Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Small ; 20(1): e2304824, 2024 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-37653618

RESUMO

Mesenchymal stem cells (MSCs) are becoming hotspots for application in disease therapies recently, combining with biomaterials and drug delivery system. A major advantage of MSCs applied in drug delivery system is that these cells enable specific targeting and releasing of cargos to the disease sites. However, the potential tumor tropic effects of MSCs raised concerns on biosafety. To solve this problem, there are emerging methods of isolating cell membranes and developing nanoformulations to perform drug delivery, which avoids concerns on biosafety without disturbing the membrane functions of specific polarizing and locating. These cargoes are so called "nanoghosts." This review article summarizes the current applications of nanoghosts, the promising potential of MSCs to be applied in membrane isolation and nanoghost construction, and possible approaches to develop better drug delivery system harnessing from MSC ghost cell membranes.


Assuntos
Células-Tronco Mesenquimais , Neoplasias , Humanos , Biomimética , Neoplasias/metabolismo , Sistemas de Liberação de Medicamentos , Membrana Celular , Células-Tronco Mesenquimais/metabolismo
2.
Chemosphere ; 306: 135578, 2022 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-35798154

RESUMO

Overexpression of proteins/antigens and other gene-related sequences in the bodies could lead to significant mutations and refractory diseases. Detection and identification of assorted trace concentrations of such proteins/antigens and/or gene-related sequences remain challenging, affecting different pathogens and making viruses stronger. Correspondingly, coronavirus (SARS-CoV-2) mutations/alterations and spread could lead to overexpression of ssDNA and the related antigens in the population and brisk activity in gene-editing technologies in the treatment/detection may lead to the presence of pCRISPR in the blood. Therefore, the detection and evaluation of their trace concentrations are of critical importance. CaZnO-based nanoghosts (NGs) were synthesized with the assistance of a high-gravity technique at a 1,800 MHz field, capitalizing on the use of Rosmarinus officinalis leaf extract as the templating agent. A complete chemical, physical and biological investigation revealed that the synthesized NGs presented similar morphological features to the mesenchymal stem cells (MSCs), resulting in excellent biocompatibility, interaction with ssDNA- and/or pCRISPR-surface, through various chemical and physical mechanisms. This comprise the unprecedented synthesis of a fully inorganic nanostructure with behavior that is similar to MSCs. Furthermore, the endowed exceptional ability of inorganic NGs for detective sensing/folding of ssDNA and pCRISPR and recombinant SARS-CoV-2 spike antigen (RSCSA), along with in-situ hydrogen peroxide detection on the HEK-293 and HeLa cell lines, was discerned. On average, they displayed a high drug loading capacity of 55%, and the acceptable internalizations inside the HT-29 cell lines affirmed the anticipated MSCs-like behavior of these inorganic-NGs.


Assuntos
DNA de Cadeia Simples , Doxorrubicina , Sistemas de Liberação de Fármacos por Nanopartículas , SARS-CoV-2 , Glicoproteína da Espícula de Coronavírus , Cálcio , DNA de Cadeia Simples/análise , Doxorrubicina/administração & dosagem , Células HEK293 , Células HeLa , Humanos , Glicoproteína da Espícula de Coronavírus/análise , Glicoproteína da Espícula de Coronavírus/genética , Óxido de Zinco
3.
J Control Release ; 333: 28-40, 2021 05 10.
Artigo em Inglês | MEDLINE | ID: mdl-33741386

RESUMO

Antisense oligonucleotides (ASOs) carry an enormous therapeutic potential in different research areas, however, the lack of appropriate carriers for their delivery to the target tissues is hampering their clinical translation. The present study investigates the application of novel biomimetic nano-vesicles, Nano-Ghosts (NGs), for the delivery of ASOs to human mesenchymal stem cells (MSCs), using a microRNA inhibitor (antimiR) against miR-221 as proof-of-concept. The integration of this approach with a hyaluronic acid-fibrin (HA-FB) hydrogel scaffold is also studied, thus expanding the potential of NGs applications in regenerative medicine. The study shows robust antimiR encapsulation in the NGs using electroporation and the NGs ability to be internalized in MSCs and to deliver their cargo while avoiding endo-lysosomal degradation. This leads to rapid and strong knock-down of miR-221 in hMSCs in vitro, both in 2D and 3D hydrogel culture conditions (>90% and > 80% silencing efficiency, respectively). Finally, in vivo studies performed with an osteochondral defect model demonstrate the NGs ability to effectively deliver antimiR to endogenous cells. Altogether, these results prove that the NGs can operate as stand-alone system or as integrated platform in combination with scaffolds for the delivery of ASOs for a wide range of applications in drug delivery and regenerative medicine.


Assuntos
Células-Tronco Mesenquimais , MicroRNAs , Biomimética , Humanos , Hidrogéis , Oligonucleotídeos Antissenso
4.
Methods ; 177: 126-134, 2020 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-31794834

RESUMO

Currently, nano-carriers for anti-cancer drug delivery are complex systems, which struggle with immunogenicity and enhanced permeability effect (EPR)-related problems that halt the clinical translation of many therapeutics. Consequently, a rapidly growing field of research has been focusing on biomimetic nano-vesicles (BNVs) as an effective delivery alternative. Nevertheless, the translation of many BNVs is limited due to scalability problems, inconsistent production process, and insufficient loading efficiency. Here we discuss the process of our previously published BNVs, termed Nano-Ghosts (NGs), which are produced from the membrane of mesenchymal stem cells. We demonstrate the flexibility of the process, while alternating physical methodologies (sonication or extrusion) to produce the NGs while preserving their desired characteristics. We also show that our NGs can be labeled using multiple methods (fluorescence, radiolabeling, and genetic engineering) for tracking and diagnostic purposes. Lastly, we demonstrate that the loading efficiency can be improved by using electroporation to accommodate a range of therapeutics (small molecules, peptides and DNA) that can be delivered by the NGs. Our results emphasize the robustness of the NGs technology, its versatility and a vast range of applications, differentiating it from other BNVs and leading the way towards clinical translation.


Assuntos
Materiais Biomiméticos/química , Composição de Medicamentos/métodos , Sistemas de Liberação de Medicamentos/métodos , Vesículas Extracelulares/metabolismo , Células-Tronco Mesenquimais/química , Células A549 , Bioengenharia/métodos , Transporte Biológico , Materiais Biomiméticos/metabolismo , Membrana Celular/química , Membrana Celular/metabolismo , Liberação Controlada de Fármacos , Eletroporação/métodos , Vesículas Extracelulares/química , Vesículas Extracelulares/transplante , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Humanos , Mesilato de Imatinib/metabolismo , Mesilato de Imatinib/farmacologia , Cinética , Células-Tronco Mesenquimais/metabolismo , Nanoestruturas/química , Peptídeos/metabolismo , Peptídeos/farmacologia , Sonicação/métodos , Coloração e Rotulagem/métodos
5.
Adv Healthc Mater ; 8(10): e1801589, 2019 05.
Artigo em Inglês | MEDLINE | ID: mdl-30963725

RESUMO

Nanoghosts (NGs) are nanovesicles reconstructed from the cytoplasmic membranes of mesenchymal stem cells (MSCs). By retaining MSC membranes, the NGs retain the ability of these cells to home in on multiple tumors, laying the foundations, thereby, for the development of a targeted drug delivery platform. The susceptibility of MSCs to functional changes, following their exposure to cytokines or cancer-derived conditioned-media (CM), presents the opportunity to modify the NGs by conditioning their source cells. This opportunity is investigated by comparing the membrane protein composition and the tumor uptake of NGs derived from naïve MSCs (N-NG) against conditioned NGs made from MSCs pre-treated with conditioned-media (CM-NG) or with a mix of the proinflammatory cytokines TNF-α and IL-1ß (Cyto-NG). CM-NGs are found to be more targeted towards immune cells than Cyto- or N-NGs, while Cyto-NGs are the most tumor-targeted ones, with similar immune-targeting capacity as N-NGs but with a higher affinity towards endothelial cells. Proteomic variations were wider in the CM-NGs, with exceptionally higher levels of ICAM-1 compared to N- and Cyto-NGs. From a translational point of view, the data show that the tumor-targeting ability of the NGs, and possibly that of other MSC-derived extracellular vesicles, can be enhanced by simple conditioning of their source cells.


Assuntos
Membrana Celular/metabolismo , Meios de Cultivo Condicionados/farmacologia , Citocinas/farmacologia , Células-Tronco Mesenquimais/efeitos dos fármacos , Animais , Linhagem Celular , Membrana Celular/química , Humanos , Integrinas/metabolismo , Molécula 1 de Adesão Intercelular/metabolismo , Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/metabolismo , Camundongos , Camundongos Nus , Nanoestruturas/química , Neoplasias/imunologia , Neoplasias/metabolismo , Neoplasias/patologia , Proteoma/metabolismo
6.
Nanomedicine ; 18: 169-178, 2019 06.
Artigo em Inglês | MEDLINE | ID: mdl-30853651

RESUMO

Mammalian cell membranes are often incompatible with chemical modifications typically used to increase circulation half-life. Using cellular nanoghosts as a model, we show that proline-alanine-serine (PAS) peptide sequences expressed on the membrane surface can extend the circulation time of a cell membrane derived nanotherapeutic. Membrane expression of a PAS 40 repeat sequence decreased protein binding and resulted in a 90% decrease in macrophage uptake when compared with non-PASylated controls (P ≤ 0.05). PASylation also extended circulation half-life (t1/2 = 37 h) compared with non-PASylated controls (t1/2 = 10.5 h) (P ≤ 0.005), resulting in ~7-fold higher in vivo serum concentrations at 24 h and 48 h (P ≤ 0.005). Genetically engineered membrane expression of PAS repeats may offer an alternative to PEGylation and provide extended circulation times for cellular membrane-derived nanotherapeutics.


Assuntos
Membrana Celular/metabolismo , Nanopartículas/uso terapêutico , Engenharia de Proteínas , Adsorção , Animais , Proteínas Sanguíneas/metabolismo , Difusão Dinâmica da Luz , Células HEK293 , Humanos , Camundongos Endogâmicos BALB C , Nanopartículas/química , Nanopartículas/ultraestrutura , Copolímero de Ácido Poliláctico e Ácido Poliglicólico/química , Ratos , Propriedades de Superfície , Distribuição Tecidual
7.
J Control Release ; 293: 215-223, 2019 01 10.
Artigo em Inglês | MEDLINE | ID: mdl-30527755

RESUMO

The rapid development of biomimetic cell membrane-based nanoparticles is still overshadowed by many practical challenges, one of which is the difficulty to precisely measure the biodistribution of such nanoparticles. Currently, this challenge is mostly addressed using fluorescent techniques with limited sensitivity, or radioactive labeling methods, which rarely account for the nanoparticles themselves, but their payloads instead. Here we report the development of a robust method for the innate radioactive labeling of cells and membrane-based nanoparticles and their consequent sensitive detection and biodistribution measurements. The preclinical potential of this method was demonstrated with Nano-Ghosts (NGs), manufactured from the cytoplasmic membranes of mesenchymal stem cells cultured with radioactively-labeled linoleic acid and achieving a cell labeling efficiency of 36%. Radiolabeling did not affect the physiochemical properties of the NGs, which stably retained their radiolabels. Using radioactivity measurements, we are now able to determine precisely the amount of NGs uptaken by tissues and cells, thereby providing further support to our presumed active NG targeting mechanisms. Biodistribution studies comparing radiolabeled NGs to fluorescently-labeled ones have validated our method and revealed new information, which could not be obtained otherwise, regarding the NGs' unique kinetics and rapid clearance, supporting their excellent safety profiles. The reported approach may be expanded to other membrane-based entities to facilitate and hasten their preclinical development and be used in parallel with other labeling methods to provide different and additional information.


Assuntos
Membrana Celular , Células-Tronco Mesenquimais , Nanoestruturas/administração & dosagem , Células A549 , Animais , Radioisótopos de Carbono , Humanos , Ácido Linoleico/administração & dosagem , Masculino , Camundongos Endogâmicos C57BL , Camundongos Nus , Distribuição Tecidual
8.
Cancer Lett ; 414: 239-249, 2018 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-29175461

RESUMO

Mesenchymal stem cells (MSCs) are multipotent stromal cells which can differentiate into a variety of cell types including osteoblasts, adipocytes and chondrocytes. They are normally resident in adipose tissue, bone marrow and the umbilical cord, but can also be found in other tissues and are known to be recruited to sites of wound healing as well as growing tumours. The therapeutic potential of MSCs has been explored in a number of phase I/II and III clinical trials, of which several were targeted against graft-versus-host disease and to support engraftment of haematopoietic stem cells (HSCs), but currently only very few in the oncology field. There are now three clinical trials either ongoing or recruiting patients that use MSCs to treat tumour disease. In these, MSCs target gastrointestinal, lung and ovarian cancer, respectively. The first study uses MSCs loaded with a HSV-TK expression construct under the control of the CCL5 promoter, and has recently reported successful completion of Phase I/II. While no adverse side effects were seen during this study, no outcomes with respect to therapeutic benefits have been published. The other clinical trials targeting lung and ovarian cancer will be using MSCs expressing cytokines as therapeutic payload. Despite these encouraging early steps towards their clinical use, many questions are still unanswered regarding the biology of MSCs in normal and pathophysiological settings. In this review, in addition to summarising the current state of MSC-based therapeutic approaches for cancer, we will describe the remaining questions, obstacles and risks, as well as novel developments such as MSC-derived nanoghosts.


Assuntos
Transplante de Células-Tronco Mesenquimais/métodos , Células-Tronco Mesenquimais/citologia , Neoplasias/terapia , Diferenciação Celular , Ensaios Clínicos como Assunto , Sistemas de Liberação de Medicamentos/métodos , Terapia Genética/métodos , Humanos , Células-Tronco Mesenquimais/metabolismo , Modelos Biológicos
9.
Nano Lett ; 16(3): 1574-82, 2016 Mar 09.
Artigo em Inglês | MEDLINE | ID: mdl-26901695

RESUMO

Nanoghosts derived from mesenchymal stem cells and retaining their unique surface-associated tumor-targeting capabilities were redesigned as a selective and safe universal nonviral gene-therapy platform. pDNA-loaded nanoghosts efficiently targeted and transfected diverse cancer cells, in vitro and in vivo, in subcutaneous and metastatic orthotopic tumor models, leading to no adverse effects. Nanoghosts loaded with pDNA encoding for a cancer-toxic gene inhibited the growth of metastatic orthotopic lung cancer and subcutaneous prostate cancer models and dramatically prolonged the animals' survival.


Assuntos
DNA/administração & dosagem , Técnicas de Transferência de Genes , Terapia Genética , Neoplasias Pulmonares/terapia , Células-Tronco Mesenquimais , Nanoestruturas , Neoplasias da Próstata/terapia , Animais , Linhagem Celular Tumoral , DNA/genética , DNA/uso terapêutico , Humanos , Pulmão/metabolismo , Pulmão/patologia , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patologia , Masculino , Células-Tronco Mesenquimais/citologia , Camundongos Endogâmicos C57BL , Nanoestruturas/administração & dosagem , Nanoestruturas/efeitos adversos , Nanoestruturas/ultraestrutura , Metástase Neoplásica/genética , Metástase Neoplásica/patologia , Metástase Neoplásica/terapia , Plasmídeos/administração & dosagem , Plasmídeos/genética , Plasmídeos/uso terapêutico , Próstata/metabolismo , Próstata/patologia , Neoplasias da Próstata/genética , Neoplasias da Próstata/patologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA