Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
Curr Top Med Chem ; 13(19): 2484-90, 2013.
Article in English | MEDLINE | ID: mdl-24059335

ABSTRACT

An emerging common feature of the age-associated neurodegenerative disorders like Alzheimer's disease (AD) and Creutzfeldt-Jakob disease (CJD) is the ability of many disease-associated protein aggregates to induce conversion of a normal counterpart conformer leading to an acceleration of disease progression. Curative pharmacotherapy has not been achieved so far despite successes in elucidating pathomechanisms. Here, we review the pharmaceutical strategy of generating hybrid compounds, i.e. compounds consisting of several independently acting moieties with synergistic effects, on key molecular players in AD and CJD. For prion diseases, we review hybrid compounds consisting of two different heterocyclic compounds, their synergistic effects on prion replication in a cell culture model and their ability to prolong survival of experimentally prion-infected mice in vivo. While a combination therapy of several antiprion compounds including quinacrine, clomipramine, simvastatin and tocopherol prolonged survival time to 10-25%, administration of hybrid compound quinpramine alone, a chimera of acridine and iminodibenzyl scaffolds, led to 10% survival time extension. For AD, we review a hybrid compound consisting of an Aß recognizing D-peptide fused to a small molecule ß-sheet breaker, an aminopyrazole. This molecule was able to diminish Aß oligomers in cell culture and significantly decrease synaptotoxicity as measured by miniature excitatory postsynaptic responses in vitro. Hybrid compounds can dramatically increase potency of their single moieties and lead to novel functions when they act in a simultaneous or sequential manner thereby revealing synergistic properties. Their systematic generation combining different classes of compounds from peptides to small molecules has the potential to significantly accelerate drug discovery.


Subject(s)
Heterocyclic Compounds/pharmacology , Peptides/pharmacology , PrPSc Proteins/antagonists & inhibitors , Prion Diseases/drug therapy , Protein Folding/drug effects , Small Molecule Libraries/pharmacology , Animals , Drug Synergism , Heterocyclic Compounds/chemistry , Heterocyclic Compounds/therapeutic use , Humans , Models, Molecular , Molecular Structure , Peptides/chemistry , Peptides/therapeutic use , PrPSc Proteins/metabolism , Prion Diseases/diagnosis , Small Molecule Libraries/chemistry , Small Molecule Libraries/therapeutic use
2.
Neurosci Lett ; 469(1): 122-6, 2010 Jan 18.
Article in English | MEDLINE | ID: mdl-19945507

ABSTRACT

Beneficial effects of tocopherols, or vitamin E, on degenerative brain conditions have been attributed mainly to their antioxidant effects. Non-antioxidant effects of the tocopherols have been shown to be mediated by inhibition of protein kinase C (PKC) signaling. Prion disease is a paradigmatic protein conformational disease characterized by the induced conversion of a normal host protein PrP(C) to adopt a pathogenic conformation PrP(Sc). The molecular regulation of prion replication is poorly understood. Here, we show that tocopherols inhibit prion replication by a structure-activity relationship for antiprion activity independent of antioxidant activity with tocopherol succinate (TS) posessing highest EC(50) at 7 microM. Only TS but not an equally antiprion active PKC inhibitor could be partially antagonized by substochiometric 1 nM rapamycin suggesting that there are pathways via mammalian target of rapamycin (mTOR) that interfere with tocopherol's biological effects. Interaction with the mTOR pathway is a yet undescribed characteristic of tocopherol derivatives, potentially significant for pathophysiological processes other than prion propagation.


Subject(s)
Prions/antagonists & inhibitors , Tocopherols/chemistry , Tocopherols/pharmacology , Animals , Cell Death/drug effects , Cell Line, Tumor , Intracellular Signaling Peptides and Proteins/metabolism , Mice , Phosphorylation , Protein Serine-Threonine Kinases/metabolism , Ribosomal Protein S6 Kinases/metabolism , Structure-Activity Relationship , TOR Serine-Threonine Kinases , alpha-Tocopherol/chemistry , alpha-Tocopherol/pharmacology
3.
Exp Neurol ; 215(2): 397-400, 2009 Feb.
Article in English | MEDLINE | ID: mdl-18996373

ABSTRACT

Acridine-iminodibenzyl chimeric compounds were previously introduced as a class of cholesterol-redistributing substances with antiprion effects. Here, we show that administration of the lead compound quinpramine to mice with experimental autoimmune encephalitis, an animal model of multiple sclerosis (MS), significantly ameliorates disease in preventive and therapeutic paradigms. Quinpramine treatment decreased the number of inflammatory CNS lesions, antigen-specific T-cell proliferation, and pro-inflammatory cytokines IFNgamma and IL-17. Quinpramine is thus an immunoregulatory drug that is a candidate pharmaceutical for MS.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/drug therapy , Quinolinium Compounds/therapeutic use , Animals , Cell Proliferation/drug effects , Cytokines/metabolism , Disease Models, Animal , Encephalomyelitis, Autoimmune, Experimental/chemically induced , Female , Glycoproteins , Mice , Mice, Inbred C57BL , Myelin-Oligodendrocyte Glycoprotein , Peptide Fragments , T-Lymphocytes/drug effects
4.
J Med Chem ; 49(22): 6591-5, 2006 Nov 02.
Article in English | MEDLINE | ID: mdl-17064077

ABSTRACT

Human transmissible neurodegenerations including Creutzfeldt-Jakob disease are unique, since they are caused by prions, an infectious agent that replicates without nucleic acids but instead by inducing conversion of a host-resident normal prion protein to a misfolded conformational isoform. For pharmacotherapy of these unusual diseases, tricyclic heterocyclic compounds such as quinacrine have been considered, but with ambiguous success in vivo, so far. On the basis of the synergistic antiprion effects of quinacrine and iminodibenzyl derivatives, we introduce a novel class of potential pharmaceuticals representing structural chimeras of quinacrine and imipramine analogues. We describe the chemical synthesis and bioassays of a focused library of these compounds. The most potent target compound 2a revealed an EC(50) value of 20 nM determined with a cell model of prion disease, thus substantially improving the antiprion efficacy of quinacrine.


Subject(s)
Acridines/chemical synthesis , Acridines/pharmacology , Imipramine/analogs & derivatives , Imipramine/chemical synthesis , Prions/drug effects , Quinacrine/analogs & derivatives , Quinacrine/chemical synthesis , Animals , Blotting, Western , Cell Line, Tumor , Densitometry , Drug Design , Imipramine/pharmacology , Indicators and Reagents , Ligands , Mice , PrPSc Proteins/chemistry , PrPSc Proteins/drug effects , PrPSc Proteins/genetics , Quinacrine/pharmacology , Structure-Activity Relationship
5.
J Biol Chem ; 281(43): 32451-60, 2006 Oct 27.
Article in English | MEDLINE | ID: mdl-16905746

ABSTRACT

Saposin A (Sap-A) is one of five known sphingolipid activator proteins required for the lysosomal degradation of sphingolipids and for the loading of lipid antigens onto antigen-presenting molecules of the CD1 type. Sap-A assists in the degradation of galactosylceramide by galactosylceramide-beta-galactosidase in vivo, which takes place at the surface of intraendosomal/intralysosomal vesicles. Sap-A is believed to mediate the interaction between the enzyme and its membrane-bound substrate. Its dysfunction causes a variant form of Krabbe disease. In the present study we prepared glycosylated Sap-A free of other Saps, taking advantage of the Pichia pastoris expression system. Using liposomes and surface plasmon resonance spectroscopy, we tested the binding and lipid mobilization capacity of Sap-A under different conditions. Along the endocytic pathway, the pH value decreases, and the lipid composition of intraendosomal and intralysosomal membranes changes drastically. In the inner membranes the cholesterol concentration decreases, and that of the anionic phospholipid bis(monoacylglycero)phosphate increases. Here, we show that Sap-A is able to bind to liposomes and to mobilize lipids out of them at acidic pH values below pH 4.7. Low cholesterol levels and increasing concentrations of bis(monoacylglycero)phosphate favor lipid extraction significantly. Galactosylceramide as a bilayer component is not essential for lipid mobilization by Sap-A, which requires intact disulfide bridges for activity. We also show for the first time that glycosylation of Sap-A is essential for its lipid extraction activity. Variant Sap-A proteins, which cause storage of galactosylceramide in humans (Krabbe disease, Spiegel, R., Bach, G., Sury, V., Mengistu, G., Meidan, B., Shalev, S., Shneor, Y., Mandel, H., and Zeigler, M. (2005) Mol. Genet. Metab. 84, 160-166) and in mutant mice (Matsuda, J., Vanier, M. T., Saito, Y., Tohyama, J., and Suzuki, K. (2001) Hum. Mol. Genet. 10, 1191-1199) are deficient in lipid extraction capacity.


Subject(s)
Cholesterol/chemistry , Genetic Variation , Lipid Metabolism , Membrane Lipids/metabolism , Monoglycerides/chemistry , Saposins/metabolism , Glycosylation , Humans , Hydrogen-Ion Concentration , Lipid Bilayers/chemistry , Lipid Bilayers/metabolism , Liposomes/chemistry , Liposomes/metabolism , Membrane Lipids/chemistry , Pichia/genetics , Saposins/genetics , Saposins/isolation & purification , Surface Plasmon Resonance
6.
J Med Chem ; 49(17): 5300-8, 2006 Aug 24.
Article in English | MEDLINE | ID: mdl-16913719

ABSTRACT

Prion diseases are invariably fatal neurodegenerative diseases, in which the infectious agent consists of PrP(Sc), a pathogenic misfolded isoform of the normal cellular prion protein (PrP(C)). Until now, no pharmacological options exist for these novel pathogens. Here we describe the screening of a series of polyquinolines and quinolines linked to a large variety of terminal groups for their ability to cure a persistently prion infected cell line (ScN2a). Several compounds showed antiprion activity in the nanomolar range. The most active molecule, named 42, had a half-effective concentration (EC50) for antiprion activity of 50 nM. In a library of quinoline derivatives we were able to identify several structure-activity relationships (SAR). Remarkably, antiprion SAR in ScN2a cells were similar to antimalarial SAR in a cell model of malaria, particularly for the sulfonamide quinoline derivatives, suggesting that some molecular targets of antiprion and antimalarial substances overlap.


Subject(s)
Antimalarials/pharmacology , Prions/antagonists & inhibitors , Quinolines/pharmacology , Animals , Antimalarials/chemical synthesis , Antimalarials/chemistry , Cell Line, Tumor , Chloroquine/analogs & derivatives , Chloroquine/chemical synthesis , Chloroquine/pharmacology , Drug Evaluation, Preclinical , Mice , Molecular Structure , Quinolines/chemical synthesis , Quinolines/chemistry , Stereoisomerism , Structure-Activity Relationship
7.
J Neurochem ; 98(3): 748-59, 2006 Aug.
Article in English | MEDLINE | ID: mdl-16749906

ABSTRACT

Prion diseases are invariably fatal, neurodegenerative diseases transmitted by an infectious agent, PrPSc, a pathogenic, conformational isoform of the normal prion protein (PrPC). Heterocyclic compounds such as acridine derivatives like quinacrine abolish prion infectivity in a cell culture model of prion disease. Here, we report that these compounds execute their antiprion activity by redistributing cholesterol from the plasma membrane to intracellular compartments, thereby destabilizing membrane domains. Our findings are supported by the fact that structurally unrelated compounds with known cholesterol-redistributing effects - U18666A, amiodarone, and progesterone - also possessed high antiprion potency. We show that tricyclic antidepressants (e.g. desipramine), another class of heterocyclic compounds, displayed structure-dependent antiprion effects and enhanced the antiprion effects of quinacrine, allowing lower doses of both drugs to be used in combination. Treatment of ScN2a cells with quinacrine or desipramine induced different ultrastructural and morphological changes in endosomal compartments. We synthesized a novel drug from quinacrine and desipramine, termed quinpramine, that led to a fivefold increase in antiprion activity compared to quinacrine with an EC50 of 85 nm. Furthermore, simvastatin, an inhibitor of cholesterol biosynthesis, acted synergistically with both heterocyclic compounds to clear PrPSc. Our data suggest that a cocktail of drugs targeting the lipid metabolism that controls PrP conversion may be the most efficient in treating Creutzfeldt-Jakob disease.


Subject(s)
Antidepressive Agents, Tricyclic/pharmacology , Detergents/pharmacokinetics , Intracellular Membranes/drug effects , Intracellular Membranes/metabolism , Prions/antagonists & inhibitors , Prions/metabolism , Quinacrine/pharmacology , Amiodarone/pharmacology , Androstenes/pharmacology , Animals , Antidepressive Agents, Tricyclic/chemistry , Cell Line, Tumor , Cell Membrane/drug effects , Cell Membrane/genetics , Cell Membrane/metabolism , Chickens , Cholesterol/metabolism , Drug Combinations , Humans , Mice , Progesterone/pharmacology , Quinacrine/chemistry
8.
J Lipid Res ; 46(10): 2254-64, 2005 Oct.
Article in English | MEDLINE | ID: mdl-16061947

ABSTRACT

A mass spectrometric method is described for monitoring cerebrosides in the presence of excess concentrations of alkali metal salts. This method has been adapted for use in the assay of arylsulfatase A (ASA) and the cerebroside sulfate activator protein (CSAct or saposin B). Detection of the neutral glycosphingolipid cerebroside product was achieved via enhancement of ionization efficiency in the presence of lithium ions. Assay samples were extracted into the chloroform phase as for the existing assays, dried, and diluted in methanol-chloroform-containing lithium chloride. Samples were analyzed by electrospray ionization mass spectrometry with a triple quadrupole mass spectrometer in the multiple reaction monitoring tandem mass spectrometric mode. The assay has been used to demonstrate several previously unknown or ambiguous aspects of the coupled ASA/CSAct reaction, including an absolute in vitro preference for CSAct over the other saposins (A, C, and D) and a preference for the non-hydroxylated species of the sulfatide substrate over the corresponding hydroxylated species. The modified assay for the coupled ASA/CSAct reaction could find applicability in settings in which the assay could not be performed previously because of the need for radiolabeled substrate, which is now not required.


Subject(s)
Cerebroside-Sulfatase/analysis , Saposins/analysis , Spectrometry, Mass, Electrospray Ionization/methods , Animals , Cattle , Lithium/chemistry , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization
9.
Biol Chem ; 384(9): 1293-8, 2003 Sep.
Article in English | MEDLINE | ID: mdl-14515991

ABSTRACT

Acid sphingomyelinase (A-SMase, EC 3.1.4.12) catalyzes the lysosomal degradation of sphingomyelin to phosphorylcholine and ceramide. Inherited deficiencies of acid sphingomyelinase activity result in various clinical forms of Niemann-Pick disease, which are characterised by massive lysosomal accumulation of sphingomyelin. Sphingomyelin hydrolysis by both, acid sphingomyelinase and membrane-associated neutral sphingomyelinase, plays also an important role in cellular signaling systems regulating proliferation, apoptosis and differentiation. Here, we present a potent and selective novel inhibitor of A-SMase, L-alpha-phosphatidyl-D-myo-inositol-3,5-bisphosphate (PtdIns3,5P2), a naturally occurring substance detected in mammalian, plant and yeast cells. The inhibition constant Ki for the new A-SMase inhibitor PtdIns3,5P2 is 0.53 microM as determined in a micellar assay system with radiolabeled sphingomyelin as substrate and recombinant human A-SMase purified from insect cells. Even at concentrations of up to 50 microM, PtdIns3,5P2 neither decreased plasma membrane-associated, magnesium-dependent neutral sphingomyelinase activity, nor was it an inhibitor of the lysosomal hydrolases beta-hexosaminidase A and acid ceramidase. Other phosphoinositides tested had no or a much weaker effect on acid sphingomyelinase. Different inositol-bisphosphates were studied to elucidate structure-activity relationships for A-SMase inhibition. Our investigations provide an insight into the structural features required for selective, efficient inhibition of acid sphingomyelinase and may also be used as starting point for the development of new potent A-SMase inhibitors optimised for diverse applications.


Subject(s)
Phosphatidylinositol Phosphates/pharmacology , Sphingomyelin Phosphodiesterase/antagonists & inhibitors , Dose-Response Relationship, Drug , Humans , Kinetics , Magnesium , Membrane Proteins/drug effects , Phosphatidylinositol Phosphates/chemistry , Recombinant Proteins , Structure-Activity Relationship , Substrate Specificity
10.
Biochimie ; 85(3-4): 439-48, 2003.
Article in English | MEDLINE | ID: mdl-12770782

ABSTRACT

The physiological degradation of several membrane-bound glycosphingolipids (GSLs) by water-soluble lysosomal exohydrolases requires the assistance of sphingolipid activator proteins (SAPs). Four of these SAPs are synthesized from a single precursor protein (prosaposin). Inherited deficiency of this precursor results in a rare disease in humans with an accumulation of ceramide (Cer) and glycolipids such as glucosylceramide and lactosylceramide (LacCer). In a previous study, we have shown that human SAP-D stimulates the lysosomal degradation of Cer in precursor deficient cells. In order to study the role of SAPs (or saposins) A-D in cellular GSL catabolism, we recently investigated the catabolism of exogenously added [(3)H]labeled ganglioside GM1, Forssman lipid, and endogenously [(14)C]labeled GSLs in SAP-precursor deficient human fibroblasts after the addition of recombinant SAP-A, -B, -C and -D. We found that activator protein deficient cells are still able to slowly degrade gangliosides GM1 and GM3, Forssman lipid and globotriaosylceramide to a significant extent, while LacCer catabolism critically depends on the presence of SAPs. The addition of either of the SAPs, SAP-A, SAP-B or SAP-C, resulted in an efficient hydrolysis of LacCer.


Subject(s)
Glycoproteins/metabolism , Glycosphingolipids/metabolism , Base Sequence , Cells, Cultured , Endocytosis , Fibroblasts/metabolism , G(M1) Ganglioside/metabolism , G(M3) Ganglioside/metabolism , Globosides/metabolism , Glycoproteins/deficiency , Glycosphingolipids/chemistry , Humans , Molecular Sequence Data , Saposins , Sphingolipid Activator Proteins , Trihexosylceramides/metabolism
11.
J Mol Biol ; 328(3): 669-81, 2003 May 02.
Article in English | MEDLINE | ID: mdl-12706724

ABSTRACT

Human lysosomal beta-hexosaminidases are dimeric enzymes composed of alpha and beta-chains, encoded by the genes HEXA and HEXB. They occur in three isoforms, the homodimeric hexosaminidases B (betabeta) and S (alphaalpha), and the heterodimeric hexosaminidase A (alphabeta), where dimerization is required for catalytic activity. Allelic variations in the HEXA and HEXB genes cause the fatal inborn errors of metabolism Tay-Sachs disease and Sandhoff disease, respectively. Here, we present the crystal structure of a complex of human beta-hexosaminidase B with a transition state analogue inhibitor at 2.3A resolution (pdb 1o7a). On the basis of this structure and previous studies on related enzymes, a retaining double-displacement mechanism for glycosyl hydrolysis by beta-hexosaminidase B is proposed. In the dimer structure, which is derived from an analysis of crystal packing, most of the mutations causing late-onset Sandhoff disease reside near the dimer interface and are proposed to interfere with correct dimer formation. The structure reported here is a valid template also for the dimeric structures of beta-hexosaminidase A and S.


Subject(s)
Acetylglucosamine/analogs & derivatives , Models, Molecular , Sandhoff Disease/genetics , beta-N-Acetylhexosaminidases/chemistry , Acetylglucosamine/chemistry , Acetylglucosamine/metabolism , Acute Disease , Alleles , Bacteria/enzymology , Binding Sites , Catalysis , Chronic Disease , Crystallography, X-Ray , Dimerization , Hexosaminidase A , Hexosaminidase B , Hot Temperature , Humans , Mutation , Point Mutation , Protein Structure, Quaternary , RNA Splicing , Sequence Homology, Amino Acid , Substrate Specificity , beta-N-Acetylhexosaminidases/genetics , beta-N-Acetylhexosaminidases/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...