Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 45
Filter
Add more filters










Publication year range
1.
Scand J Pain ; 14: 25-38, 2017 01.
Article in English | MEDLINE | ID: mdl-28850427

ABSTRACT

BACKGROUND: The first line pharmacological treatment of cancer pain is morphine and surrogates but a significant pain relief and a reduction of the side-effects of these compounds makes it necessary to combine them with other drugs acting on different targets. The aim of this study was to measure the antinociceptive effect on cancer-induced bone pain resulting from the association of the endogenous opioids enkephalin and non-opioid analgesic drugs. For this purpose, PL265 a new orally active single dual inhibitor of the two degrading enkephalins enzymes, neprilysin (NEP) and aminopeptidase N (APN) was used. It strictly increased the levels of enkephalin at their sites of releases. The selected non-opioid compounds are: gabapentin, A-317491 (P2X3 receptor antagonist), ACEA (CB1 receptor antagonist), AM1241 (CB2 receptor antagonist), JWH-133 (CB2 receptor antagonist), URB937 (FAAH inhibitor), and NAV26 (Nav1.7 channel blocker). METHODS: Experiments. Experiments were performed in 5-6 weeks old (26-33g weight) C57BL/6 mice. Cell culture and cell inoculation. B16-F10 melanoma cells were cultured and when preconfluent, treated and detached. Finally related cells were resuspended to obtain a concentration of 2×106 cells/100µL. Then 105 cells were injected into the right tibial medullar cavity. Control mice were treated by killed cells by freezing. Behavioural studies. Thermal withdrawal latencies were measured on a unilatered hot plate (UHP) maintained at 49±0.2°C. Mechanical threshold values were obtained by performing the von Frey test using the "up and down" method. To evaluate the nature (additive or synergistic) of the interactions between PL265 and different drugs, an isobolographic analysis following the method described by Tallarida was performed. RESULTS: The results demonstrate the ability of PL265, a DENKI that prevents the degradation of endogenous ENKs, to counteract cancer-induced bone thermal hyperalgesia in mice, by exclusively stimulating peripheral opioid receptors as demonstrated by used of an opioid antagonist unable to enter the brain. The development of such DENKIs, endowed with druggable pharmacokinetic characteristics, such as good absorption by oral route, can be considered as an important step in the development of much needed novel antihyperalgesic drugs. Furthermore, all the tested combinations resulted in synergistic antihyperalgesic effects. As shown here, the greatest synergistic antinociceptive effect (doses could be lowered by 70%) was produced by the combination of PL265 with the P2X3 receptor antagonist (A-317491), cannabinoid CB1 receptor agonist (exogenous, ACEA and endogenous URB937-protected-AEA) and Nav1.7 blocker (NAV26) whose mechanism of action involves the direct activation of the enkephalinergic system. CONCLUSIONS: These multi-target-based antinociceptive strategies using combinations of non-opioid drugs with dual inhibitors of enkephalin degrading enzymes may bring therapeutic advantages in terms of efficacy and safety by allowing the reduction of doses of one of the compounds or of both, which is of the utmost interest in the chronic treatment of cancer pain. IMPLICATIONS: This article presents synergistic antinociceptive effect produced by the combination of PL265 with non-opioid analgesic drugs acting via unrelated mechanisms. These multi-target-based antinociceptive strategies may bring therapeutic advantages by allowing the reduction of doses, which is of great interest in the chronic treatment of cancer pain.


Subject(s)
Analgesics/pharmacology , Bone and Bones/drug effects , Cancer Pain/drug therapy , Neprilysin/antagonists & inhibitors , Propionates/pharmacology , Administration, Oral , Animals , Bone and Bones/physiopathology , Cancer Pain/physiopathology , Cell Line, Tumor , Cell Survival/drug effects , Dose-Response Relationship, Drug , Drug Synergism , Drug Therapy, Combination , Enkephalins/metabolism , Hyperalgesia/drug therapy , Hyperalgesia/physiopathology , Mice, Inbred C57BL , Morphine/administration & dosage , Neoplasm Transplantation , Neprilysin/metabolism , Random Allocation
2.
Bioorg Med Chem Lett ; 27(16): 3883-3890, 2017 08 15.
Article in English | MEDLINE | ID: mdl-28676269

ABSTRACT

New neprilysin inhibitors containing an α-mercaptoketone HSC(R1R2)CO group, as zinc ligand were designed. Two parameters were explored for potency optimization: the size of the inhibitor which could interact with the S1, S1' or S2' domain of the enzyme and the nature of the substituents R1, R2 of the mercaptoketone group. Introduction of a cyclohexyl chain in R1, R2 position and a (3-thiophen)benzyl group in position R3 (compound 12n) yielded to the most potent inhibitor of this series with a Ki value of 2±0.3nM. This result suggests that this new inhibitor interacts within the S1, S1' domain of NEP allowing a pentacoordination of the catalytic Zn2+ ion by the mercaptoketone moiety.


Subject(s)
Enzyme Inhibitors/pharmacology , Ketones/pharmacology , Neprilysin/antagonists & inhibitors , Dose-Response Relationship, Drug , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/chemistry , Humans , Ketones/chemical synthesis , Ketones/chemistry , Molecular Structure , Neprilysin/metabolism , Structure-Activity Relationship
3.
Eur J Pharmacol ; 788: 176-182, 2016 Oct 05.
Article in English | MEDLINE | ID: mdl-27266667

ABSTRACT

Neuropathic pain remains difficult to treat due to the involvement of various pathophysiological mechanisms in its pathogeny. Among the different opioidergic systems the enkephalinergic one is primarily recruited via activation of delta opioid receptor (DOP) in chronic pain and of mu opioid receptor (MOP) in acute pain. To investigate the role of their endogenous ligands Met and Leu-enkephalin in neuropathic pain control, a dual inhibitor of their degrading enzymes, PL265, which acts restrictively at the level of peripheral nociceptors, was administered per os to assess its efficacy in pain prevention and alleviation using a partial sciatic nerve ligation model (PSNL) in mice. We demonstrated here that the pre-injury oral administration of PL265 (50mg/kg) during the 9 days of neuropathy development reduces thermal hyperalgesia and mechanical allodynia for two weeks after the end of treatment. The repeated administration (50mg/kg daily, during 10 days) does not induce tolerance. Therefore, protecting the enkephalins released at the peripheral level during neuropathic pain with oral PL265 seems to be a promising approach to prevent and alleviate the painful symptoms of neuropathic pain in humans without the unwanted effects of exogenous opiates such as morphine.


Subject(s)
Alanine/analogs & derivatives , Biphenyl Compounds/pharmacology , Neprilysin/antagonists & inhibitors , Neuralgia/diet therapy , Neuralgia/prevention & control , Protease Inhibitors/pharmacology , Alanine/administration & dosage , Alanine/adverse effects , Alanine/pharmacology , Alanine/therapeutic use , Animals , Biphenyl Compounds/administration & dosage , Biphenyl Compounds/adverse effects , Biphenyl Compounds/therapeutic use , Body Weight/drug effects , Disease Models, Animal , Hyperalgesia/prevention & control , Male , Mice , Neuralgia/enzymology , Protease Inhibitors/administration & dosage , Protease Inhibitors/adverse effects , Protease Inhibitors/therapeutic use , Safety , Time Factors , Touch/drug effects
4.
Eur J Med Chem ; 102: 58-67, 2015 Sep 18.
Article in English | MEDLINE | ID: mdl-26241877

ABSTRACT

The endogenous opioid system, essentially constituted by two opioid receptors which are stimulated by the natural internal effectors enkephalins (Met-enkephalin and Leu-enkephalin), is present at the different sites (peripheral, spinal, central) of the control of pain. We have demonstrated that the protection of the enkephalin inactivation by the two metallopeptidases (neprilysin and neutral aminopeptidase) increases their local concentration selectively induced by pain stimuli triggering analgesic responses. With the aim of increasing the orally antinociceptive responses of the previously described disulfide DENKIs ( [Formula: see text] CH(R1)CH2-S-S-CH2-C(R2R3)CONHCH(R4)COOR5), we designed new pro-drugs, in the same chemical series, with a transient protection of the free amino group by an acyloxyalkyl carbamate, giving rise to ((CH3)2CHCO2CH(CH3)OCONHCH(R1)CH2-S-S-CH2-C(R2R3)CONHCH(R4)COOR5) pro-drugs 2a-2g. These compounds were easily prepared from their parent analogs, with a good yield. They were tested per os and shown to be highly efficient in peripherally-controlled inflammatory and neuropathic pain with long lasting effects but completely inactive in the acute centrally-controlled hot plate test, a model of pain by excess of nociception. This demonstrates that DENKIs are able to relieve pain at its source thanks to the increase of enkephalin levels.


Subject(s)
Analgesics/pharmacology , Disulfides/pharmacology , Neprilysin/antagonists & inhibitors , Pain/drug therapy , Protease Inhibitors/pharmacology , Administration, Oral , Analgesics/administration & dosage , Analgesics/chemistry , Animals , Disulfides/administration & dosage , Disulfides/chemistry , Male , Mice , Mice, Inbred Strains , Neprilysin/metabolism , Pain Measurement , Protease Inhibitors/administration & dosage , Protease Inhibitors/chemistry
5.
Pharmacol Res Perspect ; 3(2): e00116, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25692029

ABSTRACT

The peripheral endogenous opioid system is critically involved in neuropathic and inflammatory pain generation as suggested by the modulation of opioid receptors expression and enkephalins (ENKs) release observed in these painful conditions. Accordingly, an innovative approach in the treatment of these nocifensive events is to increase and maintain high local concentrations of extracellular pain-evoked ENKs, by preventing their physiological enzymatic inactivation by two Zn metallopeptidases, the neutral endopeptidase (NEP, neprilysin, EC 3.4.24.11) and the neutral aminopeptidase (APN, EC 3.4.11.2). With this aim, new orally active dual ENKephalinase inhibitors (DENKIs) were designed as soluble prodrugs by introducing a N-terminal cleavable carbamate in the previously described aminophosphinic inhibitors. This induces long-lasting antinociceptive responses after oral administration, in various rodent models of inflammatory and neuropathic pain. These responses are mediated through stimulation of peripheral opioid receptors by DENKIs-protected ENKs as demonstrated by naloxone methiodide reversion. In all tested models, the most efficient prodrug 2a (PL265) was active, at least during 150-180 min, after single oral administration of 25-50 mg/kg in mice and of 100-200 mg/kg in rats. In models of neuropathic pain, both hyperalgesia and allodynia were markedly reduced. Interestingly, combination of inactive doses of 2a (PL265) and of the anti-epileptic drug gabapentin had synergistic effect on neuropathic pain. Pharmacokinetic studies of 2a (PL265) in rats show that the active drug is the only generated metabolite produced. These encouraging results have made 2a (PL265) a suitable candidate for clinical development.

6.
J Med Chem ; 57(13): 5748-63, 2014 Jul 10.
Article in English | MEDLINE | ID: mdl-24927250

ABSTRACT

Protecting enkephalins, endogenous opioid peptides released in response to nociceptive stimuli, is an innovative approach for acute and neuropathic pain alleviation. This is achieved by inhibition of their enzymatic degradation by two membrane-bound Zn-metallopeptidases, neprilysin (NEP, EC 3.4.24.11) and aminopeptidase N (APN, EC 3.4.11.2). Selective and efficient inhibitors of both enzymes, designated enkephalinases, have been designed that markedly increase extracellular concentrations and half-lives of enkephalins, inducing potent antinociceptive effects. Several chemical families of Dual ENKephalinase Inhibitors (DENKIs) have previously been developed but devoid of oral activity. We report here the design and synthesis of new pro-drugs, derived from co-drugs combining a NEP and an APN inhibitor through a disulfide bond with side chains improving oral bioavailability. Their pharmacological properties were assessed in various animal models of pain targeting central and/or peripheral opioid systems. Considering its efficacy in acute and neuropathic pain, one of these new DENKIs, 19-IIIa, was selected for clinical development.


Subject(s)
Disulfides/chemical synthesis , Neuralgia/drug therapy , Phenylpropionates/chemical synthesis , Protease Inhibitors/administration & dosage , Administration, Oral , Analgesics/administration & dosage , Analgesics/chemical synthesis , Analgesics/pharmacokinetics , Animals , CD13 Antigens/antagonists & inhibitors , Disulfides/administration & dosage , Disulfides/pharmacokinetics , Enkephalins/metabolism , Humans , Male , Mice , Neprilysin/antagonists & inhibitors , Neuralgia/enzymology , Phenylpropionates/administration & dosage , Phenylpropionates/pharmacokinetics , Prodrugs/administration & dosage , Prodrugs/chemical synthesis , Prodrugs/pharmacokinetics , Prodrugs/therapeutic use , Propylamines , Protease Inhibitors/chemical synthesis , Protease Inhibitors/pharmacokinetics , Rats
7.
Anal Biochem ; 441(2): 152-61, 2013 Oct 15.
Article in English | MEDLINE | ID: mdl-23851339

ABSTRACT

Leukotriene A4 hydrolase (LTA4H) is a bifunctional zinc-dependent metalloprotease bearing both an epoxide hydrolase, producing the pro-inflammatory LTB4 leukotriene, and an aminopeptidase activity, whose physiological relevance has long been ignored. Distinct substrates are commonly used for each activity, although none is completely satisfactory; LTA4, substrate for the hydrolase activity, is unstable and inactivates the enzyme, whereas aminoacids ß-naphthylamide and para-nitroanilide, used as aminopeptidase substrates, are poor and nonselective. Based on the three-dimensional structure of LTA4H, we describe a new, specific, and high-affinity fluorigenic substrate, PL553 [L-(4-benzoyl)phenylalanyl-ß-naphthylamide], with both in vitro and in vivo applications. PL553 possesses a catalytic efficiency (k(cat)/K(m)) of 3.8±0.5×104 M⁻¹ s⁻¹ using human recombinant LTA4H and a limit of detection and quantification of less than 1 to 2 ng. The PL553 assay was validated by measuring the inhibitory potency of known LTA4H inhibitors and used to characterize new specific amino-phosphinic inhibitors. The LTA4H inhibition measured with PL553 in mouse tissues, after intravenous administration of inhibitors, was also correlated with a reduction in LTB4 levels. This authenticates the assay as the first allowing the easy measurement of endogenous LTA4H activity and in vitro specific screening of new LTA4H inhibitors.


Subject(s)
Enzyme Assays/methods , Epoxide Hydrolases/antagonists & inhibitors , Epoxide Hydrolases/metabolism , Naphthalenes/metabolism , Peptides/metabolism , Animals , Enzyme Inhibitors/pharmacology , Epoxide Hydrolases/chemistry , Humans , Male , Mice , Molecular Docking Simulation , Naphthalenes/analysis , Peptides/analysis , Protein Conformation , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Spectrometry, Fluorescence/methods , Substrate Specificity
8.
J Biomol Screen ; 18(6): 726-35, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23427044

ABSTRACT

Detection and quantification of low doses of botulinum toxin serotype A (BoNT/A) in medicinal preparations require precise and sensitive methods. With mounting pressure from governmental authorities to replace the mouse LD50 assay, interest in alternative methods such as the endopeptidase assay, quantifying the toxin active moiety, is growing. Using internal collision-induced fluorescence quenching, Pharmaleads produced peptides encompassing the SNAP-25 cleavage site: a 17-mer (PL63) and a 48-mer (PL50) reaching the previously identified α-exosite, with PL50 showing higher apparent affinity for BoNT/A. Peptide mapping experiments revealed that this increased affinity is mainly due to a connecting peptide sequence between the N-terminus of PL63 and the α-exosite, identifying a new cooperative exosite on BoNT/A. Other endopeptidase substrates available, including SNAPTide and BoTest A/E, are both based on fluorescent resonance energy transfer (FRET) technology. To compare these assays, their limits of detection and quantification were determined using light chain or 150-kDa BoNT/A. Detection limits of PL50 and BoTest were over 100 times better than those using SNAPtide in standard conditions. Although the BoTest possessed a detection limit around 0.2 pM for either BoNT/A form, its quantification limit (9.7 pM) using purified BoNT/A was 12 times inferior to PL50, estimated at 0.8 pM, suitable for medicinal preparation quantification.


Subject(s)
Botulinum Toxins, Type A/chemistry , Fluorescent Dyes/chemistry , Peptides/chemistry , Pharmaceutical Preparations/chemistry , Animals , Biological Assay/methods , Lethal Dose 50 , Limit of Detection , Mice , Sensitivity and Specificity
9.
FASEB J ; 26(12): 5161-71, 2012 Dec.
Article in English | MEDLINE | ID: mdl-22923332

ABSTRACT

Inflammatory pain can be controlled by endogenous opioid peptides. Here we blocked the degradation of opioids in peripheral injured tissue to locally augment this physiological system. In rats with hindpaw inflammation, inhibitors of aminopeptidase N (APN; bestatin) or neutral endopeptidase (NEP; thiorphan), and a dual inhibitor, NH(2)-CH-Ph-P(O)(OH)CH(2)-CH-CH(2)Ph(p-Ph)-CONH-CH-CH(3)-COOH (P8B), were applied to injured paws. Combined bestatin (1.25-5 mg)/thiorphan (0.2-0.8 mg) or P8B (0.0625-1 mg) alone elevated mechanical nociceptive thresholds to 307 and 227% of vehicle-treated controls, respectively. This analgesia was abolished by antibodies to methionine-enkephalin, leucine-enkephalin, and dynorphin A 1-17, by peripherally restricted and by selective µ-, δ-, and κ-opioid receptor antagonists. Flow cytometry and photospectrometry revealed expression and metabolic activity of APN and NEP on macrophages, granulocytes, and sciatic nerves from inflamed tissue. Radioimmunoassays showed that inhibition of leukocytic APN and NEP by bestatin (5-500 µM)/thiorphan (1-100 µM) combinations or by P8B (1-100 µM) prevented the degradation of enkephalins. Blockade of neuronal peptidases by bestatin (0.5-10 mM)/thiorphan (0.1-5 mM) or by P8B (0.1-10 mM) additionally hindered dynorphin A 1-17 catabolism. Thus, leukocytes and peripheral nerves are important sources of APN and NEP in inflamed tissue, and their blockade promotes peripheral opioid analgesia.


Subject(s)
CD13 Antigens/antagonists & inhibitors , Enzyme Inhibitors/pharmacology , Inflammation/prevention & control , Neprilysin/antagonists & inhibitors , Pain/prevention & control , Alanine/analogs & derivatives , Alanine/pharmacology , Amino Acid Sequence , Animals , Antibodies/immunology , Antibodies/pharmacology , CD13 Antigens/metabolism , Dose-Response Relationship, Drug , Dynorphins/immunology , Dynorphins/metabolism , Dynorphins/pharmacology , Enkephalin, Leucine/immunology , Enkephalin, Leucine/metabolism , Enkephalin, Leucine/pharmacology , Enkephalin, Methionine/immunology , Enkephalin, Methionine/metabolism , Enkephalin, Methionine/pharmacology , Flow Cytometry , Hindlimb/drug effects , Hindlimb/innervation , Hindlimb/physiopathology , Inflammation/complications , Inflammation/enzymology , Leucine/analogs & derivatives , Leucine/pharmacology , Leukocytes/drug effects , Leukocytes/enzymology , Male , Narcotic Antagonists , Neprilysin/metabolism , Neurons/drug effects , Neurons/enzymology , Opioid Peptides/immunology , Opioid Peptides/metabolism , Opioid Peptides/pharmacology , Pain/complications , Pain/enzymology , Pain Threshold/drug effects , Phosphinic Acids/pharmacology , Rats , Rats, Wistar , Receptors, Opioid/metabolism , Thiorphan/pharmacology
11.
J Biol Chem ; 287(24): 20221-30, 2012 Jun 08.
Article in English | MEDLINE | ID: mdl-22528499

ABSTRACT

Legionella pneumophila has been shown to secrete a protease termed major secretory protein (Msp). This protease belongs to the M4 family of metalloproteases and shares 62.9% sequence similarity with pseudolysin (EC 3.4.24.26). With the aim of developing a specific enzymatic assay for the detection and quantification of Msp, the Fluofast substrate library was screened using both enzymes in parallel. Moreover, based on the crystal structure of pseudolysin, a model of the Msp structure was built. Screening of the peptide library identified a lead substrate specifically cleaved by Msp that was subsequently optimized by rational design. The proposed model for Msp is consistent with the enzymatic characteristics of the studied peptide substrates and provides new structural information useful for the characterization of the protease. This study leads to the identification of the first selective and high affinity substrate for Msp that is able to detect picomolar concentrations of the purified enzyme. The identified substrate could be useful for the development of a novel method for the rapid detection of Legionella.


Subject(s)
Bacterial Proteins/chemistry , Legionella pneumophila/enzymology , Metalloproteases/chemistry , Models, Molecular , Structural Homology, Protein
12.
Nat Rev Drug Discov ; 11(4): 292-310, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22460123

ABSTRACT

Chronic pain remains unsatisfactorily treated, and few novel painkillers have reached the market in the past century. Increasing the levels of the main endogenous opioid peptides - enkephalins - by inhibiting their two inactivating ectopeptidases, neprilysin and aminopeptidase N, has analgesic effects in various models of inflammatory and neuropathic pain. Stemming from the same pharmacological concept, fatty acid amide hydrolase (FAAH) inhibitors have also been found to have analgesic effects in pain models by preventing the breakdown of endogenous cannabinoids. Dual enkephalinase inhibitors and FAAH inhibitors are now in early-stage clinical trials. In this Review, we compare the effects of these two potential classes of novel analgesics and describe the progress in their rational design. We also consider the challenges in their clinical development and opportunities for combination therapies.


Subject(s)
Analgesics/pharmacology , Analgesics/pharmacokinetics , Cannabinoid Receptor Modulators/antagonists & inhibitors , Cannabinoid Receptor Modulators/pharmacokinetics , Chronic Pain/drug therapy , Enkephalins/antagonists & inhibitors , Amidohydrolases/antagonists & inhibitors , Animals , Cannabinoid Receptor Modulators/pharmacology , Chronic Pain/metabolism , Humans
13.
Anal Biochem ; 419(2): 95-105, 2011 Dec 15.
Article in English | MEDLINE | ID: mdl-21893023

ABSTRACT

Protease inhibitors represent a major class of drugs, even though a large number of proteases remain unexplored. Consequently, a great interest lies in the identification of highly sensitive substrates useful for both the characterization and the validation of these enzyme targets and for the design of inhibitors as potential therapeutic agents through high-throughput screening (HTS). With this aim, a synthetic substrate library, in which the highly fluorescent (L)-pyrenylalanine residue (Pya) is efficiently quenched by its proximity with the p-nitro-(L)-phenylalanine (Nop) moiety, was designed. The cleavage between Pya and Nop leads to a highly fluorescent metabolite providing the required sensitivity. This library, characterized by a water-soluble primary sequence Ac-SGK-Pya-(X)(n)(-)Nop-GGK-NH(2), X being a mixture of 10 natural amino acids (A, I, L, K, F, W, E, Q, T, P) and n varying from 0 to 3, was validated using enzymes belonging to the four main types of hydrolases: serine-, metallo-, cystein-, and aspartyl-proteases. The selectivity of substrates belonging to this library was evidenced by characterizing specific substrates for the isoenzymes NEP-1 and NEP-2. This library easily synthesized is of great interest for the identification and development of selective and specific substrates for still uncharacterized endoproteases.


Subject(s)
Biochemistry/methods , Peptide Hydrolases/metabolism , Peptide Library , Peptides/metabolism , Amino Acid Sequence , Animals , Chromatography, Liquid , Humans , Hydrolysis , Isoenzymes/metabolism , Mass Spectrometry , Molecular Sequence Data , Peptides/chemistry , Spectrometry, Fluorescence , Spectrophotometry, Ultraviolet , Substrate Specificity
14.
J Biol Chem ; 285(45): 34390-400, 2010 Nov 05.
Article in English | MEDLINE | ID: mdl-20807771

ABSTRACT

Endothelin-converting enzyme-2 (ECE-2) is a membrane-bound zinc-dependent metalloprotease that shares a high degree of sequence homology with ECE-1, but displays an acidic pH optimum characteristic of maturing enzymes acting late in the secretory pathway. Although ECE-2, like ECE-1, can cleave the big endothelin intermediate to produce the vasoconstrictive endothelin peptide, its true physiological function remains to be elucidated, a task that is hampered by the lack of specific tools to study and discriminate ECE-2 from ECE-1, i.e. specific substrates and/or specific inhibitors. To fill this gap, we searched for novel ECE-specific peptide substrates. To this end, peptides derived from the big endothelin intermediate were tested using ECE-1 and ECE-2, leading to the identification of an ECE-1-specific substrate. Moreover, screening of our proprietary fluorigenic peptide Fluofast® libraries using ECE-1 and ECE-2 allowed the identification of Ac-SKG-Pya-F-W-Nop-GGK-NH(2) (PL405), as a specific and high affinity ECE-2 substrate. Indeed, ECE-2 cleaved PL405 at the Pya-F amide bond with a specificity constant (k(cat)/K(m)) of 8.1 ± 0.9 × 10(3) M(-1) s(-1). Using this novel substrate, we also characterized the first potent (K(i) = 7.7 ± 0.3 nM) and relatively selective ECE-2 inhibitor and developed a quantitative fluorigenic ECE-2 assay. The assay was used to study the ex vivo ECE-2 activity in wild type and ECE-2 knock-out tissues and was found to truly reflect ECE-2 expression patterns. The PL405 assay is thus the first tool to study ECE-2 inhibition using high throughput screening or for ex vivo ECE-2 quantification.


Subject(s)
Aspartic Acid Endopeptidases/biosynthesis , Aspartic Acid Endopeptidases/chemistry , Endothelins/chemistry , Enzyme Assays/methods , Fluorescent Dyes/chemistry , Gene Expression Regulation, Enzymologic/physiology , Metalloendopeptidases/biosynthesis , Metalloendopeptidases/chemistry , Peptides/chemistry , Animals , Aspartic Acid Endopeptidases/administration & dosage , Aspartic Acid Endopeptidases/genetics , Endothelin-Converting Enzymes , Endothelins/genetics , Endothelins/metabolism , Enzyme Inhibitors/chemistry , Humans , Metalloendopeptidases/administration & dosage , Metalloendopeptidases/genetics , Mice , Organ Specificity , Peptide Library , Peptides/genetics , Peptides/metabolism , Substrate Specificity
15.
FEBS Lett ; 584(15): 3446-51, 2010 Aug 04.
Article in English | MEDLINE | ID: mdl-20609366

ABSTRACT

Leukotriene A4 hydrolase (LTA4H) is a key enzyme in the inflammatory process of mammals. It is an epoxide hydrolase and an aminopeptidase of the M1 family of the MA clan of Zn-metallopeptidases. We have solved the crystal structure of LTA4H in complex with N-[3(R)-[(hydroxyamino)carbonyl]-2-benzyl-1-oxopropyl]-L-alanine, a potent inhibitor of several Zn-metalloenzymes, both endopeptidases and aminopeptidases. The inhibitor binds along the sequence signature for M1 aminopeptidases, GXMEN. It exhibits bidentate chelation of the catalytic zinc and binds to LTA4H's enzymatically essential carboxylate recognition site. The structure gives clues to the binding of this inhibitor to related enzymes and thereby identifies residues of their S1' sub sites as well as strategies for design of inhibitors.


Subject(s)
Dipeptides/chemistry , Drug Design , Epoxide Hydrolases/chemistry , Metalloproteases/antagonists & inhibitors , Protease Inhibitors/chemistry , Zinc/metabolism , Amino Acid Sequence , Conserved Sequence , Crystallography, X-Ray , Enzyme Assays , Epoxide Hydrolases/isolation & purification , Models, Molecular , Molecular Sequence Data , Protease Inhibitors/pharmacology , Protein Binding/drug effects , Sequence Alignment
16.
Hypertension ; 55(2): 474-80, 2010 Feb.
Article in English | MEDLINE | ID: mdl-19996063

ABSTRACT

The preferred ligand of angiotensin (Ang) II type 2 (AT(2)R)-mediated natriuresis is Ang III. The major enzyme responsible for the metabolism of Ang III is aminopeptidase N, which is selectively inhibited by compound PC-18. In this study, urine sodium excretion rates (U(Na)V), fractional excretion of sodium, fractional excretion of lithium, glomerular filtration rate, and mean arterial pressures were studied in prehypertensive and hypertensive spontaneously hypertensive rats (SHRs) and compared with age-matched Wistar-Kyoto rats (WKYs). Although renal interstitial infusion of Ang II type 1 receptor blocker candesartan increased U(Na)V in WKYs from a baseline of 0.05+/-0.01 to 0.17+/-0.04 micromol/min (P<0.01), identical infusions failed to increase U(Na)V in hypertensive SHRs. Coinfusion of AT(2)R antagonist PD-123319 abolished the natriuretic responses to candesartan in WKYs, indicating an AT(2)R-mediated effect. AT(2)R-mediated natriuresis was enabled in hypertensive SHRs by inhibiting the metabolism of Ang III with PC-18 (0.05+/-0.01 to 0.11+/-0.03 micromol/min; P<0.05). The defects in sodium excretion were present before the onset of hypertension in SHRs, because young WKYs demonstrated double the U(Na)V of SHRs (0.04+/-0.006 versus 0.02+/-0.003 micromol/min; P<0.01) at baseline. The increased U(Na)V of young WKYs was attributed to reduced renal proximal tubule sodium reabsorption, because increases in fractional excretion of sodium were paralleled by increases in fractional excretion of lithium. Renal interstitial PC-18 infusion ameliorated defective AT(2)R-mediated natriuresis in young SHRs by increasing fractional excretion of sodium and fractional excretion of lithium without changing the glomerular filtration rate. Thus, increased renal proximal tubule sodium retention is observed before the onset of hypertension in SHRs, and inhibition of the metabolism of Ang III ameliorates this pathophysiologic defect in sodium excretion.


Subject(s)
Benzimidazoles/pharmacology , CD13 Antigens/antagonists & inhibitors , Imidazoles/pharmacology , Natriuresis/drug effects , Pyridines/pharmacology , Receptor, Angiotensin, Type 2/metabolism , Tetrazoles/pharmacology , Analysis of Variance , Angiotensin II Type 1 Receptor Blockers/pharmacology , Animals , Biphenyl Compounds , CD13 Antigens/metabolism , Disease Models, Animal , Female , Glomerular Filtration Rate/drug effects , Hypertension/drug therapy , Hypertension/physiopathology , Lithium/metabolism , Natriuresis/physiology , Probability , Random Allocation , Rats , Rats, Inbred SHR , Rats, Inbred WKY , Sodium/metabolism
17.
Neurosci Lett ; 465(3): 285-9, 2009 Nov 20.
Article in English | MEDLINE | ID: mdl-19765404

ABSTRACT

Although previous studies describe the up-regulation of purinergic P2X(3) receptors expressed at peripheral nociceptive fibers in experimental painful neoplastic processes, the analgesic efficacy of P2X(3) receptor antagonists has not been tested in these settings. We study here the effect of the P2X(3) receptor antagonist, A-317491, on thermal hyperalgesia produced by the intratibial inoculation of NCTC 2472 fibrosarcoma cells to C3H/HeJ mice. The peritumoral administration of A-317491 (10-100 microg) dose-dependently attenuated osteosarcoma-induced thermal hyperalgesia without modifying thermal latencies measured in the contralateral paws. This antihyperalgesic effect was inhibited by the coadministration of naloxone-methiodide (0.1-1 microg) or the systemic injection of the selective mu-opioid receptor antagonist cyprodime (1 mg/kg), demonstrating the involvement of peripheral mu-opioid receptors. Furthermore, the antihyperalgesic effect induced by A-317491, was antagonised by the coadministration of an anti-enkephalin antibody supporting the participation of endogenous enkephalins. Consistent with this result, the antihyperalgesic effect induced by A-317491 was dramatically enhanced by the administration of an enkephalin-degrading inhibitor, Debio 0827, as demonstrated by isobolographic analysis. This synergism opens the theoretical possibility that the combination of both types of drugs could be useful to counteract some nociceptive symptoms derived from tumor development.


Subject(s)
Enkephalins/metabolism , Hyperalgesia/etiology , Hyperalgesia/metabolism , Osteosarcoma/complications , Osteosarcoma/metabolism , Receptors, Purinergic P2/metabolism , Animals , Cell Line, Tumor , Hot Temperature , Male , Mice , Mice, Inbred C3H , Purinergic P2 Receptor Antagonists , Receptors, Purinergic P2X3
18.
Acta Crystallogr D Biol Crystallogr ; 65(Pt 8): 814-22, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19622865

ABSTRACT

Aminopeptidase N (APN; EC 3.4.11.2) purified from Escherichia coli has been crystallized with the optically pure aminophosphinic inhibitor PL250, H(3)N(+)-CH(CH(3))-P(O)(OH)-CH(2)-CH(CH(2)Ph)-CONH-CH(CH(2)Ph)CO(2)(-), which mimics the transition state of the hydrolysis reaction. PL250 inhibits APN with a K(i) of 1.5-2.2 nM and its three-dimensional structure in complex with E. coli APN showed its interaction with the S(1), S'(1) and S'(2) subsites of the catalytic site. In this structure, the Zn ion was shown to be pentacoordinated by His297, His301 and Glu320 of APN and the two O atoms of the phosphinic moiety of PL250. One of these O atoms is also involved in a hydrogen bond to Tyr381, supporting the proposed role of this amino acid in the stabilization of the transition state of the enzymatic process. The strength of the phosphinic zinc binding and the occupancy of the S'(2) subsite account for the 100-fold increase in affinity of PL250 compared with the dipeptide-derived inhibitor bestatin (K(i) = 4.1 x 10(-6) M). Accordingly, the removal of the C-terminal phenylalanine of PL250 resulted in a large decrease in affinity (K(i) = 2.17 x 10(-7) M). Furthermore, it was observed that the C-terminal carboxyl group of the inhibitor makes no direct interactions with the amino acids of the APN active site. Interestingly, PL250 exhibits the same inhibitory potency for E. coli APN and for mammalian enzymes, suggesting that the structure of the complex could be used as a template for the rational design of various human APN inhibitors needed to study the role of this aminopeptidase in various pathologies.


Subject(s)
CD13 Antigens/chemistry , Escherichia coli Proteins/chemistry , Escherichia coli/enzymology , Phenylalanine/analogs & derivatives , Phosphinic Acids/chemistry , Animals , Binding Sites , CD13 Antigens/antagonists & inhibitors , CD13 Antigens/metabolism , Crystallization , Crystallography, X-Ray , Drug Design , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/metabolism , Enzyme Inhibitors/pharmacology , Escherichia coli/drug effects , Escherichia coli Proteins/metabolism , Humans , Hydrogen Bonding , Phenylalanine/chemistry , Phenylalanine/metabolism , Phosphinic Acids/metabolism , Protein Binding , Protein Conformation , Zinc/metabolism
19.
Appl Environ Microbiol ; 75(13): 4382-90, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19429547

ABSTRACT

Botulinum neurotoxin type A (BoNT/A), the most poisonous substance known to humans, is a potential bioterrorism agent. The light-chain protein induces a flaccid paralysis through cleavage of the 25-kDa synaptosome-associated protein (SNAP-25), involved in acetylcholine release at the neuromuscular junction. BoNT/A is widely used as a therapeutic agent and to reduce wrinkles. The toxin is used at very low doses, which have to be accurately quantified. With this aim, internally quenched fluorescent substrates containing the fluorophore/repressor pair pyrenylalanine (Pya)/4-nitrophenylalanine (Nop) were developed. Nop and Pya were, respectively, introduced at positions 197 and 200 of the cleavable fragment (amino acids 187 to 203) of SNAP-25 (with norleucine at position 202 [Nle(202)]), which is acetylated at its N terminus and amidated at its C terminus. Cleavage of this peptide occurred between positions 197 and 198, as in SNAP-25, and was easily quantified by the strong fluorescence emission of the metabolite. To increase the assay sensitivity, the peptide sequence of the previous substrate was lengthened to account for exosite binding to BoNT/A. We synthesized the peptide PL50 (SNAP-25-NH(2) acetylated at positions 156 to 203 [Nop(197), Pya(200), Nle(202)]) and its analogue PL51, in which all methionines were replaced by nonoxidizable Nle. Consistent with a large increase in affinity for BoNT/A, PL50 and PL51 exhibit catalytic efficiencies of 2.6 x 10(6) M(-1) s(-1) and 8.85 x 10(6) M(-1) s(-1), respectively, and behave as the best fluorigenic substrates of BoNT/A reported to date. Under optimized assay conditions, they allow simple quantification of as little as 100 and 60 pg of BoNT/A, respectively, within 2 h with a classical fluorimeter. Calibration of the method against the mouse 50% lethal dose assay unequivocally validates the enzymatic assay.


Subject(s)
Botulinum Toxins, Type A/analysis , Fluorescent Dyes/metabolism , Fluorometry/methods , Animals , Mice , Sensitivity and Specificity
20.
J Neurochem ; 109(1): 248-56, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19187443

ABSTRACT

Several lines of data previously indicated that N-terminally truncated forms of amyloid-beta (Abeta) peptides are likely the earliest and more abundant species immunohistochemically detectable in Alzheimer's disease-affected brains. It is noteworthy that the free N-terminal residue of full-length Abeta (fl-Abeta) is an aspartyl residue, suggesting that Abeta could be susceptible to exopeptidasic attack by aminopeptidase A (APA)-like proteases. In this context, we have examined whether APA could target Abeta peptides in both cell-free and cellular models. We first show that the general aminopeptidase inhibitor amastatin as well as two distinct aminopeptidase A inhibitors EC33 and pl302 both significantly increase the recovery of genuine fl-Abeta peptides generated by cells over-expressing Swedish-mutated beta amyloid precursor protein (APP) while the aminopeptidase N blocker pl250 did not modify fl-Abeta recovery. In agreement with this observation, we establish that over-expressed APA drastically reduces, in a calcium dependent manner, fl-Abeta but not APP IntraCellular Domain in a cell-free model of Abeta production. In agreement with the above data, we show that recombinant APA degrades fl-Abeta in a pl302-sensitive manner. Interestingly, we also show that EC33 and pl302 lower staurosporine-stimulated activation of caspase-3 in wild-type fibroblasts but not in betaAPP/beta-amyloid precursor protein-like protein 2 (APLP2) double knockout fibroblasts, suggesting that protecting endogenous fl-Abeta physiological production triggers neuroprotective phenotype. By contrast, EC33 does not modify staurosporine-induced caspase-3 activation in wild-type and Swedish-mutated betaAPP-HEK293 expressing cells that display exacerbated production of Abeta. Overall, our data establish that APA contributes to the N-terminal truncation of Abeta and suggest that this cleavage is likely abrogating a protective function associated with physiological but not supraphysiological levels of genuine fl-Abeta peptides.


Subject(s)
Amyloid beta-Peptides/chemistry , Amyloid beta-Peptides/metabolism , Glutamyl Aminopeptidase/physiology , Peptide Fragments/chemistry , Peptide Fragments/metabolism , Animals , Cell Line , Glutamyl Aminopeptidase/antagonists & inhibitors , Glutamyl Aminopeptidase/genetics , Humans , Mice , Mice, Knockout , Peptides/physiology , Protein Structure, Tertiary
SELECTION OF CITATIONS
SEARCH DETAIL
...