Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 38
Filter
Add more filters











Publication year range
1.
Biomaterials ; 285: 121536, 2022 06.
Article in English | MEDLINE | ID: mdl-35533442

ABSTRACT

Soft polymer nanoparticles designed to disassemble and release an antagonist of the neurokinin 1 receptor (NK1R) in endosomes provide efficacious yet transient relief from chronic pain. These micellar nanoparticles are unstable and rapidly release cargo, which may limit the duration of analgesia. We examined the efficacy of stable star polymer nanostars containing the NK1R antagonist aprepitant-amine for the treatment of chronic pain in mice. Nanostars continually released cargo for 24 h, trafficked through the endosomal system, and disrupted NK1R endosomal signaling. After intrathecal injection, nanostars accumulated in endosomes of spinal neurons. Nanostar-aprepitant reversed mechanical, thermal and cold allodynia and normalized nociceptive behavior more efficaciously than free aprepitant in preclinical models of neuropathic and inflammatory pain. Analgesia was maintained for >10 h. The sustained endosomal delivery of antagonists from slow-release nanostars provides effective and long-lasting reversal of chronic pain.


Subject(s)
Chronic Pain , Neurokinin-1 Receptor Antagonists , Animals , Aprepitant/pharmacology , Aprepitant/therapeutic use , Chronic Pain/drug therapy , Endosomes , Mice , Neurokinin-1 Receptor Antagonists/pharmacology , Neurokinin-1 Receptor Antagonists/therapeutic use , Polymers/pharmacology
2.
J Biol Chem ; 296: 100345, 2021.
Article in English | MEDLINE | ID: mdl-33515548

ABSTRACT

G-protein-coupled receptors (GPCRs) are traditionally known for signaling at the plasma membrane, but they can also signal from endosomes after internalization to control important pathophysiological processes. In spinal neurons, sustained endosomal signaling of the neurokinin 1 receptor (NK1R) mediates nociception, as demonstrated in models of acute and neuropathic pain. An NK1R antagonist, Spantide I (Span), conjugated to cholestanol (Span-Chol), accumulates in endosomes, inhibits endosomal NK1R signaling, and causes prolonged antinociception. However, the extent to which the Chol-anchor influences long-term location and activity is poorly understood. Herein, we used fluorescent correlation spectroscopy and targeted biosensors to characterize Span-Chol over time. The Chol-anchor increased local concentration of probe at the plasma membrane. Over time we observed an increase in NK1R-binding affinity and more potent inhibition of NK1R-mediated calcium signaling. Span-Chol, but not Span, caused a persistent decrease in NK1R recruitment of ß-arrestin and receptor internalization to early endosomes. Using targeted biosensors, we mapped the relative inhibition of NK1R signaling as the receptor moved into the cell. Span selectively inhibited cell surface signaling, whereas Span-Chol partitioned into endosomal membranes and blocked endosomal signaling. In a preclinical model of pain, Span-Chol caused prolonged antinociception (>9 h), which is attributable to a three-pronged mechanism of action: increased local concentration at membranes, a prolonged decrease in NK1R endocytosis, and persistent inhibition of signaling from endosomes. Identifying the mechanisms that contribute to the increased preclinical efficacy of lipid-anchored NK1R antagonists is an important step toward understanding how we can effectively target intracellular GPCRs in disease.


Subject(s)
Analgesics/pharmacology , Cholestanol/pharmacology , Neurokinin-1 Receptor Antagonists/pharmacology , Pain/drug therapy , Substance P/analogs & derivatives , Analgesics/chemistry , Analgesics/therapeutic use , Animals , Cell Membrane/drug effects , Cell Membrane/metabolism , Cholestanol/analogs & derivatives , Cholestanol/therapeutic use , Endosomes/drug effects , Endosomes/metabolism , HEK293 Cells , Humans , Male , Mice, Inbred C57BL , Neurokinin-1 Receptor Antagonists/chemistry , Neurokinin-1 Receptor Antagonists/therapeutic use , Pain/metabolism , Pain Management , Substance P/chemistry , Substance P/pharmacology , Substance P/therapeutic use
3.
Nat Commun ; 11(1): 4482, 2020 09 08.
Article in English | MEDLINE | ID: mdl-32901011

ABSTRACT

Intracellular trafficking governs receptor signaling, pathogenesis, immune responses and fate of nanomedicines. These processes are typically tracked by observing colocalization of fluorescent markers using confocal microscopy. However, this method is low throughput, limited by the resolution of microscopy, and can miss fleeting interactions. To address this, we developed a localization sensor composed of a quenched SNAP-tag substrate (SNAPSwitch) that can be conjugated to biomolecules using click chemistry. SNAPSwitch enables quantitative detection of trafficking to locations of interest within live cells using flow cytometry. Using SNAPSwitch, we followed the trafficking of DNA complexes from endosomes into the cytosol and nucleus. We show that antibodies against the transferrin or hyaluronan receptor are initially sorted into different compartments following endocytosis. In addition, we can resolve which side of the cellular membrane material was located. These results demonstrate SNAPSwitch is a high-throughput and broadly applicable tool to quantitatively track localization of materials in cells.


Subject(s)
DNA/metabolism , Molecular Probes/chemistry , Nanoparticles/metabolism , Proteins/metabolism , Animals , Biological Transport, Active , Biosensing Techniques/methods , Click Chemistry , Flow Cytometry , Fluorescent Dyes , HEK293 Cells , Humans , Mice , Microscopy, Confocal , Molecular Probe Techniques , Molecular Probes/metabolism , NIH 3T3 Cells
4.
ACS Chem Biol ; 15(3): 718-727, 2020 03 20.
Article in English | MEDLINE | ID: mdl-32022538

ABSTRACT

Cathepsin X/Z/P is cysteine cathepsin with unique carboxypeptidase activity. Its expression is associated with cancer and neurodegenerative diseases, although its roles during normal physiology are still poorly understood. Advances in our understanding of its function have been hindered by a lack of available tools that can specifically measure the proteolytic activity of cathepsin X. We present a series of activity-based probes that incorporate a sulfoxonium ylide warhead, which exhibit improved specificity for cathepsin X compared to previously reported probes. We apply these probes to detect cathepsin X activity in cell and tissue lysates, in live cells and in vivo, and to localize active cathepsin X in mouse tissues by microscopy. Finally, we utilize an improved method to generate chloromethylketones, necessary intermediates for synthesis of acyloxymethylketones probes, by way of sulfoxonium ylide intermediates. In conclusion, the probes presented in this study will be valuable for investigating cathepsin X pathophysiology.


Subject(s)
Cathepsins/antagonists & inhibitors , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/metabolism , Fluorescent Dyes/chemistry , Amino Acids/chemistry , Animals , Cell Culture Techniques , Cell Line , Diazomethane/chemistry , Humans , Hydrocarbons, Fluorinated/chemistry , Ketones/chemistry , Kidney/cytology , Kidney/diagnostic imaging , Kinetics , Male , Mice , Mice, Inbred C57BL , Optical Imaging , Protein Domains , Small Molecule Libraries/chemistry , Structure-Activity Relationship , Substrate Specificity
5.
Article in English | Sec. Est. Saúde SP, SESSP-IBPROD, Sec. Est. Saúde SP | ID: but-ib17422

ABSTRACT

Granzyme A (GzmA) is secreted by cytotoxic lymphocytes and has traditionally been viewed as a mediator of cell death. However, a growing body of data suggests the physiological role of GzmA is promotion of inflammation. Here, we show that GzmA is significantly elevated in the sera of chikungunya virus (CHIKV) patients and that GzmA levels correlated with viral loads and disease scores in these patients. Serum GzmA levels were also elevated in CHIKV mouse models, with NK cells the likely source. Infection of mice deficient in type I interferon responses with CHIKV, Zika virus, or dengue virus resulted in high levels of circulating GzmA. We also show that subcutaneous injection of enzymically active recombinant mouse GzmA was able to mediate inflammation, both locally at the injection site as well as at a distant site. Protease activated receptors (PARs) may represent targets for GzmA, and we show that treatment with PAR antagonist ameliorated GzmA- and CHIKV-mediated inflammation.

6.
Front Immunol, v. 10, 3083, jan. 2020
Article in English | Sec. Est. Saúde SP, SESSP-IBPROD, Sec. Est. Saúde SP | ID: bud-2931

ABSTRACT

Granzyme A (GzmA) is secreted by cytotoxic lymphocytes and has traditionally been viewed as a mediator of cell death. However, a growing body of data suggests the physiological role of GzmA is promotion of inflammation. Here, we show that GzmA is significantly elevated in the sera of chikungunya virus (CHIKV) patients and that GzmA levels correlated with viral loads and disease scores in these patients. Serum GzmA levels were also elevated in CHIKV mouse models, with NK cells the likely source. Infection of mice deficient in type I interferon responses with CHIKV, Zika virus, or dengue virus resulted in high levels of circulating GzmA. We also show that subcutaneous injection of enzymically active recombinant mouse GzmA was able to mediate inflammation, both locally at the injection site as well as at a distant site. Protease activated receptors (PARs) may represent targets for GzmA, and we show that treatment with PAR antagonist ameliorated GzmA- and CHIKV-mediated inflammation.

7.
Sci Rep ; 9(1): 13295, 2019 09 16.
Article in English | MEDLINE | ID: mdl-31527638

ABSTRACT

Neutrophil elastase is a serine protease that has been implicated in the pathogenesis of inflammatory bowel disease. Due to post-translational control of its activation and high expression of its inhibitors in the gut, measurements of total expression poorly reflect the pool of active, functional neutrophil elastase. Fluorogenic substrate probes have been used to measure neutrophil elastase activity, though these tools lack specificity and traceability. PK105 is a recently described fluorescent activity-based probe, which binds to neutrophil elastase in an activity-dependent manner. The irreversible nature of this probe allows for accurate identification of its targets in complex protein mixtures. We describe the reactivity profile of PK105b, a new analogue of PK105, against recombinant serine proteases and in tissue extracts from healthy mice and from models of inflammation induced by oral cancer and Legionella pneumophila infection. We apply PK105b to measure neutrophil elastase activation in an acute model of experimental colitis. Neutrophil elastase activity is detected in inflamed, but not healthy, colons. We corroborate this finding in mucosal biopsies from patients with ulcerative colitis. Thus, PK105b facilitates detection of neutrophil elastase activity in tissue lysates, and we have applied it to demonstrate that this protease is unequivocally activated during colitis.


Subject(s)
Colitis, Ulcerative/immunology , Colitis, Ulcerative/pathology , Leukocyte Elastase/metabolism , Neutrophil Activation/immunology , Neutrophils/immunology , Animals , Cells, Cultured , Female , Humans , Legionella pneumophila/immunology , Legionnaires' Disease/pathology , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Nude , Mouth Neoplasms/pathology
8.
Front Immunol ; 10: 3083, 2019.
Article in English | MEDLINE | ID: mdl-31993061

ABSTRACT

Granzyme A (GzmA) is secreted by cytotoxic lymphocytes and has traditionally been viewed as a mediator of cell death. However, a growing body of data suggests the physiological role of GzmA is promotion of inflammation. Here, we show that GzmA is significantly elevated in the sera of chikungunya virus (CHIKV) patients and that GzmA levels correlated with viral loads and disease scores in these patients. Serum GzmA levels were also elevated in CHIKV mouse models, with NK cells the likely source. Infection of mice deficient in type I interferon responses with CHIKV, Zika virus, or dengue virus resulted in high levels of circulating GzmA. We also show that subcutaneous injection of enzymically active recombinant mouse GzmA was able to mediate inflammation, both locally at the injection site as well as at a distant site. Protease activated receptors (PARs) may represent targets for GzmA, and we show that treatment with PAR antagonist ameliorated GzmA- and CHIKV-mediated inflammation.


Subject(s)
Arbovirus Infections/immunology , Chikungunya Fever/immunology , Granzymes/immunology , Inflammation/immunology , Killer Cells, Natural/immunology , Animals , Granzymes/blood , Humans , Mice , Mice, Inbred C57BL
9.
J Med Chem ; 61(18): 8444-8456, 2018 09 27.
Article in English | MEDLINE | ID: mdl-30156410

ABSTRACT

A series of 5-substituted tetrahydroisoquinolines was synthesized via a 10-step linear synthesis to assess whether replacement of noscapine's southern isobenzofuranone with other moieties resulted in retained cytotoxic activity. One such molecule, 18g, bearing a para-methoxybenzyl functionality with N-ethylcarbamoyl substitution, produced cell-cycle arrest at the G2/M phase with an EC50 of 2.7 µM in the MCF-7 breast-cancer cell line, a 7-fold increase compared with that of noscapine (5). This molecule had similar activity (EC50 of 2.5 µM) against the resistant NCI/AdrRES cell line, demonstrating its potential to overcome or avert known resistance mechanisms, unlike current cytotoxic agents. Compound 18g was found to modify the drug-efflux activity of P-gp and, in combination studies, potentiate the antiproliferative activity of vinblastine. These results provide insights into structural modifications to noscapine that will guide future development toward more potent cytotoxic agents that are active against resistant cancer cells.


Subject(s)
Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/pharmacology , Breast Neoplasms/pathology , Cytotoxins/chemical synthesis , Cytotoxins/pharmacology , Noscapine/chemistry , Pancreatic Neoplasms/pathology , Tetrahydroisoquinolines/chemistry , Apoptosis , Breast Neoplasms/drug therapy , Cell Division , Cell Proliferation , Female , Humans , Models, Molecular , Molecular Structure , Pancreatic Neoplasms/drug therapy , Protein Conformation , Structure-Activity Relationship , Tumor Cells, Cultured
10.
Proc Natl Acad Sci U S A ; 115(31): E7438-E7447, 2018 07 31.
Article in English | MEDLINE | ID: mdl-30012612

ABSTRACT

Once activated at the surface of cells, G protein-coupled receptors (GPCRs) redistribute to endosomes, where they can continue to signal. Whether GPCRs in endosomes generate signals that contribute to human disease is unknown. We evaluated endosomal signaling of protease-activated receptor-2 (PAR2), which has been proposed to mediate pain in patients with irritable bowel syndrome (IBS). Trypsin, elastase, and cathepsin S, which are activated in the colonic mucosa of patients with IBS and in experimental animals with colitis, caused persistent PAR2-dependent hyperexcitability of nociceptors, sensitization of colonic afferent neurons to mechanical stimuli, and somatic mechanical allodynia. Inhibitors of clathrin- and dynamin-dependent endocytosis and of mitogen-activated protein kinase kinase-1 prevented trypsin-induced hyperexcitability, sensitization, and allodynia. However, they did not affect elastase- or cathepsin S-induced hyperexcitability, sensitization, or allodynia. Trypsin stimulated endocytosis of PAR2, which signaled from endosomes to activate extracellular signal-regulated kinase. Elastase and cathepsin S did not stimulate endocytosis of PAR2, which signaled from the plasma membrane to activate adenylyl cyclase. Biopsies of colonic mucosa from IBS patients released proteases that induced persistent PAR2-dependent hyperexcitability of nociceptors, and PAR2 association with ß-arrestins, which mediate endocytosis. Conjugation to cholestanol promoted delivery and retention of antagonists in endosomes containing PAR2 A cholestanol-conjugated PAR2 antagonist prevented persistent trypsin- and IBS protease-induced hyperexcitability of nociceptors. The results reveal that PAR2 signaling from endosomes underlies the persistent hyperexcitability of nociceptors that mediates chronic pain of IBS. Endosomally targeted PAR2 antagonists are potential therapies for IBS pain. GPCRs in endosomes transmit signals that contribute to human diseases.


Subject(s)
Chronic Pain/etiology , Endosomes/physiology , Irritable Bowel Syndrome/physiopathology , Receptor, PAR-2/physiology , Signal Transduction/physiology , Animals , Endocytosis , Extracellular Signal-Regulated MAP Kinases/physiology , Humans , Nociception , Nociceptors/physiology , Trypsin/pharmacology
11.
ACS Sens ; 3(6): 1182-1189, 2018 06 22.
Article in English | MEDLINE | ID: mdl-29676153

ABSTRACT

The ability to determine the amount of material endocytosed by a cell is important for our understanding of cell biology and in the design of effective carriers for drug delivery. To quantify internalization by fluorescence, the signal from material remaining on the cell surface must be differentiated from endocytosed material. Sensors for internalization offer advantages over traditional methods for achieving this as they exhibit improved sensitivity, allow for multiple fluorescent markers to be used simultaneously, and are amenable to high-throughput analysis. We have developed a small fluorescent internalization sensor, similar in size to a standard fluorescent dye, that can be conjugated to proteins and uses the rapid and highly specific bio-orthogonal reaction between a tetrazine and a trans-cyclooctene group to switch off the surface signal. The sensor can be attached to a variety of materials using simple chemistry and is compatible with flow cytometry and fluorescence microscopy, making it a useful tool to study the uptake of material into cells.


Subject(s)
Fluorescent Dyes/chemistry , Oligonucleotides/chemistry , Cells, Cultured , Click Chemistry , Endocytosis , Flow Cytometry , Fluorescence , Humans , Microscopy, Fluorescence , Molecular Structure , Surface Properties
12.
J Med Chem ; 61(5): 2087-2103, 2018 03 08.
Article in English | MEDLINE | ID: mdl-29446948

ABSTRACT

The adenosine A1 receptor (A1AR) is a potential novel therapeutic target for myocardial ischemia-reperfusion injury. However, to date, clinical translation of prototypical A1AR agonists has been hindered due to dose limiting adverse effects. Recently, we demonstrated that the biased bitopic agonist 1, consisting of an adenosine pharmacophore linked to an allosteric moiety, could stimulate cardioprotective A1AR signaling in the absence of unwanted bradycardia. Therefore, this study aimed to investigate the structure-activity relationship of compound 1 biased agonism. A series of novel derivatives of 1 were synthesized and pharmacologically profiled. Modifications were made to the orthosteric adenosine pharmacophore, linker, and allosteric 2-amino-3-benzoylthiophene pharmacophore to probe the structure-activity relationships, particularly in terms of biased signaling, as well as A1AR activity and subtype selectivity. Collectively, our findings demonstrate that the allosteric moiety, particularly the 4-(trifluoromethyl)phenyl substituent of the thiophene scaffold, is important in conferring bitopic ligand bias at the A1AR.


Subject(s)
Adenosine A1 Receptor Agonists , Adenosine/analogs & derivatives , Adenosine/chemical synthesis , Adenosine/pharmacology , Adenosine A1 Receptor Agonists/adverse effects , Adenosine A1 Receptor Agonists/chemical synthesis , Allosteric Regulation , Animals , Cricetinae , Humans , Ligands , Phenols/chemistry , Structure-Activity Relationship , Thiophenes/chemistry
13.
Chemistry ; 24(8): 1922-1930, 2018 Feb 06.
Article in English | MEDLINE | ID: mdl-29171692

ABSTRACT

Dihydropteroate synthase (DHPS) is an enzyme of the folate biosynthesis pathway, which catalyzes the formation of 7,8-dihydropteroate (DHPt) from 6-hydroxymethyl-7,8-dihydropterin pyrophosphate (DHPPP) and para-aminobenzoic acid (pABA). DHPS is the long-standing target of the sulfonamide class of antibiotics that compete with pABA. In the wake of sulfa drug resistance, targeting the structurally rigid (and more conserved) pterin site has been proposed as an alternate strategy to inhibit DHPS in wild-type and sulfa drug resistant strains. Following the work on developing pterin-site inhibitors of the adjacent enzyme 6-hydroxymethyl-7,8-dihydropterin pyrophosphokinase (HPPK), we now present derivatives of 8-mercaptoguanine, a fragment that binds weakly within both enzymes, and quantify sub-µm binding using surface plasmon resonance (SPR) to Escherichia coli DHPS (EcDHPS). Eleven ligand-bound EcDHPS crystal structures delineate the structure-activity relationship observed providing a structural framework for the rational development of novel, substrate-envelope-compliant DHPS inhibitors.


Subject(s)
Dihydropteroate Synthase/antagonists & inhibitors , Enzyme Inhibitors/chemistry , Guanine/analogs & derivatives , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/metabolism , Binding Sites , Catalytic Domain , Crystallography, X-Ray , Dihydropteroate Synthase/metabolism , Enzyme Inhibitors/metabolism , Escherichia coli/enzymology , Guanine/metabolism , Hydrogen Bonding , Ligands , Protein Binding , Protein Structure, Tertiary , Structure-Activity Relationship , Substrate Specificity , Sulfonamides/chemistry , Surface Plasmon Resonance
14.
Proc Natl Acad Sci U S A ; 114(46): 12309-12314, 2017 11 14.
Article in English | MEDLINE | ID: mdl-29087309

ABSTRACT

G protein-coupled receptors (GPCRs) are considered to function primarily at the plasma membrane, where they interact with extracellular ligands and couple to G proteins that transmit intracellular signals. Consequently, therapeutic drugs are designed to target GPCRs at the plasma membrane. Activated GPCRs undergo clathrin-dependent endocytosis. Whether GPCRs in endosomes control pathophysiological processes in vivo and are therapeutic targets remains uncertain. We investigated the contribution of endosomal signaling of the calcitonin receptor-like receptor (CLR) to pain transmission. Calcitonin gene-related peptide (CGRP) stimulated CLR endocytosis and activated protein kinase C (PKC) in the cytosol and extracellular signal regulated kinase (ERK) in the cytosol and nucleus. Inhibitors of clathrin and dynamin prevented CLR endocytosis and activation of cytosolic PKC and nuclear ERK, which derive from endosomal CLR. A cholestanol-conjugated antagonist, CGRP8-37, accumulated in CLR-containing endosomes and selectively inhibited CLR signaling in endosomes. CGRP caused sustained excitation of neurons in slices of rat spinal cord. Inhibitors of dynamin, ERK, and PKC suppressed persistent neuronal excitation. CGRP8-37-cholestanol, but not unconjugated CGRP8-37, prevented sustained neuronal excitation. When injected intrathecally to mice, CGRP8-37-cholestanol inhibited nociceptive responses to intraplantar injection of capsaicin, formalin, or complete Freund's adjuvant more effectively than unconjugated CGRP8-37 Our results show that CLR signals from endosomes to control pain transmission and identify CLR in endosomes as a therapeutic target for pain. Thus, GPCRs function not only at the plasma membrane but also in endosomes to control complex processes in vivo. Endosomal GPCRs are a drug target that deserve further attention.


Subject(s)
Calcitonin Receptor-Like Protein/genetics , Endocytosis/drug effects , Endosomes/metabolism , Nociception/physiology , Pain/physiopathology , Synaptic Transmission/drug effects , Adrenergic Antagonists/pharmacology , Animals , Calcitonin Gene-Related Peptide/pharmacology , Calcitonin Receptor-Like Protein/antagonists & inhibitors , Calcitonin Receptor-Like Protein/metabolism , Capsaicin/antagonists & inhibitors , Capsaicin/pharmacology , Cholestanols/pharmacology , Clathrin/antagonists & inhibitors , Clathrin/genetics , Clathrin/metabolism , Dynamins/genetics , Dynamins/metabolism , Endosomes/drug effects , Formaldehyde/antagonists & inhibitors , Formaldehyde/pharmacology , Freund's Adjuvant/antagonists & inhibitors , Freund's Adjuvant/pharmacology , Gene Expression Regulation , Injections, Spinal , Male , Mice , Microtomy , Mitogen-Activated Protein Kinase 1/genetics , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/genetics , Mitogen-Activated Protein Kinase 3/metabolism , Nociception/drug effects , Pain/chemically induced , Pain/genetics , Pain/prevention & control , Peptide Fragments/pharmacology , Protein Kinase C/genetics , Protein Kinase C/metabolism , Rats , Spinal Cord/cytology , Spinal Cord/drug effects , Spinal Cord/metabolism , Tissue Culture Techniques
15.
J Phys Chem Lett ; 8(21): 5277-5282, 2017 Nov 02.
Article in English | MEDLINE | ID: mdl-28990781

ABSTRACT

Distance measurements by pulse electron paramagnetic resonance techniques, such as double electron-electron resonance (DEER, also called PELDOR), have become an established tool to explore structural properties of biomacromolecules and their assemblies. In such measurements a pair of spin labels provides a single distance constraint. Here we show that by employing three different types of spin labels that differ in their spectroscopic and spin dynamics properties it is possible to extract three independent distances from a single sample. We demonstrate this using the Antennapedia homeodomain orthogonally labeled with Gd3+ and Mn2+ tags in complex with its cognate DNA binding site labeled with a nitroxide.

16.
Chemistry ; 23(53): 13033-13036, 2017 Sep 21.
Article in English | MEDLINE | ID: mdl-28763128

ABSTRACT

Intermolecular 1 H-1 H nuclear Overhauser effects (NOE) present a powerful tool to assess contacts between proteins and binding partners, but are difficult to identify for complexes of high molecular weight. This report shows that intermolecular NOEs can readily be observed following chemical labeling with tert-butyl or trimethylsilyl (TMS) groups. Proteins can be furnished with tert-butyl or TMS groups site-specifically using genetically encoded unnatural amino acids or by chemical modification of single cysteine residues. No isotope labeling is required. The approach is demonstrated with the 95 kDa complex between tetrameric E. coli single-stranded DNA binding protein (SSB) and single-stranded DNA.


Subject(s)
DNA, Single-Stranded/chemistry , DNA-Binding Proteins/chemistry , Trimethylsilyl Compounds/chemistry , Amino Acid Sequence , Binding Sites , Escherichia coli , Isotope Labeling/methods , Ligands , Models, Molecular , Molecular Structure , Nuclear Magnetic Resonance, Biomolecular/methods , Protein Binding , Structure-Activity Relationship
17.
Sci Transl Med ; 9(392)2017 05 31.
Article in English | MEDLINE | ID: mdl-28566424

ABSTRACT

Typically considered to be cell surface sensors of extracellular signals, heterotrimeric GTP-binding protein (G protein)-coupled receptors (GPCRs) control many pathophysiological processes and are the target of 30% of therapeutic drugs. Activated receptors redistribute to endosomes, but researchers have yet to explore whether endosomal receptors generate signals that control complex processes in vivo and are viable therapeutic targets. We report that the substance P (SP) neurokinin 1 receptor (NK1R) signals from endosomes to induce sustained excitation of spinal neurons and pain transmission and that specific antagonism of the NK1R in endosomes with membrane-anchored drug conjugates provides more effective and sustained pain relief than conventional plasma membrane-targeted antagonists. Pharmacological and genetic disruption of clathrin, dynamin, and ß-arrestin blocked SP-induced NK1R endocytosis and prevented SP-stimulated activation of cytosolic protein kinase C and nuclear extracellular signal-regulated kinase, as well as transcription. Endocytosis inhibitors prevented sustained SP-induced excitation of neurons in spinal cord slices in vitro and attenuated nociception in vivo. When conjugated to cholestanol to promote endosomal targeting, NK1R antagonists selectively inhibited endosomal signaling and sustained neuronal excitation. Cholestanol conjugation amplified and prolonged the antinociceptive actions of NK1R antagonists. These results reveal a critical role for endosomal signaling of the NK1R in the complex pathophysiology of pain and demonstrate the use of endosomally targeted GPCR antagonists.


Subject(s)
Endosomes/metabolism , Molecular Targeted Therapy , Nociception , Pain/drug therapy , Receptors, Neurokinin-1/metabolism , Signal Transduction , Animals , Cell Compartmentation , Clathrin/metabolism , Dynamins/metabolism , Endocytosis/drug effects , Endosomes/drug effects , GTP-Binding Proteins/metabolism , HEK293 Cells , Humans , Lipids/chemistry , Models, Biological , Neurokinin-1 Receptor Antagonists/pharmacology , Neurokinin-1 Receptor Antagonists/therapeutic use , Neurons/drug effects , Neurons/metabolism , Neurons/pathology , Nociception/drug effects , Pain/pathology , Protein Binding/drug effects , Rats , Signal Transduction/drug effects , Spinal Cord/pathology , Subcellular Fractions/metabolism , Substance P/metabolism , beta-Arrestins/metabolism
18.
Bioorg Med Chem Lett ; 27(2): 254-260, 2017 01 15.
Article in English | MEDLINE | ID: mdl-27923620

ABSTRACT

Activity-based probes are small molecules that covalently bind to the active site of a protease in an activity-dependent manner. We synthesized and characterized two fluorescent activity-based probes that target serine proteases with trypsin-like or elastase-like activity. We assessed the selectivity and potency of these probes against recombinant enzymes and demonstrated that while they are efficacious at labeling active proteases in complex protein mixtures in vitro, they are less valuable for in vivo studies. We used these probes to evaluate serine protease activity in two mouse models of acute inflammation, including pancreatitis and colitis. As anticipated, the activity of trypsin-like proteases was increased during pancreatitis. Levels of elastase-like proteases were low in pancreatic lysates and colonic luminal fluids, whether healthy or inflamed. Exogenously added recombinant neutrophil elastase was inhibited upon incubation with these samples, an effect that was augmented in inflamed samples compared to controls. These data suggest that endogenous inhibitors and elastase-degrading proteases are upregulated during inflammation.


Subject(s)
Fluorescent Dyes/chemistry , Inflammation/metabolism , Organophosphonates/chemistry , Serine Proteases/analysis , Animals , Colitis/metabolism , Fluorescent Dyes/chemical synthesis , Mice , Molecular Structure , Organophosphonates/chemical synthesis , Pancreatitis/metabolism , Serine Proteases/metabolism
19.
Mol Pharmacol ; 90(6): 715-725, 2016 Dec.
Article in English | MEDLINE | ID: mdl-27683013

ABSTRACT

Allosteric modulation of adenosine A1 receptors (A1ARs) offers a novel therapeutic approach for the treatment of numerous central and peripheral disorders; however, despite decades of research, there is a relative paucity of structural information regarding the A1AR allosteric site and mechanisms governing cooperativity with orthosteric ligands. We combined alanine-scanning mutagenesis of the A1AR second extracellular loop (ECL2) with radioligand binding and functional interaction assays to quantify effects on allosteric ligand affinity, cooperativity, and efficacy. Docking and molecular dynamics (MD) simulations were performed using an A1AR homology model based on an agonist-bound A2AAR structure. Substitution of E172ECL2 for alanine reduced the affinity of the allosteric modulators PD81723 and VCP171 for the unoccupied A1AR. Residues involved in cooperativity with the orthosteric agonist NECA were different in PD81723 and VCP171; positive cooperativity between PD81723 and NECA was reduced on alanine substitution of a number of ECL2 residues, including E170ECL2 and K173ECL2, whereas mutation of W146ECL2 and W156ECL2 decreased VCP171 cooperativity with NECA. Molecular modeling localized a likely allosteric pocket for both modulators to an extracellular vestibule that overlaps with a region used by orthosteric ligands as they transit into the canonical A1AR orthosteric site. MD simulations confirmed a key interaction between E172ECL2 and both modulators. Bound PD81723 is flanked by another residue, E170ECL2, which forms hydrogen bonds with adjacent K168ECL2 and K173ECL2. Collectively, our data suggest E172ECL2 is a key allosteric ligand-binding determinant, whereas hydrogen-bonding networks within the extracellular vestibule may facilitate the transmission of cooperativity between orthosteric and allosteric sites.


Subject(s)
Allosteric Site , Receptor, Adenosine A1/chemistry , Receptor, Adenosine A1/metabolism , Signal Transduction , Adenosine/pharmacology , Alanine/genetics , Allosteric Regulation/drug effects , Animals , Binding Sites , CHO Cells , Cricetinae , Cricetulus , Cyclic AMP/metabolism , Humans , Ligands , Molecular Dynamics Simulation , Mutation/genetics , Protein Structure, Secondary , Signal Transduction/drug effects , Structural Homology, Protein , Structure-Activity Relationship
20.
Am J Physiol Gastrointest Liver Physiol ; 311(3): G548-60, 2016 09 01.
Article in English | MEDLINE | ID: mdl-27514475

ABSTRACT

Pancreatitis is an inflammatory disease of the pancreas characterized by dysregulated activity of digestive enzymes, necrosis, immune infiltration, and pain. Repeated incidence of pancreatitis is an important risk factor for pancreatic cancer. Legumain, a lysosomal cysteine protease, has been linked to inflammatory diseases such as atherosclerosis, stroke, and cancer. Until now, legumain activation has not been studied during pancreatitis. We used a fluorescently quenched activity-based probe to assess legumain activation during caerulein-induced pancreatitis in mice. We detected activated legumain by ex vivo imaging, confocal microscopy, and gel electrophoresis. Compared with healthy controls, legumain activity in the pancreas of caerulein-treated mice was increased in a time-dependent manner. Legumain was localized to CD68(+) macrophages and was not active in pancreatic acinar cells. Using a small-molecule inhibitor of legumain, we found that this protease is not essential for the initiation of pancreatitis. However, it may serve as a biomarker of disease, since patients with chronic pancreatitis show strongly increased legumain expression in macrophages. Moreover, the occurrence of legumain-expressing macrophages in regions of acinar-to-ductal metaplasia suggests that this protease may influence reprogramming events that lead to inflammation-induced pancreatic cancer.


Subject(s)
Cysteine Endopeptidases/metabolism , Macrophages/enzymology , Pancreatitis/enzymology , Animals , Ceruletide/toxicity , Cysteine Endopeptidases/genetics , Enzyme Inhibitors/pharmacology , Female , Gene Expression Regulation, Enzymologic , Macrophages/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Pancreatitis/chemically induced
SELECTION OF CITATIONS
SEARCH DETAIL