Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
2.
Clin Pract Cases Emerg Med ; 1(4): 425-426, 2017 Nov.
Article in English | MEDLINE | ID: mdl-29849389
3.
Biotechnol Bioeng ; 113(10): 2228-40, 2016 10.
Article in English | MEDLINE | ID: mdl-27563853

ABSTRACT

Remote activation of specific cells of a heterogeneous population can provide a useful research tool for clinical and therapeutic applications. Here, we demonstrate that photostimulation of gold nanorods (AuNRs) using a tunable near-infrared (NIR) laser at specific longitudinal surface plasmon resonance wavelengths can induce the selective and temporal internalization of calcium in HEK 293T cells. Biotin-PEG-Au nanorods coated with streptavidin Alexa Fluor-633 and biotinylated anti-His antibodies were used to decorate cells genetically modified with His-tagged TRPV1 temperature-sensitive ion channel and AuNRs conjugated to biotinylated RGD peptide were used to decorate integrins in unmodified cells. Plasmonic activation can be stimulated at weak laser power (0.7-4.0 W/cm(2) ) without causing cell damage. Selective activation of TRPV1 channels could be controlled by laser power between 1.0 and 1.5 W/cm(2) . Integrin targeting robustly stimulated calcium signaling due to a dense cellular distribution of nanoparticles. Such an approach represents a functional tool for combinatorial activation of cell signaling in heterogeneous cell populations. Our results suggest that it is possible to induce cell activation via NIR-induced gold nanorod heating through the selective targeting of membrane proteins in unmodified cells to produce calcium signaling and downstream expression of specific genes with significant relevance for both in vitro and therapeutic applications. Biotechnol. Bioeng. 2016;113: 2228-2240. © 2016 Wiley Periodicals, Inc.


Subject(s)
Calcium Signaling/physiology , Calcium/metabolism , Gold/radiation effects , Infrared Rays , Nanotubes/radiation effects , TRPV Cation Channels/metabolism , Calcium Signaling/radiation effects , Gene Expression Regulation/physiology , Gene Expression Regulation/radiation effects , HEK293 Cells , Humans , Metal Nanoparticles/radiation effects , Radiation Dosage , Surface Plasmon Resonance/methods
4.
Nature ; 531(7596): 647-50, 2016 Mar 31.
Article in English | MEDLINE | ID: mdl-27007848

ABSTRACT

Targeted, temporally regulated neural modulation is invaluable in determining the physiological roles of specific neural populations or circuits. Here we describe a system for non-invasive, temporal activation or inhibition of neuronal activity in vivo and its use to study central nervous system control of glucose homeostasis and feeding in mice. We are able to induce neuronal activation remotely using radio waves or magnetic fields via Cre-dependent expression of a GFP-tagged ferritin fusion protein tethered to the cation-conducting transient receptor potential vanilloid 1 (TRPV1) by a camelid anti-GFP antibody (anti-GFP-TRPV1). Neuronal inhibition via the same stimuli is achieved by mutating the TRPV1 pore, rendering the channel chloride-permeable. These constructs were targeted to glucose-sensing neurons in the ventromedial hypothalamus in glucokinase-Cre mice, which express Cre in glucose-sensing neurons. Acute activation of glucose-sensing neurons in this region increases plasma glucose and glucagon, lowers insulin levels and stimulates feeding, while inhibition reduces blood glucose, raises insulin levels and suppresses feeding. These results suggest that pancreatic hormones function as an effector mechanism of central nervous system circuits controlling blood glucose and behaviour. The method we employ obviates the need for permanent implants and could potentially be applied to study other neural processes or used to regulate other, even dispersed, cell types.


Subject(s)
Blood Glucose/metabolism , Eating/physiology , Magnetic Fields , Neurons/physiology , Radio Waves , Ventromedial Hypothalamic Nucleus/cytology , Ventromedial Hypothalamic Nucleus/physiology , Animals , Ferritins/genetics , Ferritins/metabolism , Glucagon/blood , Glucokinase/metabolism , Homeostasis , Hypoglycemia/metabolism , Insulin/blood , Integrases/metabolism , Mice , Neural Inhibition , Pancreatic Hormones/metabolism , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , TRPV Cation Channels/genetics , TRPV Cation Channels/metabolism , Time Factors
6.
Nat Med ; 21(1): 92-98, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25501906

ABSTRACT

Means for temporally regulating gene expression and cellular activity are invaluable for elucidating underlying physiological processes and would have therapeutic implications. Here we report the development of a genetically encoded system for remote regulation of gene expression by low-frequency radio waves (RFs) or a magnetic field. Iron oxide nanoparticles are synthesized intracellularly as a GFP-tagged ferritin heavy and light chain fusion. The ferritin nanoparticles associate with a camelid anti-GFP-transient receptor potential vanilloid 1 fusion protein, αGFP-TRPV1, and can transduce noninvasive RF or magnetic fields into channel activation, also showing that TRPV1 can transduce a mechanical stimulus. This, in turn, initiates calcium-dependent transgene expression. In mice with stem cell or viral expression of these genetically encoded components, remote stimulation of insulin transgene expression with RF or a magnet lowers blood glucose. This robust, repeatable method for remote regulation in vivo may ultimately have applications in basic science, technology and therapeutics.


Subject(s)
Blood Glucose/radiation effects , Gene Expression Regulation/radiation effects , Glucose/metabolism , Insulin/biosynthesis , Magnetite Nanoparticles/radiation effects , Animals , Ferric Compounds/chemistry , Ferric Compounds/radiation effects , Ferritins/chemistry , Ferritins/genetics , Glucose/genetics , Green Fluorescent Proteins/chemistry , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/radiation effects , Homeostasis/radiation effects , Insulin/genetics , Insulin/radiation effects , Magnetite Nanoparticles/chemistry , Mice , Radio Waves , TRPV Cation Channels/chemistry , TRPV Cation Channels/genetics , Transgenes/radiation effects
7.
Curr Opin Biotechnol ; 28: 25-32, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24832071

ABSTRACT

Researchers over the last few years have recognized carbon nanotubes (CNTs) as promising materials for a number of biological applications. CNTs are increasingly being explored as potent drug carriers for cancer treatment, for biosensing, and as scaffolds for stem cell culture. Moreover, the integration of CNTs with proteins has led to the development of functional nanocomposites with antimicrobial properties. This review aims at understanding the critical role of CNTs in biological applications with a particular emphasis on more recent studies.


Subject(s)
Nanotubes/chemistry , Antineoplastic Agents/administration & dosage , Biosensing Techniques , Cell Culture Techniques , Cell Survival/drug effects , Drug Carriers/chemistry , Humans , Nanotubes/toxicity , Nanotubes, Carbon/chemistry , Neoplasms/drug therapy , Stem Cells/cytology , Tissue Engineering
8.
J Neurooncol ; 73(1): 29-36, 2005 May.
Article in English | MEDLINE | ID: mdl-15933813

ABSTRACT

We have recently reported that activation of tumor-specific T cells by subcutaneous vaccination with irradiated T9 glioma cells of syngeneic rats with a pre-existing, intracranial (i.c.) T9 glioma (T9+vaccination) promotes the mobilization of myeloid suppressor cells (MSC) which inhibit T cell function resulting in unregulated tumor progression. The current study investigated if this immunological paradigm could be recapitulated in T cell deficient rats, in other rat glioma models or using a dendritic cell (DC) vaccine. When nude rats were used in the T9+vaccination model, the level of MSC tumor infiltration remained low in vaccinated and control groups and there was no significant difference in tumor size between the groups. Increased tumor infiltration by MSC after vaccination with respective irradiated tumor cells was observed in the 9L, F98 and D74 gliomas. RT-2 tumors were markedly infiltrated with MSC regardless of vaccination. Enhanced tumor progression in response to immunization and T cell activation was observed in rats bearing F98 and D74 gliomas, although less pronounced than in the T9 model, and there was a trend for increased tumor size in the 9L glioma model. Increased MSC infiltrate and augmented T9 glioma growth were observed when DC pulsed with T9 cell lysate was used as a vaccine. These results suggest that MSC infiltration and unregulated tumor growth in response to vaccination is T cell-dependent; is not unique to the T9 glioma; and can be recapitulated with an alternate immunization approach.


Subject(s)
Brain Neoplasms/immunology , Glioma/immunology , Lymphocytes, Tumor-Infiltrating/immunology , Myeloid Cells/immunology , T-Lymphocyte Subsets/immunology , T-Lymphocytes, Regulatory/immunology , Animals , Antigen Presentation , Cancer Vaccines/immunology , Female , Immunosuppression Therapy , Lymphocyte Activation/immunology , Rats , Rats, Inbred F344 , Rats, Nude
SELECTION OF CITATIONS
SEARCH DETAIL
...