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










Base de dados
Intervalo de ano de publicação
1.
J Control Release ; 365: 60-73, 2024 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-37972765

RESUMO

Extracellular vesicles (EVs) are an effective tool to elucidate the bioeffect of nanomedicines. To clarify the interaction between oral nanomedicines and intestinal epithelial cells, and their bioeffects on downstream cells, polystyrene nanoparticles (PS-NPs) with different sizes were used as the model nanomedicines for EVs induction. Caco-2 monolayers were selected as the model of the intestinal epithelium and DLD-1 cells as the colorectal cancer model proximal to the gastrointestinal tract. It is found that compared with small-sized (25, 50, 100 nm) PS-NPs, the large-sized (200 and 500 nm) exhibited higher co-localization with multivesicular bodies and lysosomes, and more significant reduction of lysosomal acidification in Caco-2 cells. Proteomic and western-blotting analysis showed that the EVs remodeled by large-sized PS-NPs exhibited a higher extent of protein expression changes. The in vitro and in vivo signaling pathway detection in DLD-1 cells and DLD-1 cell xenograft nude mice showed that the remodeled EVs by large-sized PS-NPs inhibited the activation of multiple signaling pathways including Notch3, EGF/EGFR, and PI3K/Akt pathways, which resulted in the inhibition of tumor cell migration. These results primarily clarify the regulation mechanisms of nanomedicines-EVs-receptor cells chain. It provides a new perspective for the rational design and bioeffect evaluation of oral drug nanomaterials and sets up the fundamental knowledge for novel tumor therapeutics in the future.


Assuntos
Vesículas Extracelulares , Nanopartículas , Animais , Camundongos , Humanos , Células CACO-2 , Proteômica/métodos , Camundongos Nus , Fosfatidilinositol 3-Quinases/metabolismo , Mucosa Intestinal/metabolismo , Vesículas Extracelulares/metabolismo , Nanopartículas/metabolismo , Movimento Celular
2.
Polymers (Basel) ; 14(16)2022 Aug 11.
Artigo em Inglês | MEDLINE | ID: mdl-36015531

RESUMO

As a light-weight natural keratin biocomposite, Bovidae horn exhibits high mechanical properties and energy absorption. Different to the widely studied horn from subfamily Bovinae and Antilocapridae, few studies have focused on the horn sheath of subfamily Caprinae. In this work, three Caprinae horn sheathes from Cashmere goat, White goat and Black sheep were selected. Charpy pendulum impact tests were performed, and the fracture characteristics were evaluated. It was demonstrated that water plays an important role in acquiring balanced dynamic mechanical properties in all Caprinae horn sheaths. The hydrated keratin provides large plastic deformation capacity and further gives rise to a gradual generation of micro-cracks. Multi-scale structure including wavy-shaped interface, scattered voids and hierarchical micro-fibre were observed. Such a structure induced complex fracture mechanisms, such as delamination, 90° crack deflection and fibre pull-out, which were probably influenced by interfacial strength. The results are expected to endow the research and thinking of Bovidae horn.

3.
J Control Release ; 339: 430-444, 2021 11 10.
Artigo em Inglês | MEDLINE | ID: mdl-34655679

RESUMO

The intestinal barrier has always been the rate-limiting step in the oral administration process. To overcome the intestinal barrier, researchers have widely adopted nanocarriers, especially active-targeting nanocarriers strategies. However, most of these strategies focus on the ligand decoration of nanocarriers targeting specific receptors, so their applications are confined to specific receptors or specific cell types. In this study, we tried to investigate more common strategies in the field of transmembrane transport enhancement. Trans-Golgi network (TGN) is the sorting center of biosynthetic route which could achieve polarized localization of proteins in polarized epithelial cells, and the basolateral plasma membrane is where all transcytotic cargos have to pass through. Thus, it is expected that guiding nanocarriers to TGN or basolateral plasma membrane may improve the transcytosis. Hence, we choose sorting signal peptide to modify micelles to guide micelles to TGN (named as BAC decorated micelles, BAC-M) or to basolateral plasma membrane (named as STX decorated micelles, STX-M). By incorporating coumarin-6 (C6) or Cy5-PEG-PCL in the micelles to indicate the behavior of micelles, the effects of these two strategies on the transcytosis were investigated. To our surprise, BAC-M and STX-M behaved quite differently when crossing biological barriers. BAC-M showed significant superiority in colocalization with TGN, transmembrane transport and even in vivo absorption, while STX-M had no significant difference from blank micelles. Further investigation revealed that the strategy of directly guiding nanocarriers to the basolateral plasma membrane (STX-M) only caused the stack of vesicles near the basolateral plasma membrane. So, we concluded that guiding nanocarriers to TGN which related to secretion may contribute to the transmembrane transport. This common strategy based on the physiological function of TGN in polarized epithelial cells will have broad application prospects in overcoming biological barrier.


Assuntos
Células Epiteliais , Rede trans-Golgi , Transporte Biológico , Membrana Celular/metabolismo , Transporte Proteico , Rede trans-Golgi/metabolismo
4.
Asian J Pharm Sci ; 16(1): 97-109, 2021 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-33613733

RESUMO

Currently, the cancer immunotherapy has made great progress while antitumor vaccine attracts substantial attention. Still, the selection of adjuvants as well as antigens are always the most crucial issues for better vaccination. In this study, we proposed a biomimetic antitumor nanovaccine based on biocompatible nanocarriers and tumor cell membrane antigens. Briefly, endogenous calcium pyrophosphate nanogranules with possible immune potentiating effect are designed and engineered, both as delivery vehicles and adjuvants. Then, these nanocarriers are coated with lipids and B16-OVA tumor cell membranes, so the biomembrane proteins can serve as tumor-specific antigens. It was found that calcium pyrophosphate nanogranules themselves were compatible and possessed adjuvant effect, while membrane proteins including tumor associated antigen were transferred onto the nanocarriers. It was demonstrated that such a biomimetic nanovaccine could be well endocytosed by dendritic cells, promote their maturation and antigen-presentation, facilitate lymph retention, and trigger obvious immune response. It was confirmed that the biomimetic vaccine could induce strong T-cell response, exhibit excellent tumor therapy and prophylactic effects, and simultaneously possess nice biocompatibility. In general, the present investigation might provide insights for the further design and application of antitumor vaccines.

5.
J Extracell Vesicles ; 9(1): 1806444, 2020 Aug 19.
Artigo em Inglês | MEDLINE | ID: mdl-32944191

RESUMO

A long-term and huge challenge in nanomedicine is the substantial uptake and rapid clearance mediated by the mononuclear phagocyte system (MPS), which enormously hinders the development of nanodrugs. Inspired by the natural merits of extracellular vesicles, we therefore developed a combined "eat me/don't eat me" strategy in an effort to achieve MPS escape and efficient drug delivery. Methodologically, cationized mannan-modified extracellular vesicles derived from DC2.4 cells were administered to saturate the MPS (eat me strategy). Then, nanocarriers fused to CD47-enriched exosomes originated from human serum were administered to evade phagocytosis by MPS (don't eat me strategy). The nanocarriers were also loaded with antitumor drugs and functionalized with a novel homing peptide to promote the tumour tissue accumulation and cancer cell uptake (eat me strategy). The concept was proven in vitro as evidenced by the reduced endocytosis of macrophages and enhanced uptake by tumour cells, whereas prolonged circulation time and increased tumour accumulation were demonstrated in vivo. Specially, the strategy induced a 123.53% increase in tumour distribution compared to conventional nanocarrier. The study both shed light on the challenge overcoming of phagocytic evasion and provided a strategy for significantly improving therapeutic outcomes, potentially permitting active drug delivery via targeted nanomedicines.

6.
J Control Release ; 323: 600-612, 2020 07 10.
Artigo em Inglês | MEDLINE | ID: mdl-32278828

RESUMO

Nanotechniques show significant merits in terms of improving the oral bioavailability of poorly water-soluble drugs. However, the mechanisms behind are not clear yet. For instance, what is the contribution of free drug released during nanogranule transcytosis, as well as the impact of drug transporter and chylomicron? To address these issues, sorafenib nanogranules (SFN-NGs) were prepared as model by the high-gravity antisolvent precipitation method which approaches to practical mass production. Then, a multiaspect study on the transcytosis mechanism of SFN-NGs was conducted in Caco-2 cells and rats, including paracellular transport, endocytosis, intracellular trafficking, transmembrane pathway, as well as the involvement of transporter and chylomicron. Pharmacokinetics in rats demonstrated an obvious superiority of SFN-NGs in oral absorption and lymphatic transfer over SFN crude drugs. Different from free SFN, SFN-NGs could be internalized in cells in early stage by caveolin/lipid raft or clathrin induced endocytosis, and transported intactly through the polarized cell monolayers. While in late stage, transporter-mediated transport of free SFN began to play a vital role on the transmembrane of SFN-NGs. No paracellular transport of SFN-NGs was found, and the trafficking of SFN-NGs was affected by the pathway of ER-Golgi complexes. Surprisedly, the intracellular free SFN was the main source of transmembrane for SFN-NGs, which was entrapped into chylomicrons and then secreted into the extracellular space. Generally, the findings in current study may shed light on the absorption mechanism of oral nanoformulations.


Assuntos
Nanopartículas , Animais , Células CACO-2 , Endocitose , Humanos , Ratos , Sorafenibe , Transcitose
7.
Mol Pharm ; 17(1): 239-250, 2020 01 06.
Artigo em Inglês | MEDLINE | ID: mdl-31800258

RESUMO

Oral administration is an ideal alternative for drug delivery due to its convenience and safety. However, oral protein delivery is limited by biological barriers such as the mucus barrier and epithelial barrier, which hamper drugs from entering the blood successfully. Here we presented PC6/CS NPs, a thiolated-polymer-based nanodrug delivery system in the form of poly(acrylic acid)-cysteine-6-mercaptonicotinic acid (PAA-Cys-6MNA, PC6), which is a kind of preactivated thiolated polymer, coated on chitosan (CS) nanoparticles (NPs). Its ability to overcome the mucus barrier and epithelial barrier was investigated. The existence of PC6 made the NPs prone to penetrate the mucus layer as well as strengthened the transcellular transport of insulin on epithelial cells. PC6/CS NPs efficiently enhanced the oral bioavailability of insulin to 16.2%. The improvement resulted from the function of PC6: (1) "diluting" mucus to promote nanoparticle penetration, (2) opening a tight junction to help insulin transport via the paracellular pathway, (3) making the nanoparticle more electrically neutral during the penetration process, and (4) uncoating from PC6/CS NPs so that positive CS NPs were adhered and uptaken by epithelial cells. Our study proves that PC6/CS NPs, which can achieve mucus penetration and epithelial permeation efficiently, are a potential nanocarrier for oral protein delivery.


Assuntos
Diabetes Mellitus Experimental/tratamento farmacológico , Células Epiteliais/metabolismo , Insulina/administração & dosagem , Muco/metabolismo , Nanopartículas/química , Ácidos Picolínicos/química , Resinas Acrílicas/química , Administração Oral , Animais , Disponibilidade Biológica , Linhagem Celular Tumoral , Quitosana/metabolismo , Cisteína/química , Diabetes Mellitus Experimental/sangue , Diabetes Mellitus Experimental/metabolismo , Portadores de Fármacos/química , Portadores de Fármacos/metabolismo , Células Epiteliais/efeitos dos fármacos , Humanos , Insulina/metabolismo , Insulina/farmacocinética , Microscopia Eletrônica de Transmissão , Muco/efeitos dos fármacos , Nanopartículas/metabolismo , Nanopartículas/toxicidade , Nanopartículas/ultraestrutura , Ácidos Nicotínicos/química , Ácidos Picolínicos/metabolismo , Ratos , Compostos de Sulfidrila/química , Junções Íntimas/efeitos dos fármacos , Junções Íntimas/metabolismo
8.
Adv Drug Deliv Rev ; 143: 177-205, 2019 03 15.
Artigo em Inglês | MEDLINE | ID: mdl-31201837

RESUMO

Extensive studies on nanomedicines have been conducted for drug delivery and disease diagnosis (especially for cancer therapy). However, the intracellular and in vivo biofate of nanomedicines, which is significantly associated with their clinical therapeutic effect, is poorly understood at present. This is because of the technical challenges to quantify the disassembly and behaviour of nanomedicines. As a fluorescence- and distance-based approach, the Förster Resonance Energy Transfer (FRET) technique is very successful to study the interaction of nanomedicines with biological systems. In this review, principles on how to select a FRET pair and construct FRET-based nanomedicines have been described first, followed by their application to study structural integrity, biodistribution, disassembly kinetics, and elimination of nanomedicines at intracellular and in vivo levels, especially with drug nanocarriers including polymeric micelles, polymeric nanoparticles, and lipid-based nanoparticles. FRET is a powerful tool to reveal changes and interaction of nanoparticles after delivery, which will be very useful to guide future developments of nanomedicine.


Assuntos
Transferência Ressonante de Energia de Fluorescência , Nanomedicina , Animais , Sistemas de Liberação de Medicamentos , Humanos
9.
ACS Nano ; 13(5): 5058-5076, 2019 05 28.
Artigo em Inglês | MEDLINE | ID: mdl-31034211

RESUMO

Overcoming the epithelial barriers to enhance drug transport is a focused topic for gastrointestinal, intratracheal, intranasal, vaginal, and intrauterine delivery. Nanomedicines with targeting functionization promote such a process owing to specific ligand-receptor interaction. However, compared to the cell uptake of targeting nanotherapies, currently few studies concentrate on their transcytosis including endocytosis for "in" and exocytosis for "out". In fact, the cellular regulatory mechanism for these pathways as well as the principle of ligand's effect on the transcytosis are almost ignored. Here, we fabricated transferrin (Tf) functionalized nanogranules (Tf-NG) as the nanomedicine model and confirmed the difference in polar distributions of Tf receptors (TfRs) between two epithelium models (bipolarity for Caco-2 and unipolarity for MDCK cells). Compared to the nonspecific reference, Tf-conjugation boosted the endocytosis by different pathways in two cell models and transformed the intracellular route of Tf-NG in both cells differently, affecting exocytosis, recycling, and degradation but not the secretion pathway. Only bipolar cells could establish a complete transport flow from "in" to "out", leading to the enhanced transcytosis of Tf-NG. Importantly, epithelia could make responses to Tf-NG transcytosis. Based on the quantitative proteomics, the intracellular trafficking of Tf-NG altered the protein expression profiles, in which the endocytosis- and transcytosis-related proteins were specifically upregulated. Particularly, only bipolar cells could positively feed back to such trafficking via accelerating the subsequent Tf-NG transcytosis. Here, all the cell transport of Tf-NG was polarity associated. In summary, Tf modification elevated the transcytosis of Tf-NG across the epithelium by triggering the polarity-associated transport flow and positive cell feedback loop. These findings provided an insight into the targeting nanodelivery for efficient transport through epithelial barriers.


Assuntos
Polaridade Celular , Epitélio/metabolismo , Retroalimentação Fisiológica , Nanopartículas/química , Transcitose , Transferrina/metabolismo , Animais , Transporte Biológico , Bovinos , Linhagem Celular , Cães , Endocitose , Exocitose , Humanos , Masculino , Modelos Biológicos , Nanopartículas/ultraestrutura , Nanotecnologia , Ratos Wistar , Soroalbumina Bovina/metabolismo
10.
Biomaterials ; 180: 78-90, 2018 10.
Artigo em Inglês | MEDLINE | ID: mdl-30025247

RESUMO

Active-targeting nanocarriers can significantly improve the transcytosis of poorly water-soluble or bio-macromolecular drugs across biological barrier. However, reasons for the improvement are not understood enough, which hampered the reasonable design of active targeting nanocarriers. To illustrate how different factors influence the transport of active-targeting nanocarriers, we established ligand-decorated micelles targeting different receptors to study how the decorations influence the transcytosis of the micelles by comparing the endocytosis, transport pathway and exocytosis process. Three different kinds of receptors, Neonatal Fc receptor (FcRn), transferrin receptor (TfR) and αvß3 integrin were selected. They presented three different transport pathways, mainly mediating transcytosis, recycling pathway and cell binding, respectively. Their corresponding ligand FcBP, 7pep and c(RGDfK) decorated micelles with different ligand densities were prepared first. Then the effects of receptor and ligand density on the transcytosis across biological barrier were investigated. The results showed that the uptake rate of active micelles was higher than passive micelles and an optimum ligand density with most endocytosis appeared in all functional micelles. Transport pathway study showed 7pep decorated micelles were transferred into apical recycling endosome (ARE) and exocytosed to apical plasma membrane in a ligand depended way. c(RGDfK) decorated micelles were transferred through common recycling endosome (CRE) and Golgi complex to basolateral plasma membrane instead of ARE. While FcBP decorated micelles took both the recycling pathway and transcytosis through CRE, but not Golgi complex. Proper ligand density, not the higher the better, led the most uptake. Also the apical to basolateral transcytosis ratio may not be in accordance with the uptake. Among all the itineraries, transcytosis through CRE is the best itinerary for transcytosis. So, in the design of active targeting nanocarriers to overcome biological barrier, receptor character should be considered priorly, and then ligand density should be optimized.


Assuntos
Antígenos de Histocompatibilidade Classe I/metabolismo , Integrina alfaVbeta3/metabolismo , Receptores Fc/metabolismo , Receptores da Transferrina/metabolismo , Células CACO-2 , Endossomos/metabolismo , Exocitose/fisiologia , Complexo de Golgi/metabolismo , Antígenos de Histocompatibilidade Classe I/química , Humanos , Integrina alfaVbeta3/química , Micelas , Receptores Fc/química , Receptores da Transferrina/química , Transcitose/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...