Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Nanomedicine ; 39: 102459, 2022 01.
Artigo em Inglês | MEDLINE | ID: mdl-34530163

RESUMO

Combining diagnosis and treatment approaches in one entity is the goal of theranostics for cancer therapy. Magnetic nanoparticles have been extensively used as contrast agents for nuclear magnetic resonance imaging as well as drug carriers and remote actuation agents. Poly(2-oxazoline)-based polymeric micelles, which have been shown to efficiently solubilize hydrophobic drugs and drug combinations, have high loading capacity (above 40% w/w) for paclitaxel. In this study, we report the development of novel theranostic system, NanoFerrogels, which is designed to capitalize on the magnetic nanoparticle properties as imaging agents and the poly(2-oxazoline)-based micelles as drug loading compartment. We developed six formulations with magnetic nanoparticle content of 0.3%-12% (w/w), with the z-average sizes of 85-130 nm and ξ-potential of 2.7-28.3 mV. The release profiles of paclitaxel from NanoFerrogels were notably dependent on the degree of dopamine grafting on poly(2-oxazoline)-based micelles. Paclitaxel loaded NanoFerrogels showed efficacy against three breast cancer lines which was comparable to free paclitaxel. They also showed improved tumor and lymph node accumulation and signal reduction in vivo (2.7% in tumor; 8.5% in lymph node) compared to clinically approved imaging agent ferumoxytol (FERAHEME®) 24 h after administration. NanoFerrogels responded to super-low frequency alternating current magnetic field (50 kA m-1, 50 Hz) which accelerated drug release from paclitaxel-loaded NanoFerrogels or caused death of cells loaded with NanoFerrogels. These proof-of-concept experiments demonstrate that NanoFerrogels have potential as remotely actuated theranostic platform for cancer diagnosis and treatment.


Assuntos
Antineoplásicos , Neoplasias da Mama , Antineoplásicos/uso terapêutico , Neoplasias da Mama/tratamento farmacológico , Linhagem Celular Tumoral , Portadores de Fármacos/química , Sistemas de Liberação de Medicamentos , Liberação Controlada de Fármacos , Feminino , Óxido Ferroso-Férrico , Humanos , Campos Magnéticos , Micelas , Oxazóis , Paclitaxel/farmacologia , Paclitaxel/uso terapêutico , Medicina de Precisão
2.
Int J Mol Sci ; 23(19)2022 Sep 22.
Artigo em Inglês | MEDLINE | ID: mdl-36232435

RESUMO

The magneto-mechanical approach is a powerful technique used in many different applications in biomedicine, including remote control enzyme activity, cell receptors, cancer-selective treatments, mechanically-activated drug releases, etc. This approach is based on the use of a combination of magnetic nanoparticles and external magnetic fields that have led to the movement of such nanoparticles with torques and forces (enough to change the conformation of biomolecules or even break weak chemical bonds). However, despite many theoretical and experimental works on this topic, it is difficult to predict the magneto-mechanical effects in each particular case, while the important results are scattered and often cannot be translated to other experiments. The main reason is that the magneto-mechanical effect is extremely sensitive to changes in any parameter of magnetic nanoparticles and the environment and changes in the parameters of the applied magnetic field. Thus, in this review, we (1) summarize and propose a simplified theoretical explanation of the main factors affecting the efficiency of the magneto-mechanical approach; (2) discuss the nature of the MNP-mediated mechanical forces and their order of magnitude; (3) show some of the main applications of the magneto-mechanical approach in the control over the properties of biological systems.


Assuntos
Campos Magnéticos , Nanopartículas , Magnetismo
3.
J Colloid Interface Sci ; 657: 778-787, 2024 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-38081112

RESUMO

Magneto-mechanical actuation (MMA) using the low-frequency alternating magnetic fields (AMFs) of magnetic nanoparticles internalized into cancer cells can be used to irreparably damage these cells. However, nanoparticles in cells usually agglomerate, thus greatly augmenting the delivered force compared to single nanoparticles. Here, we demonstrate that MMA also decreases the cell viability, with the MMA mediated by individual, non-interacting nanoparticles. The effect was demonstrated with ferrimagnetic (i.e., permanently magnetic) barium-hexaferrite nanoplatelets (NPLs, ∼50 nm wide and 3 nm thick) with a unique, perpendicular orientation of the magnetization. Two cancer-cell lines (MDA-MB-231 and HeLa) are exposed to the NPLs in-vitro under different cell-culture conditions and actuated with a uniaxial AMF. TEM analyses show that only a small number of NPLs internalize in the cells, always situated in membrane-enclosed compartments of the endosomal-lysosomal system. Most compartments contain 1-2 NPLs and only seldom are the NPLs found in small groups, but never in close contact or mutually oriented. Even at low concentrations, the single NPLs reduce the cell viability when actuated with AMFs, which is further increased when the cells are in starvation conditions. These results pave the way for more efficient in-vivo MMA at very low particle concentrations.


Assuntos
Nanopartículas , Neoplasias , Humanos , Campos Magnéticos , Células HeLa , Lisossomos
4.
ACS Biomater Sci Eng ; 9(12): 6902-6914, 2023 Dec 11.
Artigo em Inglês | MEDLINE | ID: mdl-38014849

RESUMO

Cancer treatment is one of the major health problems that burden our society. According to the American Cancer Society, over 1.9 million new cancer cases and ∼0.6 million deaths from cancer are expected in the US in 2023. Therapeutic targeting is considered to be the gold standard in cancer treatment. However, when a tumor grows beyond a critical size, its vascular system differentiates abnormally and erratically, creating a heterogeneous endothelial barrier that further restricts drug delivery into tumors. While several methods exist, these prompt tumor migration and the appearance of new metastatic sites. Herein, we propose an innovative method based on magneto-mechanical actuation (MMA) to induce endothelial permeability. This method employs FDA-approved PEGylated superparamagnetic iron oxide nanoparticles (PEG-SPIONs) and alternating nonheating magnetic fields. MMA lies in the translation of magnetic forces into mechanical agitation. As a proof of concept, we developed a 2D cell culture model based on human umbilical vein endothelial cells (HUVEC), which were incubated with PEG-SPIONs and then exposed to different magnetic doses. After adjusting the particle concentration, incubation times, and parameters (amplitude, frequency, and exposure time) of the magnetic field generator, we induced actin filament remodeling and subsequent vascular endothelial-cadherin junction disruption. This led to transient gaps in cell monolayers, through which fluorescein isothiocyanate-dextran was translocated. We observed no cell viability reduction for 3 h of particle incubation up to a concentration of 100 µg/mL in the presence and absence of magnetic fields. For optimal permeability studies, the magnetic field parameters were adjusted to 100 mT, 65 Hz, and 30 min in a pulse mode with 5 min OFF intervals. We found that the endothelial permeability reached the highest value (33%) when 2 h postmagnetic field treatment was used. To explain these findings, a magneto-mechanical transduced stress mechanism mediated by intracellular forces was proposed. This method can open new avenues for targeted drug delivery into anatomic regions within the body for a broad range of disease interventions.


Assuntos
Sistemas de Liberação de Medicamentos , Neoplasias , Estados Unidos , Humanos , Células Endoteliais da Veia Umbilical Humana , Permeabilidade
5.
ACS Nano ; 17(21): 21690-21707, 2023 11 14.
Artigo em Inglês | MEDLINE | ID: mdl-37862095

RESUMO

Abnormal mechanical loading often leads to the progressive degradation of cartilage and causes osteoarthritis (OA). Although multiple mechanoresponsive strategies based on biomaterials have been designed to restore healthy cartilage microenvironments, methods to remotely control the on-demand mechanical forces for cartilage repair pose significant challenges. Here, a magneto-mechanically controlled mesenchymal stem cell (MSC) platform, based on the integration of intercellular mechanical communication and intracellular mechanosignaling processes, is developed for OA treatment. MSCs loaded with antioxidative melanin@Fe3O4 magnetic nanoparticles (Magcells) rapidly assemble into highly ordered cell clusters with enhanced cell-cell communication under a time-varying magnetic field, which enables long-term retention and differentiation of Magcells in the articular cavity. Subsequently, via mimicking the gait cycle, chondrogenesis can be further enhanced by the dynamic activation of mechanical signaling processes in Magcells. This sophisticated magneto-mechanical actuation strategy provides a paradigm for developing mechano-therapeutics to repair cartilage in OA treatment.


Assuntos
Cartilagem Articular , Células-Tronco Mesenquimais , Osteoartrite , Humanos , Condrogênese , Condrócitos/metabolismo , Osteoartrite/terapia , Diferenciação Celular
6.
ACS Appl Mater Interfaces ; 15(16): 19877-19891, 2023 Apr 26.
Artigo em Inglês | MEDLINE | ID: mdl-37040569

RESUMO

Engineered cells used as smart vehicles for delivery of secreted therapeutic proteins enable effective treatment of cancer and certain degenerative, autoimmune, and genetic disorders. However, current cell-based therapies use mostly invasive tools for tracking proteins and do not allow for controlled secretion of therapeutic proteins, which could result in unconstrained killing of surrounding healthy tissues or ineffective killing of host cancer cells. Regulating the expression of therapeutic proteins after success of therapy remains elusive. In this study, a noninvasive therapeutic approach mediated by magneto-mechanical actuation (MMA) was developed to remotely regulate the expression of the tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) protein, which is secreted by transduced cells. Stem cells, macrophages, and breast cancer cells were transduced with a lentiviral vector encoding the SGpL2TR protein. SGpL2TR comprises TRAIL and GpLuc domains optimized for cell-based applications. Our approach relies on the remote actuation of cubic-shape highly magnetic field responsive superparamagnetic iron oxide nanoparticles (SPIONs) coated with nitrodopamine PEG (ND-PEG), which are internalized within the cells. Cubic ND-PEG-SPIONs actuated by superlow frequency alternating current magnetic fields can translate magnetic forces into mechanical motion and in turn spur mechanosensitive cellular responses. Cubic ND-PEG-SPIONs were artificially designed to effectively operate at low magnetic field strengths (<100 mT) retaining approximately 60% of their saturation magnetization. Compared to other cells, stems cells were more sensitive to the interaction with actuated cubic ND-PEG-SPIONs, which clustered near the endoplasmic reticulum (ER). Luciferase, ELISA, and RT-qPCR analyses revealed a marked TRAIL downregulation (secretion levels were depleted down to 30%) when intracellular particles at 0.100 mg/mL Fe were actuated by magnetic fields (65 mT and 50 Hz for 30 min). Western blot studies indicated actuated, intracellular cubic ND-PEG-SPIONs can cause mild ER stress at short periods (up to 3 h) of postmagnetic field treatment thus leading to the unfolded protein response. We observed that the interaction of TRAIL polypeptides with ND-PEG can also contribute to this response. To prove the applicability of our approach, we used glioblastoma cells, which were exposed to TRAIL secreted from stem cells. We demonstrated that in the absence of MMA treatment, TRAIL essentially killed glioblastoma cells indiscriminately, but when treated with MMA, we were able to control the cell killing rate by adjusting the magnetic doses. This approach can expand the capabilities of stem cells to serve as smart vehicles for delivery of therapeutic proteins in a controlled manner without using interfering and expensive drugs, while retaining their potential to regenerate damaged tissue after treatment. This approach brings forth new alternatives to regulate protein expression noninvasively for cell therapy and other cancer therapies.


Assuntos
Glioblastoma , Nanopartículas de Magnetita , Humanos , Nanopartículas de Magnetita/química , Polietilenoglicóis/química , Fenômenos Magnéticos
7.
Nanomaterials (Basel) ; 13(22)2023 Nov 14.
Artigo em Inglês | MEDLINE | ID: mdl-37999295

RESUMO

Magnetic nanoparticles (MPs) are emerging as powerful and versatile tools for biotechnology, including cancer research and theranostic applications. Stem cell-mediated magnetic particle delivery has been previously recognized as a modality to target sites of malignancies. Here, we propose the use of adipose-derived mesenchymal cells (ADSC) for the targeted delivery of Fe-Cr-Nb-B magnetic particles to human osteosarcoma (HOS) cells and magneto-mechanical actuation (MMA) for targeting and destroying HOS cells. We show that MPs are easily incorporated by ADSCs and HOS cells, as confirmed by TEM images and a ferrozine assay. MP-loaded ADSCs display increased motility towards tumor cells compared with their unloaded counterparts. MMA of MP-loaded ADSCs induces HOS destruction, as confirmed by the MTT and live/dead assays. MMA enables the release of the MPs towards cancer cells, producing a significant decrease (about 80%) in HOS viability immediately after application. In contrast, normal human dermal fibroblasts' (NHDFs) viability exposed to similar conditions remains high, showing a differential behavior of normal and malignant cells to MP load and MMA exposure. Taken together, the method could derive successful strategies for in vivo applications in targeting and destroying malignant cells while protecting normal cells.

8.
ACS Nano ; 15(12): 20020-20031, 2021 12 28.
Artigo em Inglês | MEDLINE | ID: mdl-34807565

RESUMO

Magnetic nanomotors (MNMs), powered by a magnetic field, are ideal platforms to achieve versatile biomedical applications in a collective and spatiotemporal fashion. Although the programmable swarm of MNMs that mimics the highly ordered behaviors of living creatures has been extensively studied at the microscale, it is of vital importance to manipulate MNM swarms at the nanoscale for on-demand tasks at the cellular level. In this work, a Cy5-tagged caspase-3-specific peptide-modified MNM is designed, and the adaptive control behaviors of MNM swarms are revealed in lysosomes to induce the cancer cell apoptosis under a rotating magnetic field (RMF). A magneto-programmed vortex is predicted to occur with swarms under RMF by the finite element method model and verified in vitro. According to the dynamic model and numerical simulation, the critical rotating frequency under which MNMs are out of step is strongly correlated to their assembling and swarming properties. The adaptivity of swarms maximizes the synchronous rotation to achieve an optimal energy conversion rate. The frequency-adapted controllability of MNM swarms for cancer cell apoptosis is observed in real time in vitro and in vivo. This work provides theoretical and experimental insights to adaptively control MNM swarms for cancer treatment.


Assuntos
Campos Magnéticos , Neoplasias , Simulação por Computador , Magnetismo , Rotação
9.
Colloids Surf B Biointerfaces ; 206: 111931, 2021 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-34171621

RESUMO

This work is devoted to studying the effects of non-magnetic shell coating on nanoparticles in a low frequency alternating magnetic field (LF AMF) on tumor cells in vitro. Two types of iron oxide nanoparticles with the same magnetic core with and without silica shells were synthesized. Nanoparticles with silica shells significantly decreased the viability of PC3 cancer cells in a low frequency alternating magnetic field according to the cytotoxicity test, unlike uncoated nanoparticles. We showed that cell death results from the intracellular membrane integrity failure, and the calcium ions concentration increase with the subsequent necrosis. Transmission electron microscopy images showed that the uncoated silica nanoparticles are primarily found in an aggregated form in cells. We believe that uncoated nanoparticles lose their colloidal stability in an acidic endosomal environment after internalization into the cell due to surface etching and the formation of aggregates. As a result, they encounter high endosomal macromolecular viscosity and become unable to rotate efficiently. We assume that effective rotation of nanoparticles causes cell death. In turn, silica shell coating increases nanoparticles stability, preventing aggregation in endosomes. Thus, we propose that the colloidal stability of magnetic nanoparticles inside cells is one of the key factors for effective magneto-mechanical actuation.


Assuntos
Nanopartículas de Magnetita , Neoplasias , Campos Magnéticos , Magnetismo , Nanopartículas de Magnetita/toxicidade , Dióxido de Silício
10.
ACS Appl Bio Mater ; 3(12): 8172-8187, 2020 Dec 21.
Artigo em Inglês | MEDLINE | ID: mdl-35019598

RESUMO

The positive response of superparamagnetic iron oxide nanoparticles (SPIONs), in terms of biodegradability, circulation, elimination, toxicity, and manipulation of their structure/activity relationship, has enabled them to find their way into commercialization as an iron supplement, MRI contrast agents, MPI tracers, and hyperthermia and magneto-mechanical actuators. This Review focuses on the most current progress regarding the application of SPIONs as magnetic therapeutic agents for cancer treatment and tissue engineering. Because of their superior magnetic anisotropy, irreversibility of high- and low-field magnetization, and superparamagnetic ordering at corporal temperatures, they exhibit the unique ability to respond to theraputic doses (e.g., in magnetic hyperthermia and targeted drug delivery). This Review discusses the role of SPIONs to enhance chemotherapy and radiotherapy efficiency and specificity and how this enhancement could mitigate some side effects. SPIONs applied as tools for gene delivery, immunotherapy, and tissue engineering are also reviewed in the context of their potential to translational medicine. Lastly, some emerging issues concerning SPION toxicity are summarized and how they are being addressed to achieve success in clinical applications is discussed.

11.
J Colloid Interface Sci ; 579: 508-519, 2020 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-32623117

RESUMO

HYPOTHESIS: The magneto-mechanical actuation (MMA) of magnetic nanoparticles with a low-frequency alternating magnetic field (AMF) can be used to destroy cancer cells. So far, MMA was tested on different cells using different nanoparticles and different field characteristics, which makes comparisons and any generalizations about the results of MMA difficult. In this paper we propose the use of giant unilamellar vesicles (GUVs) as a simple model system to study the effect of MMA on a closed lipid bilayer membrane, i.e., a basic building block of any cell. EXPERIMENTS: The GUVs were exposed to barium-hexaferrite nanoplatelets (NPLs, ~50 nm wide and 3 nm thick) with unique magnetic properties dominated by a permanent magnetic moment that is perpendicular to the platelet, at different concentrations (1-50 µg/mL) and pH values (4.2-7.4) of the aqueous suspension. The GUVs were observed with an optical microscope while being exposed to a uniaxial AMF (3-100 Hz, 2.2-10.6 mT). FINDINGS: When the NPLs were electrostatically attached to the GUV membranes, the MMA induced cyclic fluctuations of the GUVs' shape corresponding to the AMF frequency at the low NPL concentration (1 µm/mL), whereas the GUVs were bursting at the higher concentration (10 µg/mL). Theoretical considerations suggested that the bursting of the GUVs is a consequence of the local action of an assembly of several NPLs, rather than a collective effect of all the absorbed NPLs.


Assuntos
Nanopartículas , Fosfolipídeos , Bário , Bicamadas Lipídicas , Lipossomas Unilamelares
12.
J Clin Med ; 8(10)2019 Sep 22.
Artigo em Inglês | MEDLINE | ID: mdl-31546701

RESUMO

There is currently an interest in "active" implantable biomedical devices that include mechanical stimulation as an integral part of their design. This paper reports the experimental use of a porous scaffold made of interconnected networks of slender ferromagnetic fibers that can be actuated in vivo by an external magnetic field applying strains to in-growing cells. Such scaffolds have been previously characterized in terms of their mechanical and cellular responses. In this study, it is shown that the shape changes induced in the scaffolds can be used to promote osteogenesis in vitro. In particular, immunofluorescence, gene and protein analyses reveal that the actuated networks exhibit higher mineralization and extracellular matrix production, and express higher levels of osteocalcin, alkaline phosphatase, collagen type 1α1, runt-related transcription factor 2 and bone morphogenetic protein 2 than the static controls at the 3-week time point. The results suggest that the cells filling the inter-fiber spaces are able to sense and react to the magneto-mechanically induced strains facilitating osteogenic differentiation and maturation. This work provides evidence in support of using this approach to stimulate bone ingrowth around a device implanted in bone and can pave the way for further applications in bone tissue engineering.

13.
Theranostics ; 7(6): 1735-1748, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28529648

RESUMO

Magnetic nanoparticles (MNPs) functionalized with targeting moieties can recognize specific cell components and induce mechanical actuation under magnetic field. Their size is adequate for reaching tumors and targeting cancer cells. However, due to the nanometric size, the force generated by MNPs is smaller than the force required for largely disrupting key components of cells. Here, we show the magnetic assembly process of the nanoparticles inside the cells, to form elongated aggregates with the size required to produce elevated mechanical forces. We synthesized iron oxide nanoparticles doped with zinc, to obtain high magnetization, and functionalized with the epidermal growth factor (EGF) peptide for targeting cancer cells. Under a low frequency rotating magnetic field at 15 Hz and 40 mT, the internalized EGF-MNPs formed elongated aggregates and generated hundreds of pN to dramatically damage the plasma and lysosomal membranes. The physical disruption, including leakage of lysosomal hydrolases into the cytosol, led to programmed cell death and necrosis. Our work provides a novel strategy of designing magnetic nanomedicines for mechanical destruction of cancer cells.


Assuntos
Fator de Crescimento Epidérmico/metabolismo , Compostos Férricos/metabolismo , Glioblastoma , Magnetoterapia/métodos , Nanopartículas Metálicas , Terapia de Alvo Molecular/métodos , Morte Celular , Linhagem Celular Tumoral , Humanos
14.
J Control Release ; 219: 43-60, 2015 Dec 10.
Artigo em Inglês | MEDLINE | ID: mdl-26407671

RESUMO

The paper describes the concept of magneto-mechanical actuation of single-domain magnetic nanoparticles (MNPs) in super-low and low frequency alternating magnetic fields (AMFs) and its possible use for remote control of nanomedicines and drug delivery systems. The applications of this approach for remote actuation of drug release as well as effects on biomacromolecules, biomembranes, subcellular structures and cells are discussed in comparison to conventional strategies employing magnetic hyperthermia in a radio frequency (RF) AMF. Several quantitative models describing interaction of functionalized MNPs with single macromolecules, lipid membranes, and proteins (e.g. cell membrane receptors, ion channels) are presented. The optimal characteristics of the MNPs and an AMF for effective magneto-mechanical actuation of single molecule responses in biological and bio-inspired systems are discussed. Altogether, the described studies and phenomena offer opportunities for the development of novel therapeutics both alone and in combination with magnetic hyperthermia.


Assuntos
Campos Magnéticos , Nanopartículas/administração & dosagem , Sistemas de Liberação de Medicamentos , Hipertermia Induzida , Nanomedicina
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA