Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 3 de 3
Filter
Add more filters











Database
Language
Publication year range
1.
Drug Deliv ; 19(3): 149-54, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22353012

ABSTRACT

CONTEXT: Growth differentiation factor 5 (GDF5), in addition to its role in bone and joint development, protects dopaminergic (DA) neurons from degeneration, and is a potential therapeutic agent for Parkinson's disease. Its large size and insolubility at physiologic pH are obstacles for drug administration to the central nervous system (CNS) in humans. OBJECTIVE: In this study, formulations to deliver GDF5 to the brain using intranasal (IN) administration were developed. MATERIALS AND METHODS: IN administration of GDF5 in acidic buffer, 20 mM sodium acetate (NaAc) at pH 4.25, was performed in rats. Also, a lipid microemulsion (LME) comprised of olive oil and phosphatidylserine (PS) was used to formulate GDF5 at neutral pH for IN administration. Tissue concentrations of GDF5 were determined by both gamma counting and enzyme-linked immunosorbent assay (ELISA). RESULTS: IN administration of GDF5 in acidic buffers bypassed the blood-brain barrier (BBB), resulting in delivery to the brain with limited systemic exposure. IN administration of GDF5-LME increased drug targeting to the midbrain eightfold when compared to IN administration of GDF5 in acidic buffer. DISCUSSION AND CONCLUSION: This study is the first to show that GDF5 can be formulated at neutral pH and can be directly delivered to the CNS via IN administration, with biologically relevant concentrations in the midbrain where it may be used to treat Parkinson's disease.


Subject(s)
Drug Delivery Systems , Growth Differentiation Factor 5/administration & dosage , Growth Differentiation Factor 5/metabolism , Mesencephalon/metabolism , Administration, Intranasal , Animals , Drug Delivery Systems/methods , Humans , Male , Mesencephalon/drug effects , Rats , Rats, Sprague-Dawley
2.
J Pharmacol Exp Ther ; 330(3): 679-86, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19509317

ABSTRACT

Deferoxamine (DFO) is a high-affinity iron chelator approved by the Food and Drug Administration for treating iron overload. Preclinical research suggests that systemically administered DFO prevents and treats ischemic stroke damage and intracerebral hemorrhage. However, translation into human trials has been limited, probably because of difficulties with DFO administration. A noninvasive method of intranasal administration has emerged recently as a rapid way to bypass the blood-brain barrier and target therapeutic agents to the central nervous system. We report here that intranasal administration targets DFO to the brain and reduces systemic exposure, and that intranasal DFO prevents and treats stroke damage after middle cerebral artery occlusion (MCAO) in rats. A 6-mg dose of DFO resulted in significantly higher DFO concentrations in the brain (0.9-18.5 microM) at 30 min after intranasal administration than after intravenous administration (0.1-0.5 microM, p < 0.05). Relative to blood concentration, intranasal delivery increased targeting of DFO to the cortex approximately 200-fold compared with intravenous delivery. Intranasal administration of three 6-mg doses of DFO did not result in clinically significant changes in blood pressure or heart rate. Pretreatment with intranasal DFO (three 6-mg doses) 48 h before MCAO significantly decreased infarct volume by 55% versus control (p < 0.05). In addition, post-treatment with intranasal administration of DFO (six 6-mg doses) immediately after reperfusion significantly decreased infarct volume by 55% (p < 0.05). These experiments suggest that intranasally administered DFO may be a useful treatment for stroke, and a prophylactic for patients at high risk for stroke.


Subject(s)
Brain Ischemia/complications , Brain/metabolism , Deferoxamine/administration & dosage , Deferoxamine/pharmacology , Neuroprotective Agents/administration & dosage , Neuroprotective Agents/pharmacology , Stroke/etiology , Stroke/prevention & control , Administration, Intranasal , Animals , Behavior, Animal/drug effects , Deferoxamine/pharmacokinetics , Dose-Response Relationship, Drug , Infarction, Middle Cerebral Artery/pathology , Infarction, Middle Cerebral Artery/prevention & control , Iron Radioisotopes , Male , Middle Cerebral Artery/physiology , Neuroprotective Agents/pharmacokinetics , Rats , Rats, Sprague-Dawley , Reperfusion Injury/pathology , Reperfusion Injury/prevention & control , Tissue Distribution
3.
J Neuroimmune Pharmacol ; 2(1): 87-92, 2007 Mar.
Article in English | MEDLINE | ID: mdl-18040830

ABSTRACT

Intranasal (IN) delivery of HIV-1 Tat in aging mice was investigated as a possible model for HIV-1 infection in the brain. After IN administration, the distribution of [(125)I]-labeled Tat in the brains of Swiss Webster mice was evaluated by autoradiography and gamma counting. [(125)I]-labeled Tat was detected at the highest concentrations in the olfactory bulb, cervical nodes, and trigeminal nerve tract. In another experiment, APPSw transgenic mice were used to model chronic Tat exposure. The mice were treated intranasally with 6 mug Tat (n = 4) or vehicle (n = 4) three times per week for 4 weeks. Total RNA was isolated from the frontal cortex, and differential gene expression analysis was performed using gene microarrays. Gene ontology profiles indicated innate immunity, inflammatory and apoptotic responses. Five genes of interest in the Tat-treated mice that were significantly elevated in the microarrays were validated by RT-PCR. One gene, the Toll-like receptor 9 (Tlr9), has previously been shown to activate signaling cascades leading to innate immunity and enhanced HIV-1 gene expression. A second gene, Fas, plays a key role in neuroinflammation. Two cysteine-rich cytokines associated with chemotaxis were elevated: MCP-1 (Ccl2), which is chemotactic for monocytes, and Ccl17 (TARC), which is chemotactic for lymphocytes. Finally, the gene sestrin was significantly elevated and has been associated with oxidative stress, in particular amyloid beta-induced oxidative stress. This IN Tat model of neuroinflammation may be useful to study HIV-1-induced neurodegeneration.


Subject(s)
Brain/metabolism , Gene Expression Regulation, Viral/physiology , Gene Products, tat/administration & dosage , Gene Products, tat/physiology , Administration, Intranasal , Animals , Blood-Brain Barrier/physiology , Brain/virology , Gene Expression Profiling , Gene Products, tat/metabolism , HIV-1/physiology , Iodine Radioisotopes/metabolism , Male , Mice , Mice, Transgenic , Oligonucleotide Array Sequence Analysis , Tissue Distribution/genetics
SELECTION OF CITATIONS
SEARCH DETAIL