Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 47
Filter
1.
J Nanobiotechnology ; 18(1): 120, 2020 Aug 31.
Article in English | MEDLINE | ID: mdl-32867843

ABSTRACT

Brain-derived neurotrophic factor (BDNF) is essential for the development and function of human neurons, therefore it is a promising target for neurodegenerative disorders treatment. Here, we studied BDNF-based electrostatic complex with dendrimer nanoparticles encapsulated in polyethylene glycol (PEG) in neurotoxin-treated, differentiated neuroblastoma SH-SY5Y cells, a model of neurodegenerative mechanisms. PEG layer was adsorbed at dendrimer-protein core nanoparticles to decrease their cellular uptake and to reduce BDNF-other proteins interactions for a prolonged time. Cytotoxicity and confocal microscopy analysis revealed PEG-ylated BDNF-dendrimer nanoparticles can be used for continuous neurotrophic factor delivery to the neurotoxin-treated cells over 24 h without toxic effect. We offer a reliable electrostatic route for efficient encapsulation and controlled transport of fragile therapeutic proteins without any covalent cross-linker; this could be considered as a safe drug delivery system. Understanding the polyvalent BDNF interactions with dendrimer core nanoparticles offers new possibilities for design of well-ordered protein drug delivery systems.


Subject(s)
Brain-Derived Neurotrophic Factor , Dendrimers/chemistry , Drug Carriers , Nanoparticles/chemistry , Neuroblastoma/metabolism , Brain-Derived Neurotrophic Factor/chemistry , Brain-Derived Neurotrophic Factor/pharmacokinetics , Cell Line, Tumor , Drug Carriers/chemistry , Drug Carriers/pharmacokinetics , Humans , Neurotoxins/adverse effects , Polyethylene Glycols/chemistry , Static Electricity
2.
Adv Exp Med Biol ; 1074: 465-471, 2018.
Article in English | MEDLINE | ID: mdl-29721977

ABSTRACT

This review discusses the therapeutic potential of brain-derived neurotrophic factor (BDNF) for retinal degeneration. BDNF, nerve growth factor (NGF), neurotrophin 3 (NT-3) and NT-4/NT-5 belong to the neurotrophin family. These neuronal modulators activate a common receptor and a specific tropomyosin-related kinase (Trk) receptor. BDNF was identified as a photoreceptor protectant in models of retinal degeneration as early as 1992. However, development of effective therapeutics that exploit this pathway has been difficult due to challenges in sustaining therapeutic levels in the retina.


Subject(s)
Brain-Derived Neurotrophic Factor/therapeutic use , Neuroprotective Agents/therapeutic use , Retinal Degeneration/drug therapy , Animals , Brain-Derived Neurotrophic Factor/genetics , Brain-Derived Neurotrophic Factor/pharmacokinetics , Cell Survival/drug effects , Dependovirus/genetics , Disease Models, Animal , Drug Evaluation, Preclinical , Genetic Therapy , Genetic Vectors/therapeutic use , Humans , Mice , Neuroprotective Agents/pharmacokinetics , Photoreceptor Cells, Vertebrate/drug effects , Rats , Recombinant Proteins/pharmacokinetics , Recombinant Proteins/therapeutic use , Retinal Degeneration/prevention & control , Retinal Degeneration/therapy
3.
Exp Hematol ; 63: 17-21.e1, 2018 07.
Article in English | MEDLINE | ID: mdl-29705266

ABSTRACT

Increased chemokine C-X-C receptor 4 (CXCR4) expression is related to unfavorable outcome in chronic lymphocytic leukemia (CLL). Brain-derived neurotrophic factor (BDNF) is a neuronal growth factor that has been shown previously to interact with CXCR4 in neuronal cells. Here, we studied the in vitro effect of BDNF on CXCR4 expression and chemotaxis toward stromal derived factor-1 (SDF-1) in freshly isolated CLL cells. We also explored the correlations between serum BDNF levels in CLL patients and disease characteristics and clinical course. Incubation of CLL cells with recombinant BDNF (50 ng/mL) resulted in a downregulation of CXCR4 surface expression and atenuated chemotaxis toward SDF-1. Higher serum BDNF levels were associated with a mutated immunoglobulin heavy chain variable (IGHV) gene, an early clinical stage, and a stable clinical course. Our findings suggest that increased circulating blood BDNF may be associated with a favorable effect in CLL. However, the exact mechanism of this favorable effect should be investigated further.


Subject(s)
Brain-Derived Neurotrophic Factor/blood , Leukemia, Lymphocytic, Chronic, B-Cell/blood , Neoplasm Proteins/blood , Receptors, CXCR4/biosynthesis , Brain-Derived Neurotrophic Factor/genetics , Brain-Derived Neurotrophic Factor/pharmacokinetics , Brain-Derived Neurotrophic Factor/physiology , Chemokine CXCL12/pharmacology , Chemotaxis/drug effects , Down-Regulation/drug effects , Gene Expression Regulation, Leukemic/drug effects , Gene Rearrangement, B-Lymphocyte, Heavy Chain , Genes, Immunoglobulin , Humans , Immunoglobulin Variable Region/genetics , Kaplan-Meier Estimate , Leukemia, Lymphocytic, Chronic, B-Cell/genetics , Leukemia, Lymphocytic, Chronic, B-Cell/mortality , Leukemia, Lymphocytic, Chronic, B-Cell/pathology , Membrane Glycoproteins/biosynthesis , Membrane Glycoproteins/genetics , Neoplasm Proteins/biosynthesis , Neoplasm Proteins/genetics , Neoplasm Proteins/physiology , Platelet Count , Prognosis , Receptor, trkB/biosynthesis , Receptor, trkB/genetics , Receptors, CXCR4/genetics , Recombinant Proteins/pharmacology , Tumor Cells, Cultured
4.
Sci Rep ; 6: 32292, 2016 08 25.
Article in English | MEDLINE | ID: mdl-27558932

ABSTRACT

Recurrent laryngeal nerve (RLN) injury remains a challenge due to the lack of effective treatments. In this study, we established a new drug delivery system consisting of a tube of Heal-All Oral Cavity Repair Membrane loaded with laminin and neurotrophic factors and tested its ability to promote functional recovery following RLN injury. We created recombinant fusion proteins consisting of brain-derived neurotrophic factor (BDNF) and glial cell line-derived neurotrophic factor (GDNF) fused to laminin-binding domains (LBDs) in order to prevent neurotrophin diffusion. LBD-BDNF, LBD-GDNF, and laminin were injected into a collagen tube that was fitted to the ends of the transected RLN in rats. Functional recovery was assessed 4, 8, and 12 weeks after injury. Although vocal fold movement was not restored until 12 weeks after injury, animals treated with the collagen tube loaded with laminin, LBD-BDNF and LBD-GDNF showed improved recovery in vocalisation, arytenoid cartilage angles, compound muscle action potentials and regenerated fibre area compared to animals treated by autologous nerve grafting (p < 0.05). These results demonstrate the drug delivery system induced nerve regeneration following RLN transection that was superior to that induced by autologus nerve grafting. It may have potential applications in nerve regeneration of RLN transection injury.


Subject(s)
Brain-Derived Neurotrophic Factor , Collagen , Glial Cell Line-Derived Neurotrophic Factor , Laminin , Laryngeal Nerves/physiology , Lingual Nerve Injuries/therapy , Nerve Regeneration/drug effects , Tissue Scaffolds/chemistry , Animals , Brain-Derived Neurotrophic Factor/chemistry , Brain-Derived Neurotrophic Factor/pharmacokinetics , Brain-Derived Neurotrophic Factor/pharmacology , Collagen/chemistry , Collagen/pharmacokinetics , Collagen/pharmacology , Glial Cell Line-Derived Neurotrophic Factor/chemistry , Glial Cell Line-Derived Neurotrophic Factor/pharmacokinetics , Glial Cell Line-Derived Neurotrophic Factor/pharmacology , Laminin/chemistry , Laminin/pharmacokinetics , Laminin/pharmacology , Lingual Nerve Injuries/metabolism , Lingual Nerve Injuries/pathology , Male , Rats , Rats, Sprague-Dawley , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/pharmacokinetics , Recombinant Fusion Proteins/pharmacology
5.
Sci Adv ; 2(5): e1600519, 2016 05.
Article in English | MEDLINE | ID: mdl-27386554

ABSTRACT

Encapsulation of therapeutic molecules within polymer particles is a well-established method for achieving controlled release, yet challenges such as low loading, poor encapsulation efficiency, and loss of protein activity limit clinical translation. Despite this, the paradigm for the use of polymer particles in drug delivery has remained essentially unchanged for several decades. By taking advantage of the adsorption of protein therapeutics to poly(lactic-co-glycolic acid) (PLGA) nanoparticles, we demonstrate controlled release without encapsulation. In fact, we obtain identical, burst-free, extended-release profiles for three different protein therapeutics with and without encapsulation in PLGA nanoparticles embedded within a hydrogel. Using both positively and negatively charged proteins, we show that short-range electrostatic interactions between the proteins and the PLGA nanoparticles are the underlying mechanism for controlled release. Moreover, we demonstrate tunable release by modifying nanoparticle concentration, nanoparticle size, or environmental pH. These new insights obviate the need for encapsulation and offer promising, translatable strategies for a more effective delivery of therapeutic biomolecules.


Subject(s)
Delayed-Action Preparations , Drug Carriers , Drug Delivery Systems , Lactic Acid , Nanoparticles , Polyglycolic Acid , Adsorption , Brain-Derived Neurotrophic Factor/administration & dosage , Brain-Derived Neurotrophic Factor/pharmacokinetics , Drug Carriers/chemistry , Drug Compounding , Drug Liberation , Hydrogen-Ion Concentration , Lactic Acid/chemistry , Monte Carlo Method , Nanoparticles/chemistry , Polyglycolic Acid/chemistry , Polylactic Acid-Polyglycolic Acid Copolymer , Proteins/administration & dosage , Proteins/chemistry , Proteins/pharmacokinetics , Static Electricity
6.
Biomaterials ; 100: 41-52, 2016 09.
Article in English | MEDLINE | ID: mdl-27240161

ABSTRACT

Ultrasound-targeted microbubble destruction (UTMD) has been shown to be a promising tool to deliver proteins to select body areas. This study aimed to analyze whether UTMD was able to deliver brain-derived neurotrophic factor (BDNF) to the brain, enhancing functional recovery and white matter repair, in an animal model of subcortical stroke induced by endothelin (ET)-1. UTMD was used to deliver BDNF to the brain 24 h after stroke. This technique was shown to be safe, given there were no cases of hemorrhagic transformation or blood brain barrier (BBB) leakage. UTMD treatment was associated with increased brain BDNF levels at 4 h after administration. Targeted ultrasound delivery of BDNF improved functional recovery associated with fiber tract connectivity restoration, increasing oligodendrocyte markers and remyelination compared to BDNF alone administration in an experimental animal model of white matter injury.


Subject(s)
Brain-Derived Neurotrophic Factor/administration & dosage , Brain/drug effects , Drug Delivery Systems/methods , Microbubbles , Stroke/drug therapy , Ultrasonics/methods , White Matter/drug effects , Animals , Brain/pathology , Brain-Derived Neurotrophic Factor/pharmacokinetics , Brain-Derived Neurotrophic Factor/therapeutic use , Cell Line , Male , Oligodendroglia/drug effects , Oligodendroglia/pathology , Rats, Sprague-Dawley , Stroke/pathology , Ultrasonic Waves , White Matter/pathology
7.
Neuroreport ; 27(2): 116-23, 2016 Jan 20.
Article in English | MEDLINE | ID: mdl-26656937

ABSTRACT

This study aimed to examine the effect of controlled release of brain-derived neurotrophic factor (BDNF) and neurotrophin-3 (NT-3) from collagen gel on rat neural stem cells (NSCs). With three groups of collagen gel, BDNF/collagen gel, and NT-3/collagen gel as controls, BDNF and NT-3 were tested in the BDNF-NT-3/collagen gel group at different time points. The enzyme-linked immunosorbent assay results showed that BDNF and NT-3 were steadily released from collagen gels for 10 days. The cell viability test and the bromodeoxyuridine incorporation assay showed that BDNF-NT-3/collagen gel supported the survival and proliferation of NSCs. The results also showed that the length of processes was markedly longer and differentiation percentage from NSCs into neurons was much higher in the BDNF-NT-3/collagen gel group than those in the collagen gel, BDNF/collagen gel, and NT-3/collagen gel groups. These findings suggest that BDNF-NT-3/collagen gel could significantly improve the ability of NSCs proliferation and differentiation.


Subject(s)
Brain-Derived Neurotrophic Factor/pharmacology , Collagen/pharmacology , Neural Stem Cells/drug effects , Neurotrophin 3/pharmacology , Animals , Brain-Derived Neurotrophic Factor/pharmacokinetics , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Cell Survival/drug effects , Cells, Cultured , Female , Neurotrophin 3/pharmacokinetics , Phenotype , Rats , Rats, Sprague-Dawley
8.
Sci Rep ; 5: 15032, 2015 Oct 14.
Article in English | MEDLINE | ID: mdl-26463268

ABSTRACT

Although Alzheimer's disease (AD) has been reported for more than 100 years, there is still a lack of effective cures for this devastating disorder. Among the various obstacles that hold back drug development, the blood-brain barrier (BBB) is one of them. Here, we constructed a novel fusion peptide by linking the active domain of brain-derived neurotrophic factor (BDNF) with an HIV-encoded transactivator of transcription (TAT) that has a strong membrane-penetrating property. After intraperitoneal injection, the eGFP-TAT could be robustly detected in different brain regions. By using scopolamine-induced rats and APPswe mice representing AD-like cholinergic deficits and amyloidosis, respectively, we found that intraperitoneal administration of the peptide significantly improved spatial memory with activation of the TrkB/ERK1/2/Akt pathway and restoration of several memory-associated proteins in both models. Administration of the peptide also modulated ß-amyloid and tau pathologies in APPswe mice, and it increased the amount of M receptor with modulation of acetylcholinesterase in scopolamine-induced rats. We conclude that intraperitoneal administration of our TAT-BDNF peptide could efficiently target multiple molecular pathways in the brain and improve the cognitive functions in AD-like rodent models.


Subject(s)
Alzheimer Disease/drug therapy , Alzheimer Disease/physiopathology , Brain-Derived Neurotrophic Factor/administration & dosage , Cognition Disorders/prevention & control , Cognition Disorders/physiopathology , Gene Products, tat/administration & dosage , Alzheimer Disease/chemically induced , Amyloid beta-Protein Precursor , Animals , Blood-Brain Barrier/metabolism , Brain-Derived Neurotrophic Factor/genetics , Brain-Derived Neurotrophic Factor/pharmacokinetics , Cognition/drug effects , Cognition Disorders/chemically induced , Disease Models, Animal , Drug Combinations , Gene Products, tat/genetics , Gene Products, tat/pharmacokinetics , Injections, Intraperitoneal , Male , Mice , Mice, Transgenic , Nerve Tissue Proteins/metabolism , Rats , Rats, Sprague-Dawley , Recombinant Fusion Proteins/administration & dosage , Recombinant Fusion Proteins/genetics , Scopolamine , Treatment Outcome
9.
Int J Nanomedicine ; 10: 3245-67, 2015.
Article in English | MEDLINE | ID: mdl-25995632

ABSTRACT

Neurodegenerative causes of blindness and deafness possess a major challenge in their clinical management as proper treatment guidelines have not yet been found. Brain-derived neurotrophic factor (BDNF) has been established as a promising therapy against neurodegenerative disorders including hearing and visual loss. Unfortunately, the blood-retinal barrier and blood-cochlear barrier, which have a comparable structure to the blood-brain barrier prevent molecules of larger sizes (such as BDNF) from exiting the circulation and reaching the targeted cells. Anatomical features of the eye and ear allow use of local administration, bypassing histo-hematic barriers. This paper focuses on highlighting a variety of strategies proposed for the local administration of the BDNF, like direct delivery, viral gene therapy, and cell-based therapy, which have been shown to successfully improve development, survival, and function of spiral and retinal ganglion cells. The similarities and controversies for BDNF treatment of posterior eye diseases and inner ear diseases have been analyzed and compared. In this review, we also focus on the possibility of translation of this knowledge into clinical practice. And finally, we suggest that using nanoparticulate drug-delivery systems may substantially contribute to the development of clinically viable techniques for BDNF delivery into the cochlea or posterior eye segment, which, ultimately, can lead to a long-term or permanent rescue of auditory and optic neurons from degeneration.


Subject(s)
Blindness , Blood-Brain Barrier/metabolism , Brain-Derived Neurotrophic Factor , Deafness , Drug Delivery Systems , Animals , Blindness/drug therapy , Blindness/physiopathology , Brain-Derived Neurotrophic Factor/pharmacokinetics , Brain-Derived Neurotrophic Factor/pharmacology , Brain-Derived Neurotrophic Factor/therapeutic use , Cell Line , Deafness/drug therapy , Deafness/physiopathology , Humans , Mice
10.
Small ; 10(21): 4244-8, 2014 Nov 12.
Article in English | MEDLINE | ID: mdl-25099026

ABSTRACT

Mesoporous silica supraparticles (MS-SPs) are prepared via self-assembly of mesoporous silica nanoparticles under capillary force action in confined droplets. The MS-SPs are effective carriers for sustained drug delivery. Animal studies show that these particles are suitable for chronic intracochlear implantation, and neurotrophins released from the MS-SPs can efficiently rescue primary auditory neurons in an in vivo sensorineural hearing loss model.


Subject(s)
Delayed-Action Preparations/administration & dosage , Drug Carriers , Ear, Inner , Nanoparticles , Silicon Dioxide/chemistry , Animals , Brain-Derived Neurotrophic Factor/administration & dosage , Brain-Derived Neurotrophic Factor/pharmacokinetics , Cochlea/drug effects , Cochlea/pathology , Deafness/drug therapy , Deafness/metabolism , Deafness/pathology , Delayed-Action Preparations/pharmacokinetics , Drug Carriers/chemical synthesis , Drug Carriers/chemistry , Drug Carriers/pharmacokinetics , Drug Delivery Systems , Ear, Inner/drug effects , Ear, Inner/metabolism , Guinea Pigs , Materials Testing , Nanoparticles/chemistry , Nanoparticles/therapeutic use
11.
PLoS One ; 8(5): e64037, 2013.
Article in English | MEDLINE | ID: mdl-23700454

ABSTRACT

Loss of huntingtin-mediated BDNF gene transcription has been shown to occur in HD and thus contribute to the degeneration of the striatum. Several studies have indicated that an increase in BDNF levels is associated with neuroprotection and amelioration of neurological signs in animal models of HD. In a recent study, an increase in BDNF mRNA and protein levels was recorded in mice administered recombinant BDNF peripherally. Chronic, indwelling osmotic mini-pumps containing either recombinant BDNF or saline were surgically placed in R6/2 or wild-type mice from 4 weeks of age until euthanasia. Neurological evaluation (paw clasping, rotarod performance, locomotor activity in an open field) was performed. After transcardial perfusion, histological and immunohistochemical studies were performed. We found that BDNF- treated R6/2 mice survived longer and displayed less severe signs of neurological dysfunction than the vehicle treated ones. Primary outcome measures such as brain volume, striatal atrophy, size and morphology of striatal neurons, neuronal intranuclear inclusions and microglial reaction confirmed a neuroprotective effect of the compound. BDNF was effective in increasing significantly the levels of activated CREB and of BDNF the striatal spiny neurons. Moreover, systemically administered BDNF increased the synthesis of BDNF as demonstrated by RT-PCR, and this might account for the beneficial effects observed in this model.


Subject(s)
Brain-Derived Neurotrophic Factor/administration & dosage , Huntington Disease/drug therapy , Neuroprotective Agents/administration & dosage , Animals , Brain-Derived Neurotrophic Factor/metabolism , Brain-Derived Neurotrophic Factor/pharmacokinetics , Cerebral Cortex/drug effects , Cerebral Cortex/metabolism , Corpus Striatum/drug effects , Corpus Striatum/metabolism , Cyclic AMP Response Element-Binding Protein/metabolism , Disease Models, Animal , Drug Evaluation, Preclinical , Extracellular Signal-Regulated MAP Kinases/metabolism , Female , Gene Expression , Huntington Disease/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Nerve Degeneration/drug therapy , Nerve Degeneration/metabolism , Neuroprotective Agents/metabolism , Neuroprotective Agents/pharmacokinetics , Phosphorylation , Protein Processing, Post-Translational , Psychomotor Performance/drug effects , RNA, Messenger/genetics , RNA, Messenger/metabolism , Recombinant Proteins/administration & dosage , Rotarod Performance Test
12.
Pharm Res ; 29(6): 1562-9, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22231987

ABSTRACT

PURPOSE: Delivery of therapeutic proteins across the blood-brain barrier (BBB) is severely limited by their size and biochemical properties. Here we showed that a 39-amino acid peptide derived from the rabies virus glycoprotein (RDP) was exploited as an efficient protein carrier for brain-targeting delivery. METHODS: Three proteins with different molecular weight and pI, ß-galactosidase (ß-Gal), luciferase (Luc) and brain-derived neurotrophic factor (BDNF), were fused to RDP and intravenously injected into the mice respectively. The slices of different tissues with X-Gal staining were used to examine whether RDP could deliver ß-Gal targeted into the CNS. The time-course relationship of RDP-Luc was studied to confirm the transport efficiency of RDP. The neuroprotective function of RDP-BDNF was examined in mouse experimental stroke to explore the pharmacological effect of RDP fusion protein. RESULTS: The results showed that the fusion proteins rapidly and specific entered the nerve cells in 15 min, and the t(1/2) was about 1 hr. Furthermore, RDP-BDNF fusion protein showed the neuroprotective properties in mouse experimental stroke including reduction of stroke volume and neural deficit. CONCLUSIONS: RDP provides an effective approach for the targeted delivery of biological active proteins into the central nervous system.


Subject(s)
Blood-Brain Barrier/metabolism , Brain-Derived Neurotrophic Factor/pharmacokinetics , Drug Carriers , Glycoproteins/pharmacokinetics , Neuroprotective Agents/pharmacokinetics , Peptide Fragments/pharmacokinetics , Rabies virus/metabolism , Viral Proteins/pharmacokinetics , Animals , Brain-Derived Neurotrophic Factor/administration & dosage , Brain-Derived Neurotrophic Factor/chemistry , Capillary Permeability , Chemistry, Pharmaceutical , Disease Models, Animal , Drug Compounding , Glycoproteins/administration & dosage , Glycoproteins/chemistry , Half-Life , Humans , Infarction, Middle Cerebral Artery/drug therapy , Infarction, Middle Cerebral Artery/metabolism , Infarction, Middle Cerebral Artery/pathology , Injections, Intravenous , Luciferases/administration & dosage , Luciferases/chemistry , Luciferases/pharmacokinetics , Male , Mice , Neuroprotective Agents/administration & dosage , Neuroprotective Agents/chemistry , Peptide Fragments/administration & dosage , Peptide Fragments/chemistry , Recombinant Fusion Proteins/administration & dosage , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/pharmacokinetics , Technology, Pharmaceutical/methods , Tissue Distribution , Viral Proteins/administration & dosage , Viral Proteins/chemistry , beta-Galactosidase/administration & dosage , beta-Galactosidase/chemistry , beta-Galactosidase/pharmacokinetics
13.
Pharm Res ; 29(2): 441-7, 2012 Feb.
Article in English | MEDLINE | ID: mdl-21879386

ABSTRACT

PURPOSE: To investigate the plausibility of delivering brain-derived neurotrophic factor (BDNF) to brain via nose-to-brain pathway using chitosan as barrier-modulating agent. METHODS: Effect of different viscosity grades chitosan at different concentrations on permeation of fluorescein isothio-cyanate dextran (FD 40 K) across bovine olfactory mucosa was studied using Franz diffusion cells. Medium viscosity chitosan was used to carry out permeation studies of BDNF. Pharmacokinetic and pharmacodynamic studies were carried out in Sprague dawley rats upon intranasal/i.v administration of different formulations. RESULTS: Medium viscosity chitosan more efficiently enhanced permeation of FD 40 K across olfactory mucosa compared to other grades. In case of BDNF, medium viscosity chitosan (0.25% w/v) enhanced permeation ~14-fold over control (18.78 ± 16.69 ng/cm(2)). Brain bioavailability of rats administered intranasally with BDNF solution containing chitosan was significantly enhanced ~13-fold compared to rats administered with same concentration of BDNF solution without chitosan. In rats subjected to immobilization stress, BDNF solution containing chitosan significantly decreased immobility time. CONCLUSIONS: Intranasal formulations containing chitosan as barrier-modulating agent significantly enhanced brain bioavailability of BDNF. Delivery of BDNF was found to counteract stress-induced depression in rats.


Subject(s)
Brain-Derived Neurotrophic Factor/administration & dosage , Brain-Derived Neurotrophic Factor/pharmacokinetics , Brain/metabolism , Chitosan/metabolism , Drug Carriers/metabolism , Olfactory Mucosa/metabolism , Administration, Intranasal , Animals , Biological Availability , Chitosan/chemistry , Drug Carriers/chemistry , Male , Rats , Rats, Sprague-Dawley
14.
Pharm Res ; 27(1): 82-91, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19921405

ABSTRACT

PURPOSE: Brain-derived neurotrophic factor (BDNF) plays an important role in neuroprotection and repair, but long-term delivery from polymer systems has been challenging. We investigated the role the chemistry of the polymer played in loading and delivery of BDNF via microspheres, which are suitable for minimally invasive administration. METHODS: We synthesized polymers based on PLGA and PEG to determine what components augmented loading and delivery. We characterized microspheres fabricated from these polymers using a battery of tests, including sizing, in vitro release, and bioactivity of the BDNF using PC12 cells engineered to express the trkB receptor. RESULTS: We found that a triblock polymer of PLGA, PLL, and PEG led to the delivery of BDNF for periods of time greater than 60 days and that the BDNF delivered was bioactive. The microsphere size was amendable to injection via a 30 gauge needle, allowing minimally invasive delivery. CONCLUSIONS: PLGA-PLL-PEG leads to greater loading and longer-term delivery of BDNF than PLGA or a blend of the polymers. We hypothesize that the introduction of an amphiphilic PLGA-based polymer increases the interaction of the BDNF with the polymer and leads to release that more closely correlates with the degradation of the polymer.


Subject(s)
Brain-Derived Neurotrophic Factor , Chemistry, Pharmaceutical/methods , Drug Carriers/chemical synthesis , Microspheres , Polymers/chemistry , Animals , Brain-Derived Neurotrophic Factor/chemistry , Brain-Derived Neurotrophic Factor/pharmacokinetics , Brain-Derived Neurotrophic Factor/pharmacology , Cell Differentiation/drug effects , Drug Carriers/chemistry , Lactic Acid/chemistry , Models, Chemical , PC12 Cells , Particle Size , Polyethylene Glycols/chemistry , Polyglycolic Acid/chemistry , Polylactic Acid-Polyglycolic Acid Copolymer , Polylysine/chemistry , Polymers/chemical synthesis , Rats , Receptor, trkB/agonists , Solubility
15.
J Control Release ; 119(2): 163-72, 2007 Jun 04.
Article in English | MEDLINE | ID: mdl-17428570

ABSTRACT

It was the aim of this study to establish triglyceride matrices as potential carriers for long-term release of brain-derived neurotrophic factor (BDNF), a potential therapeutic for Huntington's disease. First, four different manufacturing strategies were investigated with lysozyme as a model substance: either lyophilized protein was mixed with lipid powder, or suspended in organic solution thereof (s/o). Or else, an aqueous protein solution was dispersed by w/o emulsion in organic lipid solution. Alternatively, a PEG co-lyophilization was performed prior to dispersing solid protein microparticles in organic lipid solution. After removal of the solvent(s), the resulting powder formulations were compressed at 250 N to form mini-cylinders of 2 mm diameter, 2.2 mm height and 7 mg weight. Protein integrity after formulation and release was evaluated from an enzyme activity assay and SDS-PAGE. Confocal microscopy revealed that the resulting distribution of FITC-lysozyme within the matrices depended strongly on the manufacturing method, which had an important impact on matrix performance: matrices with a very fine and homogeneous protein distribution (PEG co-lyophilization) continually released protein for 2 months. The other methods did not guarantee a homogeneous distribution and either failed in sustaining release for more than 1 week (powder mixture), completely liberating the loading (s/o dispersion) or preserving protein activity during manufacturing (w/o emulsion, formation of aggregates and 25% activity loss). Based on these results, miniature-sized implants of 1 mm diameter, 0.8 mm height and 1 mg weight were successfully loaded by the PEG co-lyophilization method with 2% BDNF and 2% PEG. Release studies in phosphate buffer pH 7.4 at 4 and 37 degrees C revealed a controlled release of either 20 or 60% intact protein over one month as determined by ELISA. SDS-PAGE detected only minor aggregates in the matrix during release at higher temperature. In vivo evaluation of lipid cylinders in the striatum of rat brains revealed a biocompatibility comparable to silicone reference cylinders.


Subject(s)
Brain-Derived Neurotrophic Factor/pharmacokinetics , Brain/metabolism , Drug Delivery Systems/methods , Lipids/pharmacokinetics , Muramidase/pharmacokinetics , Animals , Brain/drug effects , Brain-Derived Neurotrophic Factor/administration & dosage , Chemistry, Pharmaceutical/methods , Chickens , Delayed-Action Preparations/administration & dosage , Delayed-Action Preparations/pharmacokinetics , Drug Evaluation, Preclinical/methods , Drug Implants , Female , Lipids/administration & dosage , Materials Testing/methods , Muramidase/administration & dosage , Rats , Rats, Inbred F344
16.
Biotechnol Bioeng ; 97(6): 1376-86, 2007 Aug 15.
Article in English | MEDLINE | ID: mdl-17286273

ABSTRACT

Neurotrophins, such as brain derived neurotrophic factor (BDNF), do not cross the blood-brain barrier (BBB). Certain monoclonal antibodies (MAb) to the human insulin receptor (HIR) do cross the BBB via receptor-mediated transport, and can act as a molecular Trojan horse to ferry across the BBB an attached drug. A genetically engineered fusion protein was produced whereby the amino terminus of human BDNF is fused to the carboxyl terminus of the heavy chain of a chimeric HIRMAb. The HIRMAb-BDNF fusion protein reacted equally with antibodies to human IgG and BDNF. The bi-functionality of the fusion protein was retained as the affinity of the fusion protein for the HIR was identical to that of the chimeric HIRMAb, and the affinity of the fusion protein for the trkB receptor was identical to that of BDNF. The fusion protein was equi-potent with BDNF in a neuroprotection assay in human neural cells. The pharmacokinetics (PK) of the fusion protein was examined in the adult Rhesus monkey. The mean residence time (MRT) of the fusion protein in blood was >100-fold longer than the MRT of BDNF. Therapeutic levels of BDNF were produced in primate brain following the intravenous administration of the fusion protein. A fusion protein tandem vector was engineered that allowed for isolation of a CHO cell line that produced the fusion protein at high levels in serum free medium. Neurotrophins, such as BDNF, can be re-formulated to enable these molecules to cross the human BBB, and such fusion proteins represent a new class of human neurotherapeutics.


Subject(s)
Antibodies, Monoclonal/pharmacokinetics , Blood-Brain Barrier/metabolism , Brain-Derived Neurotrophic Factor/metabolism , Brain-Derived Neurotrophic Factor/pharmacokinetics , Drug Delivery Systems/methods , Protein Engineering/methods , Animals , Antibodies, Monoclonal/genetics , Female , Haplorhini , Humans , Macaca mulatta , Metabolic Clearance Rate , Recombinant Fusion Proteins/pharmacokinetics
17.
Brain Res ; 1111(1): 227-9, 2006 Sep 21.
Article in English | MEDLINE | ID: mdl-16884698

ABSTRACT

Intravenous brain-derived neurotrophic factor (BDNF) causes a 65-70% reduction in stroke volume in rats with the middle cerebral artery occlusion (MCAO), provided the BDNF is conjugated to a blood-brain barrier (BBB) molecular Trojan horse. The latter may be a peptidomimetic monoclonal antibody (MAb) to the transferrin receptor. The present studies determine whether the effects on stroke volume correlate with an improvement in neuro-behavior using the rotarod test. The rotarod latency was >200 s at 16 RPM in all rats pre-MCAO. The latency was 30+/-7 s and 103+/-9 s at 24 h post-MCAO in the animals treated with BDNF alone and the BDNF-MAb conjugate, respectively. These studies show that when BDNF is formulated to enable transport across the BBB, the intravenous administration of the neurotrophin results in a reduction in stroke volume that is associated with a parallel improvement in functional outcome.


Subject(s)
Blood-Brain Barrier/drug effects , Brain-Derived Neurotrophic Factor/pharmacokinetics , Infarction, Middle Cerebral Artery/drug therapy , Movement Disorders/drug therapy , Recovery of Function/drug effects , Stroke/drug therapy , Animals , Antibodies, Monoclonal/pharmacokinetics , Antibodies, Monoclonal/therapeutic use , Blood-Brain Barrier/metabolism , Brain-Derived Neurotrophic Factor/metabolism , Brain-Derived Neurotrophic Factor/therapeutic use , Disease Models, Animal , Infarction, Middle Cerebral Artery/metabolism , Infarction, Middle Cerebral Artery/physiopathology , Injections, Intravenous , Male , Motor Activity/drug effects , Motor Activity/physiology , Motor Cortex/drug effects , Motor Cortex/metabolism , Motor Cortex/physiopathology , Movement Disorders/etiology , Movement Disorders/physiopathology , Paresis/drug therapy , Paresis/etiology , Paresis/physiopathology , Rats , Rats, Sprague-Dawley , Reaction Time/drug effects , Reaction Time/physiology , Recovery of Function/physiology , Stroke/metabolism , Stroke/physiopathology , Treatment Outcome
18.
J Surg Res ; 136(1): 85-91, 2006 Nov.
Article in English | MEDLINE | ID: mdl-16904693

ABSTRACT

BACKGROUND: Brain-derived neurotrophic factor (BDNF) is a survival factor for endothelial cells and expresses in the ischemic myocytes. The purpose of this study was to assess whether the simultaneous application of basic fibroblast growth factor (bFGF) and BDNF incorporating gelatin hydrogels improves angiogenesis and cardiac function in ischemic myocardium compared with bFGF applied alone. MATERIALS AND METHODS: Direct intramyocardial injection of 100 microg of bFGF plus 25 microg of BDNF, 100 microg of bFGF, or saline were performed in canine infarct model. Colored microspheres were injected to assess the regional myocardial blood flow. Cardiac function was evaluated by cine magnetic resonance imaging (MRI). Immunohistochemical staining and enzyme linked immunosorbent assay (ELISA) were used to observe the localization and expression of bFGF and BDNF protein, and myocardial microvessel density was assessed by von Willebrand factor staining. RESULTS: Left ventricular ejection fraction (LVEF) was higher in bFGF plus BDNF group than in saline or bFGF group. Blood flow of the peri-infarct region was increased by bFGF plus BDNF treatment. The distribution of bFGF and BDNF-positive cardiomyocytes was similar in three groups. The expression of bFGF and BDNF protein and microvessel density in bFGF plus BDNF group was higher than in the other two groups. CONCLUSIONS: This study indicates that the sustained dual release of bFGF and BDNF incorporating gelatin hydrogels can improve angiogenesis and left ventricular function in the ischemic myocardium compared with bFGF applied alone. bFGF plus BDNF administration may be a promising therapeutic strategy for the treatment of ischemic myocardium.


Subject(s)
Brain-Derived Neurotrophic Factor/pharmacology , Fibroblast Growth Factor 2/pharmacology , Myocardial Ischemia/drug therapy , Neovascularization, Physiologic/drug effects , Ventricular Dysfunction, Left/drug therapy , Animals , Brain-Derived Neurotrophic Factor/pharmacokinetics , Coronary Circulation/drug effects , Disease Models, Animal , Dogs , Drug Therapy, Combination , Enzyme-Linked Immunosorbent Assay , Fibroblast Growth Factor 2/pharmacokinetics , Hydrogels/pharmacology , Immunohistochemistry , Microcirculation/drug effects , Microspheres , Stroke Volume/drug effects
19.
Brain Res ; 1067(1): 250-5, 2006 Jan 05.
Article in English | MEDLINE | ID: mdl-16364259

ABSTRACT

Brain-derived neurotrophic factor (BDNF) has recently emerged as one of the most potent molecular mediators of not only central synaptic plasticity, but also behavioral interactions between an organism and its environment. Our previous studies on the insular cortex (IC), a region of the temporal cortex implicated in the acquisition and storage of conditioned taste aversion (CTA), have demonstrated that induction of long-term potentiation (LTP) in the projection from the basolateral nucleus of the amygdala (Bla) to the IC, previous to CTA training, enhances the retention of this task. Recently, we found that intracortical microinfusion of BDNF induces a lasting potentiation of synaptic efficacy in the Bla-IC projection of adult rats in vivo. In this work, we present experimental data showing that intracortical microinfusion of BDNF previous to CTA training enhances the retention of this task. These findings support the concept that BDNF may contribute to memory-related functions performed by a neocortical area, playing a critical role in long-term synaptic plasticity.


Subject(s)
Brain-Derived Neurotrophic Factor/pharmacology , Memory/physiology , Taste/physiology , Animals , Brain-Derived Neurotrophic Factor/administration & dosage , Brain-Derived Neurotrophic Factor/pharmacokinetics , Conditioning, Psychological , Drinking Behavior/drug effects , Drinking Behavior/physiology , Infusions, Parenteral , Memory/drug effects , Models, Animal , Rats , Synapses/drug effects , Synapses/physiology , Taste/drug effects , Tissue Distribution
20.
Biomaterials ; 27(3): 452-9, 2006 Jan.
Article in English | MEDLINE | ID: mdl-16115674

ABSTRACT

Degradable hydrogels are useful vehicles for the delivery of growth factors to promote the regeneration of diseased or damaged tissue. In the central nervous system, there are many instances where the delivery of neurotrophins has great potential in tissue repair, especially for treatment of spinal cord injury. In this work, hydrogels based on poly(ethylene glycol) that form via a photoinitiated polymerization were investigated for the delivery of neurotrophins. The release kinetics of these factors are controlled by changes in the network crosslinking density, which influences neurotrophin diffusion and subsequent release from the gels with total release times ranging from weeks to several months. The release and activity of one neurotrophic factor, ciliary-neurotrophic factor (CNTF), was assessed with a cell-based proliferation assay and an assay for neurite outgrowth from retinal explants. CNTF released from a degradable hydrogel above an explanted retina was able to stimulate outgrowth of a significantly higher number of neurites than controls without CNTF. Finally, unique microsphere/hydrogel composites were developed to simultaneously deliver multiple neurotrophins with individual release rates.


Subject(s)
Cell Enlargement/drug effects , Hydrogels/pharmacology , Nerve Growth Factors/pharmacology , Neurites/drug effects , Animals , Brain-Derived Neurotrophic Factor/pharmacokinetics , Cell Line , Cell Proliferation/drug effects , Ciliary Neurotrophic Factor/pharmacokinetics , Ciliary Neurotrophic Factor/pharmacology , Delayed-Action Preparations/chemistry , Drug Carriers/chemistry , Erythroblasts/drug effects , Ethylamines/chemistry , Glycolates/chemistry , Humans , Hydrogels/metabolism , In Vitro Techniques , Lactates/chemical synthesis , Lactates/chemistry , Lactic Acid , Mice , Microspheres , Nerve Growth Factors/pharmacokinetics , Neurites/physiology , Neurotrophin 3/pharmacokinetics , Polyethylene Glycols/chemical synthesis , Polyethylene Glycols/chemistry , Polyglycolic Acid , Polylactic Acid-Polyglycolic Acid Copolymer , Retina/cytology , Retina/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...