Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
Add more filters










Publication year range
1.
Cells ; 13(5)2024 Feb 27.
Article in English | MEDLINE | ID: mdl-38474378

ABSTRACT

BACKGROUND: Diabetic foot ulcers (DFU) pose a significant health risk in diabetic patients, with insufficient revascularization during wound healing being the primary cause. This study aimed to assess microvessel sprouting and wound healing capabilities using vascular endothelial growth factor (VEGF-A) and a modified fibroblast growth factor (FGF1). METHODS: An ex vivo aortic ring rodent model and an in vivo wound healing model in diabetic mice were employed to evaluate the microvessel sprouting and wound healing capabilities of VEGF-A and a modified FGF1 both as monotherapies and in combination. RESULTS: The combination of VEGF-A and FGF1 demonstrated increased vascular sprouting in the ex vivo mouse aortic ring model, and topical administration of a combination of VEGF-A and FGF1 mRNAs formulated in lipid nanoparticles (LNPs) in mouse skin wounds promoted faster wound closure and increased neovascularization seven days post-surgical wound creation. RNA-sequencing analysis of skin samples at day three post-wound creation revealed a strong transcriptional response of the wound healing process, with the combined treatment showing significant enrichment of genes linked to skin growth. CONCLUSION: f-LNPs encapsulating VEGF-A and FGF1 mRNAs present a promising approach to improving the scarring process in DFU.


Subject(s)
Diabetes Mellitus, Experimental , Diabetic Foot , Humans , Mice , Animals , Vascular Endothelial Growth Factor A/metabolism , Fibroblast Growth Factor 1 , Neovascularization, Physiologic/physiology , Wound Healing/physiology , Disease Models, Animal
2.
J Med Chem ; 66(13): 8782-8807, 2023 07 13.
Article in English | MEDLINE | ID: mdl-37343272

ABSTRACT

Recent clinical reports have highlighted the need for wild-type (WT) and mutant dual inhibitors of c-MET kinase for the treatment of cancer. We report herein a novel chemical series of ATP competitive type-III inhibitors of WT and D1228V mutant c-MET. Using a combination of structure-based drug design and computational analyses, ligand 2 was optimized to a highly selective chemical series with nanomolar activities in biochemical and cellular settings. Representatives of the series demonstrate excellent pharmacokinetic profiles in rat in vivo studies with promising free-brain exposures, paving the way for the design of brain permeable drugs for the treatment of c-MET driven cancers.


Subject(s)
Antineoplastic Agents , Neoplasms , Rats , Animals , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-met , Drug Design , Adenosine Triphosphate , Antineoplastic Agents/pharmacology
3.
Bioorg Med Chem Lett ; 75: 128948, 2022 11 01.
Article in English | MEDLINE | ID: mdl-35987508

ABSTRACT

The c-MET receptor tyrosine kinase has received considerable attention as a cancer drug target yet there remains a need for inhibitors which are selective for c-MET and able to target emerging drug-resistant mutants. We report here the discovery, by screening a DNA-encoded chemical library, of a highly selective c-MET inhibitor which was shown by X-ray crystallography to bind to the kinase in an unprecedented manner. These results represent a novel mode of inhibiting c-MET with a small molecule and may provide a route to targeting drug-resistant forms of the kinase whilst avoiding potential toxicity issues associated with broad kinome inhibition.


Subject(s)
Antineoplastic Agents , Proto-Oncogene Proteins c-met , Antineoplastic Agents/pharmacology , Cell Line, Tumor , DNA , Protein Kinase Inhibitors/chemistry , Small Molecule Libraries/chemistry
4.
J Biol Chem ; 296: 100551, 2021.
Article in English | MEDLINE | ID: mdl-33744286

ABSTRACT

The glucocorticoid receptor (GR) is a ligand-dependent transcription factor that plays a central role in inflammation. The GR activity is also modulated via protein-protein interactions, including binding of 14-3-3 proteins induced by GR phosphorylation. However, the specific phosphorylation sites on the GR that trigger these interactions and their functional consequences are less clear. Hence, we sought to examine this system in more detail. We used phosphorylated GR peptides, biophysical studies, and X-ray crystallography to identify key residues within the ligand-binding domain of the GR, T524 and S617, whose phosphorylation results in binding of the representative 14-3-3 protein 14-3-3ζ. A kinase screen identified misshapen-like kinase 1 (MINK1) as responsible for phosphorylating T524 and Rho-associated protein kinase 1 for phosphorylating S617; cell-based approaches confirmed the importance of both GR phosphosites and MINK1 but not Rho-associated protein kinase 1 alone in inducing GR-14-3-3 binding. Together our results provide molecular-level insight into 14-3-3-mediated regulation of the GR and highlight both MINK1 and the GR-14-3-3 axis as potential targets for future therapeutic intervention.


Subject(s)
14-3-3 Proteins/metabolism , Gene Expression Regulation , Protein Serine-Threonine Kinases/metabolism , Receptors, Glucocorticoid/metabolism , Threonine/metabolism , 14-3-3 Proteins/genetics , HEK293 Cells , Humans , Mutation , Phosphorylation , Protein Binding , Protein Serine-Threonine Kinases/genetics , Receptors, Glucocorticoid/genetics , Threonine/genetics , Transcriptional Activation
5.
ACS Med Chem Lett ; 12(1): 162-167, 2021 Jan 14.
Article in English | MEDLINE | ID: mdl-33488978

ABSTRACT

We report here a fragment screen directed toward the c-MET kinase from which we discovered a series of inhibitors able to bind to a rare conformation of the protein in which the P-loop adopts a collapsed, or folded, arrangement. Preliminary SAR exploration led to an inhibitor (7) with nanomolar biochemical activity against c-MET and promising cell activity and kinase selectivity. These findings increase our structural understanding of the folded P-loop conformation of c-MET and provide a sound structural and chemical basis for further investigation of this underexplored yet potentially therapeutically exploitable conformational state.

6.
ACS Chem Biol ; 15(11): 2885-2895, 2020 11 20.
Article in English | MEDLINE | ID: mdl-33164499

ABSTRACT

The alarming growth of antibiotic resistance that is currently ongoing is a serious threat to human health. One of the most promising novel antibiotic targets is MraY (phospho-MurNAc-pentapeptide-transferase), an essential enzyme in bacterial cell wall synthesis. Through recent advances in biochemical research, there is now structural information available for MraY, and for its human homologue GPT (GlcNAc-1-P-transferase), that opens up exciting possibilities for structure-based drug design. The antibiotic compound tunicamycin is a natural product inhibitor of MraY that is also toxic to eukaryotes through its binding to GPT. In this work, we have used tunicamycin and modified versions of tunicamycin as tool compounds to explore the active site of MraY and to gain further insight into what determines inhibitor potency. We have investigated tunicamycin variants where the following motifs have been modified: the length and branching of the tunicamycin fatty acyl chain, the saturation of the fatty acyl chain, the 6″-hydroxyl group of the GlcNAc ring, and the ring structure of the uracil motif. The compounds are analyzed in terms of how potently they bind to MraY, inhibit the activity of the enzyme, and affect the protein thermal stability. Finally, we rationalize these results in the context of the protein structures of MraY and GPT.


Subject(s)
Anti-Bacterial Agents/pharmacology , Bacterial Proteins/antagonists & inhibitors , Bacterial Proteins/chemistry , Catalytic Domain/drug effects , Transferases/antagonists & inhibitors , Transferases/chemistry , Tunicamycin/pharmacology , Bacterial Infections/drug therapy , Bacterial Proteins/metabolism , Clostridium/enzymology , Clostridium Infections/drug therapy , Guanosine Triphosphate/metabolism , Humans , Molecular Docking Simulation , Transferases/metabolism , Transferases (Other Substituted Phosphate Groups)
7.
SLAS Discov ; 25(6): 535-551, 2020 Jul.
Article in English | MEDLINE | ID: mdl-32425085

ABSTRACT

Secreted proteins and their cognate plasma membrane receptors regulate human physiology by transducing signals from the extracellular environment into cells resulting in different cellular phenotypes. Systematic use of secretome proteins in assays enables discovery of novel biology and signaling pathways. Several secretome-based phenotypic screening platforms have been described in the literature and shown to facilitate target identification in drug discovery. In this review, we summarize the current status of secretome-based screening. This includes annotation, production, quality control, and sample management of secretome libraries, as well as how secretome libraries have been applied to discover novel target biology using different disease-relevant cell-based assays. A workflow for secretome-based screening is shared based on the AstraZeneca experience. The secretome library offers several advantages compared with other libraries used for target discovery: (1) screening using a secretome library directly identifies the active protein and, in many cases, its cognate receptor, enabling a rapid understanding of the disease pathway and subsequent formation of target hypotheses for drug discovery; (2) the secretome library covers significant areas of biological signaling space, although the size of this library is small; (3) secretome proteins can be added directly to cells without additional manipulation. These factors make the secretome library ideal for testing in physiologically relevant cell types, and therefore it represents an attractive approach to phenotypic target discovery.


Subject(s)
Drug Discovery/methods , High-Throughput Screening Assays , Proteome/genetics , Cellular Microenvironment/genetics , Gene Library , Humans
8.
ACS Med Chem Lett ; 10(9): 1322-1327, 2019 Sep 12.
Article in English | MEDLINE | ID: mdl-31531204

ABSTRACT

Many small molecule inhibitors of the cMET receptor tyrosine kinase have been evaluated in clinical trials for the treatment of cancer and resistance-conferring mutations of cMET are beginning to be reported for a number of such compounds. There is now a need to understand specific cMET mutations at the molecular level, particularly concerning small molecule recognition. Toward this end, we report here the first crystal structures of the recent clinically observed resistance-conferring D1228V cMET mutant in complex with small molecule inhibitors, along with a crystal structure of wild-type cMET bound by the clinical compound savolitinib and supporting cellular, biochemical, and biophysical data. Our findings indicate that the D1228V alteration induces conformational changes in the kinase, which could have implications for small molecule inhibitor design. The data we report here increases our molecular understanding of the D1228V cMET mutation and provides insight for future inhibitor design.

9.
ACS Chem Biol ; 14(9): 1913-1920, 2019 09 20.
Article in English | MEDLINE | ID: mdl-31329413

ABSTRACT

Demonstration of target binding is a key requirement for understanding the mode of action of new therapeutics. The cellular thermal shift assay (CETSA) has been introduced as a powerful label-free method to assess target engagement in physiological environments. Here, we present the application of live-cell CETSA to different classes of integral multipass transmembrane proteins using three case studies, the first showing a large and robust stabilization of the outer mitochondrial five-pass transmembrane protein TSPO, the second being a modest stabilization of SERCA2, and the last describing an atypical compound-driven stabilization of the GPCR PAR2. Our data demonstrated that using modified protocols with detergent extraction after the heating step, CETSA can reliably be applied to several membrane proteins of different complexity. By showing examples with distinct CETSA behaviors, we aim to provide the scientific community with an overview of different scenarios to expect during CETSA experiments, especially for challenging, membrane bound targets.


Subject(s)
Receptor, PAR-2/metabolism , Receptors, GABA/metabolism , Sarcoplasmic Reticulum Calcium-Transporting ATPases/metabolism , Aminoquinolines/pharmacology , Benzamides/pharmacology , Benzimidazoles/pharmacology , Benzodiazepinones/pharmacology , Benzodioxoles/pharmacology , Benzyl Alcohols/pharmacology , Biological Assay , Cell Line, Tumor , GABA Antagonists/pharmacology , HEK293 Cells , Hot Temperature , Humans , Imidazoles/pharmacology , Phase Transition/drug effects , Protein Multimerization/drug effects , Pyridines/pharmacology , Receptor, PAR-2/antagonists & inhibitors , Receptor, PAR-2/chemistry , Receptors, GABA/chemistry , Sarcoplasmic Reticulum Calcium-Transporting ATPases/antagonists & inhibitors , Sarcoplasmic Reticulum Calcium-Transporting ATPases/chemistry , Thapsigargin/pharmacology
10.
ChemMedChem ; 14(19): 1701-1709, 2019 10 04.
Article in English | MEDLINE | ID: mdl-31325352

ABSTRACT

The mitogen-activated protein kinase p38α pathway has been an attractive target for the treatment of inflammatory conditions such as rheumatoid arthritis. While a number of p38α inhibitors have been taken to the clinic, they have been limited by their efficacy and toxicological profile. A lead identification program was initiated to selectively target prevention of activation (PoA) of mitogen-activated protein kinase-activated protein kinase 2 (MK2) rather than mitogen- and stress-activated protein kinase 1 (MSK1), both immediate downstream substrates of p38α, to improve the efficacy/safety profile over direct p38α inhibition. Starting with a series of pyrazole amide PoA MK2 inhibitor leads, and guided by structural chemistry and rational design, a highly selective imidazole 9 (2-(3'-(2-amino-2-oxoethyl)-[1,1'-biphenyl]-3-yl)-N-(5-(N,N-dimethylsulfamoyl)-2-methylphenyl)-1-propyl-1H-imidazole-5-carboxamide) and the orally bioavailable imidazole 18 (3-methyl-N-(2-methyl-5-sulfamoylphenyl)-2-(o-tolyl)imidazole-4-carboxamide) were discovered. The PoA concept was further evaluated by protein immunoblotting, which showed that the optimized PoA MK2 compounds, despite their biochemical selectivity against MSK1 phosphorylation, behaved similarly to p38 inhibitors in cellular signaling. This study highlights the importance of selective tool compounds in untangling complex signaling pathways, and although 9 and 18 were not differentiated from p38α inhibitors in a cellular context, they are still useful tools for further research directed to understand the role of MK2 in the p38α signaling pathway.


Subject(s)
Anti-Inflammatory Agents/chemical synthesis , Enzyme Activation/drug effects , Imidazoles/chemical synthesis , Intracellular Signaling Peptides and Proteins/metabolism , Mitogen-Activated Protein Kinase 14/antagonists & inhibitors , Protein Kinase Inhibitors/chemical synthesis , Protein Serine-Threonine Kinases/metabolism , Pyrazoles/chemistry , Anti-Inflammatory Agents/pharmacology , Cells, Cultured , Humans , Imidazoles/pharmacology , Molecular Targeted Therapy , Protein Kinase Inhibitors/pharmacology , Signal Transduction/drug effects
11.
J Biol Chem ; 294(35): 13106-13116, 2019 08 30.
Article in English | MEDLINE | ID: mdl-31308176

ABSTRACT

Forkhead box protein O1 (FOXO1) is a transcription factor involved in various cellular processes such as glucose metabolism, development, stress resistance, and tumor suppression. FOXO1's transcriptional activity is controlled by different environmental cues through a myriad of posttranslational modifications. In response to growth factors, the serine/threonine kinase AKT phosphorylates Thr24 and Ser256 in FOXO1 to stimulate binding of 14-3-3 proteins, causing FOXO1 inactivation. In contrast, low nutrient and energy levels induce FOXO1 activity. AMP-activated protein kinase (AMPK), a master regulator of cellular energy homeostasis, partly mediates this effect through phosphorylation of Ser383 and Thr649 in FOXO1. In this study, we identified Ser22 as an additional AMPK phosphorylation site in FOXO1's N terminus, with Ser22 phosphorylation preventing binding of 14-3-3 proteins. The crystal structure of a FOXO1 peptide in complex with 14-3-3 σ at 2.3 Å resolution revealed that this is a consequence of both steric hindrance and electrostatic repulsion. Furthermore, we found that AMPK-mediated Ser22 phosphorylation impairs Thr24 phosphorylation by AKT in a hierarchical manner. Thus, numerous mechanisms maintain FOXO1 activity via AMPK signaling. AMPK-mediated Ser22 phosphorylation directly and indirectly averts binding of 14-3-3 proteins, whereas phosphorylation of Ser383 and Thr649 complementarily stimulates FOXO1 activity. Our results shed light on a mechanism that integrates inputs from both AMPK and AKT signaling pathways in a small motif to fine-tune FOXO1 transcriptional activity.


Subject(s)
14-3-3 Proteins/metabolism , AMP-Activated Protein Kinases/metabolism , Forkhead Box Protein O1/metabolism , Proto-Oncogene Proteins c-akt/metabolism , 14-3-3 Proteins/chemistry , Cells, Cultured , Forkhead Box Protein O1/chemistry , Forkhead Box Protein O1/genetics , HEK293 Cells , Humans , Models, Molecular , Phosphorylation , Signal Transduction
12.
Pain ; 160(9): 1989-2003, 2019 09.
Article in English | MEDLINE | ID: mdl-31045747

ABSTRACT

P2X4 is a ligand-gated ion channel implicated in neuropathic pain. Drug discovery efforts targeting P2X4 have been unsuccessful largely because of the difficulty in engineering specificity and selectivity. Here, we describe for the first time the generation of a panel of diverse monoclonal antibodies (mAbs) to human and mouse P2X4, capable of both positive and negative modulation of channel function. The affinity-optimised anti-P2X4 mAb IgG#151-LO showed exquisite selectivity for human P2X4 and induced potent and complete block of P2X4 currents. Site-directed mutagenesis of P2X4 revealed the head domain as a key interaction site for inhibitory mAbs. Inhibition of spinal P2X4 either by intrathecal delivery of an anti-P2X4 mAb or by systemic delivery of an anti-P2X4 bispecific mAb with enhanced blood-spinal cord barrier permeability produced long-lasting (>7 days) analgesia in a mouse model of neuropathic pain. We therefore propose that inhibitory mAbs binding the head domain of P2X4 have therapeutic potential for the treatment of neuropathic pain.


Subject(s)
Antibodies, Monoclonal/administration & dosage , Antibodies, Monoclonal/metabolism , Neuralgia/metabolism , Neuralgia/prevention & control , Receptors, Purinergic P2X4/metabolism , Animals , Cells, Cultured , Female , HEK293 Cells , Humans , Injections, Spinal , Mice , Mice, Inbred C57BL , Protein Binding/physiology , Purinergic P2X Receptor Antagonists/administration & dosage , Purinergic P2X Receptor Antagonists/metabolism , Rats , Rats, Sprague-Dawley
13.
SLAS Discov ; 23(5): 429-436, 2018 06.
Article in English | MEDLINE | ID: mdl-29316408

ABSTRACT

The discovery of ligands via affinity-mediated selection of DNA-encoded chemical libraries is driven by the quality and concentration of the protein target. G-protein-coupled receptors (GPCRs) and other membrane-bound targets can be difficult to isolate in their functional state and at high concentrations, and therefore have been challenging for affinity-mediated selection. Here, we report a successful selection campaign against protease-activated receptor 2 (PAR2). Using a thermo-stabilized mutant of PAR2, we conducted affinity selection using our >100-billion-compound DNA-encoded library. We observed a number of putative ligands enriched upon selection, and subsequent cellular profiling revealed these ligands to comprise both agonists and antagonists. The agonist series shared structural similarity with known agonists. The antagonists were shown to bind in a novel allosteric binding site on the PAR2 protein. This report serves to demonstrate that cell-free affinity selection against GPCRs can be achieved with mutant stabilized protein targets.


Subject(s)
DNA/genetics , Mutation/drug effects , Receptors, G-Protein-Coupled/agonists , Receptors, G-Protein-Coupled/antagonists & inhibitors , Small Molecule Libraries/pharmacology , Allosteric Site/drug effects , Cell Line , HEK293 Cells , Humans , Ligands , Proteins/genetics , Receptor, PAR-2 , Receptors, G-Protein-Coupled/genetics
14.
Nature ; 545(7652): 112-115, 2017 05 04.
Article in English | MEDLINE | ID: mdl-28445455

ABSTRACT

Protease-activated receptors (PARs) are a family of G-protein-coupled receptors (GPCRs) that are irreversibly activated by proteolytic cleavage of the N terminus, which unmasks a tethered peptide ligand that binds and activates the transmembrane receptor domain, eliciting a cellular cascade in response to inflammatory signals and other stimuli. PARs are implicated in a wide range of diseases, such as cancer and inflammation. PARs have been the subject of major pharmaceutical research efforts but the discovery of small-molecule antagonists that effectively bind them has proved challenging. The only marketed drug targeting a PAR is vorapaxar, a selective antagonist of PAR1 used to prevent thrombosis. The structure of PAR1 in complex with vorapaxar has been reported previously. Despite sequence homology across the PAR isoforms, discovery of PAR2 antagonists has been less successful, although GB88 has been described as a weak antagonist. Here we report crystal structures of PAR2 in complex with two distinct antagonists and a blocking antibody. The antagonist AZ8838 binds in a fully occluded pocket near the extracellular surface. Functional and binding studies reveal that AZ8838 exhibits slow binding kinetics, which is an attractive feature for a PAR2 antagonist competing against a tethered ligand. Antagonist AZ3451 binds to a remote allosteric site outside the helical bundle. We propose that antagonist binding prevents structural rearrangements required for receptor activation and signalling. We also show that a blocking antibody antigen-binding fragment binds to the extracellular surface of PAR2, preventing access of the tethered ligand to the peptide-binding site. These structures provide a basis for the development of selective PAR2 antagonists for a range of therapeutic uses.


Subject(s)
Receptor, PAR-2/chemistry , Receptor, PAR-2/metabolism , Allosteric Regulation/drug effects , Allosteric Site/drug effects , Antibodies, Blocking/chemistry , Antibodies, Blocking/pharmacology , Benzimidazoles/chemistry , Benzimidazoles/pharmacology , Benzodioxoles/chemistry , Benzodioxoles/pharmacology , Benzyl Alcohols/chemistry , Benzyl Alcohols/pharmacology , Crystallography, X-Ray , Humans , Imidazoles/chemistry , Imidazoles/pharmacology , Immunoglobulin Fab Fragments/chemistry , Immunoglobulin Fab Fragments/pharmacology , Kinetics , Ligands , Models, Molecular , Receptor, PAR-2/antagonists & inhibitors , Signal Transduction/drug effects
15.
Nat Chem Biol ; 13(3): 265-267, 2017 03.
Article in English | MEDLINE | ID: mdl-28068312

ABSTRACT

The rapid increase of antibiotic resistance has created an urgent need to develop novel antimicrobial agents. Here we describe the crystal structure of the promising bacterial target phospho-N-acetylmuramoyl-pentapeptide translocase (MraY) in complex with the nucleoside antibiotic tunicamycin. The structure not only reveals the mode of action of several related natural-product antibiotics but also gives an indication on the binding mode of the MraY UDP-MurNAc-pentapeptide and undecaprenyl-phosphate substrates.


Subject(s)
Anti-Bacterial Agents/pharmacology , Bacterial Proteins/drug effects , Clostridium/drug effects , Transferases/drug effects , Tunicamycin/pharmacology , Anti-Bacterial Agents/chemistry , Bacterial Proteins/chemistry , Clostridium/enzymology , Microbial Sensitivity Tests , Models, Molecular , Structure-Activity Relationship , Transferases/chemistry , Transferases (Other Substituted Phosphate Groups) , Tunicamycin/chemistry
16.
Biochemistry ; 56(3): 458-467, 2017 01 24.
Article in English | MEDLINE | ID: mdl-28029774

ABSTRACT

N1-Acetylspermine oxidase (APAO) catalyzes the conversion of N1-acetylspermine or N1-acetylspermidine to spermidine or putrescine, respectively, with concomitant formation of N-acetyl-3-aminopropanal and hydrogen peroxide. Here we present the structure of murine APAO in its oxidized holo form and in complex with substrate. The structures provide a basis for understanding molecular details of substrate interaction in vertebrate APAO, highlighting a key role for an asparagine residue in coordinating the N1-acetyl group of the substrate. We applied computational methods to the crystal structures to rationalize previous observations with regard to the substrate charge state. The analysis suggests that APAO features an active site ideally suited for binding of charged polyamines. We also reveal the structure of APAO in complex with the irreversible inhibitor MDL72527. In addition to the covalent adduct, a second MDL72527 molecule is bound in the active site. Binding of MDL72527 is accompanied by altered conformations in the APAO backbone. On the basis of structures of APAO, we discuss the potential for development of specific inhibitors.


Subject(s)
Oxidoreductases/chemistry , Putrescine/chemistry , Spermidine/analogs & derivatives , Spermidine/chemistry , Spermine/analogs & derivatives , Aldehydes/chemistry , Aldehydes/metabolism , Animals , Catalytic Domain , Gene Expression , Hydrogen Peroxide/chemistry , Hydrogen Peroxide/metabolism , Kinetics , Mice , Models, Molecular , Oxidation-Reduction , Oxidoreductases/genetics , Oxidoreductases/metabolism , Propylamines/chemistry , Propylamines/metabolism , Protein Structure, Secondary , Putrescine/analogs & derivatives , Putrescine/metabolism , Spermidine/metabolism , Spermine/chemistry , Spermine/metabolism
17.
ChemistryOpen ; 5(5): 445-449, 2016 Oct.
Article in English | MEDLINE | ID: mdl-27777836

ABSTRACT

Investigations of ligand-binding kinetics to membrane proteins are hampered by their poor stability and low expression levels, which often translates into sensitivity-related limitations impaired by low signal-to-noise ratios. Inspired by affinity capturing of water-soluble proteins, which utilizes water as the mobile phase, we demonstrate affinity capturing and local enrichment of membrane proteins by using a fluid lipid bilayer as the mobile phase. Specific membrane-protein capturing and enrichment in a microfluidic channel was accomplished by immobilizing a synthesized trivalent nitrilotriacetic acid (tris-NTA)-biotin conjugate. A polymer-supported lipid bilayer containing His6-tagged ß-secretase (BACE) was subsequently laterally moved over the capture region by using a hydrodynamic flow. Specific enrichment of His6-BACE in the Ni2+-NTA-modified region of the substrate resulted in a stationary three-fold increase in surface coverage, and an accompanied increase in ligand-binding response.

18.
Anal Chem ; 87(18): 9194-203, 2015 Sep 15.
Article in English | MEDLINE | ID: mdl-26268463

ABSTRACT

Supported lipid bilayers (SLBs) have contributed invaluable information about the physiochemical properties of cell membranes, but their compositional simplicity often limits the level of knowledge that can be gained about the structure and function of transmembrane proteins in their native environment. Herein, we demonstrate a generic protocol for producing polymer-supported lipid bilayers on glass surfaces that contain essentially all naturally occurring cell-membrane components of a cell line while still retaining transmembrane protein mobility and activity. This was achieved by merging vesicles made from synthetic lipids (PEGylated lipids and POPC lipids) with native cell-membrane vesicles to generate hybrid vesicles which readily rupture into a continuous polymer-supported lipid bilayer. To investigate the properties of these complex hybrid SLBs and particularly the behavior of their integral membrane-proteins, we used total internal reflection fluorescence imaging to study a transmembrane protease, ß-secretase 1 (BACE1), whose ectoplasmic and cytoplasmic domains could both be specifically targeted with fluorescent reporters. By selectively probing the two different orientations of BACE1 in the resulting hybrid SLBs, the role of the PEG-cushion on transmembrane protein lateral mobility was investigated. The results reveal the necessity of having the PEGylated lipids present during vesicle adsorption to prevent immobilization of transmembrane proteins with protruding domains. The proteolytic activity of BACE1 was unadulterated by the sonication process used to merge the synthetic and native membrane vesicles; importantly it was also conserved in the SLB. The presented strategy could thus serve both fundamental studies of membrane biophysics and the production of surface-based bioanalytical sensor platforms.


Subject(s)
Cell Membrane/chemistry , Dimethylpolysiloxanes/chemistry , Lipid Bilayers/chemistry , Membrane Proteins/metabolism , Movement , Phosphatidylcholines/chemistry , Polyethylene Glycols/chemistry , Amino Acid Sequence , Aspartic Acid Endopeptidases/chemistry , Aspartic Acid Endopeptidases/metabolism , Cell Line , Glass/chemistry , Membrane Proteins/chemistry , Models, Molecular , Molecular Conformation , Molecular Sequence Data , Surface Properties
19.
Anal Chem ; 87(8): 4100-3, 2015 Apr 21.
Article in English | MEDLINE | ID: mdl-25855499

ABSTRACT

Inhibition-in-solution assays (ISA) employing surface-based biosensors such as surface plasmon resonance (SPR) are an effective screening approach in drug discovery. However, analysis of potent binders remains a significant hurdle due to limited sensitivity and accompanied depletion of the inhibiting compounds due to high protein concentrations needed for detectable binding signals. To overcome this limitation, we explored a microscopy-based single-molecule ISA compatible with liposome-reconstituted membrane proteins. Using a set of validated small molecule inhibitors against ß-secretase 1 (BACE1), the assay was benchmarked with respect to sensitivity and dynamic range against SPR. We demonstrate that the dynamic range of measurable affinities is greatly extended by more than 2 orders of magnitude as compared to SPR, thus facilitating measurements of highly potent (Kd < nM) compounds.


Subject(s)
Amyloid Precursor Protein Secretases/antagonists & inhibitors , Aspartic Acid Endopeptidases/antagonists & inhibitors , Drug Discovery , Enzyme Inhibitors/pharmacology , Molecular Imaging , Amyloid Precursor Protein Secretases/metabolism , Aspartic Acid Endopeptidases/metabolism , Enzyme Inhibitors/chemistry , Humans , Solutions , Structure-Activity Relationship , Surface Plasmon Resonance
20.
PLoS One ; 9(8): e104001, 2014.
Article in English | MEDLINE | ID: mdl-25089527

ABSTRACT

Neprilysin is a transmembrane zinc metallopeptidase that degrades a wide range of peptide substrates. It has received attention as a potential therapy for Alzheimer's disease due to its ability to degrade the peptide amyloid beta. However, its broad range of peptide substrates has the potential to limit its therapeutic use due to degradation of additional peptides substrates that tightly regulate many physiological processes. We sought to generate a soluble version of the ectodomain of neprilysin with improved activity and specificity towards amyloid beta as a potential therapeutic for Alzheimer's disease. Extensive amino acid substitutions were performed at positions surrounding the active site and inner surface of the enzyme and variants screened for activity on amyloid beta 1-40, 1-42 and a variety of other physiologically relevant peptides. We identified several mutations that modulated and improved both enzyme selectivity and intrinsic activity. Neprilysin variant G399V/G714K displayed an approximately 20-fold improved activity on amyloid beta 1-40 and up to a 3,200-fold reduction in activity on other peptides. Along with the altered peptide substrate specificity, the mutant enzyme produced a markedly altered series of amyloid beta cleavage products compared to the wild-type enzyme. Crystallisation of the mutant enzyme revealed that the amino acid substitutions result in alteration of the shape and size of the pocket containing the active site compared to the wild-type enzyme. The mutant enzyme offers the potential for the more efficient degradation of amyloid beta in vivo as a therapeutic for the treatment of Alzheimer's disease.


Subject(s)
Amyloid beta-Peptides/chemistry , Neprilysin/chemistry , Peptide Fragments/chemistry , Peptides/chemistry , Protein Engineering , Recombinant Proteins/chemistry , Alzheimer Disease/drug therapy , Amino Acid Sequence , Amino Acid Substitution , Gene Expression , Humans , Models, Molecular , Molecular Sequence Data , Mutation , Neprilysin/genetics , Peptides/genetics , Protein Structure, Tertiary , Proteolysis , Recombinant Proteins/genetics , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Structure-Activity Relationship
SELECTION OF CITATIONS
SEARCH DETAIL
...