Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 624
Filter
1.
J Phys Chem Lett ; 12(37): 9020-9025, 2021 Sep 23.
Article in English | MEDLINE | ID: mdl-34516127

ABSTRACT

Ribonucleotide reductase (RNR), which supplies the building blocks for DNA biosynthesis and its repair, has been linked to human diseases and is emerging as a therapeutic target. Here, we present a mechanistic investigation of triapine (3AP), a clinically relevant small molecule that inhibits the tyrosyl radical within the RNR ß2 subunit. Solvent kinetic isotope effects reveal that proton transfer is not rate-limiting for inhibition of Y122· of E. coli RNR ß2 by the pertinent 3AP-Fe(II) adduct. Vibrational spectroscopy further demonstrates that unlike inhibition of the ß2 tyrosyl radical by hydroxyurea, a carboxylate containing proton wire is not at play. Binding measurements reveal a low nanomolar affinity (Kd ∼ 6 nM) of 3AP-Fe(II) for ß2. Taken together, these data should prompt further development of RNR inactivators based on the triapine scaffold for therapeutic applications.


Subject(s)
Enzyme Inhibitors/chemistry , Escherichia coli Proteins/metabolism , Escherichia coli/enzymology , Ferrous Compounds/chemistry , Pyridines/chemistry , Ribonucleotide Reductases/metabolism , Thiosemicarbazones/chemistry , Enzyme Inhibitors/metabolism , Escherichia coli Proteins/antagonists & inhibitors , Free Radicals/chemistry , Free Radicals/metabolism , Hydroxyurea/chemistry , Protein Binding , Protein Subunits/antagonists & inhibitors , Protein Subunits/chemistry , Protein Subunits/metabolism , Ribonucleotide Reductases/antagonists & inhibitors , Spectrophotometry, Ultraviolet , Spectroscopy, Fourier Transform Infrared
2.
J Med Chem ; 64(14): 10230-10245, 2021 07 22.
Article in English | MEDLINE | ID: mdl-34228444

ABSTRACT

Proteasomes are broadly expressed key components of the ubiquitin-dependent protein degradation pathway containing catalytically active subunits (ß1, ß2, and ß5). LMP7 (ß5i) is a subunit of the immunoproteasome, an inducible isoform that is predominantly expressed in hematopoietic cells. Clinically effective pan-proteasome inhibitors for the treatment of multiple myeloma (MM) nonselectively target LMP7 and other subunits of the constitutive proteasome and immunoproteasome with comparable potency, which can limit the therapeutic applicability of these drugs. Here, we describe the discovery and structure-based hit optimization of novel amido boronic acids, which selectively inhibit LMP7 while sparing all other subunits. The exploitation of structural differences between the proteasome subunits culminated in the identification of the highly potent, exquisitely selective, and orally available LMP7 inhibitor 50 (M3258). Based on the strong antitumor activity observed with M3258 in MM models and a favorable preclinical data package, a phase I clinical trial was initiated in relapsed/refractory MM patients.


Subject(s)
Drug Discovery , Proteasome Endopeptidase Complex/metabolism , Proteasome Inhibitors/pharmacology , Dose-Response Relationship, Drug , Humans , Molecular Structure , Proteasome Inhibitors/chemical synthesis , Proteasome Inhibitors/chemistry , Protein Subunits/antagonists & inhibitors , Protein Subunits/metabolism , Structure-Activity Relationship
3.
Indian J Pharmacol ; 53(2): 132-142, 2021.
Article in English | MEDLINE | ID: mdl-34100397

ABSTRACT

OBJECTIVES: Water contaminated with arsenic affected millions of people worldwide and arsenic exposure is related to various neurological disorders. Hence, the current study was planned to investigate the neuroprotective activity of diosmin (DSN) against arsenic induced neurotoxicity as an attempt to identify therapeutic intervention to combat arsenicism. MATERIALS AND METHODS: Sodium arsenite an inducer of neurotoxicity was administered orally (13 mg/kg) and DSN treatment at two selected doses (50 and 100 mg/kg) was done for 21 days. Behavioral and biochemical variations were examined by various parameters. Furthermore, histopathological and immunohistochemistry studies were done with the brain sections. RESULTS: The behavioral studies evidenced that arsenic has suppressed the exploratory behavior and motor coordination in rats and DSN treatment has recovered the behavioral changes to normal. Arsenic administration has also found to induce oxidative stress and DSN co-treatment has ameliorated the oxidative stress markers. Interestingly, depleted levels of neurotransmitters were observed with the arsenic and it was restored back by the DSN treatment. Histopathological alterations like pyknosis of the neuronal cells were identified with arsenic exposure and subsided upon DSN co administration. Immunohistochemical studies have revealed the expression of NOX4 and its gp91phox and P47phox subunits and its suppression by DSN treatment may be the key therapeutic factor of it. CONCLUSIONS: Treatment with DSN showed a beneficial effect in protecting against arsenic-induced neurotoxicity by suppressing the toxicity changes and the antioxidant effect of DSN might be attributed to its ability of suppressing NOX4 and its subunits.


Subject(s)
Arsenic/toxicity , Diosmin/therapeutic use , NADPH Oxidase 4/antagonists & inhibitors , Neuroprotective Agents/therapeutic use , Neurotoxicity Syndromes/drug therapy , Animals , Antioxidants/analysis , Arsenic/analysis , Brain/drug effects , Brain/pathology , Brain Chemistry/drug effects , Female , Maze Learning/drug effects , Neurotoxicity Syndromes/metabolism , Neurotoxicity Syndromes/pathology , Neurotransmitter Agents/analysis , Oxidative Stress/drug effects , Protein Subunits/antagonists & inhibitors , Rats , Rats, Wistar
4.
J Neurochem ; 159(1): 90-100, 2021 10.
Article in English | MEDLINE | ID: mdl-34008858

ABSTRACT

α-Conotoxins are small disulfide-rich peptides found in the venom of marine cone snails and are potent antagonists of nicotinic acetylcholine receptors (nAChRs). They are valuable pharmacological tools and have potential therapeutic applications for the treatment of chronic pain or neurological diseases and disorders. In the present study, we synthesized and functionally characterized a novel α-conotoxin Bt1.8, which was cloned from Conus betulinus. Bt1.8 selectively inhibited ACh-evoked currents in Xenopus oocytes expressing rat(r) α6/α3ß2ß3 and rα3ß2 nAChRs with an IC50 of 2.1 nM and 9.4 nM, respectively, and similar potency for human (h) α6/α3ß2ß3 and hα3ß2 nAChRs. Additionally, Bt1.8 had higher binding affinity with a slower dissociation rate for the rα6/α3ß2ß3 subtype compared to rα3ß2. The amino acid sequence of Bt1.8 is significantly different from other reported α-conotoxins targeting the two nAChR subtypes. Further Alanine scanning analyses demonstrated that residues Ile9, Leu10, Asn11, Asn12 and Asn14 are critical for its inhibitory activity at the α6/α3ß2ß3 and α3ß2 subtypes. Moreover, the NMR structure of Bt1.8 indicated the presence of a relatively larger hydrophobic zone than other α4/7-conotoxins which may explain its potent inhibition at α6/α3ß2ß3 nAChRs.


Subject(s)
Conotoxins/pharmacology , Nicotinic Antagonists/pharmacology , Receptors, Nicotinic/metabolism , Animals , Conotoxins/chemistry , Conotoxins/isolation & purification , Conus Snail , Dose-Response Relationship, Drug , Female , Humans , Nicotinic Antagonists/chemistry , Nicotinic Antagonists/isolation & purification , Oocytes , Protein Structure, Tertiary , Protein Subunits/antagonists & inhibitors , Protein Subunits/genetics , Protein Subunits/metabolism , Rats , Receptors, Nicotinic/genetics , Xenopus laevis
5.
J Med Chem ; 64(8): 4857-4869, 2021 04 22.
Article in English | MEDLINE | ID: mdl-33821636

ABSTRACT

LONP1 is an AAA+ protease that maintains mitochondrial homeostasis by removing damaged or misfolded proteins. Elevated activity and expression of LONP1 promotes cancer cell proliferation and resistance to apoptosis-inducing reagents. Despite the importance of LONP1 in human biology and disease, very few LONP1 inhibitors have been described in the literature. Herein, we report the development of selective boronic acid-based LONP1 inhibitors using structure-based drug design as well as the first structures of human LONP1 bound to various inhibitors. Our efforts led to several nanomolar LONP1 inhibitors with little to no activity against the 20S proteasome that serve as tool compounds to investigate LONP1 biology.


Subject(s)
ATP-Dependent Proteases/antagonists & inhibitors , Drug Design , Mitochondrial Proteins/antagonists & inhibitors , Protease Inhibitors/chemistry , ATP-Dependent Proteases/metabolism , Binding Sites , Boronic Acids/chemistry , Boronic Acids/metabolism , Boronic Acids/pharmacology , Bortezomib/chemistry , Bortezomib/metabolism , Cell Line , Cell Survival/drug effects , Humans , Mitochondrial Proteins/metabolism , Molecular Docking Simulation , Protease Inhibitors/metabolism , Protease Inhibitors/pharmacology , Proteasome Endopeptidase Complex/chemistry , Proteasome Endopeptidase Complex/metabolism , Protein Subunits/antagonists & inhibitors , Protein Subunits/metabolism , Structure-Activity Relationship
6.
Eur J Med Chem ; 219: 113455, 2021 Jul 05.
Article in English | MEDLINE | ID: mdl-33894528

ABSTRACT

Proteasomes contribute to maintaining protein homeostasis and their inhibition is beneficial in certain types of cancer and in autoimmune diseases. However, the inhibition of the proteasomes in healthy cells leads to unwanted side-effects and significant effort has been made to identify inhibitors specific for the immunoproteasome, especially to treat diseases which manifest increased levels and activity of this proteasome isoform. Here, we report our efforts to discover fragment-sized inhibitors of the human immunoproteasome. The screening of an in-house library of structurally diverse fragments resulted in the identification of benzo[d]oxazole-2(3H)-thiones, benzo[d]thiazole-2(3H)-thiones, benzo[d]imidazole-2(3H)-thiones, and 1-methylbenzo[d]imidazole-2(3H)-thiones (with a general term benzoXazole-2(3H)-thiones) as inhibitors of the chymotrypsin-like (ß5i) subunit of the immunoproteasome. A subsequent structure-activity relationship study provided us with an insight regarding growing vectors. Binding to the ß5i subunit was shown and selectivity against the ß5 subunit of the constitutive proteasome was determined. Thorough characterization of these compounds suggested that they inhibit the immunoproteasome by forming a disulfide bond with the Cys48 available specifically in the ß5i active site. To obtain fragments with biologically more tractable covalent interactions, we performed a warhead scan, which yielded benzoXazole-2-carbonitriles as promising starting points for the development of selective immunoproteasome inhibitors with non-peptidic scaffolds.


Subject(s)
Proteasome Endopeptidase Complex/metabolism , Proteasome Inhibitors/chemistry , Drug Evaluation, Preclinical , Humans , Inhibitory Concentration 50 , Oxazoles/chemistry , Proteasome Endopeptidase Complex/chemistry , Proteasome Inhibitors/metabolism , Protein Subunits/antagonists & inhibitors , Protein Subunits/metabolism , Structure-Activity Relationship , Thiazoles/chemistry , Thiones/chemistry
7.
Front Immunol ; 12: 586521, 2021.
Article in English | MEDLINE | ID: mdl-33717067

ABSTRACT

Antibodies recognizing the amino-terminal domain of receptor subunit proteins modify the receptor efficiency to controlling transmitter release in isolated nerve endings (e.g., synaptosomes) indirectly confirming their presence in these particles but also allowing to speculate on their subunit composition. Western blot analysis and confocal microscopy unveiled the presence of the GluA1, GluA2, GluA3, and GluA4 receptor subunits in cortical synaptosomes. Functional studies confirmed the presence of presynaptic release-regulating AMPA autoreceptors in these terminals, whose activation releases [3H]D-aspartate ([3H]D-Asp, here used as a marker of glutamate) in a NBQX-dependent manner. The AMPA autoreceptors traffic in a constitutive manner, since entrapping synaptosomes with the pep2-SVKI peptide (which interferes with the GluA2-GRIP1/PICK1 interaction) amplified the AMPA-evoked releasing activity, while the inactive pep2-SVKE peptide was devoid of activity. Incubation of synaptosomes with antibodies recognizing the NH2 terminus of the GluA2 and the GluA3 subunits increased, although to a different extent, the GluA2 and 3 densities in synaptosomal membranes, also amplifying the AMPA-evoked glutamate release in a NBQX-dependent fashion. We then analyzed the releasing activity of complement (1:300) from both treated and untreated synaptosomes and found that the complement-induced overflow occurred in a DL-t-BOA-sensitive, NBQX-insensitive fashion. We hypothesized that anti-GluA/GluA complexes in neuronal membranes could trigger the classic pathway of activation of the complement, modifying its releasing activity. Accordingly, the complement-evoked release of [3H]D-Asp from antiGluA2 and anti-GluA3 antibody treated synaptosomes was significantly increased when compared to untreated terminals and facilitation was prevented by omitting the C1q component of the immunocomplex. Antibodies recognizing the NH2 terminus of the GluA1 or the GluA4 subunits failed to affect both the AMPA and the complement-evoked tritium overflow. Our results suggest the presence of GluA2/GluA3-containing release-regulating AMPA autoreceptors in cortical synaptosomes. Incubation of synaptosomes with commercial anti-GluA2 or anti-GluA3 antibodies amplifies the AMPA-evoked exocytosis of glutamate through a complement-independent pathway, involving an excessive insertion of AMPA autoreceptors in plasma membranes but also affects the complement-dependent releasing activity, by promoting the classic pathway of activation of the immunocomplex. Both events could be relevant to the development of autoimmune diseases typified by an overproduction of anti-GluA subunits.


Subject(s)
Antibodies/pharmacology , Protein Interaction Domains and Motifs/drug effects , Protein Subunits/antagonists & inhibitors , Receptors, AMPA/antagonists & inhibitors , Animals , Cell Membrane/drug effects , Cell Membrane/metabolism , Cerebral Cortex/metabolism , Complement C1q/immunology , Fluorescent Antibody Technique , Male , Mice , Receptors, AMPA/chemistry , Synaptosomes/drug effects , Synaptosomes/metabolism
8.
Molecules ; 26(4)2021 Feb 14.
Article in English | MEDLINE | ID: mdl-33673017

ABSTRACT

Influenza A virus (IAV) encodes a polymerase composed of three subunits: PA, with endonuclease activity, PB1 with polymerase activity and PB2 with host RNA five-prime cap binding site. Their cooperation and stepwise activation include a process called cap-snatching, which is a crucial step in the IAV life cycle. Reproduction of IAV can be blocked by disrupting the interaction between the PB2 domain and the five-prime cap. An inhibitor of this interaction called pimodivir (VX-787) recently entered the third phase of clinical trial; however, several mutations in PB2 that cause resistance to pimodivir were observed. First major mutation, F404Y, causing resistance was identified during preclinical testing, next the mutation M431I was identified in patients during the second phase of clinical trials. The mutation H357N was identified during testing of IAV strains at Centers for Disease Control and Prevention. We set out to provide a structural and thermodynamic analysis of the interactions between cap-binding domain of PB2 wild-type and PB2 variants bearing these mutations and pimodivir. Here we present four crystal structures of PB2-WT, PB2-F404Y, PB2-M431I and PB2-H357N in complex with pimodivir. We have thermodynamically analysed all PB2 variants and proposed the effect of these mutations on thermodynamic parameters of these interactions and pimodivir resistance development. These data will contribute to understanding the effect of these missense mutations to the resistance development and help to design next generation inhibitors.


Subject(s)
Drug Resistance, Viral/drug effects , Influenza A virus/enzymology , Protein Subunits/antagonists & inhibitors , Pyridines/chemistry , Pyridines/pharmacology , Pyrimidines/chemistry , Pyrimidines/pharmacology , Pyrroles/chemistry , Pyrroles/pharmacology , RNA-Dependent RNA Polymerase/metabolism , Viral Proteins/antagonists & inhibitors , Crystallography, X-Ray , Influenza A virus/drug effects , Models, Molecular , Mutant Proteins/metabolism , Mutation/genetics , Protein Domains , Protein Subunits/chemistry , Protein Subunits/metabolism , Quantum Theory , RNA-Dependent RNA Polymerase/antagonists & inhibitors , RNA-Dependent RNA Polymerase/chemistry , Thermodynamics , Viral Proteins/chemistry , Viral Proteins/metabolism
9.
Neuropharmacology ; 189: 108528, 2021 05 15.
Article in English | MEDLINE | ID: mdl-33773999

ABSTRACT

N-methyl-d-aspartate receptors (NMDARs) play an essential role in regulating glutamatergic neurotransmission. Recently, pathogenic missense mutations were identified in genes encoding NMDAR subunits; however, their effect on NMDAR activity is often poorly understood. Here, we examined whether three previously identified pathogenic mutations (M641I, A645S, and Y647S) in the M3 domain of the GluN1 subunit affect the receptor's surface delivery, agonist sensitivity, Mg2+ block, and/or inhibition by the FDA-approved NMDAR blocker memantine. When expressed in HEK293 cells, we found reduced surface expression of GluN1-M641I/GluN2A, GluN1-Y647S/GluN2A, and GluN1-Y647S/GluN2B receptors; other mutation-bearing NMDAR combinations, including GluN1/GluN3A receptors, were expressed at normal surface levels. When expressed in rat hippocampal neurons, we consistently found reduced surface expression of the GluN1-M641I and GluN1-Y647S subunits when compared with wild-type GluN1 subunit. At the functional level, we found that GluN1-M641I/GluN2 and GluN1-A645S/GluN2 receptors expressed in HEK293 cells have wild-type EC50 values for both glutamate and glycine; in contrast, GluN1-Y647S/GluN2 receptors do not produce glutamate-induced currents. In the presence of a physiological concentration of Mg2+, we found that GluN1-M641I/GluN2 receptors have a lower memantine IC50 and slower offset kinetics, whereas GluN1-A645S/GluN2 receptors have a higher memantine IC50 and faster offset kinetics when compared to wild-type receptors. Finally, we found that memantine was the most neuroprotective in hippocampal neurons expressing GluN1-M641I subunits, followed by neurons expressing wild-type GluN1 and then GluN1-A645S subunits in an NMDA-induced excitotoxicity assay. These results indicate that specific pathogenic mutations in the M3 domain of the GluN1 subunit differentially affect the trafficking and functional properties of NMDARs.


Subject(s)
Excitatory Amino Acid Agonists/administration & dosage , Excitatory Amino Acid Antagonists/administration & dosage , Mutation/genetics , Nerve Tissue Proteins/genetics , Protein Subunits/genetics , Receptors, N-Methyl-D-Aspartate/genetics , Animals , Cells, Cultured , Dose-Response Relationship, Drug , Female , HEK293 Cells , Hippocampus/drug effects , Hippocampus/physiology , Humans , Male , Mutation/drug effects , Nerve Tissue Proteins/agonists , Nerve Tissue Proteins/antagonists & inhibitors , Protein Subunits/agonists , Protein Subunits/antagonists & inhibitors , Rats , Rats, Wistar , Receptors, N-Methyl-D-Aspartate/agonists , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , Surface Properties/drug effects
10.
EBioMedicine ; 65: 103242, 2021 Mar.
Article in English | MEDLINE | ID: mdl-33684886

ABSTRACT

BACKGROUND: Type 2 diabetes (T2D) increases the risk of many types of cancer. Dysregulation of proteasome-related protein degradation leads to tumorigenesis, while Exendin-4, a glucagon-like peptide 1 receptor (GLP-1R) agonist, possesses anti-cancer effects. METHODS: We explored the co-expression of proteasome alpha 2 subunit (PSMA2) and GLP-1R in the Cancer Genome Atlas (TCGA) database and human cervical cancer specimens, supplemented by in vivo and in vitro studies using multiple cervical cancer cell lines. FINDINGS: PSMA2 expression was increased in 12 cancer types in TCGA database and cervical cancer specimens from patients with T2D (T2D vs non-T2D: 3.22 (95% confidence interval CI: 1.38, 5.05) vs 1.00 (0.66, 1.34) fold change, P = 0.01). psma2-shRNA decreased cell proliferation in vitro, and tumour volume and Ki67 expression in vivo. Exendin-4 decreased psma2 expression, tumour volume and Ki67 expression in vivo. There was no change in GLP-1R expression in 12 cancer types in TCGA database. However, GLP-1R expression (T2D vs non-T2D: 5.49 (3.0, 8.1) vs 1.00 (0.5, 1.5) fold change, P < 0.001) was increased and positively correlated with PSMA2 expression in T2D-related (r = 0.68)  but not in non-T2D-related cervical cancer specimens. This correlation was corroborated by in vitro experiments where silencing glp-1r decreased psma2 expression. Exendin-4 attenuated phospho-p65 and -IκB expression in the NF-κB pathway. INTERPRETATION: PSMA2 and GLP-1R expression in T2D-related cervical cancer specimens was increased and positively correlated, suggesting hyperglycaemia might promote cancer growth by increasing PSMA2 expression which could be attenuated by Exendin-4. FUNDING: This project was supported by Postdoctoral Fellowship Scheme, Direct Grant, Diabetes Research and Education Fund from the Chinese University of Hong Kong (CUHK).


Subject(s)
Diabetes Mellitus, Type 2/pathology , Exenatide/pharmacology , Glucagon-Like Peptide-1 Receptor/metabolism , Proteasome Endopeptidase Complex/metabolism , Up-Regulation/drug effects , Uterine Cervical Neoplasms/pathology , Case-Control Studies , Cell Line, Tumor , Cell Proliferation/drug effects , Databases, Genetic , Diabetes Mellitus, Type 2/complications , Female , Glucagon-Like Peptide-1 Receptor/genetics , Humans , I-kappa B Proteins/metabolism , Proteasome Endopeptidase Complex/chemistry , Proteasome Endopeptidase Complex/genetics , Protein Subunits/antagonists & inhibitors , Protein Subunits/genetics , Protein Subunits/metabolism , RNA Interference , RNA, Small Interfering/metabolism , Signal Transduction/drug effects , Transcription Factor RelA/metabolism , Uterine Cervical Neoplasms/complications
11.
Amyloid ; 28(1): 24-29, 2021 Mar.
Article in English | MEDLINE | ID: mdl-32811187

ABSTRACT

Transthyretin (TTR) tetramer dissociation is rate limiting for aggregation and subunit exchange. Slowing of TTR tetramer dissociation via kinetic stabiliser binding slows cardiomyopathy progression. Quadruplicate subunit exchange comparisons of the drug candidate AG10, and the drugs tolcapone, diflunisal, and tafamidis were carried out at 1, 5, 10, 20 and 30 µM concentrations in 4 distinct pooled wild type TTR (TTRwt) human plasma samples. These experiments reveal that the concentration dependence of the efficacy of each compound at inhibiting TTR dissociation was primarily determined by the ratio between the stabiliser's dissociation constants from TTR and albumin, which competes with TTR to bind kinetic stabilisers. The best stabilisers, tafamidis (80 mg QD), AG10 (800 mg BID), and tolcapone (3 x 100 mg over 12 h), exhibit very similar kinetic stabilisation at the plasma concentrations resulting from these doses. At a 10 µM plasma concentration, AG10 is slightly more potent as a kinetic stabiliser vs. tolcapone and tafamidis (which are similar), which are substantially more potent than diflunisal. Dissociation of TTR can be limited to 10% of its normal rate at concentrations of 5.7 µM AG10, 10.3 µM tolcapone, 12.0 µM tafamidis, and 188 µM diflunisal. The potency similarities revealed by our study suggest that differences in safety, adsorption and metabolism, pharmacokinetics, and tissue distribution become important for kinetic stabiliser clinical use decisions.


Subject(s)
Amyloid Neuropathies, Familial/drug therapy , Amyloid/genetics , Cardiomyopathies/drug therapy , Prealbumin/genetics , Amyloid/antagonists & inhibitors , Amyloid/blood , Amyloid/chemistry , Amyloid Neuropathies, Familial/blood , Amyloid Neuropathies, Familial/genetics , Amyloid Neuropathies, Familial/pathology , Benzoates/pharmacology , Benzoxazoles/pharmacology , Cardiomyopathies/blood , Cardiomyopathies/genetics , Cardiomyopathies/pathology , Diflunisal/pharmacology , Humans , Kinetics , Prealbumin/chemistry , Protein Aggregates/drug effects , Protein Binding/drug effects , Protein Multimerization/drug effects , Protein Subunits/antagonists & inhibitors , Protein Subunits/blood , Protein Subunits/chemistry , Protein Subunits/genetics , Pyrazoles/pharmacology , Tolcapone/pharmacology
12.
J Med Chem ; 63(21): 13103-13123, 2020 11 12.
Article in English | MEDLINE | ID: mdl-33095579

ABSTRACT

The immunoproteasome (i-20S) has emerged as a therapeutic target for autoimmune and inflammatory disorders and hematological malignancies. Inhibition of the chymotryptic ß5i subunit of i-20S inhibits T cell activation, B cell proliferation, and dendritic cell differentiation in vitro and suppresses immune responses in animal models of autoimmune disorders and allograft rejection. However, cytotoxicity to immune cells has accompanied the use of covalently reactive ß5i inhibitors, whose activity against the constitutive proteasome (c-20S) is cumulative with the time of exposure. Herein, we report a structure-activity relationship study of a class of noncovalent proteasome inhibitors with picomolar potencies and 1000-fold selectivity for i-20S over c-20S. Furthermore, these inhibitors are specific for ß5i over the other five active subunits of i-20S and c-20S, providing useful tools to study the functions of ß5i in immune responses. The potency of these compounds in inhibiting human T cell activation suggests that they may have therapeutic potential.


Subject(s)
Dipeptides/chemistry , Proteasome Endopeptidase Complex/metabolism , Proteasome Inhibitors/chemistry , Binding Sites , Cell Proliferation/drug effects , Dipeptides/metabolism , Dipeptides/pharmacology , HeLa Cells , Humans , Inhibitory Concentration 50 , Kinetics , Lymphocyte Activation/drug effects , Proteasome Endopeptidase Complex/chemistry , Proteasome Inhibitors/metabolism , Proteasome Inhibitors/pharmacology , Protein Binding , Protein Subunits/antagonists & inhibitors , Protein Subunits/metabolism , Structure-Activity Relationship , T-Lymphocytes/cytology , T-Lymphocytes/drug effects , T-Lymphocytes/metabolism
13.
Proc Natl Acad Sci U S A ; 117(41): 25851-25858, 2020 10 13.
Article in English | MEDLINE | ID: mdl-32999066

ABSTRACT

Ionotropic glutamate receptors (iGluRs) are key molecules for synaptic signaling in the central nervous system, which makes them promising drug targets. Intensive efforts are being devoted to the development of subunit-selective ligands, which should enable more precise pharmacologic interventions while limiting the effects on overall neuronal circuit function. However, many AMPA and kainate receptor complexes in vivo are heteromers composed of different subunits. Despite their importance, little is known about how subunit-selective ligands affect the gating of heteromeric iGluRs, namely their activation and desensitization properties. Using fast ligand application experiments, we studied the effects of competitive antagonists that block glutamate from binding at part of the four subunits. We found that UBP-310, a kainate receptor antagonist with high selectivity for GluK1 subunits, reduces the desensitization of GluK1/GluK2 heteromers and fully abolishes the desensitization of GluK1/GluK5 heteromers. This effect is mirrored by subunit-selective agonists and heteromeric receptors that contain binding-impaired subunits, as we show for both kainate and GluA2 AMPA receptors. These findings are consistent with a model in which incomplete agonist occupancy at the four receptor subunits can provide activation without inducing desensitization. However, we did not detect significant steady-state currents during UBP-310 dissociation from GluK1 homotetramers, indicating that antagonist dissociation proceeds in a nonuniform and cooperativity-driven manner, which disfavors nondesensitizing occupancy states. Besides providing mechanistic insights, these results have direct implications for the use of subunit-selective antagonists in neuroscience research and envisioned therapeutic interventions.


Subject(s)
Protein Subunits/antagonists & inhibitors , Receptors, Ionotropic Glutamate/chemistry , Receptors, Ionotropic Glutamate/metabolism , Dimerization , HEK293 Cells , Humans , Ligands , Protein Subunits/chemistry , Protein Subunits/genetics , Protein Subunits/metabolism , Receptors, Ionotropic Glutamate/antagonists & inhibitors , Receptors, Ionotropic Glutamate/genetics
14.
Signal Transduct Target Ther ; 5(1): 220, 2020 10 06.
Article in English | MEDLINE | ID: mdl-33024075
15.
Nat Commun ; 11(1): 4370, 2020 09 01.
Article in English | MEDLINE | ID: mdl-32873792

ABSTRACT

BRAF kinase, a critical effector of the ERK signaling pathway, is hyperactivated in many cancers. Oncogenic BRAFV600E signals as an active monomer in the absence of active RAS, however, in many tumors BRAF dimers mediate ERK signaling. FDA-approved RAF inhibitors poorly inhibit BRAF dimers, which leads to tumor resistance. We found that Ponatinib, an FDA-approved drug, is an effective inhibitor of BRAF monomers and dimers. Ponatinib binds the BRAF dimer and stabilizes a distinct αC-helix conformation through interaction with a previously unrevealed allosteric site. Using these structural insights, we developed PHI1, a BRAF inhibitor that fully uncovers the allosteric site. PHI1 exhibits discrete cellular selectivity for BRAF dimers, with enhanced inhibition of the second protomer when the first protomer is occupied, comprising a novel class of dimer selective inhibitors. This work shows that Ponatinib and BRAF dimer selective inhibitors will be useful in treating BRAF-dependent tumors.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , MAP Kinase Signaling System/drug effects , Neoplasms/drug therapy , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins B-raf/antagonists & inhibitors , Allosteric Site/drug effects , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Cell Line, Tumor , Crystallography, X-Ray , Drug Design , Drug Screening Assays, Antitumor , Humans , Imidazoles/pharmacology , Imidazoles/therapeutic use , MAP Kinase Signaling System/genetics , Molecular Docking Simulation , Mutation , Neoplasms/genetics , Neoplasms/pathology , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/therapeutic use , Protein Multimerization/drug effects , Protein Subunits/antagonists & inhibitors , Protein Subunits/genetics , Protein Subunits/metabolism , Proto-Oncogene Proteins B-raf/genetics , Proto-Oncogene Proteins B-raf/metabolism , Proto-Oncogene Proteins B-raf/ultrastructure , Pyridazines/pharmacology , Pyridazines/therapeutic use , Small Molecule Libraries , Structure-Activity Relationship
16.
Neuropharmacology ; 177: 108256, 2020 10 15.
Article in English | MEDLINE | ID: mdl-32738308

ABSTRACT

Nicotine is a highly addictive drug found in tobacco that drives its continued use despite the harmful consequences. The initiation of nicotine abuse involves the mesolimbic dopamine system, which contributes to the rewarding sensory stimuli and associative learning processes in the beginning stages of addiction. Nicotine binds to neuronal nicotinic acetylcholine receptors (nAChRs), which come in a diverse collection of subtypes. The nAChRs that contain the α4 and ß2 subunits, often in combination with the α6 subunit, are particularly important for nicotine's ability to increase midbrain dopamine neuron firing rates and phasic burst firing. Chronic nicotine exposure results in numerous neuroadaptations, including the upregulation of particular nAChR subtypes associated with long-term desensitization of the receptors. When nicotine is no longer present, for example during attempts to quit smoking, a withdrawal syndrome develops. The expression of physical withdrawal symptoms depends mainly on the α2, α3, α5, and ß4 nicotinic subunits in the epithalamic habenular complex and its target regions. Thus, nicotine affects diverse neural systems and an array of nAChR subtypes to mediate the overall addiction process. This article is part of the special issue on 'Contemporary Advances in Nicotine Neuropharmacology'.


Subject(s)
Brain/metabolism , Nicotine/metabolism , Receptors, Nicotinic/metabolism , Tobacco Use Disorder/metabolism , Animals , Brain/drug effects , Humans , Nicotine/administration & dosage , Nicotinic Agonists/administration & dosage , Nicotinic Agonists/metabolism , Nicotinic Antagonists/administration & dosage , Nicotinic Antagonists/metabolism , Protein Subunits/agonists , Protein Subunits/antagonists & inhibitors , Protein Subunits/chemistry , Protein Subunits/metabolism , Receptors, Nicotinic/chemistry , Tobacco Use Disorder/psychology
17.
Proc Natl Acad Sci U S A ; 117(28): 16283-16291, 2020 07 14.
Article in English | MEDLINE | ID: mdl-32611810

ABSTRACT

The difficulty of achieving robust functional expression of insect nicotinic acetylcholine receptors (nAChRs) has hampered our understanding of these important molecular targets of globally deployed neonicotinoid insecticides at a time when concerns have grown regarding the toxicity of this chemotype to insect pollinators. We show that thioredoxin-related transmembrane protein 3 (TMX3) is essential to enable robust expression in Xenopus laevis oocytes of honeybee (Apis mellifera) and bumblebee (Bombus terrestris) as well as fruit fly (Drosophila melanogaster) nAChR heteromers targeted by neonicotinoids and not hitherto robustly expressed. This has enabled the characterization of picomolar target site actions of neonicotinoids, findings important in understanding their toxicity.


Subject(s)
Insect Proteins/metabolism , Insecticides/pharmacology , Neonicotinoids/pharmacology , Nicotinic Agonists/pharmacology , Receptors, Nicotinic/metabolism , Acetylcholine/pharmacology , Animals , Bees/metabolism , Dose-Response Relationship, Drug , Drosophila melanogaster/metabolism , Insect Proteins/agonists , Insect Proteins/genetics , Oocytes/metabolism , Protein Subunits/antagonists & inhibitors , Protein Subunits/genetics , Protein Subunits/metabolism , Receptors, Nicotinic/genetics , Thioredoxins/genetics , Thioredoxins/metabolism , Xenopus laevis
18.
Cell ; 182(2): 357-371.e13, 2020 07 23.
Article in English | MEDLINE | ID: mdl-32610085

ABSTRACT

Excitatory neurotransmission meditated by glutamate receptors including N-methyl-D-aspartate receptors (NMDARs) is pivotal to brain development and function. NMDARs are heterotetramers composed of GluN1 and GluN2 subunits, which bind glycine and glutamate, respectively, to activate their ion channels. Despite importance in brain physiology, the precise mechanisms by which activation and inhibition occur via subunit-specific binding of agonists and antagonists remain largely unknown. Here, we show the detailed patterns of conformational changes and inter-subunit and -domain reorientation leading to agonist-gating and subunit-dependent competitive inhibition by providing multiple structures in distinct ligand states at 4 Å or better. The structures reveal that activation and competitive inhibition by both GluN1 and GluN2 antagonists occur by controlling the tension of the linker between the ligand-binding domain and the transmembrane ion channel of the GluN2 subunit. Our results provide detailed mechanistic insights into NMDAR pharmacology, activation, and inhibition, which are fundamental to the brain physiology.


Subject(s)
Receptors, N-Methyl-D-Aspartate/metabolism , Binding Sites , Binding, Competitive , Cryoelectron Microscopy , Crystallography, X-Ray , Dimerization , Glutamic Acid/chemistry , Glutamic Acid/metabolism , Glycine/chemistry , Glycine/metabolism , Humans , Ligands , Molecular Dynamics Simulation , Protein Structure, Quaternary , Protein Subunits/agonists , Protein Subunits/antagonists & inhibitors , Protein Subunits/genetics , Protein Subunits/metabolism , Receptors, N-Methyl-D-Aspartate/agonists , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , Receptors, N-Methyl-D-Aspartate/genetics , Recombinant Proteins/biosynthesis , Recombinant Proteins/chemistry , Recombinant Proteins/isolation & purification
19.
Annu Rev Biochem ; 89: 45-75, 2020 06 20.
Article in English | MEDLINE | ID: mdl-32569524

ABSTRACT

Ribonucleotide reductases (RNRs) catalyze the de novo conversion of nucleotides to deoxynucleotides in all organisms, controlling their relative ratios and abundance. In doing so, they play an important role in fidelity of DNA replication and repair. RNRs' central role in nucleic acid metabolism has resulted in five therapeutics that inhibit human RNRs. In this review, we discuss the structural, dynamic, and mechanistic aspects of RNR activity and regulation, primarily for the human and Escherichia coli class Ia enzymes. The unusual radical-based organic chemistry of nucleotide reduction, the inorganic chemistry of the essential metallo-cofactor biosynthesis/maintenance, the transport of a radical over a long distance, and the dynamics of subunit interactions all present distinct entry points toward RNR inhibition that are relevant for drug discovery. We describe the current mechanistic understanding of small molecules that target different elements of RNR function, including downstream pathways that lead to cell cytotoxicity. We conclude by summarizing novel and emergent RNR targeting motifs for cancer and antibiotic therapeutics.


Subject(s)
Anti-Bacterial Agents/chemistry , Antineoplastic Agents/chemistry , Escherichia coli Infections/drug therapy , Neoplasms/drug therapy , Nucleotides/metabolism , Ribonucleotide Reductases/chemistry , Anti-Bacterial Agents/therapeutic use , Antineoplastic Agents/therapeutic use , Biocatalysis , Drug Discovery/methods , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/therapeutic use , Escherichia coli/drug effects , Escherichia coli/enzymology , Escherichia coli/genetics , Escherichia coli Infections/enzymology , Escherichia coli Infections/genetics , Escherichia coli Infections/microbiology , Humans , Molecular Docking Simulation , Neoplasms/enzymology , Neoplasms/genetics , Neoplasms/pathology , Nucleotides/chemistry , Oxidation-Reduction , Protein Structure, Secondary , Protein Subunits/antagonists & inhibitors , Protein Subunits/chemistry , Protein Subunits/genetics , Protein Subunits/metabolism , Ribonucleotide Reductases/antagonists & inhibitors , Ribonucleotide Reductases/genetics , Ribonucleotide Reductases/metabolism , Small Molecule Libraries/chemistry , Small Molecule Libraries/therapeutic use , Structure-Activity Relationship
20.
Biochem Pharmacol ; 181: 114124, 2020 11.
Article in English | MEDLINE | ID: mdl-32593612

ABSTRACT

The α9α10 nicotinic acetylcholine receptor (nAChR) has been characterized as an effective anti-pain target that functions through a non-opioid mechanism. However, as a pentameric ion channel comprised of two different subunits, the specific targeting of α9α10 nAChRs has proven challenging. Previously the 13-amino-acid peptide, RgIA, was shown to block α9α10 nAChRs with high potency and specificity. This peptide, characterized from the venom of the carnivorous marine snail, Conus regius, produced analgesia in several rodent models of chronic pain. Despite promising pre-clinical data in behavioral assays, the number of specific α9α10 nAChR antagonists remains small and the physiological mechanisms of analgesia remain cryptic. In this study, we implement amino-acid substitutions to definitively characterize the chemical properties of RgIA that contribute to its activity against α9α10 nAChRs. Using this mutational approach, we determined the vital role of biochemical side-chain properties and amino acids in the second loop that are amenable to substitutions to further engineer next-generation analogs for the blockade of α9α10 nAChRs.


Subject(s)
Amino Acid Substitution , Amino Acids/genetics , Conotoxins/genetics , Receptors, Nicotinic/metabolism , Amino Acid Sequence , Amino Acids/chemistry , Amino Acids/metabolism , Animals , Binding Sites/genetics , Conotoxins/metabolism , Conotoxins/pharmacology , Humans , Mollusk Venoms/chemistry , Mollusk Venoms/metabolism , Nicotinic Antagonists/metabolism , Nicotinic Antagonists/pharmacology , Oocytes/drug effects , Oocytes/metabolism , Oocytes/physiology , Protein Subunits/antagonists & inhibitors , Protein Subunits/genetics , Protein Subunits/metabolism , Rats , Receptors, Nicotinic/genetics , Sequence Homology, Amino Acid , Xenopus laevis
SELECTION OF CITATIONS
SEARCH DETAIL
...