Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
Add more filters










Publication year range
1.
J Pharmacol Exp Ther ; 362(1): 119-130, 2017 07.
Article in English | MEDLINE | ID: mdl-28446518

ABSTRACT

Numerous studies suggest that the majority of amyloid-ß (Aß) peptides deposited in Alzheimer's disease (AD) are truncated and post-translationally modified at the N terminus. Among these modified species, pyroglutamyl-Aß (pE-Aß, including N3pE-Aß40/42 and N11pE-Aß40/42) has been identified as particularly neurotoxic. The N-terminal modification renders the peptide hydrophobic, accelerates formation of oligomers, and reduces degradation by peptidases, leading ultimately to the accumulation of the peptide and progression of AD. It has been shown that the formation of pyroglutamyl residues is catalyzed by glutaminyl cyclase (QC). Here, we present data about the pharmacological in vitro and in vivo efficacy of the QC inhibitor (S)-1-(1H-benzo[d]imidazol-5-yl)-5-(4-propoxyphenyl)imidazolidin-2-one (PQ912), the first-in-class compound that is in clinical development. PQ912 inhibits human, rat, and mouse QC activity, with Ki values ranging between 20 and 65 nM. Chronic oral treatment of hAPPSLxhQC double-transgenic mice with approximately 200 mg/kg/day via chow shows a significant reduction of pE-Aß levels and concomitant improvement of spatial learning in a Morris water maze test paradigm. This dose results in a brain and cerebrospinal fluid concentration of PQ912 which relates to a QC target occupancy of about 60%. Thus, we conclude that >50% inhibition of QC activity in the brain leads to robust treatment effects. Secondary pharmacology experiments in mice indicate a fairly large potency difference for Aß cyclization compared with cyclization of physiologic substrates, suggesting a robust therapeutic window in humans. This information constitutes an important translational guidance for predicting the therapeutic dose range in clinical studies with PQ912.


Subject(s)
Alzheimer Disease/drug therapy , Aminoacyltransferases/antagonists & inhibitors , Benzimidazoles/therapeutic use , Enzyme Inhibitors/therapeutic use , Imidazolines/therapeutic use , Nootropic Agents/therapeutic use , Alzheimer Disease/psychology , Amyloid beta-Peptides/chemistry , Amyloid beta-Peptides/drug effects , Animals , Benzimidazoles/cerebrospinal fluid , Benzimidazoles/pharmacokinetics , Binding Sites , Cyclization , Drug Delivery Systems , Enzyme Inhibitors/cerebrospinal fluid , Enzyme Inhibitors/pharmacokinetics , Female , HEK293 Cells , Humans , Imidazolines/cerebrospinal fluid , Imidazolines/pharmacokinetics , Male , Maze Learning/drug effects , Mice , Mice, Transgenic , Nootropic Agents/cerebrospinal fluid , Nootropic Agents/pharmacokinetics , Protein Binding , Rats , Spatial Learning/drug effects
2.
Sci Rep ; 6: 31441, 2016 08 11.
Article in English | MEDLINE | ID: mdl-27511630

ABSTRACT

Rheumatoid arthritis is characterised by synovial inflammation and proliferation of fibroblast-like synoviocytes. The induction of apoptosis has long been proposed as a target for proliferative autoimmune diseases, and has further been shown to act as a successful treatment of experimental models of arthritis, such as collagen-induced arthritis. Here we examined the effects of specific oral small-molecule inhibitors of the transcription regulating cyclin-dependent kinase 9 on the development and progression of collagen-induced arthritis. DBA/1 mice were immunised with bovine collagen type II and treated orally with specific CDK9 inhibitors. The effects of CDK9 inhibition on RNA levels and protein expression, apoptosis induction, caspase activation and lymphocyte phenotype were further analysed. Mice showed a significant delay in disease onset and a reduction in disease severity following treatment with CDK9 inhibitors. Inhibiting CDK9 activity in peripheral blood mononuclear cells resulted in the loss of Mcl-1 expression at both the protein and RNA levels, along with a subsequent increase in apoptosis. CDK9 specific inhibitors may be a potential alternative treatment not only of cancer, but also for autoimmune- and inflammatory diseases. Taken together, these results show that transient inhibition of CDK9 induces apoptosis in leukocyte subsets and modulates the immune response.


Subject(s)
Arthritis, Rheumatoid/drug therapy , Cyclin-Dependent Kinase 9/antagonists & inhibitors , Enzyme Inhibitors/administration & dosage , Immunologic Factors/administration & dosage , Administration, Oral , Animals , Apoptosis , Collagen Type II/administration & dosage , Disease Models, Animal , Gene Expression Profiling , Lymphocytes/physiology , Mice, Inbred DBA , Proteome/analysis , Treatment Outcome
3.
Neuropeptides ; 57: 21-34, 2016 Jun.
Article in English | MEDLINE | ID: mdl-26988064

ABSTRACT

BACKGROUND: Dipeptidyl peptidase 4 (DPP4; EC 3.4.14.5; CD26) is a membrane-bound or shedded serine protease that hydrolyzes dipeptides from the N-terminus of peptides with either proline or alanine at the penultimate position. Substrates of DPP4 include several stress-related neuropeptides implicated in anxiety, depression and schizophrenia. A decline of DPP4-like activity has been reported in sera from depressed patient, but not fully characterized regarding DPP4-like enzymes, therapeutic interventions and protein. METHODS: Sera from 16 melancholic- and 16 non-melancholic-depressed patients were evaluated for DPP4-like activities and the concentration of soluble DPP4 protein before and after treatment by anti-depressive therapies. Post-translational modification of DPP4-isoforms and degradation of NPY, Peptide YY (PYY), Galanin-like peptide (GALP), Orexin B (OrxB), OrxA, pituitary adenylate cyclase-activating polypeptide (PACAP) and substance P (SP) were studied in serum and in ex vivo human blood. N-terminal truncation of biotinylated NPY by endothelial membrane-bound DPP4 versus soluble DPP4 was determined in rat brain perfusates and spiked sera. RESULTS: Lower DPP4 activities in depressed patients were reversed by anti-depressive treatment. In sera, DPP4 contributed to more than 90% of the overall DPP4-like activity and correlated with its protein concentration. NPY displayed equal degradation in serum and blood, and was equally truncated by serum and endothelial DPP4. In addition, GALP and rat OrxB were identified as novel substrates of DPP4. CONCLUSION: NPY is the best DPP4-substrate in blood, being truncated by soluble and membrane DPP4, respectively. The decline of soluble DPP4 in acute depression could be reversed upon anti-depressive treatment. Peptidases from three functional compartments regulate the bioactivity of NPY in blood.


Subject(s)
Depressive Disorder/blood , Depressive Disorder/enzymology , Dipeptidyl Peptidase 4/blood , Neuropeptide Y/blood , Stress, Psychological/blood , Adult , Animals , Antidepressive Agents/therapeutic use , Depressive Disorder/drug therapy , Endothelium/metabolism , Female , Humans , Hydrolysis , Isoenzymes/blood , Male , Middle Aged , Orexins/blood , Pituitary Adenylate Cyclase-Activating Polypeptide/blood , Protein Processing, Post-Translational , Proteolysis , Rats , Substance P/blood
4.
Alzheimers Dement (N Y) ; 1(3): 182-195, 2015 Nov.
Article in English | MEDLINE | ID: mdl-29854937

ABSTRACT

INTRODUCTION: Pyroglutamate-amyloid-ß (pE-Aß) peptides are major components of Aß-oligomers and Aß-plaques, which are regarded as key culprits of Alzheimer's disease (AD) pathology. PQ912 is a competitive inhibitor of the enzyme glutaminyl cyclase (QC), essential for the formation of pE-Aß peptides. METHODS: A randomized, double-blind, placebo-controlled, single- and multiple-ascending oral dose study investigated the safety, pharmacokinetics, and pharmacodynamics of PQ912 in healthy nonelderly and elderly subjects. RESULTS: PQ912 was considered safe and well tolerated with dose-proportional pharmacokinetics up to doses of 200 mg. At higher doses up to 1800 mg, exposure was supraproportional and exposure in elderly subjects was approximately 1.5- to 2.1-fold higher. Exposure in cerebrospinal fluid (CSF) was approximately 20% of the unbound drug in plasma, and both serum and CSF QC activity was inhibited in a dose-related manner. DISCUSSION: This first-in-man study of a compound-targeting QC inhibition justifies further development of PQ912 for the treatment of AD.

5.
J Med Chem ; 56(17): 6613-25, 2013 Sep 12.
Article in English | MEDLINE | ID: mdl-23886302

ABSTRACT

Glutaminyl cyclase (hQC) has emerged as a new potential target for the treatment of Alzheimer's disease (AD). The inhibition of hQC prevents of the formation of the Aß3(pE)-40,42 species which were shown to be of elevated neurotoxicity and are likely to act as a seeding core, leading to an accelerated formation of Aß-oligomers and fibrils. This work presents a new class of inhibitors of hQC, resulting from a pharmacophore-based screen. Hit molecules were identified, containing benzimidazole as the metal binding group connected to 1,3,4-oxadiazole as the central scaffold. The subsequent optimization resulted in benzimidazolyl-1,3,4-thiadiazoles and -1,2,3-triazoles with an inhibitory potency in the nanomolar range. Further investigation into the potential binding mode of the new compound classes combined molecular docking and site directed mutagenesis studies.


Subject(s)
Aminoacyltransferases/antagonists & inhibitors , Benzimidazoles/chemistry , Benzimidazoles/pharmacology , Aminoacyltransferases/chemistry , Molecular Docking Simulation , Spectrometry, Mass, Electrospray Ionization , Structure-Activity Relationship
6.
Int J Exp Pathol ; 94(3): 217-25, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23560443

ABSTRACT

Inflammation is an integral part of non-alcoholic fatty liver disease (NAFLD), the most prevalent form of hepatic pathology found in the general population. In this context, recently we have examined the potential role of Glutaminyl Cyclases (QC and isoQC), and their inhibitors, in the maturation of chemokines, for example, monocyte chemoattractant protein 1 (MCP-1, CCL2), to generate their bioactive conformation. Catalysis by isoQC leads to the formation of an N-terminal pyroglutamate residue protecting CCL2 against degradation by aminopeptidases. This is of importance because truncated forms possess a reduced potential to attract immune cells. Since liver inflammation is characterized by the up-regulation of different chemokine pathways, and within this CCL2 is known to be a prominent example, we hypothesised that application of QC/isoQC inhibitors may alleviate liver inflammation by destabilizing CCL2. Therefore, we investigated the role of QC/isoQC inhibition, in comparison with the angiotensin receptor blocker Telmisartan, during development of pathology in a mouse model of non-alcoholic fatty liver disease. Application of a QC/isoQC inhibitor led to a significant reduction in circulating alanine aminotransferase and NAFLD activity score accompanied by an inhibitory effect on hepatocyte ballooning. Further analysis revealed a specific reduction of inflammation by decreasing the number of F4/80-positive macrophages, which is in agreement with the proposed CCL2-related mechanism of action of QC/isoQC inhibitors. Finally, QC/isoQC inhibitor application attenuated liver fibrosis as characterized by reduced collagen deposition in the liver parenchyma. Thus in conclusion, QC/isoQC inhibitors are a promising novel class of anti-non-alcoholic steatohepatitis drugs which have a comparable disease-modifying effect to that of Telmisartan, which is probably mediated via specific interference with a comparable monocyte/macrophage infiltration that occurs under inflammatory conditions.


Subject(s)
Aminoacyltransferases/antagonists & inhibitors , Chemokine CCL2/metabolism , Enzyme Inhibitors/pharmacology , Fatty Liver , Hepatitis , Aminoacyltransferases/metabolism , Angiotensin-Converting Enzyme Inhibitors/pharmacology , Animals , Benzimidazoles/pharmacology , Benzoates/pharmacology , Cell Line, Tumor , Chemokine CCL2/immunology , Disease Models, Animal , Fatty Liver/drug therapy , Fatty Liver/enzymology , Fatty Liver/immunology , Hepatitis/drug therapy , Hepatitis/enzymology , Hepatitis/immunology , Human Umbilical Vein Endothelial Cells , Humans , Leukemia, Monocytic, Acute/pathology , Macrophages/cytology , Mice , Mice, Inbred C57BL , Monocytes/cytology , Non-alcoholic Fatty Liver Disease , Telmisartan
7.
J Infect Dis ; 207(5): 768-77, 2013 Mar 01.
Article in English | MEDLINE | ID: mdl-23204180

ABSTRACT

BACKGROUND: Septic arthritis is a severe and rapidly debilitating disease mainly caused by Staphylococcus aureus. Here, we assess the antiarthritic efficiency of glutaminyl cyclase (QC) inhibitors. METHODS: Mice were inoculated with an arthritogenic amount of S. aureus intravenously or by local administration into the knee joint. Animals were treated with QC inhibitors (PBD155 and PQ529) via chow during the experiment. QC and isoQC knockout mice were also analyzed for arthritis symptoms after local administration of bacteria. RESULTS: Both QC inhibitors significantly delayed the onset of clinical signs of arthritis, and inhibitors significantly decreased weight loss in treated animals. Following intraarticular injection of S. aureus, PBD155-treated mice had lower levels of synovitis and bone erosion, as well as less myeloperoxidase in synovial tissue. Fluorescence-activated cell sorter analysis revealed that PBD155 treatment affected the expression pattern of adhesion molecules, preventing the upregulation of cells expressing CD11b/CD18. CONCLUSION: The compounds investigated here represent a novel class of small molecular antiarthritic inhibitors. In our studies, they exerted strong antiinflammatory actions, and therefore they might be suited for disease-modifying treatment of infectious arthritis.


Subject(s)
Aminoacyltransferases/antagonists & inhibitors , Arthritis, Infectious/drug therapy , Enzyme Inhibitors/therapeutic use , Staphylococcal Infections/drug therapy , Staphylococcus aureus/pathogenicity , Administration, Oral , Animals , Arthritis, Infectious/microbiology , Arthritis, Infectious/pathology , Disease Models, Animal , Mice , Mice, Knockout , Staphylococcal Infections/microbiology , Staphylococcal Infections/pathology , Treatment Outcome
8.
Chem Biol Drug Des ; 80(6): 937-46, 2012 Dec.
Article in English | MEDLINE | ID: mdl-22967026

ABSTRACT

Glutaminyl cyclases (QCs) catalyze the formation of pyroglutamate-modified amyloid peptides deposited in neurodegenerative disorders such as Alzheimer's disease. Inhibitors of QC are currently in development as potential therapeutics. The crystal structures of the potent inhibitor PBD150 bound to human and murine QC (hQC, mQC) have been described recently. The binding modes of a dimethoxyphenyl moiety of the inhibitor are significantly different between the structures, which contrasts with a similar K(i) value. We show the conformation of PBD150 prone to disturbance by protein-protein interactions within the crystals. Semi-empirical calculations of the enzyme-inhibitor interaction within the crystal suggest significant differences in the dissociation constants between the binding modes. To probe for interactions in solution, a site-directed mutagenesis on hQC was performed. The replacement of F325 and I303 by alanine or asparagine resulted in a 800-fold lower activity of the inhibitor, whereas the exchange of S323 by alanine or valine led to a 20-fold higher activity of PBD150. The results provide an example of deciphering the interaction mode between a target enzyme and lead substance in solution, if co-crystallization does not mirror such interactions properly. Thus, the study might provide implications for rapid screening of binding modes also for other drug targets.


Subject(s)
Aminoacyltransferases/antagonists & inhibitors , Enzyme Inhibitors/chemistry , Aminoacyltransferases/genetics , Aminoacyltransferases/metabolism , Animals , Binding Sites , Catalytic Domain , Circular Dichroism , Crystallography, X-Ray , Drug Design , Enzyme Inhibitors/chemical synthesis , Humans , Mice , Molecular Docking Simulation , Mutagenesis, Site-Directed , Recombinant Proteins/antagonists & inhibitors , Recombinant Proteins/genetics , Recombinant Proteins/metabolism
9.
EMBO Mol Med ; 3(9): 545-58, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21774078

ABSTRACT

Acute and chronic inflammatory disorders are characterized by detrimental cytokine and chemokine expression. Frequently, the chemotactic activity of cytokines depends on a modified N-terminus of the polypeptide. Among those, the N-terminus of monocyte chemoattractant protein 1 (CCL2 and MCP-1) is modified to a pyroglutamate (pE-) residue protecting against degradation in vivo. Here, we show that the N-terminal pE-formation depends on glutaminyl cyclase activity. The pE-residue increases stability against N-terminal degradation by aminopeptidases and improves receptor activation and signal transduction in vitro. Genetic ablation of the glutaminyl cyclase iso-enzymes QC (QPCT) or isoQC (QPCTL) revealed a major role of isoQC for pE(1) -CCL2 formation and monocyte infiltration. Consistently, administration of QC-inhibitors in inflammatory models, such as thioglycollate-induced peritonitis reduced monocyte infiltration. The pharmacologic efficacy of QC/isoQC-inhibition was assessed in accelerated atherosclerosis in ApoE3*Leiden mice, showing attenuated atherosclerotic pathology following chronic oral treatment. Current strategies targeting CCL2 are mainly based on antibodies or spiegelmers. The application of small, orally available inhibitors of glutaminyl cyclases represents an alternative therapeutic strategy to treat CCL2-driven disorders such as atherosclerosis/restenosis and fibrosis.


Subject(s)
Aminoacyltransferases/metabolism , Cell Movement , Chemokine CCL2/metabolism , Inflammation/immunology , Inflammation/pathology , Isoenzymes/metabolism , Monocytes/metabolism , Animals , Atherosclerosis/immunology , Atherosclerosis/pathology , Cell Line , Chemokine CCL2/antagonists & inhibitors , Female , Gene Silencing , Humans , Mice , Mice, Inbred C57BL , Monocytes/enzymology
10.
Biochemistry ; 50(28): 6280-8, 2011 Jul 19.
Article in English | MEDLINE | ID: mdl-21671571

ABSTRACT

Formation of N-terminal pyroglutamate (pGlu or pE) from glutaminyl or glutamyl precursors is catalyzed by glutaminyl cyclases (QC). As the formation of pGlu-amyloid has been linked with Alzheimer's disease, inhibitors of QCs are currently the subject of intense development. Here, we report three crystal structures of N-glycosylated mammalian QC from humans (hQC) and mice (mQC). Whereas the overall structures of the enzymes are similar to those reported previously, two surface loops in the neighborhood of the active center exhibit conformational variability. Furthermore, two conserved cysteine residues form a disulfide bond at the base of the active center that was not present in previous reports of hQC structure. Site-directed mutagenesis suggests a structure-stabilizing role of the disulfide bond. At the entrance to the active center, the conserved tryptophan residue, W(207), which displayed multiple orientations in previous structure, shows a single conformation in both glycosylated human and murine QCs. Although mutagenesis of W(207) into leucine or glutamine altered substrate conversion significantly, the binding constants of inhibitors such as the highly potent PQ50 (PBD150) were minimally affected. The crystal structure of PQ50 bound to the active center of murine QC reveals principal binding determinants provided by the catalytic zinc ion and a hydrophobic funnel. This study presents a first comparison of two mammalian QCs containing typical, conserved post-translational modifications.


Subject(s)
Aminoacyltransferases/chemistry , Aminoacyltransferases/metabolism , Amino Acid Sequence , Aminoacyltransferases/genetics , Animals , Cattle , Conserved Sequence , Crystallography, X-Ray , Enzyme Activation/genetics , Escherichia coli/enzymology , Escherichia coli/genetics , Glycosylation , Humans , Mice , Molecular Sequence Data , Pichia/enzymology , Pichia/genetics , Protein Conformation , Protein Processing, Post-Translational/genetics , Rats , Sheep
11.
J Med Chem ; 52(22): 7069-80, 2009 Nov 26.
Article in English | MEDLINE | ID: mdl-19863057

ABSTRACT

The inhibition of human glutaminyl cyclase (hQC) has come into focus as a new potential approach for the treatment of Alzheimer's disease. The hallmark of this principle is the prevention of the formation of Abeta(3,11(pE)-40,42), as these Abeta-species were shown to be of elevated neurotoxicity and likely to act as a seeding core leading to an accelerated formation of Abeta-oligomers and fibrils. Starting from 1-(3-(1H-imidazol-1-yl)propyl)-3-(3,4-dimethoxyphenyl)thiourea, bioisosteric replacements led to the development of new classes of inhibitors. The optimization of the metal-binding group was achieved by homology modeling and afforded a first insight into the probable binding mode of the inhibitors in the hQC active site. The efficacy assessment of the hQC inhibitors was performed in cell culture, directly monitoring the inhibition of Abeta(3,11(pE)-40,42) formation.


Subject(s)
Aminoacyltransferases/antagonists & inhibitors , Drug Design , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Aminoacyltransferases/chemistry , Amyloid beta-Peptides/antagonists & inhibitors , Amyloid beta-Peptides/chemistry , Amyloid beta-Peptides/metabolism , Catalytic Domain , Cell Line , Enzyme Inhibitors/chemical synthesis , Humans , Models, Molecular , Pyrrolidonecarboxylic Acid/metabolism , Structure-Activity Relationship , Thiourea/chemistry
12.
Nat Med ; 14(10): 1106-11, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18836460

ABSTRACT

Because of their abundance, resistance to proteolysis, rapid aggregation and neurotoxicity, N-terminally truncated and, in particular, pyroglutamate (pE)-modified Abeta peptides have been suggested as being important in the initiation of pathological cascades resulting in the development of Alzheimer's disease. We found that the N-terminal pE-formation is catalyzed by glutaminyl cyclase in vivo. Glutaminyl cyclase expression was upregulated in the cortices of individuals with Alzheimer's disease and correlated with the appearance of pE-modified Abeta. Oral application of a glutaminyl cyclase inhibitor resulted in reduced Abeta(3(pE)-42) burden in two different transgenic mouse models of Alzheimer's disease and in a new Drosophila model. Treatment of mice was accompanied by reductions in Abeta(x-40/42), diminished plaque formation and gliosis and improved performance in context memory and spatial learning tests. These observations are consistent with the hypothesis that Abeta(3(pE)-42) acts as a seed for Abeta aggregation by self-aggregation and co-aggregation with Abeta(1-40/42). Therefore, Abeta(3(pE)-40/42) peptides seem to represent Abeta forms with exceptional potency for disturbing neuronal function. The reduction of brain pE-Abeta by inhibition of glutaminyl cyclase offers a new therapeutic option for the treatment of Alzheimer's disease and provides implications for other amyloidoses, such as familial Danish dementia.


Subject(s)
Alzheimer Disease/drug therapy , Aminoacyltransferases/antagonists & inhibitors , Amyloid beta-Peptides/metabolism , Enzyme Inhibitors/therapeutic use , Pyrrolidonecarboxylic Acid/metabolism , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Aminoacyltransferases/physiology , Animals , Brain/enzymology , Cells, Cultured , Disease Models, Animal , Dose-Response Relationship, Drug , Enzyme Inhibitors/pharmacology , Female , Humans , Memory/drug effects , Mice , Mice, Transgenic
13.
J Neurochem ; 106(3): 1225-36, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18627432

ABSTRACT

Modified amyloid beta (Abeta) peptides represent major constituents of the amyloid deposits in Alzheimer's disease and Down's syndrome. In particular, N-terminal pyroglutamate (pGlu) following truncation renders Abeta more stable, increases hydrophobicity and the aggregation velocity. Recent evidence based on in vitro studies suggests that the cyclization of glutamic acid, leading to pGlu-Abeta, is catalyzed by the enzyme glutaminyl cyclase (QC) following limited proteolysis of Abeta at the N-terminus. Here, we studied the pGlu-formation by rat QC in vitro as well as after microinjection of Abeta(1-40) and Abeta(3-40) into the rat cortex in vivo/in situ with and without pharmacological QC inhibition. Significant pGlu-Abeta formation was observed following injection of Abeta(3-40) after 24 h, indicating a catalyzed process. The generation of pGlu-Abeta from Abeta(3-40) was significantly inhibited by intracortical microinjection of a QC inhibitor. The study provides first evidence that generation of pGlu-Abeta is a QC-catalyzed process in vivo. The approach per se offers a strategy for a rapid evaluation of compounds targeting a reduction of pGlu formation at the N-terminus of amyloid peptides.


Subject(s)
Aminoacyltransferases/antagonists & inhibitors , Amyloid beta-Peptides/metabolism , Cerebral Cortex/enzymology , Hippocampus/enzymology , Pyrrolidonecarboxylic Acid/metabolism , Aminoacyltransferases/chemistry , Aminoacyltransferases/genetics , Aminoacyltransferases/metabolism , Amyloid beta-Peptides/antagonists & inhibitors , Animals , Cell Line , Cerebral Cortex/drug effects , Cerebral Cortex/metabolism , Enzyme Inhibitors/administration & dosage , Female , Hippocampus/drug effects , Hippocampus/metabolism , Humans , Microinjections , Pyrrolidonecarboxylic Acid/antagonists & inhibitors , Rats , Rats, Sprague-Dawley , Sequence Homology, Amino Acid
14.
J Mol Biol ; 375(3): 708-19, 2008 Jan 18.
Article in English | MEDLINE | ID: mdl-18042490

ABSTRACT

A new inhibitor, H-Ala-Ile-pyrrolidin-2-yl boronic acid, was developed as an inhibitor against prolyl tripeptidyl aminopeptidase with a K(i) value of 88.1 nM. The structure of the prolyl tripeptidyl aminopeptidase complexed with the inhibitor (enzyme-inhibitor complex) was determined at 2.2 A resolution. The inhibitor was bound to the active site through a covalent bond between Ser603 and the boron atom of the inhibitor. This structure should closely mimic the structure of the reaction intermediate between the enzyme and substrate. We previously proposed that two glutamate residues, Glu205 and Glu636, are involved in the recognition of substrates. In order to clarify the function of these glutamate residues in substrate recognition, three mutant enzymes, E205A, E205Q, and E636A were generated by site-directed mutagenesis. The E205A mutant was expressed as an inclusion body. The E205Q mutant was expressed in soluble form, but no activity was detected. Here, the structures of the E636A mutant and its complex with the inhibitor were determined. The inhibitor was located at almost the same position as in the wild-type enzyme-inhibitor complex. The amino group of the inhibitor interacted with Glu205 and the main-chain carbonyl group of Gln203. In addition, a water molecule in the place of Glu636 of the wild-type enzyme interacted with the amino group of the inhibitor. This water molecule was located near the position of Glu636 in the wild-type and formed a hydrogen bond with Gln203. The k(cat)/K(M) values of the E636A mutant toward the two substrates used were smaller than those of the wild-type by two orders of magnitude. The K(i) value of our inhibitor for the E636A mutant was 48.8 microM, which was 554-fold higher than that against the wild-type enzyme. Consequently, it was concluded that Glu205 and Glu636 are significant residues for the N-terminal recognition of a substrate.


Subject(s)
Enzyme Inhibitors/metabolism , Porphyromonas gingivalis/enzymology , Amino Acid Sequence , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Binding Sites , Boron/metabolism , Catalytic Domain , Crystallography, X-Ray , Dimerization , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/chemistry , Glutamic Acid/genetics , Glutamic Acid/metabolism , Hydrogen Bonding , Hydrolysis , Hydrophobic and Hydrophilic Interactions , Kinetics , Models, Chemical , Models, Molecular , Molecular Sequence Data , Mutation , Porphyromonas gingivalis/chemistry , Protein Binding , Protein Structure, Secondary , Protein Structure, Tertiary , Serine/metabolism , Substrate Specificity , Water/chemistry , X-Ray Diffraction
15.
Peptides ; 28(2): 257-68, 2007 Feb.
Article in English | MEDLINE | ID: mdl-17223229

ABSTRACT

N-terminal truncation of NPY has important physiological consequences, because the truncated peptides lose their capability to activate the Y1-receptor. The sources of N-terminally truncated NPY and related peptides are unknown and several proline specific peptidases may be involved. First, we therefore provide an overview on the peptidases, belonging to structural and functional homologues of dipeptidyl peptidase 4 (DP4) as well as aminopeptidase P (APP) and thus, represent potential candidates of NPY cleavage in vivo. Second, applying selective inhibitors against DP4, DP8/9 and DP2, respectively, the enzymatic distribution was analyzed in brain extracts from wild type and DP4 deficient F344 rat substrains and human plasma samples in activity studies as well as by matrix assisted laser desorption/ionisation-time of flight (MALDI-TOF)-mass spectrometry. Third, co-transfection of Cos-1 cells with Dpp4 and Npy followed by confocal lasermicroscopy illustrated that hNPY-dsRed1-N1 was transported in large dense core vesicles towards the membrane while rDP4-GFP-C1 was transported primarily in different vesicles thereby providing no clear evidence for co-localization of NPY and DP4. Nevertheless, the review and experimental results of activity and mass spectrometry studies support the notion that at least five peptidases (DP4, DP8, DP9, XPNPEP1, XPNPEP2) are potentially involved in NPY cleavage while the serine protease DP4 (CD26) could be the principal peptidase involved in the N-terminal truncation of NPY. However, DP8 and DP9 are also capable of cleaving NPY, whereas no cleavage could be demonstrated for DP2.


Subject(s)
Dipeptidyl Peptidase 4/metabolism , Neuropeptide Y/metabolism , Animals , Base Sequence , COS Cells , Chlorocebus aethiops , DNA Primers , Hydrolysis , Microscopy, Confocal , Microscopy, Fluorescence , Rats , Rats, Inbred F344 , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization
16.
Biol Chem ; 388(2): 155-62, 2007 Feb.
Article in English | MEDLINE | ID: mdl-17261078

ABSTRACT

Mutations in the mouse ATRN gene, which encodes attractin, offer links between this protein and pigmentation, metabolism, immune status and neurodegeneration. However, the mechanisms of attractin action are not understood. The protein was first identified in humans in a circulating form in serum. A protease activity was postulated similar to the membrane-bound ectoenzyme DP4/CD26. In the last decade, both DP4/CD26 and attractin were controversially described to be the major source of human serum DP4 activity. We purified attractin from human plasma, and found that the DP4-like activity of the preparation shows nearly identical kinetic properties to that of recombinant human DP4. In contrast, the native electrophoretic behavior of this activity is clearly different from human and porcine DP4, but co-migrates with the protein band identified as attractin by Western blotting and N-terminal sequencing. Nevertheless, a DP4 impurity could be demonstrated in purified plasma attractin and the activity could be removed by ADA affinity chromatography, resulting in a homogenous attractin preparation without DP4 activity. These results are substantiated by expression of different attractin isoforms, in which no DP4 activity was found either. This indicates that the multidomain protein attractin acts as a receptor or adhesion protein rather than a protease.


Subject(s)
Adenosine Deaminase/physiology , Dipeptidyl Peptidase 4/physiology , Glycoproteins/physiology , Membrane Proteins/physiology , Adenosine Deaminase/analysis , Adenosine Deaminase/blood , Blotting, Western , Cell Line , Chromatography, Affinity/methods , Cloning, Molecular , Dipeptidyl Peptidase 4/analysis , Dipeptidyl Peptidase 4/blood , Electrophoresis, Polyacrylamide Gel , Enzyme Activation/physiology , Glycoproteins/analysis , Glycoproteins/blood , Humans , Isoenzymes/analysis , Isoenzymes/blood , Isoenzymes/physiology , Membrane Proteins/blood , Membrane Proteins/isolation & purification , Recombinant Proteins/blood , Recombinant Proteins/isolation & purification , Recombinant Proteins/metabolism , Reference Values , Sensitivity and Specificity
17.
Biochim Biophys Acta ; 1764(10): 1618-25, 2006 Oct.
Article in English | MEDLINE | ID: mdl-17005457

ABSTRACT

Mammalian cell lines were examined concerning their Glutaminyl Cyclase (QC) activity using a HPLC method. The enzyme activity was suppressed by a QC specific inhibitor in all homogenates. Aim of the study was to prove whether inhibition of QC modifies the posttranslational maturation of N-glutamine and N-glutamate peptide substrates. Therefore, the impact of QC-inhibition on amino-terminal pyroglutamate (pGlu) formation of the modified amyloid peptides Abeta(N3E-42) and Abeta(N3Q-42) was investigated. These amyloid-beta peptides were expressed as fusion proteins with either the pre-pro sequence of TRH, to be released by a prohormone convertase, or as engineered amyloid precursor protein for subsequent liberation of Abeta(N3Q-42) after beta- and gamma-secretase cleavage during posttranslational processing. Inhibition of QC leads in both expression systems to significantly reduced pGlu-formation of differently processed Abeta-peptides. This reveals the importance of QC-activity during cellular maturation of pGlu-containing peptides. Thus, QC-inhibition should impact bioactivity, stability or even toxicity of pyroglutamyl peptides preventing glutamine and glutamate cyclization.


Subject(s)
Aminoacyltransferases/antagonists & inhibitors , Amyloid beta-Peptides/metabolism , Peptide Fragments/metabolism , Pyrrolidonecarboxylic Acid/metabolism , Aminoacyltransferases/analysis , Amyloid beta-Peptides/chemistry , Amyloid beta-Peptides/genetics , Animals , Cell Line , Chromatography, High Pressure Liquid , Humans , Mice , Peptide Fragments/chemistry , Peptide Fragments/genetics , Pyrrolidonecarboxylic Acid/analysis
18.
J Med Chem ; 49(2): 664-77, 2006 Jan 26.
Article in English | MEDLINE | ID: mdl-16420052

ABSTRACT

The first effective inhibitors for human glutaminyl cyclase (QC) are described. The structures are developed by applying a ligand-based optimization approach starting from imidazole. Screening of derivatives of that heterocycle led to compounds of the imidazol-1-yl-alkyl thiourea type as a lead scaffold. A library of thiourea derivatives was synthesized, resulting in an inhibitory improvement by 2 orders of magnitude, leading to 1-(3-(1H-imidazol-1-yl)propyl)-3-(3,4-dimethoxyphenyl)thiourea as a potent inhibitor. Systematic exploitation of the scaffold revealed a strong impact on the inhibitory efficacy and resulted in the development of imidazole-propyl-thioamides as another new class of potent inhibitors. A flexible alignment of the most potent compounds of the thioamide and thiourea class and a QC substrate revealed a good match of characteristic features of the molecules, which suggests a similar binding mode of both inhibitors and the substrate to the active site of QC.


Subject(s)
Aminoacyltransferases/antagonists & inhibitors , Imidazoles/chemical synthesis , Thioamides/chemical synthesis , Thiourea/analogs & derivatives , Aminoacyltransferases/chemistry , Chelating Agents/chemical synthesis , Chelating Agents/chemistry , Humans , Imidazoles/chemistry , Models, Molecular , Structure-Activity Relationship , Thioamides/chemistry , Thiourea/chemical synthesis , Thiourea/chemistry , Zinc/chemistry
19.
J Mol Biol ; 355(4): 768-83, 2006 Jan 27.
Article in English | MEDLINE | ID: mdl-16330047

ABSTRACT

Dipeptidyl peptidase IV (DPIV) is an alpha,beta-hydrolase-like serine exopeptidase, which removes dipeptides, preferentially with a C-terminal l-Pro residue, from the N terminus of longer peptide substrates. Previously, we determined the tetrameric 1.8A crystal structure of native porcine DPIV. Each monomer is composed of a beta-propeller and a catalytic domain, which together embrace an internal cavity housing the active centre. This cavity is connected to the bulk solvent by a "propeller opening" and a "side opening". Here, we analyse DPIV complexes with a t-butyl-Gly-Pro-Ile tripeptide, Pro-boroPro, a piperazine purine compound, and aminoethyl phenyl sulfonylfluoride. The latter two compounds bind to the active-site groove in a compact and a quite bulky manner, respectively, causing considerable shifts of the catalytic Ser630 side-chain and of the Tyr547 phenolic group, which forms the oxyanion hole. The tripeptide, mimicking a peptide substrate, is clamped to the active site through tight interactions via its N-terminal alpha-ammonium group, the P2 carbonyl group, the P1-l-Pro side-chain, the C-terminal carboxylate group, and the stable orthoacid ester amide formed between the scissile peptide carbonyl group and Ser630 O(gamma). This stable trapping of the tripeptide could be due to stabilization of the protonated His740 imidazolium cation by the adjacent negatively charged C-terminal carboxylate group, preventing proton transfer to the leaving group nitrogen atom. Docking experiments with the compact rigid 58 residue protein aprotinin, which had been shown to be processed by DPIV, indicate that the Arg1-Pro2 N terminus can access the DPIV active site only upon widening of its side openings, probably by separation of the first and the last propeller blades, and/or of the catalytic and the propeller domain.


Subject(s)
Dipeptidyl Peptidase 4/chemistry , Dipeptidyl Peptidase 4/metabolism , Animals , Crystallography, X-Ray , Models, Molecular , Peptides/chemistry , Peptides/metabolism , Pliability , Protease Inhibitors/chemistry , Protease Inhibitors/metabolism , Protein Structure, Quaternary , Protein Structure, Tertiary , Structural Homology, Protein , Sulfones/chemistry , Sulfones/metabolism , Swine
20.
Biochemistry ; 44(40): 13415-24, 2005 Oct 11.
Article in English | MEDLINE | ID: mdl-16201766

ABSTRACT

Murine glutaminyl cyclase (mQC) was identified in the insulinoma cell line beta-TC 3 by determination of enzymatic activity and RT-PCR. The cloned cDNA was expressed in the secretory pathway of the methylotrophic yeast Pichia pastoris and purified after fermentation using a new three-step protocol. mQC converted a set of various substrates with very similar specificity to human QC, indicating a virtually identical catalytic competence. Furthermore, mQC was competitively inhibited by imidazole derivatives. A screen of thiol reagents revealed cysteamine as a competitive inhibitor of mQC bearing a Ki value of 42 +/-2 microM. Substitution of the thiol or the amino group resulted in a drastic loss of inhibitory potency. The pH dependence of catalysis and inhibition support that an uncharged nitrogen of the inhibitors and the substrate is necessary in order to bind to the active site of the enzyme. In contrast to imidazole and cysteamine, the heterocyclic chelators 1,10-phenanthroline, 2,6-dipicolinic acid, and 8-hydroxyquinoline inactivated mQC in a time-dependent manner. In addition, citric acid inactivated the enzyme at pH 5.5. Inhibition by citrate was abolished in the presence of zinc ions. A determination of the metal content by total reflection X-ray fluorescence spectrometry and atomic absorption spectroscopy in mQC revealed stoichiometric amounts of zinc bound to the protein. Metal ion depletion appeared to have no significant effect on protein structure as shown by fluorescence spectroscopy, suggesting a catalytic role of zinc. The results demonstrate that mQC and probably all animal QCs are zinc-dependent catalysts. Apparently, during evolution from an ancestral protease, a switch occurred in the catalytic mechanism which is mainly based on a loss of one metal binding site.


Subject(s)
Aminoacyltransferases/chemistry , Zinc/chemistry , Amino Acid Sequence , Animals , Binding Sites , Catalysis , Cattle , Citric Acid/pharmacology , Cloning, Molecular , DNA, Complementary/metabolism , Dose-Response Relationship, Drug , Electrophoresis, Polyacrylamide Gel , Escherichia coli , Evolution, Molecular , Humans , Hydrogen-Ion Concentration , Imidazoles/chemistry , Kinetics , Mice , Models, Chemical , Molecular Sequence Data , Nitrogen/chemistry , Photons , Pichia/metabolism , Recombinant Proteins/chemistry , Reverse Transcriptase Polymerase Chain Reaction , Sequence Homology, Amino Acid , Spectrometry, Fluorescence , Spectrophotometry , Substrate Specificity , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...