Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
J Med Chem ; 66(1): 251-265, 2023 01 12.
Article in English | MEDLINE | ID: mdl-36540942

ABSTRACT

The mitochondrial rhomboid protease PARL regulates mitophagy by balancing intramembrane proteolysis of PINK1 and PGAM5. It has been implicated in the pathogenesis of Parkinson's disease, but its investigation as a possible therapeutic target is challenging in this context because genetic deficiency of PARL may result in compensatory mechanisms. To address this problem, we undertook a hitherto unavailable chemical biology strategy. We developed potent PARL-targeting ketoamide inhibitors and investigated the effects of acute PARL suppression on the processing status of PINK1 intermediates and on Parkin activation. This approach revealed that PARL inhibition leads to a robust activation of the PINK1/Parkin pathway without major secondary effects on mitochondrial properties, which demonstrates that the pharmacological blockage of PARL to boost PINK1/Parkin-dependent mitophagy is a feasible approach to examine novel therapeutic strategies for Parkinson's disease. More generally, this study showcases the power of ketoamide inhibitors for cell biological studies of rhomboid proteases.


Subject(s)
Parkinson Disease , Peptide Hydrolases , Humans , Metalloproteases/genetics , Metalloproteases/metabolism , Mitophagy , Parkinson Disease/drug therapy , Protein Kinases/metabolism , Mitochondrial Proteins/metabolism , Endopeptidases , Ubiquitin-Protein Ligases/metabolism
2.
Trends Biochem Sci ; 43(9): 726-739, 2018 09.
Article in English | MEDLINE | ID: mdl-30055896

ABSTRACT

The rhomboid superfamily of transmembrane (TM) proteins includes intramembrane serine proteases and several classes of pseudoprotease. Rhomboid-like proteins occur widely across evolution and comprise biologically important regulators of fate of membrane proteins, influencing their proteolysis, trafficking, or degradation. In this review, we discuss how structural and mechanistic insights into the action of rhomboid proteases can inform on the mechanism of the pseudoproteases, and discuss the impact of structural understanding on the development of inhibitors and other chemical biology tools for these proteins. Development of modulators would be particularly relevant for the iRhoms, which are key regulators of ADAM17 and, hence, tumor necrosis factor (TNF) and epidermal growth factor receptor (EGFR) signaling, two medically important pathways.


Subject(s)
ADAM17 Protein/metabolism , Membrane Proteins/metabolism , Proteolysis , Signal Transduction/physiology , ADAM17 Protein/genetics , Animals , Humans , Membrane Proteins/genetics , Protein Transport/physiology , Structure-Activity Relationship , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism
3.
Bioorg Med Chem Lett ; 28(8): 1423-1427, 2018 05 01.
Article in English | MEDLINE | ID: mdl-29506958

ABSTRACT

Rhomboid proteases form one of the most widespread intramembrane protease families. They have been implicated in variety of human diseases. The currently reported rhomboid inhibitors display some selectivity, but their construction involves multistep synthesis protocols. Here, we report benzoxazin-4-ones as novel inhibitors of rhomboid proteases with a covalent, but slow reversible inhibition mechanism. Benzoxazin-4-ones can be synthesized from anthranilic acid derivatives in a one-step synthesis, making them easily accessible. We demonstrate that an alkoxy substituent at the 2-position is crucial for potency and results in low micromolar inhibitors of rhomboid proteases. Hence, we expect that these compounds will allow rapid synthesis and optimization of inhibitors of rhomboids from different organisms.


Subject(s)
Benzoxazines/pharmacology , DNA-Binding Proteins/antagonists & inhibitors , Escherichia coli Proteins/antagonists & inhibitors , Membrane Proteins/antagonists & inhibitors , Serine Proteinase Inhibitors/pharmacology , Animals , Bacillus subtilis/enzymology , Benzoxazines/chemical synthesis , Benzoxazines/chemistry , Cattle , Chymotrypsin/antagonists & inhibitors , Endopeptidases , Enzyme Assays , Escherichia coli/enzymology , Molecular Structure , Serine Proteinase Inhibitors/chemical synthesis , Serine Proteinase Inhibitors/chemistry , Structure-Activity Relationship , Trypsin/chemistry , Trypsin Inhibitors/chemical synthesis , Trypsin Inhibitors/chemistry , Trypsin Inhibitors/pharmacology , ortho-Aminobenzoates/chemistry
4.
Bioorg Med Chem Lett ; 28(8): 1417-1422, 2018 05 01.
Article in English | MEDLINE | ID: mdl-29463448

ABSTRACT

Rhomboids are intramembrane serine proteases with diverse physiological functions in organisms ranging from archaea to humans. Crystal structure analysis has provided a detailed understanding of the catalytic mechanism, and rhomboids have been implicated in various disease contexts. Unfortunately, the design of specific rhomboid inhibitors has lagged behind, and previously described small molecule inhibitors displayed insufficient potency and/or selectivity. Using a computer-aided approach, we focused on the discovery of novel scaffolds with reduced liabilities and the possibility for broad structural variations. Docking studies with the E. coli rhomboid GlpG indicated that 2-styryl substituted benzoxazinones might comprise novel rhomboid inhibitors. Protease in vitro assays confirmed activity of 2-styryl substituted benzoxazinones against GlpG but not against the soluble serine protease α-chymotrypsin. Furthermore, mass spectrometry analysis demonstrated covalent modification of the catalytic residue Ser201, corroborating the predicted mechanism of inhibition and the formation of an acyl enzyme intermediate. In conclusion, 2-styryl substituted benzoxazinones are a novel rhomboid inhibitor scaffold with ample opportunity for optimization.


Subject(s)
Benzoxazines/chemistry , Serine Proteinase Inhibitors/chemistry , Styrenes/chemistry , Animals , Benzoxazines/chemical synthesis , Catalytic Domain , Cattle , Chymotrypsin/chemistry , DNA-Binding Proteins/antagonists & inhibitors , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/genetics , Drosophila/chemistry , Drosophila Proteins/metabolism , Drug Discovery , Endopeptidases/chemistry , Endopeptidases/genetics , Enzyme Assays , Escherichia coli/enzymology , Escherichia coli Proteins/antagonists & inhibitors , Escherichia coli Proteins/chemistry , Escherichia coli Proteins/genetics , Humans , Membrane Proteins/antagonists & inhibitors , Membrane Proteins/chemistry , Membrane Proteins/genetics , Molecular Docking Simulation , Mutation , Serine/chemistry , Serine Proteinase Inhibitors/chemical synthesis , Styrenes/chemical synthesis , Transforming Growth Factor alpha/metabolism
5.
Cell Chem Biol ; 24(12): 1523-1536.e4, 2017 12 21.
Article in English | MEDLINE | ID: mdl-29107700

ABSTRACT

Rhomboid-family intramembrane proteases regulate important biological processes and have been associated with malaria, cancer, and Parkinson's disease. However, due to the lack of potent, selective, and pharmacologically compliant inhibitors, the wide therapeutic potential of rhomboids is currently untapped. Here, we bridge this gap by discovering that peptidyl α-ketoamides substituted at the ketoamide nitrogen by hydrophobic groups are potent rhomboid inhibitors active in the nanomolar range, surpassing the currently used rhomboid inhibitors by up to three orders of magnitude. Such peptidyl ketoamides show selectivity for rhomboids, leaving most human serine hydrolases unaffected. Crystal structures show that these compounds bind the active site of rhomboid covalently and in a substrate-like manner, and kinetic analysis reveals their reversible, slow-binding, non-competitive mechanism. Since ketoamides are clinically used pharmacophores, our findings uncover a straightforward modular way for the design of specific inhibitors of rhomboid proteases, which can be widely applicable in cell biology and drug discovery.


Subject(s)
Drug Design , Peptide Hydrolases/metabolism , Serine Proteinase Inhibitors/pharmacology , Gram-Negative Bacteria/enzymology , Gram-Positive Bacteria/enzymology , Models, Molecular , Molecular Conformation , Serine Proteinase Inhibitors/chemical synthesis , Serine Proteinase Inhibitors/chemistry
6.
Biochemistry ; 56(51): 6713-6725, 2017 12 26.
Article in English | MEDLINE | ID: mdl-29185711

ABSTRACT

Rhomboids are intramembrane serine proteases and belong to the group of structurally and biochemically most comprehensively characterized membrane proteins. They are highly conserved and ubiquitously distributed in all kingdoms of life and function in a wide range of biological processes, including epidermal growth factor signaling, mitochondrial dynamics, and apoptosis. Importantly, rhomboids have been associated with multiple diseases, including Parkinson's disease, type 2 diabetes, and malaria. However, despite a thorough understanding of many structural and functional aspects of rhomboids, potent and selective inhibitors of these intramembrane proteases are still not available. In this study, we describe the computer-based rational design, chemical synthesis, and biological evaluation of novel N-methylene saccharin-based rhomboid protease inhibitors. Saccharin inhibitors displayed inhibitory potency in the submicromolar range, effectiveness against rhomboids both in vitro and in live Escherichia coli cells, and substantially improved selectivity against human serine hydrolases compared to those of previously known rhomboid inhibitors. Consequently, N-methylene saccharins are promising new templates for the development of rhomboid inhibitors, providing novel tools for probing rhomboid functions in physiology and disease.


Subject(s)
Drug Design , Saccharin/analogs & derivatives , Serine Proteases/metabolism , Serine Proteinase Inhibitors/pharmacology , Computer-Aided Design , HEK293 Cells , Humans , Membrane Proteins , Saccharin/pharmacology , Serine Proteinase Inhibitors/chemistry
7.
J Biol Chem ; 292(7): 2703-2713, 2017 02 17.
Article in English | MEDLINE | ID: mdl-28069810

ABSTRACT

Rhomboid proteases are increasingly being explored as potential drug targets, but their potent and specific inhibitors are not available, and strategies for inhibitor development are hampered by the lack of widely usable and easily modifiable in vitro activity assays. Here we address this bottleneck and report on the development of new fluorogenic transmembrane peptide substrates, which are cleaved by several unrelated rhomboid proteases, can be used both in detergent micelles and in liposomes, and contain red-shifted fluorophores that are suitable for high-throughput screening of compound libraries. We show that nearly the entire transmembrane domain of the substrate is important for efficient cleavage, implying that it extensively interacts with the enzyme. Importantly, we demonstrate that in the detergent micelle system, commonly used for the enzymatic analyses of intramembrane proteolysis, the cleavage rate strongly depends on detergent concentration, because the reaction proceeds only in the micelles. Furthermore, we show that the catalytic efficiency and selectivity toward a rhomboid substrate can be dramatically improved by targeted modification of the sequence of its P5 to P1 region. The fluorogenic substrates that we describe and their sequence variants should find wide use in the detection of activity and development of inhibitors of rhomboid proteases.


Subject(s)
Fluorescent Dyes/chemistry , Peptide Hydrolases/metabolism , Peptides/metabolism , Fluorescence Resonance Energy Transfer , Kinetics , Liposomes , Substrate Specificity
8.
Brain Res ; 1498: 33-40, 2013 Mar 01.
Article in English | MEDLINE | ID: mdl-23291266

ABSTRACT

Neuropeptide FF (NPFF) belongs to the RF-amide family of peptides bearing the identical C-terminal amino acid sequence (R-F-NH2). In addition to NPFF, prolactin-releasing peptide (PrRP), another RF-amide, binds to NPFF receptors with high affinity. A selective antagonist of PrRP has not yet been identified, but a selective antagonist of NPFF, 1-adamantanecarbonyl-RF-NH2 (RF9), was recently reported to antagonize the hyperalgesic effect of NPFF after central administration to mice. In the present study, RF9 competed with NPFF analog D-Y-L-(N-Me)-F-Q-P-Q-R-F-NH2 (1DMe) in binding to CHO-K1 cell membranes transfected with the human NPFF2 receptor. In rat pituitary RC-4B/C cells, where the expression of the NPFF2 receptor was proved by immunodetection, RF9 did not reverse the phosphorylation of MAPK/ERK1/2 induced by [Tyr(1)]NPFF. In vivo experiments with fasted mice confirmed that centrally injected [Tyr(1)]NPFF significantly lowered food intake. However, RF9, a putative NPFF2 antagonist, did not reverse the anorectic effect of [Tyr(1)]NPFF. Paradoxically, RF9 itself exhibited an anorectic effect in fasted mice not only after intracerebroventricular but also after subcutaneous administration. This finding casts doubt on claims that RF9 is an NPFF antagonist.


Subject(s)
Adamantane/analogs & derivatives , Appetite Depressants/pharmacology , Dipeptides/pharmacology , Eating/drug effects , Receptors, Neuropeptide/metabolism , Adamantane/pharmacology , Animals , Binding, Competitive , CHO Cells , Cell Line, Tumor , Cricetulus , Extracellular Signal-Regulated MAP Kinases/metabolism , Humans , Male , Mice, Inbred C57BL , Oligopeptides/metabolism , Phosphorylation/drug effects , Rats
SELECTION OF CITATIONS
SEARCH DETAIL
...