Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 20 de 53
Filtrar
1.
Circulation ; 149(22): 1729-1748, 2024 May 28.
Artículo en Inglés | MEDLINE | ID: mdl-38487879

RESUMEN

BACKGROUND: Myocardial infarction (MI) and heart failure are associated with an increased incidence of cancer. However, the mechanism is complex and unclear. Here, we aimed to test our hypothesis that cardiac small extracellular vesicles (sEVs), particularly cardiac mesenchymal stromal cell-derived sEVs (cMSC-sEVs), contribute to the link between post-MI left ventricular dysfunction (LVD) and cancer. METHODS: We purified and characterized sEVs from post-MI hearts and cultured cMSCs. Then, we analyzed cMSC-EV cargo and proneoplastic effects on several lines of cancer cells, macrophages, and endothelial cells. Next, we modeled heterotopic and orthotopic lung and breast cancer tumors in mice with post-MI LVD. We transferred cMSC-sEVs to assess sEV biodistribution and its effect on tumor growth. Finally, we tested the effects of sEV depletion and spironolactone treatment on cMSC-EV release and tumor growth. RESULTS: Post-MI hearts, particularly cMSCs, produced more sEVs with proneoplastic cargo than nonfailing hearts did. Proteomic analysis revealed unique protein profiles and higher quantities of tumor-promoting cytokines, proteins, and microRNAs in cMSC-sEVs from post-MI hearts. The proneoplastic effects of cMSC-sEVs varied with different types of cancer, with lung and colon cancers being more affected than melanoma and breast cancer cell lines. Post-MI cMSC-sEVs also activated resting macrophages into proangiogenic and protumorigenic states in vitro. At 28-day follow-up, mice with post-MI LVD developed larger heterotopic and orthotopic lung tumors than did sham-MI mice. Adoptive transfer of cMSC-sEVs from post-MI hearts accelerated the growth of heterotopic and orthotopic lung tumors, and biodistribution analysis revealed accumulating cMSC-sEVs in tumor cells along with accelerated tumor cell proliferation. sEV depletion reduced the tumor-promoting effects of MI, and adoptive transfer of cMSC-sEVs from post-MI hearts partially restored these effects. Finally, spironolactone treatment reduced the number of cMSC-sEVs and suppressed tumor growth during post-MI LVD. CONCLUSIONS: Cardiac sEVs, specifically cMSC-sEVs from post-MI hearts, carry multiple protumorigenic factors. Uptake of cMSC-sEVs by cancer cells accelerates tumor growth. Treatment with spironolactone significantly reduces accelerated tumor growth after MI. Our results provide new insight into the mechanism connecting post-MI LVD to cancer and propose a translational option to mitigate this deadly association.


Asunto(s)
Vesículas Extracelulares , Insuficiencia Cardíaca , Infarto del Miocardio , Animales , Vesículas Extracelulares/metabolismo , Insuficiencia Cardíaca/metabolismo , Insuficiencia Cardíaca/patología , Insuficiencia Cardíaca/etiología , Infarto del Miocardio/patología , Infarto del Miocardio/metabolismo , Ratones , Humanos , Femenino , Neoplasias Pulmonares/patología , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/tratamiento farmacológico , Línea Celular Tumoral , Células Madre Mesenquimatosas/metabolismo , Ratones Endogámicos C57BL , Modelos Animales de Enfermedad , Masculino , Proliferación Celular/efectos de los fármacos
2.
Nanomedicine ; 46: 102596, 2022 11.
Artículo en Inglés | MEDLINE | ID: mdl-36031044

RESUMEN

Genetically engineered T cells are a powerful new modality for cancer immunotherapy. However, their clinical application for solid tumors is challenging, and crucial knowledge on cell functionality in vivo is lacking. Here, we fabricated a nanoprobe composed of dendrimers incorporating a calcium sensor and gold nanoparticles, for dual-modal monitoring of engineered T cells within a solid tumor. T cells engineered to express a melanoma-specific T-cell receptor and loaded with the nanoprobe were longitudinally monitored within melanoma xenografts in mice. Fluorescent imaging of the nanoprobe's calcium sensor revealed increased intra-tumoral activation of the T cells over time, up to 24 h. Computed tomography imaging of the nanoprobe's gold nanoparticles revealed the cells' intra-tumoral distribution pattern. Quantitative analysis revealed the intra-tumoral T cell quantities. Thus, this nanoprobe reveals intra-tumoral persistence, penetration and functional status of genetically engineered T cells, which can advance T cell-based immunotherapy and promote next-generation live cell imaging.


Asunto(s)
Melanoma , Nanopartículas del Metal , Humanos , Ratones , Animales , Oro , Calcio , Linfocitos T
3.
Int J Mol Sci ; 24(1)2022 Dec 21.
Artículo en Inglés | MEDLINE | ID: mdl-36613511

RESUMEN

Gold-containing nanoparticles are proven to be an effective radiosensitizer in the radiotherapy of tumors. Reliable imaging of nanoparticles in a tumor and surrounding normal tissues is crucial both for diagnostics and for nanoparticle application as radiosensitizers. The Fe3O4 core was introduced into gold nanoparticles to form a core/shell structure suitable for MRI imaging. The aim of this study was to assess the in vivo bimodal CT and MRI enhancement ability of novel core/shell Fe3O4@Au theranostic nanoparticles. Core/shell Fe3O4@Au nanoparticles were synthesized and coated with PEG and glucose. C57Bl/6 mice bearing Ca755 mammary adenocarcinoma tumors received intravenous injections of the nanoparticles. CT and MRI were performed at several timepoints between 5 and 102 min, and on day 17 post-injection. Core/shell Fe3O4@Au nanoparticles provided significant enhancement of the tumor and tumor blood vessels. Nanoparticles also accumulated in the liver and spleen and were retained in these organs for 17 days. Mice did not show any signs of toxicity over the study duration. These results indicate that theranostic bimodal Fe3O4@Au nanoparticles are non-toxic and serve as effective contrast agents both for CT and MRI diagnostics. These nanoparticles have potential for future biomedical applications in cancer diagnostics and beyond.


Asunto(s)
Nanopartículas del Metal , Nanopartículas , Neoplasias , Animales , Ratones , Oro , Medicina de Precisión , Nanopartículas del Metal/uso terapéutico , Nanopartículas del Metal/química , Neoplasias/diagnóstico por imagen , Neoplasias/terapia , Imagen por Resonancia Magnética/métodos , Tomografía Computarizada por Rayos X , Nanomedicina Teranóstica/métodos
4.
Small ; 15(45): e1904203, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31482695

RESUMEN

Nerve growth factor (NGF) plays a vital role in reducing the loss of cholinergic neurons in Alzheimer's disease (AD). However, its delivery to the brain remains a challenge. Herein, NGF is loaded into degradable oxidized porous silicon (PSiO2 ) carriers, which are designed to carry and continuously release the protein over a 1 month period. The released NGF exhibits a substantial neuroprotective effect in differentiated rat pheochromocytoma PC12 cells against amyloid-beta (Aß)-induced cytotoxicity, which is associated with Alzheimer's disease. Next, two potential localized administration routes of the porous carriers into murine brain are investigated: implantation of PSiO2 chips above the dura mater, and biolistic bombardment of PSiO2 microparticles through an opening in the skull using a pneumatic gene gun. The PSiO2 -implanted mice are monitored for a period of 8 weeks and no inflammation or adverse effects are observed. Subsequently, a successful biolistic delivery of these highly porous microparticles into a live-mouse brain is demonstrated for the first time. The bombarded microparticles are observed to penetrate the brain and reach a depth of 150 µm. These results pave the way for using degradable PSiO2 carriers as potential localized delivery systems for NGF to the brain.


Asunto(s)
Enfermedad de Alzheimer/tratamiento farmacológico , Enfermedad de Alzheimer/metabolismo , Encéfalo/metabolismo , Nanoestructuras/química , Factor de Crecimiento Nervioso/química , Factor de Crecimiento Nervioso/uso terapéutico , Silicio/química , Animales , Supervivencia Celular/efectos de los fármacos , Ratones , Microscopía Confocal , Microscopía Fluorescente , Nanoestructuras/uso terapéutico , Factor de Crecimiento Nervioso/farmacocinética , Células PC12 , Porosidad , Ratas , Microtomografía por Rayos X
5.
Harefuah ; 157(4): 232-236, 2018 Apr.
Artículo en Hebreo | MEDLINE | ID: mdl-29688641

RESUMEN

INTRODUCTION: Nanomedicine is a rapidly evolving medical domain utilizing 1-100nm nanoscale particles to achieve medical goals in either one or more medical aspects - diagnosis, imaging and therapy. Nanomedicine employs a combination of methods stemming from life and exact sciences. This review deals briefly with the principles behind the scenes guiding the design, manufacture and employment of these nanoparticles. Some representative examples of the various applications are provided from the abundance of existing nanoparticles. The main topics discussed are those related to composition, characteristics of nanoparticles, usage in cancer, drug delivery and various central nervous system applications. Possible toxicity and future teratogenicity research pertaining to nanoparticles are also elaborated upon.


Asunto(s)
Nanomedicina , Sistemas de Liberación de Medicamentos , Predicción , Humanos , Nanopartículas , Neoplasias
6.
Nanomedicine ; 13(2): 421-429, 2017 02.
Artículo en Inglés | MEDLINE | ID: mdl-27720990

RESUMEN

Contradictory results in clinical trials are preventing the advancement and implementation of cell-based therapy. To explain such results, there is a need to uncover the mystery regarding the fate of the transplanted cells. To answer this need, we developed a technique for noninvasive in vivo cell tracking, which uses gold nanoparticles as contrast agents for CT imaging. Herein, we investigate the design principles of this technique for intramuscular transplantation of therapeutic cells. Longitudinal studies were performed, displaying the ability to track cells over long periods of time. As few as 500 cells could be detected and a way to quantify the number of cells visualized by CT was demonstrated. Moreover, monitoring of cell functionality was demonstrated on a mouse model of Duchenne muscular dystrophy. This cell-tracking technology has the potential to become an essential tool in pre-clinical as well as clinical trials and to advance the future of cell therapy.


Asunto(s)
Rastreo Celular , Nanopartículas , Tomografía Computarizada por Rayos X/métodos , Animales , Medios de Contraste , Modelos Animales de Enfermedad , Oro , Ratones , Distrofia Muscular de Duchenne
7.
J Nanobiotechnology ; 13: 19, 2015 Mar 04.
Artículo en Inglés | MEDLINE | ID: mdl-25880565

RESUMEN

BACKGROUND: During the last decade nanoparticles have gained attention as promising drug delivery agents that can transport through the blood brain barrier. Recently, several studies have demonstrated that specifically targeted nanoparticles which carry a large payload of therapeutic agents can effectively enhance therapeutic agent delivery to the brain. However, it is difficult to draw definite design principles across these studies, owing to the differences in material, size, shape and targeting agents of the nanoparticles. Therefore, the main objective of this study is to develop general design principles that link the size of the nanoparticle with the probability to cross the blood brain barrier. Specifically, we investigate the effect of the nanoparticle size on the probability of barbiturate coated GNPs to cross the blood brain barrier by using bEnd.3 brain endothelial cells as an in vitro blood brain barrier model. RESULTS: The results show that GNPs of size 70 nm are optimal for the maximum amount of gold within the brain cells, and that 20 nm GNPs are the optimal size for maximum free surface area. CONCLUSIONS: These findings can help understand the effect of particle size on the ability to cross the blood brain barrier through the endothelial cell model, and design nanoparticles for brain imaging/therapy contrast agents.


Asunto(s)
Barbitúricos/farmacocinética , Barrera Hematoencefálica/efectos de los fármacos , Nanopartículas del Metal , Tamaño de la Partícula , Animales , Barbitúricos/química , Encéfalo/citología , Células Cultivadas , Células Endoteliales/efectos de los fármacos , Nanopartículas del Metal/química , Ratones , Microscopía Electrónica de Transmisión , Espectrofotometría Ultravioleta
8.
Nano Lett ; 14(5): 2681-7, 2014 May 14.
Artículo en Inglés | MEDLINE | ID: mdl-24697682

RESUMEN

In this study we report the use of gold nanorods (GNRs) as absorption contrast agents in the diffusion reflection (DR) method for the in vivo detection of atherosclerotic injury. The early detection and characterization of atherosclerotic vascular disease is considered to be one of the greatest medical challenges today. We show that macrophage cells, which are major components of unstable active atherosclerotic plaques, uptake gold nanoparticles, resulting in a change in the optical properties of tissue-like phantoms and a unique DR profile. In vivo DR measurements of rats that underwent injury of the carotid artery showed a clear difference between the DR profiles of the injured compared with healthy arteries. The results suggest that DR measurements following GNRs administration represent a potential novel method for the early detection of atherosclerotic vascular disease.


Asunto(s)
Aterosclerosis/diagnóstico por imagen , Medios de Contraste/química , Imagen Molecular/métodos , Nanotubos/química , Animales , Aterosclerosis/fisiopatología , Medios de Contraste/administración & dosificación , Oro/administración & dosificación , Oro/química , Humanos , Radiografía , Ratas
9.
Discov Nano ; 19(1): 18, 2024 Jan 25.
Artículo en Inglés | MEDLINE | ID: mdl-38270794

RESUMEN

The near-infrared (NIR) range of the electromagnetic (EM) spectrum offers a nearly transparent window for imaging tissue. Despite the significant potential of NIR fluorescence-based imaging, its establishment in basic research and clinical applications remains limited due to the scarcity of fluorescent molecules with absorption and emission properties in the NIR region, especially those suitable for biological applications. In this study, we present a novel approach by combining the widely used IRdye 800NHS fluorophore with gold nanospheres (GNSs) and gold nanorods (GNRs) to create Au nanodyes, with improved quantum yield (QY) and distinct lifetimes. These nanodyes exhibit varying photophysical properties due to the differences in the separation distance between the dye and the gold nanoparticles (GNP). Leveraging a rapid and highly sensitive wide-field fluorescence lifetime imaging (FLI) macroscopic set up, along with phasor based analysis, we introduce multiplexing capabilities for the Au nanodyes. Our approach showcases the ability to differentiate between NIR dyes with very similar, short lifetimes within a single image, using the combination of Au nanodyes and wide-field FLI. Furthermore, we demonstrate the uptake of Au nanodyes by mineral-oil induced plasmacytomas (MOPC315.bm) cells, indicating their potential for in vitro and in vivo applications.

10.
Materials (Basel) ; 15(21)2022 Nov 04.
Artículo en Inglés | MEDLINE | ID: mdl-36363372

RESUMEN

Hyperphosphatemia is a typical complication of end-stage renal disease, characterized by elevated and life-threatening serum phosphate levels. Hemodialysis does not enable sufficient clearance of phosphate, due to slow cell-to-plasma kinetics of phosphate ions; moreover, dietary restrictions and conventional treatment with oral phosphate binders have low success rates, together with adverse effects. Here, we developed a new concept of phosphate-trapping liposomes, to improve and prolong the control over serum phosphate levels. We designed liposomes modified with polyethylene glycol and encapsulated with the phosphate binder ferric citrate (FC liposomes). These liposomes were found to trap phosphate ions in their inner core, and thereby lower free phosphate ion concentrations in solution and in serum. The FC liposomes showed higher phosphate binding ability as phosphate concentrations increased. Moreover, these liposomes showed a time-dependent increase in uptake of phosphate, up to 25 h in serum. Thus, our findings demonstrate effective long-term phosphate trapping by FC liposomes, indicating their potential to reduce serum phosphate toxicity and improve current management of hyperphosphatemia.

11.
Diagnostics (Basel) ; 12(3)2022 Feb 23.
Artículo en Inglés | MEDLINE | ID: mdl-35328130

RESUMEN

Cardiovascular disease (CVD) is a major cause of death and disability worldwide. A real need exists in the development of new, improved therapeutic methods for treating CVD, while major advances in nanotechnology have opened new avenues in this field. In this paper, we report the use of gold nanoparticles (GNPs) coated with high-density lipoprotein (HDL) (GNP-HDL) for the simultaneous detection and therapy of unstable plaques. Based on the well-known HDL cardiovascular protection, by promoting the reverse cholesterol transport (RCT), injured rat carotids, as a model for unstable plaques, were injected with the GNP-HDL. Noninvasive detection of the plaques 24 h post the GNP injection was enabled using the diffusion reflection (DR) method, indicating that the GNP-HDL particles had accumulated in the injured site. Pathology and noninvasive CT measurements proved the recovery of the injured artery treated with the GNP-HDL. The DR of the GNP-HDL presented a simple and highly sensitive method at a low cost, resulting in simultaneous specific unstable plaque diagnosis and recovery.

12.
J Nanotheranostics ; 3(4): 177-188, 2022 Sep 30.
Artículo en Inglés | MEDLINE | ID: mdl-36324626

RESUMEN

Background: Glioblastoma is the most lethal primary brain malignancy in adults. Standard of care treatment, consisting of temozolomide (TMZ) and adjuvant radiotherapy (RT), mostly does not prevent local recurrence. The inability of drugs to enter the brain, in particular antibody-based drugs and radiosensitizers, is a crucial limitation to effective glioblastoma therapy. Methods: Here, we developed a combined strategy using radiosensitizer gold nanoparticles coated with insulin to cross the blood-brain barrier and shuttle tumor-targeting antibodies (cetuximab) into the brain. Results: Following intravenous injection to an orthotopic glioblastoma mouse model, the nanoparticles specifically accumulated within the tumor. Combining targeted nanoparticle injection with TMZ and RT standard of care significantly inhibited tumor growth and extended survival, as compared to standard of care alone. Histological analysis of tumors showed that the combined treatment eradicated tumor cells, and decreased tumor vascularization, proliferation, and repair. Conclusions: Our findings demonstrate radiosensitizer nanoparticles that effectively deliver antibodies into the brain, target the tumor, and effectively improve standard of care treatment outcome in glioblastoma.

13.
Pharmaceutics ; 14(7)2022 Jun 21.
Artículo en Inglés | MEDLINE | ID: mdl-35890207

RESUMEN

Recent research points to mesenchymal stem cells' potential for treating neurological disorders, especially drug addiction. We examined the longitudinal effect of placenta-derived mesenchymal stromal-like cells (PLX-PAD) in a rat model for cocaine addiction. Sprague-Dawley male rats were trained to self-administer cocaine or saline daily until stable maintenance. Before the extinction phase, PLX-PAD cells were administered by intracerebroventricular or intranasal routes. Neurogenesis was evaluated, as was behavioral monitoring for craving. We labeled the PLX-PAD cells with gold nanoparticles and followed their longitudinal migration in the brain parallel to their infiltration of essential peripheral organs both by micro-CT and by inductively coupled plasma-optical emission spectrometry. Cell locations in the brain were confirmed by immunohistochemistry. We found that PLX-PAD cells attenuated cocaine-seeking behavior through their capacity to migrate to specific mesolimbic regions, homed on the parenchyma in the dentate gyrus of the hippocampus, and restored neurogenesis. We believe that intranasal cell therapy is a safe and effective approach to treating addiction and may offer a novel and efficient approach to rehabilitation.

14.
ACS Omega ; 6(43): 28507-28514, 2021 Nov 02.
Artículo en Inglés | MEDLINE | ID: mdl-34746546

RESUMEN

Natural killer (NK)-cell-based immunotherapy is emerging as an attractive approach for cancer treatment. However, to facilitate and expedite clinical implementation, important questions must be answered regarding the in vivo functionality and trafficking patterns of the transferred cells. We have recently developed a noninvasive cell-tracking technique, based on gold nanoparticles (GNPs) as cell-labeling and contrast agents for whole-body computed tomography (CT) imaging. Herein, we report the implementation of this technique for longitudinal and quantitative tracking of NK cell kinetics, the migration and biodistribution in tumor-bearing mice. NK cells were successfully labeled with GNPs, without impairing their biological function, as assessed both in vitro, by cytokine release and cytotoxicity assays, and in vivo, using a xenograft model of human tumors. Using CT, we longitudinally tracked the migration of intravenously injected NK cells and observed an accumulation of effector cell clusters at the tumor site, up to 72 h. Fluorescence imaging of the cells over time correlated with ex vivo quantitative analysis of gold content in the tumor, validating the accuracy and reliability of our technique. Our cell-tracking approach thus offers a valuable tool for preclinical studies, as well as for clinical applications, to elucidate the fate of NK cells and promote the implementation of NK-cell-based immunotherapy.

15.
ACS Nano ; 15(1): 1301-1309, 2021 01 26.
Artículo en Inglés | MEDLINE | ID: mdl-33356143

RESUMEN

Fluorodeoxyglucose-positron emission tomography (18F-FDG-PET) is a powerful tool for cancer detection, staging, and follow-up. However, 18F-FDG-PET imaging has high rates of false positives, as it cannot distinguish between tumor and inflammation regions that both feature increased glucose metabolic activity. In the present study, we engineered liposomes coated with glucose and the chelator dodecane tetraacetic acid (DOTA) complexed with copper, to serve as a diagnostic technology for differentiating between cancer and inflammation. This liposome technology is based on FDA-approved materials and enables complexation with metal cations and radionuclides. We found that these liposomes were preferentially uptaken by cancer cell lines with high metabolic activity, mediated via glucose transporter-1. In vivo, these liposomes were avidly uptaken by tumors, as compared to liposomes without glucose coating. Moreover, in a combined tumor-inflammation mouse model, these liposomes accumulated in the tumor tissue and not in the inflammation region. Thus, this technology shows high specificity for tumors while evading inflammation and has potential for rapid translation to the clinic and integration with existing PET imaging systems, for effective reduction of false positives in cancer diagnosis.


Asunto(s)
Liposomas , Neoplasias , Animales , Fluorodesoxiglucosa F18 , Glucosa , Ratones , Neoplasias/diagnóstico por imagen , Tomografía de Emisión de Positrones , Radiofármacos , Sensibilidad y Especificidad
16.
ACS Omega ; 6(43): 29071-29077, 2021 Nov 02.
Artículo en Inglés | MEDLINE | ID: mdl-34746596

RESUMEN

Nanoplasmonic biosensors incorporating noble metal nanocavity arrays are widely used for the detection of various biomarkers. Gold nanorods (GNRs) have unique properties that can enhance spectroscopic detection capabilities of such nanocavity-based biosensors. However, the contribution of the physical properties of multiple GNRs to resonance enhancement of gold nanocavity arrays requires further characterization and elucidation. In this work, we study how GNR aspect ratio (AR) and surface area (SA) modify the plasmonic resonance spectrum of a gold triangular nanocavity array by both simulations and experiments. The finite integration technique (FIT) simulated the extinction spectrum of the gold nanocavity array with 300 nm periodicity onto which the GNRs of different ARs and SAs are placed. Simulations showed that matching of the GNRs longitudinal peak, which is affected by AR, to the nanocavity array's spectrum minima can optimize signal suppression and shifting. Moreover, increasing SA of the matched GNRs increased the spectral variations of the array. Experiments confirmed that GNRs conjugated to a gold triangular nanocavity array of 300 nm periodicity caused spectrum suppression and redshift. Our findings demonstrate that tailoring of the GNR AR and SA parameters to nanoplasmonic arrays has the potential to greatly improve spectral variations for enhanced plasmonic biosensing.

17.
Biomater Sci ; 9(6): 2103-2114, 2021 Mar 21.
Artículo en Inglés | MEDLINE | ID: mdl-33475633

RESUMEN

Exosomes are promising vectors for anti-tumor therapy, due to their biocompatibility, low immunogenicity, and innate ability to interact with target cells. However, promoting clinical application of exosome-based therapeutics requires elucidation of key issues, including exosome biodistribution, tumor targeting and accumulation, and the ability to overcome tumor barriers that limit the penetration of various nano-carriers and drugs. Here, we examined these parameters in exosomes derived from mesenchymal stem cells (MSC-exo) and from the A431 squamous cell carcinoma line (A431-exo), which both have potential use in cancer therapy. Using our novel technique combining gold nanoparticle (GNP) labeling of exosomes and non-invasive computed tomography imaging (CT), we longitudinally and quantitatively tracked the two intravenously-injected exosome types in A431 tumor-bearing mice. CT imaging up to 48 h and subsequent ex vivo analysis revealed tumor homing abilities of both exosome types, yet there was significantly higher tumor accumulation of MSC-exo as compared to A431-exo. Moreover, MSC-exo demonstrated the ability to penetrate the tumor and distribute throughout its bulk, while non-encapsulated GNPs remained concentrated at the tumor periphery. Histological analysis showed penetration of MSC-exo not only into the tumor tissue, but also into tumor cell cytoplasm. While the proportion of biodistribution between organs at 48 h was similar for both exosome types, more rapid clearance was indicated for A431-exo. Thus, our findings demonstrate an effect of exosome type on tumor targeting abilities and biodistribution, and suggest that MSC-exo may have superior abilities for tumor-targeted therapy.


Asunto(s)
Exosomas , Neoplasias de Cabeza y Cuello , Nanopartículas del Metal , Animales , Exosomas/metabolismo , Oro/metabolismo , Neoplasias de Cabeza y Cuello/metabolismo , Ratones , Distribución Tisular
18.
Nanomedicine (Lond) ; 14(14): 1857-1871, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-31339056

RESUMEN

Aim: Longitudinal tracking of transplanted cells in clinical and experimental setups is crucial for evaluating the efficiency of retinal cell replacement therapies. Materials & methods: Gold nanoparticle-labeled photoreceptor precursors were transplanted in the vitreous and subretinal space of rats and were longitudinally tracked for over a month using optical coherence tomography, computed tomography and fluorescence fundus imaging. Results: This multimodal imaging approach enabled high-resolution long-term tracking and estimation of cell survival in the retina and vitreous, while displaying no toxic effects on the cells or the retina. Conclusion: These observations highlight the applicability of using gold nanoparticle for retinal cell tracking in existing experimental settings and its translational potential for providing more efficient retinal cell therapy in humans.


Asunto(s)
Oro/análisis , Nanopartículas del Metal/análisis , Células Fotorreceptoras de Vertebrados/trasplante , Retina/citología , Animales , Línea Celular , Supervivencia Celular , Rastreo Celular , Humanos , Imagen Óptica , Células Fotorreceptoras de Vertebrados/citología , Ratas , Ratas Long-Evans , Retina/diagnóstico por imagen , Tomografía de Coherencia Óptica , Tomografía Computarizada por Rayos X
19.
ACS Appl Mater Interfaces ; 11(49): 45368-45380, 2019 Dec 11.
Artículo en Inglés | MEDLINE | ID: mdl-31755692

RESUMEN

Photodynamic therapy (PDT) is a promising recognized treatment for cancer. To date, PDT drugs are injected systemically, and the tumor area is irradiated to induce cell death. Current clinical protocols have several drawbacks, including limited accessibility to solid tumors and insufficient selectivity of drugs. Herein, we propose an alternative approach to improve PDT effectiveness by magnetic targeting of responsive carriers conjugated to the PDT drug. We coordinatively attached a meso-tetrahydroxyphenylchlorin (mTHPC) photosensitizer to Ce-doped-γ-Fe2O3 maghemite nanoparticles (MNPs). These MNPs are superparamagnetic and biocompatible, and the resulting mTHPC-MNPs nanocomposites are stable in aqueous suspensions. MDA-MB231 (human breast cancer) cells incubated with the mTHPC-MNPs showed high uptake and high death rates in cell population after PDT. The exposure to external magnetic forces during the incubation period directed the nanocomposites to selected sites enhancing drug accumulation that was double that of cells with no magnetic exposure. Next, breast cancer tumors were induced subcutaneously in mice and treated magnetically. In vivo results showed accelerated drug accumulation in tumors of mice injected with mTHPC-MNP nanocomposites, compared to the free drug. PDT irradiation led to a decrease in tumor size of both groups, whereas treatment with the focused magnetic nanocomposites led to significant tumor regression. Our results demonstrate a method to improve the current PDT treatments by applying magnetic forces to effectively direct the drug to cancerous tissue. This approach leads to a highly localized and effective PDT process, opening new directions for clinical PDT protocols.


Asunto(s)
Nanopartículas de Magnetita/química , Mesoporfirinas/farmacología , Fotoquimioterapia , Fármacos Fotosensibilizantes/farmacología , Animales , Materiales Biocompatibles/química , Materiales Biocompatibles/farmacología , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Cerio/química , Compuestos Férricos/química , Compuestos Férricos/farmacología , Humanos , Magnetismo , Nanopartículas de Magnetita/uso terapéutico , Mesoporfirinas/química , Ratones , Neoplasias/tratamiento farmacológico , Fármacos Fotosensibilizantes/química , Ensayos Antitumor por Modelo de Xenoinjerto
20.
J Biophotonics ; 12(1): e201800218, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30141260

RESUMEN

Atherosclerosis (AS), the leading cause of morbidity and mortality in cardiovascular disease, needs an early detection for treatment and prevention of fatal events. Here, for the first time, we applied gold nanorods (GNRs)-assisted diffusion reflection (DR), a noninvasive technique for in vivo detection of AS in a high-fat-diet-induced c57bl mouse model, which resembles the manifestation of AS in humans. DR simply detects the change in light reflection profile of tissue due to the accumulation of GNRs in the AS plaques and enables clear detection of AS lesions in carotid and femoral arteries of these hyperlipidemic mice. After 24 hours post-GNRs injection, DR showed the highest efficiency of AS detection. Moreover, the sensitivity of the DR method is much higher than computed tomography (CT) and is comparable to ex vivo high-resolution CT. Our results strongly suggest that the DR method can detect early atherosclerotic lesions in a sensitive and specific manner.


Asunto(s)
Aterosclerosis/diagnóstico , Oro/química , Hiperlipidemias/diagnóstico , Nanomedicina/métodos , Nanotubos/química , Animales , Difusión , Modelos Animales de Enfermedad , Hiperlipidemias/diagnóstico por imagen , Hiperlipidemias/patología , Ratones , Ratones Endogámicos C57BL , Sensibilidad y Especificidad , Tomografía Computarizada por Rayos X
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda