the generation of reactive oxygen species (ROS) flares by a sub-population of cells that are loaded with Nano-ZnO and upregulation of several pro-inflammatory transcripts. Further studies revealed activation of the nuclear factor (erythroid-derived 2)-like 2 (Nrf-2) stress response pathway and the associated downstream sustained augmented level of chymotrypsin-like 20 s proteasome activity to be the major mechanism underpinning this phenomenon. Interestingly, these cytoprotective responses can further aid the Nano-ZnO conditioned HaCaT cells to cross-adapt to harmful effects of ultraviolet-A (UVA) by reducing radiation-induced DNA damage. Our findings have unveiled a range of previously undocumented potent and exploitable bioeffects of Nano-ZnO induced ROS mediated signaling within the framework of nano-adaptation.


Asunto(s)
Queratinocitos/efectos de los fármacos , Nanopartículas del Metal/toxicidad , Factor 2 Relacionado con NF-E2/metabolismo , Comunicación Paracrina/efectos de los fármacos , Complejo de la Endopetidasa Proteasomal/metabolismo , Óxido de Zinc/toxicidad , Línea Celular , Relación Dosis-Respuesta a Droga , Humanos , Queratinocitos/metabolismo , Estrés Oxidativo/efectos de los fármacos , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal/efectos de los fármacos
11.
Nat Commun ; 3: 1240, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-23212369

RESUMEN

Protein quality control is essential for cellular survival. Failure to eliminate pathogenic proteins leads to their intracellular accumulation in the form of protein aggregates. Autophagy can recognize protein aggregates and degrade them in lysosomes. However, some aggregates escape the autophagic surveillance. Here we analyse the autophagic degradation of different types of aggregates of synphilin-1, a protein often found in pathogenic protein inclusions. We show that small synphilin-1 aggregates and large aggresomes are differentially targeted by constitutive and inducible autophagy. Furthermore, we identify a region in synphilin-1, necessary for its own basal and inducible aggrephagy and sufficient for the degradation of other pro-aggregating proteins. Although the presence of this peptide is sufficient for basal aggrephagy, inducible aggrephagy requires its ubiquitination, which diminishes protein mobility on the surface of the aggregate and favours the recruitment and assembly of the protein complexes required for autophagosome formation. Our study reveals different mechanisms for cells to cope with aggregate proteins via autophagy and supports the idea that autophagic susceptibility of prone-to-aggregate proteins may not depend on the nature of the aggregating proteins per se, but on their dynamic properties in the aggregate.


Asunto(s)
Autofagia/fisiología , Proteínas/metabolismo , Ancirinas/fisiología , Proteínas Portadoras/metabolismo , Línea Celular , Fenómenos Fisiológicos Celulares/fisiología , Cuerpos de Inclusión/metabolismo , Péptidos y Proteínas de Señalización Intracelular , Proteínas del Tejido Nervioso/metabolismo , Péptidos/metabolismo , Péptidos/fisiología , Fagosomas/metabolismo , Fagosomas/fisiología , Proteínas/fisiología , Ubiquitinación
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...