Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
ACS Appl Mater Interfaces ; 16(37): 49092-49103, 2024 Sep 18.
Artículo en Inglés | MEDLINE | ID: mdl-39252643

RESUMEN

Nanoparticles engineered to combat cancer and other life-threatening diseases may significantly improve patient outcomes. However, inefficient nanoparticle delivery to tumors limits their use and necessitates the development of complex delivery approaches. Here, we examine this issue by harnessing the tumor-homing abilities of human mesenchymal stem cells (MSCs) to deliver a decoupled theranostic complex of rare earth-doped nanoparticles (dNPs) and photosensitizer chlorin e6 (Ce6) to tumors. We show that both bone-marrow- and skin-derived MSCs can transport the dNP-Ce6 complex inside tumor spheroids, which is challenging to accomplish by passive delivery alone. MSCs deliver the dNP-Ce6 complex across the tumor spheroid, facilitating more effective photodynamic damage and tumor destruction than passively accumulated dNP-Ce6. The dNP-Ce6 complex also provides the built-in ability to monitor the MSC migration without causing undesired phototoxicity, which is essential for maximal and side-effect-free delivery of nanoparticles. Our results demonstrate how MSCs can be used as delivery vehicles for the transportation of the dNP-Ce6 complex, addressing the limitations of passive nanoparticle delivery and providing light-based theranostics.


Asunto(s)
Clorofilidas , Células Madre Mesenquimatosas , Nanopartículas , Fotoquimioterapia , Fármacos Fotosensibilizantes , Nanomedicina Teranóstica , Células Madre Mesenquimatosas/citología , Humanos , Nanopartículas/química , Fármacos Fotosensibilizantes/química , Fármacos Fotosensibilizantes/farmacología , Animales , Porfirinas/química , Porfirinas/farmacología , Ratones , Línea Celular Tumoral , Neoplasias/terapia , Neoplasias/patología , Neoplasias/tratamiento farmacológico
2.
J Nanobiotechnology ; 17(1): 39, 2019 Mar 13.
Artículo en Inglés | MEDLINE | ID: mdl-30866960

RESUMEN

BACKGROUND: Human mesenchymal stem cells (MSCs) have drawn much attention in the field of regenerative medicine for their immunomodulatory and anti-inflammatory effects. MSCs possess specific tumor-oriented migration and incorporation highlighting the potential for MSCs to be used as an ideal carrier for anticancer agents. Bone marrow is the main source of MSCs for clinical applications. MSCs tracking in vivo is a critical component of the safety and efficacy evaluation of therapeutic cell products; therefore, cells must be labeled with contrast agents to enable visualization of the MSCs migration in vivo. Due to their unique properties, quantum dots (QDs) are emerging as optimal tools in long-term MSC optical imaging applications. The aim of this study was to investigate the uptake dynamics, cytotoxity, subcellular and extracellular distribution of non-targeted carboxylated quantum dots in human bone marrow MSCs at different cell growing densities. RESULTS: QDs had no negative impact on MSC viability throughout the experiment and accumulated in all observed cells efficiently; however, in some MSCs QDs induced formation of lipid droplets. At low cell growing densities QDs distribute within MSCs cytoplasm already after 1 h of incubation reaching saturation after 6 h. After 24 h QDs localize mainly in the perinuclear region of the cells in endosomes. Interestingly, in more confluent culture QDs localize mostly outside MSCs. QDs abundantly mark MSC long filopodia-like structures attaching neighboring cells. At high cell density cultivation, we for the first time demonstrated that carboxylated QDs localize in human bone marrow MSC extracellular matrix. Moreover, we observed that average photoluminescence lifetime of QDs distributed in extracellular matrix are longer than lifetimes of QDs entrapped in endocytic vesicles; thus, for the first time showing the possibility to identify and distinguish localization of QDs in various extracellular and intracellular structures using fluorescence-lifetime imaging microscopy without additional staining assays. CONCLUSION: Carboxylated QDs can be used as nonspecific and effective dye for staining of human bone marrow MSCs and their specific extracellular structures. These results are promising in fundamental stem cell biology as well as in cellular therapy, anticancer drug delivery and tissue engineering.


Asunto(s)
Ácidos Carboxílicos/química , Colorantes Fluorescentes/metabolismo , Células Madre Mesenquimatosas/metabolismo , Puntos Cuánticos/metabolismo , Transporte Biológico , Movimiento Celular , Supervivencia Celular , Matriz Extracelular/metabolismo , Humanos , Imagen Óptica , Coloración y Etiquetado , Factores de Tiempo
3.
Int J Nanomedicine ; 12: 8129-8142, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-29158674

RESUMEN

PURPOSE: Cell-mediated delivery of nanoparticles is emerging as a new method of cancer diagnostics and treatment. Due to their inherent regenerative properties, adult mesenchymal stem cells (MSCs) are naturally attracted to wounds and sites of inflammation, as well as tumors. Such characteristics enable MSCs to be used in cellular hitchhiking of nanoparticles. In this study, MSCs extracted from the skin connective tissue were investigated as transporters of semiconductor nanocrystals quantum dots (QDs). MATERIALS AND METHODS: Cytotoxicity of carboxylated CdSe/ZnS QDs was assessed by lactate dehydrogenase cell viability assay. Quantitative uptake of QDs was determined by flow cytometry; their intracellular localization was evaluated by confocal microscopy. In vitro tumor-tropic migration of skin-derived MSCs was verified by Transwell migration assay. For in vivo migration studies of QD-loaded MSCs, human breast tumor-bearing immunodeficient mice were used. RESULTS: QDs were found to be nontoxic to MSCs in concentrations no more than 16 nM. The uptake studies showed a rapid QD endocytosis followed by saturating effects after 6 h of incubation and intracellular localization in the perinuclear region. In vitro migration of MSCs toward MDA-MB-231 breast cancer cells and their conditioned medium was up to nine times greater than the migration toward noncancerous breast epithelial cells MCF-10A. In vivo, systemically administered QD-labeled MSCs were mainly located in the tumor and metastatic tissues, evading most healthy organs with the exception being blood clearance organs (spleen, kidneys, liver). CONCLUSION: Skin-derived MSCs demonstrate applicability in cell-mediated delivery of nanoparticles. The findings presented in this study promise further development of a cell therapy and nanotechnology-based tool for early cancer diagnostics and therapy.


Asunto(s)
Neoplasias de la Mama/patología , Células Madre Mesenquimatosas/citología , Puntos Cuánticos/química , Piel/citología , Animales , Muerte Celular , Línea Celular Tumoral , Movimiento Celular , Forma de la Célula , Dispersión Dinámica de Luz , Endocitosis , Femenino , Humanos , Ratones SCID , Nanopartículas/química , Tamaño de la Partícula
4.
Beilstein J Nanotechnol ; 8: 1218-1230, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28685122

RESUMEN

Nanotechnology-based drug design offers new possibilities for the use of nanoparticles in imaging and targeted therapy of tumours. Due to their tumour-homing ability, nano-engineered mesenchymal stem cells (MSCs) could be utilized as vectors to deliver diagnostic and therapeutic nanoparticles into a tumour. In the present study, uptake and functional effects of carboxyl-coated quantum dots QD655 were studied in human skin MSCs. The effect of QD on MSCs was examined using a cell viability assay, Ki67 expression analysis, and tri-lineage differentiation assay. The optimal conditions for QD uptake in MSCs were determined using flow cytometry. The QD uptake route in MSCs was examined via fluorescence imaging using endocytosis inhibitors for the micropinocytosis, phagocytosis, lipid-raft, clathrin- and caveolin-dependent endocytosis pathways. These data showed that QDs were efficiently accumulated in the cytoplasm of MSCs after incubation for 6 h. The main uptake route of QDs in skin MSCs was clathrin-mediated endocytosis. QDs were mainly localized in early endosomes after 6 h as well as in late endosomes and lysosomes after 24 h. QDs in concentrations ranging from 0.5 to 64 nM had no effect on cell viability and proliferation. The expression of MSC markers, CD73 and CD90, and hematopoietic markers, CD34 and CD45, as well as the ability to differentiate into adipocytes, chondrocytes, and osteocytes, were not altered in the presence of QDs. We observed a decrease in the QD signal from labelled MSCs over time that could partly reflect QD excretion. Altogether, these data suggest that QD-labelled MSCs could be used for targeted drug delivery studies.

5.
Medicina (Kaunas) ; 50(4): 237-44, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25458961

RESUMEN

BACKGROUND AND OBJECTIVE: Superparamagnetic iron oxide nanoparticles (SPIONs) emerge as a promising tool for early cancer diagnostics and targeted therapy. However, both toxicity and biological activity of SPIONs should be evaluated in detail. The aim of this study was to synthesize superparamagnetic cobalt ferrite nanoparticles (Co-SPIONs), and to investigate their uptake, toxicity and effects on cancer stem-like properties in human pancreatic cancer cell line MiaPaCa2 and human ovarian cancer cell line A2780. MATERIALS AND METHODS: Co-SPIONs were produced by Massart's co-precipitation method. The cells were treated with Co-SPIONs at three different concentrations (0.095, 0.48, and 0.95µg/mL) for 24 and 48h. Cell viability and proliferation were analyzed after treatment. The stem-like properties of cells were assessed by investigating the cell clonogenicity and expression of cancer stem cell-associated markers, including CD24/ESA in A2780 cell line and CD44/ALDH1 in MiaPaCa2 cell line. Magnetically activated cell sorting was used for the separation of magnetically labeled and unlabeled cells. RESULTS: Both cancer cell lines accumulated Co-SPIONs, however differences in response to nanoparticles were observed between MiaPaCa2 and A2780 cell. In particular, A2780 cells were more sensitive to exposition to Co-SPIONs than MiaPaCa2 cells, indicating that a safe concentration of nanoparticles must be estimated individually for a particular cell type. Higher doses of Co-SPIONs decreased both the clonogenicity and ESA marker expression in A2780 cells. CONCLUSIONS: Co-SPIONs are not cytotoxic to cancer cells, at least when used at a concentration of up to 0.95µg/mL. Co-SPIONs have a dose-dependent effect on the clonogenic potential and ESA marker expression in A2780 cells. Magnetic detection of low concentrations of Co-SPIONS in cancer cells is a promising tool for further applications of these nanoparticles in cancer diagnosis and treatment; however, extensive research in this field is needed.


Asunto(s)
Cobalto/metabolismo , Compuestos Férricos/metabolismo , Nanopartículas/metabolismo , Neoplasias Ováricas/metabolismo , Neoplasias Pancreáticas/metabolismo , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Separación Celular , Supervivencia Celular/efectos de los fármacos , Cobalto/farmacología , Femenino , Compuestos Férricos/farmacología , Citometría de Flujo , Humanos , Tamaño de la Partícula
6.
J Biomed Nanotechnol ; 10(4): 679-86, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24734520

RESUMEN

Recently it has been suggested that quantum dots could be used in the photodynamic therapy of cancer as resonant energy donors for conventional porphyrin type photosensitizers. Here we summarize our results obtained by studying a non-covalent complex formed between quantum dots and a second generation photosensitizer, chlorin e6, in aqueous medium and in live pancreatic MiaPaCa2 cancer cells. Spectral changes in the absorption and photoluminescence of quantum dots and chlorin e6, as well as changes in the photoluminescence lifetime of quantum dots, revealed the formation of quantum dot-chlorin e6 complex. Fluorescence confocal microscopy with spectral imaging unit showed uptake of quantum dot-chlorin e6 complex in live cancer cells: the complex localized in plasma membrane and endocytic vesicles. Fluorescence lifetime imaging revealed Forster resonance energy transfer from quantum dots to chlorin e6 within live cells. Finally, a light-induced damage to cancer cells by the quantum dot-chlorin e6 complex was achieved.


Asunto(s)
Neoplasias Pancreáticas/tratamiento farmacológico , Neoplasias Pancreáticas/metabolismo , Fotoquimioterapia/métodos , Porfirinas/farmacocinética , Porfirinas/uso terapéutico , Puntos Cuánticos , Apoptosis/efectos de los fármacos , Apoptosis/efectos de la radiación , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/efectos de la radiación , Clorofilidas , Humanos , Neoplasias Pancreáticas/patología , Fármacos Fotosensibilizantes/farmacocinética , Fármacos Fotosensibilizantes/uso terapéutico
7.
Int J Nanomedicine ; 6: 2437-44, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-22072879

RESUMEN

BACKGROUND: In novel treatment approaches, therapeutics should be designed to target cancer stem cells (CSCs). Quantum dots (QDs) are a promising new tool in fighting against cancer. However, little is known about accumulation and cytotoxicity of QDs in CSCs. METHODS: Accumulation and cytotoxicity of CdTe-MPA (mercaptopropionic acid) QDs in CSCs were assessed using flow cytometry and fluorescence-activated cell sorting techniques as well as a colorimetric cell viability assay. RESULTS: We investigated the expression of two cell surface-associated glycoproteins, CD44 and CD133, in four different cancer cell lines (glioblastoma, melanoma, pancreatic, and prostate adenocarcinoma). Only the melanoma cells were positive to both markers of CD44 and CD133, whereas the other cells were only CD44-positive. The QDs accumulated to a similar extent in all subpopulations of the melanoma cells. The phenotypical response after QD treatment was compared with the response after ionizing radiation treatment. The percentage of the CD44(high-)CD133(high) subpopulation decreased from 72% to 55%-58% for both treatments. The stem-like subpopulation CD44(high)CD133(low/-) increased from 26%-28% in the untreated melanoma cells to 36%-40% for both treatments. CONCLUSION: Treatment of melanoma cells with QDs results in an increase of stem-like cell subpopulations. The changes in phenotype distribution of the melanoma cells after the treatment with QDs are comparable with the changes after ionizing radiation.


Asunto(s)
Antígenos CD/biosíntesis , Compuestos de Cadmio/administración & dosificación , Glicoproteínas/biosíntesis , Melanoma/terapia , Puntos Cuánticos , Telurio/administración & dosificación , Ácido 3-Mercaptopropiónico/administración & dosificación , Ácido 3-Mercaptopropiónico/química , Ácido 3-Mercaptopropiónico/farmacocinética , Antígeno AC133 , Biomarcadores de Tumor , Compuestos de Cadmio/química , Compuestos de Cadmio/farmacocinética , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Citometría de Flujo , Glioblastoma/inmunología , Glioblastoma/patología , Glioblastoma/terapia , Humanos , Receptores de Hialuranos/biosíntesis , Masculino , Melanoma/inmunología , Melanoma/metabolismo , Melanoma/patología , Células Madre Neoplásicas/patología , Neoplasias Pancreáticas/inmunología , Neoplasias Pancreáticas/patología , Neoplasias Pancreáticas/terapia , Péptidos , Fenotipo , Neoplasias de la Próstata/inmunología , Neoplasias de la Próstata/patología , Neoplasias de la Próstata/terapia , Telurio/química , Telurio/farmacocinética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA