Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
1.
J Org Chem ; 2024 Apr 24.
Article in English | MEDLINE | ID: mdl-38655880

ABSTRACT

A novel synthesis of sitagliptin based on a redox-active ester derived from the chiral pool is reported. The key step is an electrochemical nickel-catalyzed sp2-sp3 cross-coupling reaction using inexpensive nickel foam in an undivided cell. It was successfully applied to 21 examples in up to 88% yield. These sitagliptin-analogue precursors could potentially interact with the DPP4 enzyme. A full synthesis based on our new reaction pathway provided sitagliptin in an overall yield of 33%.

2.
Bioorg Med Chem Lett ; 100: 129614, 2024 Mar 01.
Article in English | MEDLINE | ID: mdl-38199329

ABSTRACT

Electrochemical transformations are a subject of increasing interest in early drug discovery due to its ability to assemble complex scaffolds under rather mild reaction conditions. In this context, we became interested in electrochemical decarboxylative cross-coupling (DCC) protocols of redox-active esters (RAEs) and halo(hetero)arenes. Starting with the one-step electrochemical synthesis of novel methylamino-substituted heterocycles we recognized the potential of this methodology to deliver a novel approach to ß- and γ- amino acids by starting from the corresponding RAEs. Our work finally resulted in the delivery of novel and highly valuable trifunctional building blocks based on ß- and γ-amino-acid scaffolds.


Subject(s)
Amino Acids , Esters , Electrochemistry , Molecular Structure , Amino Acids/chemistry , Esters/chemistry , Oxidation-Reduction
3.
Sci Rep ; 12(1): 15440, 2022 09 14.
Article in English | MEDLINE | ID: mdl-36104373

ABSTRACT

Nicotinamide N-methyltransferase (NNMT) is a metabolic regulator that catalyzes the methylation of nicotinamide (Nam) using the co-factor S-adenosyl-L-methionine to form 1-methyl-nicotinamide (MNA). Overexpression of NNMT and the presence of the active metabolite MNA is associated with a number of diseases including metabolic disorders. We conducted a high-throughput screening campaign that led to the identification of a tricyclic core as a potential NNMT small molecule inhibitor series. Elaborate medicinal chemistry efforts were undertaken and hundreds of analogs were synthesized to understand the structure activity relationship and structure property relationship of this tricyclic series. A lead molecule, JBSNF-000028, was identified that inhibits human and mouse NNMT activity, reduces MNA levels in mouse plasma, liver and adipose tissue, and drives insulin sensitization, glucose modulation and body weight reduction in a diet-induced obese mouse model of diabetes. The co-crystal structure showed that JBSNF-000028 binds below a hairpin structural motif at the nicotinamide pocket and stacks between Tyr-204 (from Hairpin) and Leu-164 (from central domain). JBSNF-000028 was inactive against a broad panel of targets related to metabolism and safety. Interestingly, the improvement in glucose tolerance upon treatment with JBSNF-000028 was also observed in NNMT knockout mice with diet-induced obesity, pointing towards the glucose-normalizing effect that may go beyond NNMT inhibition. JBSNF-000028 can be a potential therapeutic option for metabolic disorders and developmental studies are warranted.


Subject(s)
Metabolic Diseases , Nicotinamide N-Methyltransferase , Animals , Humans , Mice , Glucose , Metabolic Diseases/drug therapy , Niacinamide/metabolism , Niacinamide/pharmacology , Nicotinamide N-Methyltransferase/metabolism , Obesity/drug therapy
4.
NMR Biomed ; 34(11): e4583, 2021 11.
Article in English | MEDLINE | ID: mdl-34240478

ABSTRACT

The purpose of the current study was to investigate if hyperpolarized [1-13 C]pyruvate can inform us on the metabolic consequences for the kidney glucose metabolism upon treatment with the pyruvate kinase M2 (PKM2) activator TEPP-46, which has shown promise as a novel therapeutic target for diabetic nephropathy. A healthy male Wistar rat model was employed to study the conversion of [1-13 C]pyruvate to [1-13 C]lactate in the kidney 2 and 4 h after treatment with TEPP-46. All rats were scanned with hyperpolarized [1-13 C]pyruvate kidney MR and vital parameters and blood samples were taken after scanning. The PKM2 activator TEPP-46 increases the glycolytic activity in the kidneys, leading to an increased lactate production, as seen by hyperpolarized pyruvate-to-lactate conversion. The results are supported by an increase in blood lactate, a decreased blood glucose level and an increased pyruvate kinase (PK) activity. The metabolic changes observed in both kidneys following treatment with TEPP-46 are largely independent of renal function and could as such represent a new and extremely sensitive metabolic readout for future drugs targeting PKM2. These results warrant further studies in disease models to evaluate if [1-13 C]pyruvate-to-[1-13 C]lactate conversion can predict treatment outcome.


Subject(s)
Glucose/metabolism , Kidney/enzymology , Pyruvate Kinase/metabolism , Pyruvic Acid/metabolism , Alanine/metabolism , Animals , Bicarbonates/metabolism , Disease Models, Animal , Enzyme Activation , Glycolysis , Hematocrit , Humans , Hydrogen-Ion Concentration , Lactic Acid/metabolism , Male , Rats, Wistar
5.
Nat Rev Drug Discov ; 20(10): 789-797, 2021 10.
Article in English | MEDLINE | ID: mdl-34285415

ABSTRACT

Proteolysis-targeting chimeras (PROTACs) are an emerging drug modality that may offer new opportunities to circumvent some of the limitations associated with traditional small-molecule therapeutics. By analogy with the concept of the 'druggable genome', the question arises as to which potential drug targets might PROTAC-mediated protein degradation be most applicable. Here, we present a systematic approach to the assessment of the PROTAC tractability (PROTACtability) of protein targets using a series of criteria based on data and information from a diverse range of relevant publicly available resources. Our approach could support decision-making on whether or not a particular target may be amenable to modulation using a PROTAC. Using our approach, we identified 1,067 proteins of the human proteome that have not yet been described in the literature as PROTAC targets that offer potential opportunities for future PROTAC-based efforts.


Subject(s)
Drug Design , Genome , Animals , Humans , Research Design , Small Molecule Libraries
6.
SLAS Discov ; 26(6): 783-797, 2021 07.
Article in English | MEDLINE | ID: mdl-33955247

ABSTRACT

Classical high-throughput screening (HTS) technologies for the analysis of ionic currents across biological membranes can be performed using fluorescence-based, radioactive, and mass spectrometry (MS)-based uptake assays. These assays provide rapid results for pharmacological HTS, but the underlying, indirect analytical character of these assays can be linked to high false-positive hit rates. Thus, orthogonal and secondary assays using more biological target-based technologies are indispensable for further compound validation and optimization. Direct assay technologies for transporter proteins are electrophysiology-based, but are also complex, time-consuming, and not well applicable for automated profiling purposes. In contrast to conventional patch clamp systems, solid supported membrane (SSM)-based electrophysiology is a sensitive, membrane-based method for transporter analysis, and current technical developments target the demand for automated, accelerated, and sensitive assays for transporter-directed compound screening. In this study, the suitability of the SSM-based technique for pharmacological compound identification and optimization was evaluated performing cell-free SSM-based measurements with the electrogenic amino acid transporter B0AT1 (SLC6A19). Electrophysiological characterization of leucine-induced currents demonstrated that the observed signals were specific to B0AT1. Moreover, B0AT1-dependent responses were successfully inhibited using an established in-house tool compound. Evaluation of current stability and data reproducibility verified the robustness and reliability of the applied assay. Active compounds from primary screens of large compound libraries were validated, and false-positive hits were identified. These results clearly demonstrate the suitability of the SSM-based technique as a direct electrophysiological method for rapid and automated identification of small molecules that can inhibit B0AT1 activity.


Subject(s)
Amino Acid Transport Systems, Neutral/metabolism , Electrophysiological Phenomena , High-Throughput Screening Assays/methods , Amino Acid Transport Systems, Neutral/agonists , Amino Acid Transport Systems, Neutral/antagonists & inhibitors , Animals , Biological Assay/methods , Biological Transport/drug effects , CHO Cells , Cell Membrane/metabolism , Cricetulus , Humans , Mice , Patch-Clamp Techniques/methods , Reproducibility of Results , Small Molecule Libraries/chemistry , Small Molecule Libraries/pharmacology
7.
Molecules ; 26(4)2021 Feb 13.
Article in English | MEDLINE | ID: mdl-33668468

ABSTRACT

Nicotinamide-N-methyltransferase (NNMT) is a cytosolic enzyme catalyzing the transfer of a methyl group from S-adenosyl-methionine (SAM) to nicotinamide (Nam). It is expressed in many tissues including the liver, adipose tissue, and skeletal muscle. Its expression in several cancer cell lines has been widely discussed in the literature, and recent work established a link between NNMT expression and metabolic diseases. Here we describe our approach to identify potent small molecule inhibitors of NNMT featuring different binding modes as elucidated by X-ray crystallographic studies.


Subject(s)
Enzyme Inhibitors/therapeutic use , Metabolic Diseases/drug therapy , Metabolic Diseases/enzymology , Nicotinamide N-Methyltransferase/antagonists & inhibitors , Animals , Binding Sites , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , High-Throughput Screening Assays , Humans , Ligands , Mice , Microsomes, Liver/drug effects , Microsomes, Liver/enzymology , Models, Molecular , Niacinamide/metabolism , Nicotinamide N-Methyltransferase/metabolism , Rats , Substrate Specificity/drug effects
8.
J Chem Inf Model ; 60(12): 6120-6134, 2020 12 28.
Article in English | MEDLINE | ID: mdl-33245234

ABSTRACT

Mining the steadily increasing amount of chemical and biological data is a key challenge in drug discovery. Graph databases offer viable alternatives for capturing interrelationships between molecules and for generating novel insights for design. In a graph database, molecules and their properties are mapped to nodes, while relationships are described by edges. Here, we introduce a graph database for navigation in chemical space, analogue searching, and structure-activity relationship (SAR) analysis. We illustrate this concept using hERG channel inhibitors from ChEMBL to extract SAR knowledge. This graph database is built using different relationships, namely 2D-fingerprint similarity, matched molecular pairs, topomer distances, and structure-activity landscape indices (SALI). Typical applications include retrieving analogues linked by single or multiple edge paths to the query compound as well as detection of nonadditive SAR features. Finally, we identify triplets of linked molecules for clustering. The speed of searching and analysis allows the user to interactively navigate the database and to address complex questions in real-time.


Subject(s)
Drug Discovery , Cluster Analysis , Databases, Factual , Structure-Activity Relationship
9.
J Biol Chem ; 295(36): 12605-12617, 2020 09 04.
Article in English | MEDLINE | ID: mdl-32647007

ABSTRACT

In the heart, the serine carboxypeptidase cathepsin A (CatA) is distributed between lysosomes and the extracellular matrix (ECM). CatA-mediated degradation of extracellular peptides may contribute to ECM remodeling and left ventricular (LV) dysfunction. Here, we aimed to evaluate the effects of CatA overexpression on LV remodeling. A proteomic analysis of the secretome of adult mouse cardiac fibroblasts upon digestion by CatA identified the extracellular antioxidant enzyme superoxide dismutase (EC-SOD) as a novel substrate of CatA, which decreased EC-SOD abundance 5-fold. In vitro, both cardiomyocytes and cardiac fibroblasts expressed and secreted CatA protein, and only cardiac fibroblasts expressed and secreted EC-SOD protein. Cardiomyocyte-specific CatA overexpression and increased CatA activity in the LV of transgenic mice (CatA-TG) reduced EC-SOD protein levels by 43%. Loss of EC-SOD-mediated antioxidative activity resulted in significant accumulation of superoxide radicals (WT, 4.54 µmol/mg tissue/min; CatA-TG, 8.62 µmol/mg tissue/min), increased inflammation, myocyte hypertrophy (WT, 19.8 µm; CatA-TG, 21.9 µm), cellular apoptosis, and elevated mRNA expression of hypertrophy-related and profibrotic marker genes, without affecting intracellular detoxifying proteins. In CatA-TG mice, LV interstitial fibrosis formation was enhanced by 19%, and the type I/type III collagen ratio was shifted toward higher abundance of collagen I fibers. Cardiac remodeling in CatA-TG was accompanied by an increased LV weight/body weight ratio and LV end diastolic volume (WT, 50.8 µl; CatA-TG, 61.9 µl). In conclusion, CatA-mediated EC-SOD reduction in the heart contributes to increased oxidative stress, myocyte hypertrophy, ECM remodeling, and inflammation, implicating CatA as a potential therapeutic target to prevent ventricular remodeling.


Subject(s)
Cathepsin A/metabolism , Myocytes, Cardiac/metabolism , Proteolysis , Superoxide Dismutase/metabolism , Ventricular Remodeling , Animals , Cathepsin A/genetics , Male , Mice , Mice, Transgenic , Myocytes, Cardiac/pathology , Superoxide Dismutase/genetics
10.
Chemistry ; 26(56): 12738-12742, 2020 Oct 06.
Article in English | MEDLINE | ID: mdl-32716576

ABSTRACT

Hydrogen isotopically labelled compounds are essential diagnostic tools in drug research and development, as they provide vital information about the biological metabolism of drug candidates and their metabolites. Herein we report a photoredox-initiated hydrogen atom transfer (HAT) protocol which efficiently and selectively introduces deuterium or tritium at C(sp3 )-H bonds, utilizing heavy water (D2 O or T2 O) as the hydrogen isotope source, and a guanidine base. This protocol has been successfully applied to the incorporation of deuterium in several amino acids (lysine, glycine and proline) and small peptides. Finally, the method has been applied to tritium, because tritium-labelled peptides are essential for application in biological experiments, such as ligand-binding assays, or absorption, distribution, metabolism, and excretion (ADME) studies.


Subject(s)
Pharmaceutical Preparations , Amino Acids , Deuterium , Hydrogen , Isotope Labeling , Peptides
11.
Chem Sci ; 11(23): 6053-6057, 2020 Jun 01.
Article in English | MEDLINE | ID: mdl-34094098

ABSTRACT

Electrochemical decarboxylation of aryloxyacetic acids followed by fluorination provides easy access to fluoromethyl aryl ethers. This electrochemical fluorodecarboxylation offers a sustainable approach with electric current as traceless oxidant. Using Et3N·5HF as fluoride source and as supporting electrolyte, this simple electrosynthesis affords various fluoromethoxyarenes in yields up to 85%.

12.
JACC Basic Transl Sci ; 4(3): 332-344, 2019 Jun.
Article in English | MEDLINE | ID: mdl-31312757

ABSTRACT

After myocardial infarction, remote ventricular remodeling and atrial cardiomyopathy progress despite successful revascularization. In a rat model of ventricular ischemia/reperfusion, pharmacological inhibition of the protease activity of cathepsin A initiated at the time point of reperfusion prevented extracellular matrix remodeling in the atrium and the ventricle remote from the infarcted area. This scenario was associated with preservation of more viable ventricular myocardium and the prevention of an arrhythmogenic and functional substrate for atrial fibrillation. Remote ventricular extracellular matrix remodeling and atrial cardiomyopathy may represent a promising target for pharmacological atrial fibrillation upstream therapy following myocardial infarction.

13.
Amino Acids ; 51(1): 7-16, 2019 Jan.
Article in English | MEDLINE | ID: mdl-29922921

ABSTRACT

Carnosinase 1 (CN1) has been postulated to be a susceptibility factor for developing diabetic nephropathy (DN). Although its major substrate, carnosine, is beneficial in rodent models of DN, translation of these findings to humans has been hampered by high CN1 activity in human serum resulting in rapid degradation of carnosine. To overcome this hurdle, we screened a protease-directed small-molecule library for inhibitors of human recombinant CN1. We identified SAN9812 as a potent and highly selective inhibitor of CN1 activity with a Ki of 11 nM. It also inhibited CN1 activity in human serum and serum of transgenic mice-overexpressing human CN1. Subcutaneous administration of 30 mg/kg SAN9812 led to a sustained reduction in circulating CN1 activity in human CN1 transgenic (TG) mice. Simultaneous administration of carnosine and SAN9812 increased carnosine levels in plasma and kidney by up to 100-fold compared to treatment-naïve CN1-overexpressing mice. To our knowledge, this is the first study reporting on a potent and selective CN1 inhibitor with in vivo activity. SAN9812, also called carnostatine, may be used to increase renal carnosine concentration as a potential therapeutic modality for renal diseases linked to glycoxidative conditions.


Subject(s)
Carnosine/administration & dosage , Dipeptidases/antagonists & inhibitors , Drug Discovery , Imidazoles/pharmacology , Propionates/pharmacology , Protease Inhibitors/pharmacology , Small Molecule Libraries/pharmacology , Adult , Animals , Carnosine/blood , Dipeptidases/blood , Dipeptidases/genetics , Female , Gene Expression , Humans , Imidazoles/chemistry , Injections, Subcutaneous , Kinetics , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Propionates/chemistry , Protease Inhibitors/chemistry , Protein Binding , Recombinant Proteins/blood , Recombinant Proteins/genetics , Small Molecule Libraries/chemistry , Transgenes
14.
Bioorg Med Chem Lett ; 28(5): 922-925, 2018 03 01.
Article in English | MEDLINE | ID: mdl-29433927

ABSTRACT

Nicotinamide N-methyltransferase (NNMT) has been linked to obesity and diabetes. We have identified a novel nicotinamide (NA) analog, compound 12 that inhibited NNMT enzymatic activity and reduced the formation of 1-methyl-nicotinamide (MNA), the primary metabolite of NA by ∼80% at 2 h when dosed in mice orally at 50 mg/kg.


Subject(s)
Enzyme Inhibitors/pharmacology , Niacinamide/pharmacology , Nicotinamide N-Methyltransferase/antagonists & inhibitors , Dose-Response Relationship, Drug , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/chemistry , Humans , Molecular Structure , Niacinamide/chemical synthesis , Niacinamide/chemistry , Nicotinamide N-Methyltransferase/metabolism , Structure-Activity Relationship
15.
Sci Rep ; 8(1): 3660, 2018 02 26.
Article in English | MEDLINE | ID: mdl-29483571

ABSTRACT

Nicotinamide N-methyltransferase (NNMT) is a cytosolic enzyme that catalyzes the transfer of a methyl group from the co-factor S-adenosyl-L-methionine (SAM) onto the substrate, nicotinamide (NA) to form 1-methyl-nicotinamide (MNA). Higher NNMT expression and MNA concentrations have been associated with obesity and type-2 diabetes. Here we report a small molecule analog of NA, JBSNF-000088, that inhibits NNMT activity, reduces MNA levels and drives insulin sensitization, glucose modulation and body weight reduction in animal models of metabolic disease. In mice with high fat diet (HFD)-induced obesity, JBSNF-000088 treatment caused a reduction in body weight, improved insulin sensitivity and normalized glucose tolerance to the level of lean control mice. These effects were not seen in NNMT knockout mice on HFD, confirming specificity of JBSNF-000088. The compound also improved glucose handling in ob/ob and db/db mice albeit to a lesser extent and in the absence of weight loss. Co-crystal structure analysis revealed the presence of the N-methylated product of JBSNF-000088 bound to the NNMT protein. The N-methylated product was also detected in the plasma of mice treated with JBSNF-000088. Hence, JBSNF-000088 may act as a slow-turnover substrate analog, driving the observed metabolic benefits.


Subject(s)
Enzyme Inhibitors/therapeutic use , Metabolic Diseases/drug therapy , Metabolic Diseases/enzymology , Nicotinamide N-Methyltransferase/metabolism , Animals , Body Weight/drug effects , Diabetes Mellitus, Type 2/drug therapy , Diabetes Mellitus, Type 2/enzymology , Diet, High-Fat/adverse effects , Male , Mice , Mice, Inbred C57BL , Nicotinamide N-Methyltransferase/antagonists & inhibitors
16.
Biochem Biophys Res Commun ; 491(2): 416-422, 2017 09 16.
Article in English | MEDLINE | ID: mdl-28720493

ABSTRACT

Nicotinamide N-methyltransferase (NNMT) is a S-adenosyl-l-methionine (SAM)-dependent enzyme that catalyzes N-methylation of nicotinamide (NA) and other pyridines to form N-methyl pyridinium ions. Here we report the first ternary complex X-ray crystal structures of monkey NNMT and mouse NNMT in bound form with the primary endogenous product, 1-methyl nicotinamide (MNA) and demethylated cofactor, S-adenosyl-homocysteine (SAH) determined at 2.30 Å and 1.88 Å respectively. The structural fold of these enzymes is identical to human NNMT. It is known that the primary endogenous product catalyzed by NNMT, MNA is a specific inhibitor of NNMT. Our data clearly indicates that the MNA binds to the active site and it would be trapped in the active site due to the formation of the bridge between the pole (long helix, α3) and long C-terminal loop. This might explain the mechanism of MNA acting as a feedback inhibitor of NNMT.


Subject(s)
Feedback, Physiological , Niacinamide/analogs & derivatives , Nicotinamide N-Methyltransferase/chemistry , S-Adenosylmethionine/chemistry , Amino Acid Sequence , Animals , Catalytic Domain , Cloning, Molecular , Crystallography, X-Ray , Escherichia coli/genetics , Escherichia coli/metabolism , Gene Expression , Macaca mulatta , Mice , Models, Molecular , Niacinamide/chemistry , Niacinamide/metabolism , Nicotinamide N-Methyltransferase/antagonists & inhibitors , Nicotinamide N-Methyltransferase/genetics , Nicotinamide N-Methyltransferase/metabolism , Protein Binding , Protein Conformation, alpha-Helical , Protein Conformation, beta-Strand , Protein Interaction Domains and Motifs , Protein Structure, Tertiary , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , S-Adenosylmethionine/metabolism , Sequence Alignment , Sequence Homology, Amino Acid , Substrate Specificity
17.
J Transl Med ; 14(1): 153, 2016 05 31.
Article in English | MEDLINE | ID: mdl-27246731

ABSTRACT

BACKGROUND: Myocardial infarction (MI) is a major cause of heart failure. The carboxypeptidase cathepsin A is a novel target in the treatment of cardiac failure. We aim to show that recently developed inhibitors of the protease cathepsin A attenuate post-MI heart failure. METHODS: Mice were subjected to permanent left anterior descending artery (LAD) ligation or sham operation. 24 h post-surgery, LAD-ligated animals were treated with daily doses of the cathepsin A inhibitor SAR1 or placebo. After 4 weeks, the three groups (sham, MI-placebo, MI-SAR1) were evaluated. RESULTS: Compared to sham-operated animals, placebo-treated mice showed significantly impaired cardiac function and increased plasma BNP levels. Cathepsin A inhibition prevented the increase of plasma BNP levels and displayed a trend towards improved cardiac functionality. Proteomic profiling was performed for the three groups (sham, MI-placebo, MI-SAR1). More than 100 proteins were significantly altered in placebo-treated LAD ligation compared to the sham operation, including known markers of cardiac failure as well as extracellular/matricellular proteins. This ensemble constitutes a proteome fingerprint of myocardial infarction induced by LAD ligation in mice. Cathepsin A inhibitor treatment normalized the marked increase of the muscle stress marker CA3 as well as of Igγ 2b and fatty acid synthase. For numerous further proteins, cathepsin A inhibition partially dampened the LAD ligation-induced proteome alterations. CONCLUSIONS: Our proteomic and functional data suggest that cathepsin A inhibition has cardioprotective properties and support a beneficial effect of cathepsin A inhibition in the treatment of heart failure after myocardial infarction.


Subject(s)
Cathepsin A/antagonists & inhibitors , Heart Failure/drug therapy , Heart Failure/etiology , Myocardial Infarction/complications , Myocardial Infarction/drug therapy , Protease Inhibitors/therapeutic use , Proteomics/methods , Animals , Cathepsin A/metabolism , Cell Line , Disease Models, Animal , Heart Failure/metabolism , Heart Failure/physiopathology , Heart Ventricles/drug effects , Heart Ventricles/pathology , Ligation , Male , Mice, Inbred C57BL , Myocardial Infarction/metabolism , Myocardial Infarction/physiopathology , Organ Size/drug effects , Peptide Mapping , Protease Inhibitors/pharmacology , Proteome/metabolism , Rats
18.
Cardiovasc Res ; 110(3): 371-80, 2016 06 01.
Article in English | MEDLINE | ID: mdl-27032673

ABSTRACT

AIMS: Type 2 diabetes (T2D) is an independent risk factor for atrial fibrillation (AF) and stroke. The serine protease cathepsin A (CatA) is up-regulated in diabetes and plays an important role in the degradation of extracellular peptides. This study sought to delineate the role of CatA for the development of atrial remodelling under diabetic conditions. METHODS AND RESULTS: Zucker Diabetic Fatty rats (ZDF) were treated with vehicle (n = 20) or CatA-inhibitor (SAR; 50 mg/kg; n = 20), and compared with age-matched non-diabetic littermates (Ctr, n = 20). Left-atrial (LA) emptying function [magnetic resonance imaging (MRI)] and atrial electrophysiological parameters were measured before sacrifice for histological and biochemical analysis. The impact of enhanced cardiac CatA expression on atrial remodelling was determined using CatA-transgenic mice. At the age of 9.5 months, atrial tissues of ZDF rats showed increased CatA gene expression and CatA-activity, along with increased AF-susceptibility and impaired LA-emptying function. CatA-inhibition reduced CatA-activity in ZDF comparable to Ctr values and decreased LA-fibrosis formation and connexin 43 lateralization. This was associated with shorter median duration of LA-tachyarrhythmia (12.0 ± 1.7 vs. 1.2 ± 0.47 s, P < 0.01) induced by burst pacing and diminished regions of slow conduction. Cardiac MRI revealed better LA-emptying function parameters (active per cent emptying: 29 ± 1 vs. 23 ± 2%, P < 0.01) after CatA-inhibition. CatA-inhibition reduced LA bradykinin-degrading activity in ZDF. Transgenic mice overexpressing CatA demonstrated enhanced atrial fibrosis formation and increased AF-susceptibility. CONCLUSION: T2D leads to arrhythmogenic atrial remodelling in ZDF rats. CatA-inhibition reduces LA bradykinin-degrading activity in ZDF and suppresses the development of atrial structural changes and AF-promotion, implicating CatA as an important mediator for AF-substrate in T2D.


Subject(s)
Atrial Fibrillation/enzymology , Atrial Function, Left , Atrial Remodeling , Cathepsin A/metabolism , Diabetes Mellitus, Type 2/enzymology , Myocardium/enzymology , Action Potentials , Angiotensin II/metabolism , Animals , Atrial Fibrillation/genetics , Atrial Fibrillation/physiopathology , Atrial Fibrillation/prevention & control , Atrial Function, Left/drug effects , Atrial Remodeling/drug effects , Bradykinin/metabolism , Cathepsin A/antagonists & inhibitors , Cathepsin A/genetics , Connexin 43/metabolism , Diabetes Mellitus, Type 2/drug therapy , Diabetes Mellitus, Type 2/genetics , Diabetes Mellitus, Type 2/physiopathology , Disease Models, Animal , Fibrosis , Heart Rate , Mice, Inbred C57BL , Mice, Transgenic , Protease Inhibitors/pharmacology , Rats, Zucker , Time Factors
19.
Clin Pharmacol Drug Dev ; 5(1): 57-68, 2016 Jan.
Article in English | MEDLINE | ID: mdl-27119579

ABSTRACT

Cathepsin A (CathA) is a lysosomal protein where it forms a stable complex with neuraminidase and ß-galactosidase. CathA also has enzymatic activity and is involved in the degradation of many peptides. CathA was recently discovered as a target for heart failure, fostering the development of CathA inhibitors with SAR164653 as a frontrunner. The first-in-man study investigated single oral doses from 20 to 800 mg of SAR164653 followed by repeat dose studies at doses up to 800 mg in healthy young and elderly subjects. SAR164653 was safe and well tolerated at doses up to 800 mg in healthy subjects, and a maximum tolerated dose could not be determined from the study. Activity of ß-galactosidase measured in leukocytes did not show any abnormalities. The tmax was 1.0 to 2.5 hours, and the t1/2 was ∼5-11 after single dosing; exposure increased less than dose proportional. Following multiple dosing, accumulation was not observed, Cmax and AUC0-24 increased in a dose-proportional manner, and t1/2 was around 14-20 hours. The novel CathA inhibitor SAR164653 was found to have a favorable safety profile in these early phase 1 studies, but further studies are required to confirm if SAR164653 is equally safe in patients undergoing long-term treatment.


Subject(s)
Cathepsin A/antagonists & inhibitors , Adolescent , Adult , Age Factors , Aged , Aged, 80 and over , Area Under Curve , Dose-Response Relationship, Drug , Double-Blind Method , Female , Half-Life , Humans , Male , Middle Aged , Young Adult
20.
J Biol Chem ; 290(47): 28446-28455, 2015 Nov 20.
Article in English | MEDLINE | ID: mdl-26459563

ABSTRACT

The activation of the transcription factor NF-E2-related factor 2 (Nrf2) maintains cellular homeostasis in response to oxidative stress by the regulation of multiple cytoprotective genes. Without stressors, the activity of Nrf2 is inhibited by its interaction with the Keap1 (kelch-like ECH-associated protein 1). Here, we describe (3S)-1-[4-[(2,3,5,6-tetramethylphenyl) sulfonylamino]-1-naphthyl]pyrrolidine-3-carboxylic acid (RA839), a small molecule that binds noncovalently to the Nrf2-interacting kelch domain of Keap1 with a Kd of ∼6 µM, as demonstrated by x-ray co-crystallization and isothermal titration calorimetry. Whole genome DNA arrays showed that at 10 µM RA839 significantly regulated 105 probe sets in bone marrow-derived macrophages. Canonical pathway mapping of these probe sets revealed an activation of pathways linked with Nrf2 signaling. These pathways were also activated after the activation of Nrf2 by the silencing of Keap1 expression. RA839 regulated only two genes in Nrf2 knock-out macrophages. Similar to the activation of Nrf2 by either silencing of Keap1 expression or by the reactive compound 2-cyano-3,12-dioxooleana-1,9-dien-28-oic acid methyl ester (CDDO-Me), RA839 prevented the induction of both inducible nitric-oxide synthase expression and nitric oxide release in response to lipopolysaccharides in macrophages. In mice, RA839 acutely induced Nrf2 target gene expression in liver. RA839 is a selective inhibitor of the Keap1/Nrf2 interaction and a useful tool compound to study the biology of Nrf2.


Subject(s)
Intracellular Signaling Peptides and Proteins/metabolism , NF-E2-Related Factor 2/metabolism , Pyrrolidines/pharmacology , Signal Transduction/drug effects , Sulfonamides/pharmacology , Animals , Kelch-Like ECH-Associated Protein 1 , Male , Mice , Protein Binding , Pyrrolidines/metabolism , Sulfonamides/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...