Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
Add more filters










Publication year range
1.
J Nanotheranostics ; 3(4): 177-188, 2022 Sep 30.
Article in English | MEDLINE | ID: mdl-36324626

ABSTRACT

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.

2.
Nanomedicine ; 46: 102596, 2022 11.
Article in English | MEDLINE | ID: mdl-36031044

ABSTRACT

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.


Subject(s)
Melanoma , Metal Nanoparticles , Humans , Mice , Animals , Gold , Calcium , T-Lymphocytes
3.
Pharmaceutics ; 14(7)2022 Jun 21.
Article in English | MEDLINE | ID: mdl-35890207

ABSTRACT

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.

4.
Mater Horiz ; 8(4): 1314-1322, 2021 04 01.
Article in English | MEDLINE | ID: mdl-34821924

ABSTRACT

Acute kidney injury (AKI) is frequently associated with reactive oxygen species (ROS) and causes high mortality in clinics annually, and nanotechnology-mediated antioxidative therapy is emerging as a novel strategy for AKI treatment. Herein, four kinds of natural antioxidants are able to coordinate with iron (Fe) ions to form ultra-small coordination polymer nanodots (CPNs) with good water dispersibility and strong ROS scavenging ability. In particular, Fe-curcumin CPNs (Fe-Cur CPNs) are applied for cellular ROS scavenging and rhabdomyolysis-induced AKI relief.


Subject(s)
Acute Kidney Injury , Biological Products , Acute Kidney Injury/chemically induced , Antioxidants , Humans , Polymers , Reactive Oxygen Species
5.
Biomater Sci ; 9(6): 2103-2114, 2021 Mar 21.
Article in English | MEDLINE | ID: mdl-33475633

ABSTRACT

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.


Subject(s)
Exosomes , Head and Neck Neoplasms , Metal Nanoparticles , Animals , Exosomes/metabolism , Gold/metabolism , Head and Neck Neoplasms/metabolism , Mice , Tissue Distribution
6.
ACS Nano ; 15(1): 1301-1309, 2021 01 26.
Article in English | MEDLINE | ID: mdl-33356143

ABSTRACT

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.


Subject(s)
Liposomes , Neoplasms , Animals , Fluorodeoxyglucose F18 , Glucose , Mice , Neoplasms/diagnostic imaging , Positron-Emission Tomography , Radiopharmaceuticals , Sensitivity and Specificity
7.
Article in English | MEDLINE | ID: mdl-32441050

ABSTRACT

X-ray imaging is the most widely used diagnostic imaging method in modern medicine and several advanced forms of this technology have recently emerged. Iodinated molecules and barium sulfate suspensions are clinically approved X-ray contrast agents and are widely used. However, these existing contrast agents provide limited information, are suboptimal for new X-ray imaging techniques and are developing safety concerns. Thus, over the past 15 years, there has been a rapid growth in the development of nanoparticles as X-ray contrast agents. Nanoparticles have several desirable features such as high contrast payloads, the potential for long circulation times, and tunable physicochemical properties. Nanoparticles have also been used in a range of biomedical applications such as disease treatment, targeted imaging, and cell tracking. In this review, we discuss the principles behind X-ray contrast generation and introduce new types of X-ray imaging modalities, as well as potential elements and chemical compositions that are suitable for novel contrast agent development. We focus on the progress in nanoparticle X-ray contrast agents developed to be renally clearable, long circulating, theranostic, targeted, or for cell tracking. We feature agents that are used in conjunction with the newly developed multi-energy computed tomography and mammographic imaging technologies. Finally, we offer perspectives on current limitations and emerging research topics as well as expectations for the future development of the field. This article is categorized under: Diagnostic Tools > in vivo Nanodiagnostics and Imaging Nanotechnology Approaches to Biology > Nanoscale Systems in Biology.


Subject(s)
Contrast Media , Diagnostic Imaging , Nanoparticles , Nanotechnology , Tomography, X-Ray Computed , X-Rays
8.
Biomacromolecules ; 21(4): 1587-1595, 2020 04 13.
Article in English | MEDLINE | ID: mdl-32154709

ABSTRACT

Nanosystems for monitoring and tracking T cells provide an important basis for evaluating the functionality and efficacy of T cell-based immunotherapy. To this end, we designed herein an efficient nanoprobe for T cell monitoring and tracking using poly(amidoamine) (PAMAM) dendrimer-entrapped gold nanoparticles (Au DENPs) conjugated with Fluo-4 for dual-mode computed tomography (CT) and fluorescence imaging. In this study, PAMAM dendrimers of generation 5 (G5) were modified with hydroxyl-terminated polyethylene glycol (PEG) and then used to entrap 2.0 nm Au NPs followed by acetylation of the excess amine groups on the dendrimer surface. Subsequently, the calcium ion probe was covalently attached to the dendrimer nanohybrids through the PEG hydroxyl end groups to gain the functional {(Au0)25-G5.NHAc-(PEG)14-(Fluo-4)2} nanoprobe. This nanoprobe had excellent water solubility, high X-ray attenuation coefficient, and good cytocompatibility in the given concentration range, as well as a high T cell labeling efficiency. Confocal microscopy and flow cytometry results demonstrated that the nanoprobe was able to fluorescently sense activated T cells. Moreover, the nanoprobe was able to realize both CT and fluorescence imaging of subcutaneously injected T cells in vivo. Thus, the developed novel dendrimer-based nanosystem may hold great promise for advancing and improving the clinical application of T cell-based immunotherapy.


Subject(s)
Dendrimers , Metal Nanoparticles , Cell Line, Tumor , Gold , Optical Imaging , T-Lymphocytes , Tomography, X-Ray Computed
9.
Bio Protoc ; 10(11): e3635, 2020 Jun 05.
Article in English | MEDLINE | ID: mdl-33659306

ABSTRACT

Exosomes, a subtype of extracellular vesicles, are nanovesicles of endocytic origin. Exosomes contain a plethora of proteins, lipids, and genetic materials of parent cells to facilitate intercellular communications. Tracking exosomes in vivo is fundamentally important to understand their biodistribution pattern and the mechanism of biological actions in experimental models. Until now, a number of tracking protocols have been developed, including fluorescence labeling, bioluminescence imaging, magnetic resonance imaging, and computed tomography (CT) tracking of exosomes. Recently, we have shown the tracking and quantification of exosomes in a spinal cord injury model, by using two tracking approaches. More specifically, following intranasal administration of gold nanoparticle-encapsulated exosomes to rats bearing complete spinal cord injury, exosomes in the whole central nervous system were tracked by using microCT, and quantified by using inductively coupled plasma and flame atomic absorption spectroscopy. In addition, optical imaging of fluorescently labeled exosomes was performed to understand the abundance of migrating exosomes in the spinal cord lesion, as compared to the healthy controls, and to further examine their affinity to different cell types in the lesion. Thus, the protocol presented here aids in the study of exosome biodistribution at both cellular and organ levels, in the context of spinal cord injury. This protocol will also enable researchers to better elucidate the fate of administered exosomes in other models of interest.

10.
Article in English | MEDLINE | ID: mdl-31840427

ABSTRACT

Exosomes have many biological functions as short- and long distance nanocarriers for cell-to-cell communication. They allow the exchange of complex information between cells, and thereby modulate various processes such as homeostasis, immune response and angiogenesis, in both physiological and pathological conditions. In addition, due to their unique abilities of migration, targeting, and selective internalization into specific cells, they are promising delivery vectors. As such, they provide a potentially new field in diagnostics and treatment, and may serve as an alternative to cell-based therapeutic approaches. However, a major drawback for translating exosome treatment to the clinic is that current understanding of these endogenous vesicles is insufficient, especially in regards to their in vivo behavior. Tracking exosomes in vivo can provide important knowledge regarding their biodistribution, migration abilities, toxicity, biological role, communication capabilities, and mechanism of action. Therefore, the development of efficient, sensitive and biocompatible exosome labeling and imaging techniques is highly desired. Recent studies have developed different methods for exosome labeling and imaging, which have allowed for in vivo investigation of their bio-distribution, physiological functions, migration, and targeting mechanisms. These improved imaging capabilities are expected to greatly advance exosome-based nanomedicine applications. This article is categorized under: Therapeutic Approaches and Drug Discovery > Emerging Technologies Diagnostic Tools > In Vivo Nanodiagnostics and Imaging Nanotechnology Approaches to Biology > Nanoscale Systems in Biology.


Subject(s)
Diagnostic Imaging , Drug Delivery Systems , Exosomes , Nanomedicine , Animals , Humans , Mice , Tissue Distribution
11.
ACS Nano ; 13(9): 10015-10028, 2019 09 24.
Article in English | MEDLINE | ID: mdl-31454225

ABSTRACT

Individuals with spinal cord injury (SCI) usually suffer from permanent neurological deficits, while spontaneous recovery and therapeutic efficacy are limited. Here, we demonstrate that when given intranasally, exosomes derived from mesenchymal stem cells (MSC-Exo) could pass the blood brain barrier and migrate to the injured spinal cord area. Furthermore, MSC-Exo loaded with phosphatase and tensin homolog small interfering RNA (ExoPTEN) could attenuate the expression of PTEN in the injured spinal cord region following intranasal administrations. In addition, the loaded MSC-Exo considerably enhanced axonal growth and neovascularization, while reducing microgliosis and astrogliosis. The intranasal ExoPTEN therapy could also partly improve structural and electrophysiological function and, most importantly, significantly elicited functional recovery in rats with complete SCI. The results imply that intranasal ExoPTEN may be used clinically to promote recovery for SCI individuals.


Subject(s)
Exosomes/transplantation , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells/metabolism , PTEN Phosphohydrolase/metabolism , RNA, Small Interfering/metabolism , Recovery of Function , Spinal Cord Injuries/physiopathology , Spinal Cord Injuries/therapy , Administration, Intranasal , Animals , Axons/pathology , Blood-Brain Barrier/pathology , Chemotaxis , Electrophysiological Phenomena , Exosomes/ultrastructure , Female , Ganglia, Spinal/pathology , Gold/chemistry , Humans , Magnetic Resonance Imaging , Motor Activity , Nanoparticles/chemistry , Nanoparticles/ultrastructure , Neurons/pathology , Rats, Sprague-Dawley , Spinal Cord/pathology
12.
Nanomedicine (Lond) ; 14(14): 1857-1871, 2019 07.
Article in English | MEDLINE | ID: mdl-31339056

ABSTRACT

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.


Subject(s)
Gold/analysis , Metal Nanoparticles/analysis , Photoreceptor Cells, Vertebrate/transplantation , Retina/cytology , Animals , Cell Line , Cell Survival , Cell Tracking , Humans , Optical Imaging , Photoreceptor Cells, Vertebrate/cytology , Rats , Rats, Long-Evans , Retina/diagnostic imaging , Tomography, Optical Coherence , Tomography, X-Ray Computed
13.
Adv Mater ; 31(23): e1900730, 2019 Jun.
Article in English | MEDLINE | ID: mdl-30977247

ABSTRACT

Sonodynamic therapy (SDT) triggered by ultrasound (US) has attracted increasing attention owing to its abilities to overcome critical limitations including low tissue-penetration depth and phototoxicity in photodynamic therapy. Herein, the design of a new type of sonosensitizer is revealed, namely, ultrasmall oxygen-deficient bimetallic oxide MnWOX nanoparticles, for multimodal imaging-guided enhanced SDT against cancer. As-made MnWOX nanoparticles with poly(ethylene glycol) (PEG) modification show high physiological stability and biocompatibility. Interestingly, such MnWOX -PEG nanoparticles exhibit highly efficient US-triggered production of 1 O2 and •OH, higher than that of previously reported sonosensitizers (e.g., protoporphyrin IX and titanium dioxide), because the oxygen-deficient structure of MnWOX serves as an electron trap site to prevent electron-hole recombination. The glutathione depletion capability of MnWOX -PEG can also further favor SDT-triggered cancer cell killing. With efficient tumor homing as illustrated by computer tomography and magnetic resonance imaging, MnWOX -PEG enables effective destruction of mouse tumors under US stimulation. After accomplishing its therapeutic functions, MnWOX -PEG can be metabolized by the mouse body without any long-term toxicity. Herein, a new type of sono-sensitizing agent with high SDT efficacy, multimodal imaging functions, and rapid clearance is presented, an agent which is promising for noninvasive SDT cancer treatment.


Subject(s)
Glutathione/metabolism , Manganese Compounds/chemistry , Metal Nanoparticles/chemistry , Oxides/chemistry , Oxygen/chemistry , Tungsten/chemistry , Ultrasonic Therapy/methods , Ultrasonography/methods , Animals , Apoptosis , Carbocyanines/chemistry , Cell Line, Tumor , Cell Survival , Coloring Agents/chemistry , Humans , Metal Nanoparticles/therapeutic use , Mice , Neoplasm Transplantation , Polyethylene Glycols/chemistry
14.
Nano Lett ; 19(6): 3422-3431, 2019 06 12.
Article in English | MEDLINE | ID: mdl-30761901

ABSTRACT

Exosomes, nanovesicles that are secreted by different cell types, enable intercellular communication at local or distant sites. Alhough they have been found to cross the blood brain barrier, their migration and homing abilities within the brain remain unstudied. We have recently developed a method for longitudinal and quantitative in vivo neuroimaging of exosomes based on the superior visualization abilities of classical X-ray computed tomography (CT), combined with gold nanoparticles as labeling agents. Here, we used this technique to track the migration and homing patterns of intranasally administrated exosomes derived from bone marrow mesenchymal stem cells (MSC-exo) in different brain pathologies, including stroke, autism, Parkinson's disease, and Alzheimer's disease. We found that MSC-exo specifically targeted and accumulated in pathologically relevant murine models brains regions up to 96 h post administration, while in healthy controls they showed a diffuse migration pattern and clearance by 24 h. The neuro-inflammatory signal in pathological brains was highly correlated with MSC-exo accumulation, suggesting that the homing mechanism is inflammatory-driven. In addition, MSC-exo were selectively uptaken by neuronal cells, but not glial cells, in the pathological regions. Taken together, these findings can significantly promote the application of exosomes for therapy and targeted drug delivery in various brain pathologies.


Subject(s)
Brain/diagnostic imaging , Exosomes , Neurodegenerative Diseases/diagnostic imaging , Neurodevelopmental Disorders/diagnostic imaging , Alzheimer Disease/diagnostic imaging , Animals , Disease Models, Animal , Exosomes/chemistry , Gold/analysis , Mesenchymal Stem Cells/chemistry , Metal Nanoparticles/analysis , Neuroimaging/methods , Tomography, X-Ray Computed/methods
16.
Theranostics ; 7(16): 3962-3971, 2017.
Article in English | MEDLINE | ID: mdl-29109791

ABSTRACT

Personalized molecular profiling has an established role in selection of treatment for metastatic disease; however, its role in improving radiosensitivity and functional imaging has not been evaluated. In the current study, we examined molecular profiling as a tool for designing personalized targeted gold nanoparticles (GNP) to serve as dual-modal tumor radiosensitizers and functional imaging enhancers. To this end, molecular profiling of a patient's salivary gland adenoid cystic carcinoma (ACC) was performed, and anaplastic lymphoma kinase (ALK) mutation was detected. The extracted tumor was subcutaneously injected into mice, which were then treated either with radiation, the specific ALK inhibitor crizotinib, or a combination of therapies. One of these combinations, namely, ALK-targeted GNP (via crizotinib coating), was found to enhance radiation treatment, as demonstrated by a significant decrease in tumor volume over 24 days. In parallel, ALK-targeted GNP substantially augmented tumor visualization via computed tomography. The mechanism of radiosensitivity enhancement was mostly related to a diminished cell repair mechanism in tumors, as demonstrated by proliferating cell nuclear antigen staining. These findings indicate that personalized molecular profiling is an effective technique for enhancing cancer theranostics.


Subject(s)
Carcinoma, Adenoid Cystic/diagnostic imaging , Gold/chemistry , Metal Nanoparticles/chemistry , Anaplastic Lymphoma Kinase , Carcinoma, Adenoid Cystic/drug therapy , Carcinoma, Adenoid Cystic/metabolism , Crizotinib , Humans , Mutation/genetics , Pyrazoles/therapeutic use , Pyridines/therapeutic use , Receptor Protein-Tyrosine Kinases/metabolism
17.
ACS Nano ; 11(11): 10883-10893, 2017 11 28.
Article in English | MEDLINE | ID: mdl-28960957

ABSTRACT

Exosomes are emerging as effective therapeutic tools for various pathologies. These extracellular vesicles can bypass biological barriers, including the blood-brain barrier, and can serve as powerful drug and gene therapy transporters. However, the progress of therapy development is impeded by several challenges, including insufficient data on exosome trafficking and biodistribution and the difficulty to image deep brain structures in vivo. Herein, we established a method for noninvasive in vivo neuroimaging and tracking of exosomes, based on glucose-coated gold nanoparticle (GNP) labeling and computed tomography imaging. Labeling of exosomes with the GNPs was achieved directly, as opposed to the typical and less efficient indirect labeling mode through parent cells. On the mechanistic level, we found that the glucose-coated GNPs were uptaken into MSC-derived exosomes via an active, energy-dependent mechanism that is mediated by the glucose transporter GLUT-1 and involves endocytic proteins. Next, we determined optimal parameters of size and administration route; we demonstrated that 5 nm GNPs enabled improved exosome labeling and that intranasal, compared to intravenous, administration led to superior brain accumulation and thus enhanced in vivo neuroimaging. Furthermore, using a mouse model of focal brain ischemia, we noninvasively tracked intranasally administered GNP-labeled exosomes, which showed increased accumulation at the lesion site over 24 h, as compared to nonspecific migration and clearance from control brains over the same period. Thus, this exosome labeling technique can serve as a powerful diagnostic tool for various brain disorders and could potentially enhance exosome-based treatments for neuronal recovery.


Subject(s)
Brain/ultrastructure , Exosomes/ultrastructure , Metal Nanoparticles/administration & dosage , Neuroimaging/methods , Animals , Blood-Brain Barrier/drug effects , Blood-Brain Barrier/ultrastructure , Brain/drug effects , Exosomes/chemistry , Gold/administration & dosage , Gold/chemistry , Humans , Metal Nanoparticles/chemistry , Mice , Staining and Labeling , Tissue Distribution
18.
Theranostics ; 7(10): 2690-2703, 2017.
Article in English | MEDLINE | ID: mdl-28819456

ABSTRACT

Recent studies have proposed that abnormal glutamatergic neurotransmission and glial pathology play an important role in the etiology and manifestation of depression. It was postulated that restoration of normal glutamatergic transmission, by enhancing glutamate uptake, may have a beneficial effect on depression. We examined this hypothesis using unique human glial-like mesenchymal stem cells (MSCs), which in addition to inherent properties of migration to regions of injury and secretion of neurotrophic factors, were differentiated to express high levels of functional glutamate transporters (excitatory amino acid transporters; EAAT). Additionally, gold nanoparticles (GNPs), which serve as contrast agents for CT imaging, were loaded into the cells for non-invasive, real-time imaging and tracking of MSC migration and final location within the brain. MSC-EAAT (2×105; 10 µl) were administered (i.c.v.) to Flinder Sensitive Line rats (FSLs), a genetic model for depression, and longitudinal behavioral and molecular changes were monitored. FSL rats treated with MSC-EAAT showed attenuated depressive-like behaviors (measured by the forced swim test, novelty exploration test and sucrose self-administration paradigm), as compared to controls. CT imaging, Flame Atomic Absorption Spectroscopy analysis and immunohistochemistry showed that the majority of MSCs homed specifically to the dentate gyrus of the hippocampus, a region showing structural brain changes in depression, including loss of glial cells. mRNA and protein levels of EAAT1 and BDNF were significantly elevated in the hippocampus of MSC-EAAT-treated FSLs. Our findings indicate that MSC-EAATs effectively improve depressive-like manifestations, possibly in part by increasing both glutamate uptake and neurotropic factor secretion in the hippocampus.


Subject(s)
Amino Acid Transport System X-AG/biosynthesis , Depression/therapy , Gene Expression , Mesenchymal Stem Cell Transplantation/methods , Mesenchymal Stem Cells/physiology , Animals , Behavior, Animal , Dentate Gyrus/pathology , Depression/pathology , Disease Models, Animal , Humans , Longitudinal Studies , Rats , Therapeutic Uses
19.
Nanomedicine (Lond) ; 12(13): 1533-1546, 2017 Jul.
Article in English | MEDLINE | ID: mdl-28621578

ABSTRACT

AIM: Our goal was to develop an efficient nanoparticle-based system that can overcome the restrictive mechanism of the blood-brain barrier (BBB) by targeting insulin receptors and would thus enable drug delivery to the brain. METHODS: Insulin-coated gold nanoparticles (INS-GNPs) were synthesized to serve as a BBB transport system. The effect of nanoparticle size (20, 50 and 70 nm) on their ability to cross the BBB was quantitatively investigated in Balb/C mice. RESULTS: The most widespread biodistribution and highest accumulation within the brain were observed using 20 nm INS-GNPs, 2 h post injection. In vivo CT imaging revealed that particles migrated to specific brain regions, which are involved in neurodegenerative and neuropsychiatric disorders. CONCLUSION: These findings promote the optimization of nanovehicles for transport of drugs through the BBB. The insulin coating of the particles enabled targeting of specific brain regions, suggesting the potential use of INS-GNPs for delivery of various treatments for brain-related disorders.


Subject(s)
Blood-Brain Barrier/metabolism , Gold , Insulin/chemistry , Metal Nanoparticles/chemistry , Animals , Biological Transport , Contrast Media , Drug Delivery Systems , Humans , Iopamidol , Mice , Mice, Inbred BALB C , Molecular Targeted Therapy , Optical Imaging , Particle Size , Surface Properties , Tissue Distribution , Tomography Scanners, X-Ray Computed
20.
Nanomedicine ; 13(2): 421-429, 2017 02.
Article in English | MEDLINE | ID: mdl-27720990

ABSTRACT

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.


Subject(s)
Cell Tracking , Nanoparticles , Tomography, X-Ray Computed/methods , Animals , Contrast Media , Disease Models, Animal , Gold , Mice , Muscular Dystrophy, Duchenne
SELECTION OF CITATIONS
SEARCH DETAIL
...