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.
J Neurosci ; 34(9): 3122-9, 2014 Feb 26.
Article in English | MEDLINE | ID: mdl-24573272

ABSTRACT

Enzyme replacement therapy (ERT) is a treatment option for lysosomal storage disorders (LSDs) caused by deficiencies of soluble lysosomal enzymes. ERT depends on receptor-mediated transport of intravenously injected recombinant enzyme to lysosomes of patient cells. The blood-brain barrier (BBB) prevents efficient transfer of therapeutic polypeptides from the blood to the brain parenchyma and thus hinders effective treatment of LSDs with CNS involvement. We compared the potential of five brain-targeting peptides to promote brain delivery of the lysosomal enzyme arylsulfatase A (ASA). Fusion proteins between ASA and the protein transduction domain of the human immunodeficiency virus TAT protein (Tat), an Angiopep peptide (Ang-2), and the receptor-binding domains of human apolipoprotein B (ApoB) and ApoE (two versions, ApoE-I and ApoE-II) were generated. All ASA fusion proteins were enzymatically active and targeted to lysosomes when added to cultured cells. In contrast to wild-type ASA, which is taken up by mannose-6-phosphate receptors, all chimeric proteins were additionally endocytosed via mannose-6-phosphate-independent routes. For ASA-Ang-2, ASA-ApoE-I, and ASA-ApoE-II, uptake was partially due to the low-density lipoprotein receptor-related protein 1. Transendothelial transfer in a BBB cell culture model was elevated for ASA-ApoB, ASA-ApoE-I, and ASA-ApoE-II. Brain delivery was, however, increased only for ASA-ApoE-II. ApoE-II was also superior to wild-type ASA in reducing lysosomal storage in the CNS of ASA-knock-out mice treated by ERT. Therefore, the ApoE-derived peptide appears useful to treat metachromatic leukodystrophy and possibly other neurological disorders more efficiently.


Subject(s)
Blood-Brain Barrier/metabolism , Brain/metabolism , Cerebroside-Sulfatase/administration & dosage , Genetic Vectors/physiology , Peptides/metabolism , Animals , Apolipoproteins E/genetics , Blood-Brain Barrier/drug effects , Brain/cytology , Cells, Cultured , Cerebroside-Sulfatase/deficiency , Cerebroside-Sulfatase/genetics , Cricetulus , Culture Media, Conditioned/pharmacology , Disease Models, Animal , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Female , Humans , Leukodystrophy, Metachromatic/drug therapy , Leukodystrophy, Metachromatic/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Receptor, IGF Type 2/genetics , Receptor, IGF Type 2/metabolism , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism
2.
J Biol Chem ; 286(20): 17487-94, 2011 May 20.
Article in English | MEDLINE | ID: mdl-21454621

ABSTRACT

Enzyme replacement therapy is an option to treat lysosomal storage diseases caused by functional deficiencies of lysosomal hydrolases as intravenous injection of therapeutic enzymes can correct the catabolic defect within many organ systems. However, beneficial effects on central nervous system manifestations are very limited because the blood-brain barrier (BBB) prevents the transfer of enzyme from the circulation to the brain parenchyma. Preclinical studies in mouse models of metachromatic leukodystrophy, however, showed that arylsulfatase A (ASA) is able to cross the BBB to some extent, thus reducing lysosomal storage in brain microglial cells. The present study aims to investigate the routing of ASA across the BBB and to improve the transfer in vitro using a well established cell culture model consisting of primary porcine brain capillary endothelial cells cultured on Transwell filter inserts. Passive apical-to-basolateral ASA transfer was observed, which was not saturable up to high ASA concentrations. No active transport could be determined. The passive transendothelial transfer was, however, charge-dependent as reduced concentrations of negatively charged monosaccharides in the N-glycans of ASA or the addition of polycations increased basolateral ASA levels. Adsorptive transcytosis is therefore considered to be the major transport pathway. Partial inhibition of the transcellular ASA transfer by mannose 6-phosphate indicated a second route depending on the insulin-like growth factor II/mannose 6-phosphate receptor, MPR300. We conclude that cationization of ASA and an increase of the mannose 6-phosphate content of the enzyme may promote blood-to-brain transfer of ASA, thus leading to an improved therapeutic efficacy of enzyme replacement therapy behind the BBB.


Subject(s)
Blood-Brain Barrier/metabolism , Cerebroside-Sulfatase/metabolism , Endothelial Cells/metabolism , Mannosephosphates/metabolism , Animals , Blood-Brain Barrier/pathology , Cations/metabolism , Cerebroside-Sulfatase/therapeutic use , Disease Models, Animal , Endothelial Cells/pathology , Enzyme Replacement Therapy/methods , Humans , Leukodystrophy, Metachromatic/drug therapy , Leukodystrophy, Metachromatic/enzymology , Mice , Protein Transport/drug effects , Swine
3.
J Neurochem ; 111(1): 111-8, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19656257

ABSTRACT

ATP-driven efflux transport proteins at the blood-brain barrier protect the healthy brain but impede pharmacotherapy of the disordered CNS. To investigate the question how ATP-binding cassette (ABC)-transporters are regulated during inflammation or infection we analysed the effects of the cytokines tumour necrosis factor-alpha (TNF-alpha) and interleukin-1beta (IL-1beta) on the expression of brain multidrug resistance proteins in primary cultures of porcine brain capillary endothelial cells. We found that TNF-alpha and IL-1beta rapidly decrease Abcg2 (BMDP/BCRP) mRNA expression within 6 h. After 24 and 48 h the mRNA level came back to control values. The mRNA reduction at 6 h was counter-regulated by the anti-inflammatory glucocorticoid hydrocortisone. Abcg2 protein levels were suppressed at prolonged stimulations but not after 6 h of stimulation which correlates with Abcg2 specific substrate uptake measurements. Abcb1 (p-glycoprotein) protein expression was transiently increased after TNF-alpha addition within 6 h of incubation followed by a reduction after 24 and 48 h whereas the Abcb1 mRNA levels were not changed. IL-1beta caused a continuous decrease in protein expression of both ABC-transporters. Long-term treatment with an assumed TNF-alpha-downstream agent, the vasoconstrictor endothelin-1, induced Abcg2 protein expression but suppressed Abcb1. Other efflux pumps like multidrug resistance-associated proteins/Abcc were rarely affected. The present results imply a complex regulation of the two most abundant ABC-transporters at the blood-brain barrier during early inflammation stages suggesting that Abcb1 (p-glycoprotein) is an early target of TNF-alpha-signalling counterbalanced by Abcg2.


Subject(s)
ATP-Binding Cassette Transporters/metabolism , Blood-Brain Barrier/physiology , Endothelial Cells/physiology , Gene Expression Regulation/physiology , Inflammation/pathology , ATP-Binding Cassette Transporters/classification , ATP-Binding Cassette Transporters/genetics , Analysis of Variance , Animals , Anti-Inflammatory Agents/pharmacology , Brain/cytology , Cells, Cultured , Electric Impedance , Endothelial Cells/drug effects , Endothelin-1/adverse effects , Gene Expression Regulation/drug effects , Hydrocortisone/pharmacology , Inflammation/chemically induced , Inflammation/drug therapy , Interleukin-1beta/adverse effects , Swine , Time Factors , Tumor Necrosis Factor-alpha/adverse effects
SELECTION OF CITATIONS
SEARCH DETAIL
...