Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
J Biotechnol ; 338: 71-80, 2021 Sep 10.
Article in English | MEDLINE | ID: mdl-34271056

ABSTRACT

The advent of the CRISPR/Cas9 system has transformed the field of human genome engineering and has created new perspectives in the development of innovative cell therapies. However, the absence of a simple, fast and efficient delivery method of CRISPR/Cas9 into primary human cells has been limiting the progress of CRISPR/Cas9-based therapies. Here, we describe an optimized protocol for iTOP-mediated delivery of CRISPR/Cas9 in various human cells, including primary T cells, induced pluripotent stem cells (hiPSCs), Jurkat, ARPE-19 and HEK293 cells. We compare iTOP to other CRISPR/Cas9 delivery methods, such as electroporation and lipofection, and evaluate the corresponding gene-editing efficiencies and post-treatment cell viabilities. We demonstrate that the gene editing achieved by iTOP-mediated delivery of CRISPR/Cas9 is 40-95 % depending on the cell type, while post-iTOP cell viability remains high in the range of 70-95 %. Collectively, we present an optimized workflow for a simple, high-throughput and effective iTOP-mediated delivery of CRISPR/Cas9 to engineer difficult-to-transduce human cells. We believe that the iTOP technology® could contribute to the development of novel CRISPR/Cas9-based cell therapies.


Subject(s)
CRISPR-Cas Systems , T-Lymphocytes , CRISPR-Cas Systems/genetics , Gene Editing , Genome, Human , HEK293 Cells , Humans
2.
Drug Discov Today Technol ; 20: 59-69, 2016 Jun.
Article in English | MEDLINE | ID: mdl-27986226

ABSTRACT

The blood-brain barrier (BBB) represents a major obstacle for the delivery and development of drugs curing brain pathologies. However, this biological barrier presents numerous endogenous specialized transport systems that can be exploited by engineered nanoparticles to enable drug delivery to the brain. In particular, conjugation of glutathione (GSH) onto PEGylated liposomes (G-Technology®) showed to safely enhance delivery of encapsulated drugs to the brain. Yet, understanding of the mechanism of action remains limited and full mechanistic understanding will aid in the further optimization of the technology. In order to elucidate the mechanism of brain targeting by GSH-PEG liposomes, we here demonstrate that the in vivo delivery of liposomal ribavirin is increased in brain extracellular fluid according to the extent of GSH conjugation onto the liposomes. In vitro, using the hCMEC/D3 human cerebral microvascular endothelial (CMEC) cell line, as well as primary bovine and porcine CMEC (and in contrast to non-brain derived endothelial and epithelial cells), we show that liposomal uptake occurs through the process of endocytosis and that the brain-specific uptake is also glutathione conjugation-dependent. Interestingly, the uptake mechanism is an active process that is temperature-, time- and dose-dependent. Finally, early endocytosis events rely on cytoskeleton remodeling, as well as dynamin- and clathrin-dependent endocytosis pathways. Overall, our data demonstrate that the glutathione-dependent uptake mechanism of the G-Technology involves a specific endocytosis pathway indicative of a receptor-mediated mechanism, and supports the benefit of this drug delivery technology for the treatment of devastating brain diseases.


Subject(s)
Antiviral Agents/administration & dosage , Brain/metabolism , Glutathione/administration & dosage , Polyethylene Glycols/administration & dosage , Ribavirin/administration & dosage , Animals , Antiviral Agents/pharmacokinetics , Biological Transport , Cattle , Cell Line , Cells, Cultured , Endothelial Cells/metabolism , Glutathione/chemistry , Glutathione/pharmacokinetics , HEK293 Cells , Humans , Liposomes , Male , Polyethylene Glycols/chemistry , Polyethylene Glycols/pharmacokinetics , Rats, Wistar , Ribavirin/pharmacokinetics , Swine
3.
Acta Neuropathol Commun ; 2: 66, 2014 Jun 13.
Article in English | MEDLINE | ID: mdl-24923195

ABSTRACT

BACKGROUND: Hallmarks of CNS inflammation, including microglial and astrocyte activation, are prominent features in post-mortem tissue from amyotrophic lateral sclerosis (ALS) patients and in mice overexpressing mutant superoxide dismutase-1 (SOD1G93A). Administration of non-targeted glucocorticoids does not significantly alter disease progression, but this may reflect poor CNS delivery. Here, we sought to discover whether CNS-targeted, liposomal encapsulated glucocorticoid would inhibit the CNS inflammatory response and reduce motor neuron loss. SOD1G93A mice were treated with saline, free methylprednisolone (MP, 10 mg/kg/week) or glutathione PEGylated liposomal MP (2B3-201, 10 mg/kg/week) and compared to saline treated wild-type animals. Animals were treated weekly with intravenous injections for 9 weeks from 60 days of age. Weights and motor performance were monitored during this period. At the end of the experimental period (116 days) mice were imaged using T2-weighted MRI for brainstem pathology; brain and spinal cord tissue were then collected for histological analysis. RESULTS: All SOD1G93A groups showed a significant decrease in motor performance, compared to baseline, from ~100 days. SOD1G93A animals showed a significant increase in signal intensity on T2 weighted MR images, which may reflect the combination of neuronal vacuolation and glial activation in these motor nuclei. Treatment with 2B3-201, but not free MP, significantly reduced T2 hyperintensity observed in SOD1G93A mice. Compared to saline-treated and free-MP-treated SOD1G93A mice, those animals given 2B3-201 displayed significantly improved histopathological outcomes in brainstem motor nuclei, which included reduced gliosis and neuronal loss. CONCLUSIONS: In contrast to previous reports that employed free steroid preparations, CNS-targeted anti-inflammatory agent 2B3-201 (liposomal methylprednisolone) has therapeutic potential, reducing brainstem pathology in the SOD1G93A mouse model of ALS. 2B3-201 reduced neuronal loss and vacuolation in brainstem nuclei, and reduced activation preferentially in astrocytes compared with microglia. These data also suggest that other previously ineffective therapies could be of therapeutic value if delivered specifically to the CNS.


Subject(s)
Amyotrophic Lateral Sclerosis/drug therapy , Central Nervous System/drug effects , Methylprednisolone/administration & dosage , Methylprednisolone/pharmacology , Amyotrophic Lateral Sclerosis/genetics , Animals , Area Under Curve , Body Weight/drug effects , Cell Survival/drug effects , Central Nervous System/physiology , Disease Models, Animal , Drug Delivery Systems , Glucocorticoids/administration & dosage , Glucocorticoids/pharmacology , Glutathione/administration & dosage , Glutathione/pharmacology , Humans , Mice , Mice, Transgenic , Motor Activity/drug effects , Motor Neurons/drug effects , Motor Neurons/pathology , Neuroglia/drug effects , Neuroglia/physiology , Polyethylene Glycols/administration & dosage , Polyethylene Glycols/pharmacology , Superoxide Dismutase/genetics , Time Factors , Vacuoles/drug effects , Vacuoles/pathology
4.
Invest Ophthalmol Vis Sci ; 55(4): 2788-94, 2014 Apr 28.
Article in English | MEDLINE | ID: mdl-24692123

ABSTRACT

PURPOSE: Ocular inflammation is associated with the loss of visual acuity and subsequent blindness. Since their development, glucocorticoids have been the mainstay of therapy for ocular inflammatory diseases. However, the clinical benefit is limited by side effects due to the chronic use and generally high dosage that is required for effective treatment. We have developed the G-Technology to provide a means for sustained drug delivery, increased drug half-life, and reduced bodily drug exposure. Glutathione PEGylated liposomal methylprednisolone (2B3-201) has been developed as treatment for neuroinflammatory conditions and was evaluated in ocular inflammation. METHODS: The efficacy of 2B3-201 was investigated in rats with experimental autoimmune uveitis (EAU). Rats received 10 mg/kg of 2B3-201 intravenously at disease onset and at peak of the disease. The same dose of free methylprednisolone served as control treatment. Clinical signs of ocular inflammation were assessed by slit-lamp and immunohistochemistry. RESULTS: Whereas free methylprednisolone was ineffective, two doses of 2B3-201 almost completely abolished clinical signs of EAU. This was corroborated further by immunohistochemical analyses of isolated eyes. Treatment with 2B3-201 significantly reduced the infiltration of inflammatory cells and subsequent destruction of the retina cell layers. CONCLUSIONS: In this study, we show that systemic treatment with 2B3-201, a glutathione PEGylated liposomal methylprednisolone formulation, resulted in a superior efficacy in rats with EAU. Altogether, our findings hold promise for the development of a safe and more convenient systemic treatment for uveitis.


Subject(s)
Autoimmune Diseases/drug therapy , Glutathione/administration & dosage , Methylprednisolone/administration & dosage , Uveitis/drug therapy , Animals , Autoimmune Diseases/immunology , Autoimmune Diseases/pathology , Disease Models, Animal , Drug Combinations , Glucocorticoids/administration & dosage , Immunohistochemistry , Liposomes , Male , Rats , Rats, Inbred Lew , Treatment Outcome , Uveitis/immunology , Uveitis/pathology
5.
Epilepsy Res ; 108(3): 396-404, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24556423

ABSTRACT

It has been reported that glucocorticoids (GCs) can effectively control seizures in pediatric epilepsy syndromes, possibly by inhibition of inflammation. Since inflammation is supposed to be involved in epileptogenesis, we hypothesized that treatment with GCs would reduce brain inflammation and thereby modify epileptogenesis in a rat model for temporal lobe epilepsy, in which epilepsy gradually develops after electrically induced status epilepticus (SE). To prevent the severe adverse effects that are inevitable with long-term GC treatment, we used liposome nanotechnology (G-Technology(®)) to enhance the sustained delivery to the brain. Starting 4h after onset of SE, rats were treated with glutathione pegylated liposomal methylprednisolone (GSH-PEG liposomal MP) according to a treatment protocol (1× per week; 10mg/kg) that is effective in other models of neuroinflammation. Continuous electro-encephalogram (EEG) recordings revealed that SE duration and onset of spontaneous seizures were not affected by GSH-PEG liposomal MP treatment. The number and duration of spontaneous seizures were also not different between vehicle and GSH-PEG liposomal MP-treated animals. Six weeks after SE, brain inflammation, as assessed by quantification of microglia activation, was not reduced by GSH-PEG liposomal MP-treatment. Also, neuronal cell loss and mossy fiber sprouting were not affected. Our study shows that the selected GSH-PEG liposomal MP treatment regimen that was administered beyond the acute SE phase does not reduce brain inflammation and development of temporal lobe epilepsy.


Subject(s)
Anti-Inflammatory Agents/administration & dosage , Glutathione/administration & dosage , Methylprednisolone/administration & dosage , Phospholipids/administration & dosage , Status Epilepticus/drug therapy , Albumins , Animals , CD11b Antigen/metabolism , Disease Models, Animal , Drug Delivery Systems , Electrodes, Implanted/adverse effects , Hippocampus/pathology , Hippocampus/physiopathology , Male , Methylprednisolone/blood , Microglia/drug effects , Microglia/metabolism , Phosphopyruvate Hydratase/metabolism , Rats , Rats, Sprague-Dawley , Status Epilepticus/etiology , Time Factors
6.
J Drug Target ; 22(5): 460-7, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24524555

ABSTRACT

Partly due to poor blood-brain barrier drug penetration the treatment options for many brain diseases are limited. To safely enhance drug delivery to the brain, glutathione PEGylated liposomes (G-Technology®) were developed. In this study, in rats, we compared the pharmacokinetics and organ distribution of GSH-PEG liposomes using an autoquenched fluorescent tracer after intraperitoneal administration and intravenous administration. Although the appearance of liposomes in the circulation was much slower after intraperitoneal administration, comparable maximum levels of long circulating liposomes were found between 4 and 24 h after injection. Furthermore, 24 h after injection a similar tissue distribution was found. To investigate the effect of GSH coating on brain delivery in vitro uptake studies in rat brain endothelial cells (RBE4) and an in vivo brain microdialysis study in rats were used. Significantly more fluorescent tracer was found in RBE4 cell homogenates incubated with GSH-PEG liposomes compared to non-targeted PEG liposomes (1.8-fold, p < 0.001). In the microdialysis study 4-fold higher (p < 0.001) brain levels of fluorescent tracer were found after intravenous injection of GSH-PEG liposomes compared with PEG control liposomes. The results support further investigation into the versatility of GSH-PEG liposomes for enhanced drug delivery to the brain within a tolerable therapeutic window.


Subject(s)
Blood-Brain Barrier/drug effects , Drug Carriers/chemistry , Glutathione/chemistry , Polyethylene Glycols/chemistry , Animals , Blood-Brain Barrier/metabolism , Cell Line , Drug Carriers/administration & dosage , Drug Carriers/pharmacokinetics , Drug Stability , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Fluoresceins , Fluorescent Dyes , Glutathione/administration & dosage , Glutathione/pharmacokinetics , Injections, Intravenous , Injections, Spinal , Liposomes , Microdialysis , Particle Size , Polyethylene Glycols/administration & dosage , Polyethylene Glycols/pharmacokinetics , Rats , Rats, Wistar , Tissue Distribution
SELECTION OF CITATIONS
SEARCH DETAIL
...