Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 33
Filter
Add more filters










Publication year range
1.
J Am Chem Soc ; 145(29): 16069-16080, 2023 07 26.
Article in English | MEDLINE | ID: mdl-37450839

ABSTRACT

Electrophilic small molecules that can reversibly modify proteins are of growing interest in drug discovery. However, the ability to study reversible covalent probes in live cells can be limited by their reversible reactivity after cell lysis and in proteomic workflows, leading to scrambling and signal loss. We describe how thiomethyltetrazines function as reversible covalent warheads for cysteine modification, and this dynamic labeling behavior can be "switched off" via bioorthogonal chemistry inside live cells. Simultaneously, the tetrazine serves as a bioorthogonal reporter enabling the introduction of tags for fluorescent imaging or affinity purification. Thiomethyltetrazines can label isolated proteins, proteins in cellular lysates, and proteins in live cells with second-order rate constants spanning 2 orders of magnitude (k2, 1-100 M-1 s-1). Reversible modification by thiomethyltetrazines can be switched off upon the addition of trans-cyclooctene in live cells, converting the dynamic thiomethyltetrazine tag into a Diels-Alder adduct which is stable to lysis and proteomic workflows. Time-course quenching experiments were used to demonstrate temporal control over electrophilic modification. Moreover, it is shown that "locking in" the tag through Diels-Alder chemistry enables the identification of protein targets that are otherwise lost during sample processing. Three probes were further evaluated to identify unique pathways in a live-cell proteomic study. We anticipate that discovery efforts will be enabled by the trifold function of thiomethyltetrazines as electrophilic warheads, bioorthogonal reporters, and switches for "locking in" stability.


Subject(s)
Cysteine , Heterocyclic Compounds , Proteomics , Proteins/chemistry
2.
Nat Struct Mol Biol ; 30(1): 22-30, 2023 01.
Article in English | MEDLINE | ID: mdl-36522428

ABSTRACT

Glycerol-3-phosphate acyltransferase (GPAT)1 is a mitochondrial outer membrane protein that catalyzes the first step of de novo glycerolipid biosynthesis. Hepatic expression of GPAT1 is linked to liver fat accumulation and the severity of nonalcoholic fatty liver diseases. Here we present the cryo-EM structures of human GPAT1 in substrate analog-bound and product-bound states. The structures reveal an N-terminal acyltransferase domain that harbors important catalytic motifs and a tightly associated C-terminal domain that is critical for proper protein folding. Unexpectedly, GPAT1 has no transmembrane regions as previously proposed but instead associates with the membrane via an amphipathic surface patch and an N-terminal loop-helix region that contains a mitochondrial-targeting signal. Combined structural, computational and functional studies uncover a hydrophobic pathway within GPAT1 for lipid trafficking. The results presented herein lay a framework for rational inhibitor development for GPAT1.


Subject(s)
Liver , Mitochondrial Membranes , Humans , Liver/metabolism , Mitochondrial Membranes/metabolism , Glycerol-3-Phosphate O-Acyltransferase/chemistry , Glycerol-3-Phosphate O-Acyltransferase/metabolism , Amino Acid Sequence
3.
ACS Chem Biol ; 16(8): 1445-1455, 2021 08 20.
Article in English | MEDLINE | ID: mdl-34374519

ABSTRACT

Stability proteomics techniques that do not require drug modifications have emerged as an attractive alternative to affinity purification methods in drug target engagement studies. Two representative techniques include the chemical-denaturation-based SPROX (Stability of Proteins from Rates of Oxidation), which utilizes peptide-level quantification and thermal-denaturation-based TPP (Thermal Proteome Profiling), which utilizes protein-level quantification. Recently, the "OnePot" strategy was adapted for both SPROX and TPP to increase the throughput. When combined with the 2D setup which measures both the denaturation and the drug dose dimensions, the OnePot 2D format offers improved analysis specificity with higher resource efficiency. However, a systematic evaluation of the OnePot 2D format and a comparison between SPROX and TPP are still lacking. Here, we performed SPROX and TPP to identify protein targets of a well-studied pan-kinase inhibitor staurosporine with K562 lysate, in curve-fitting and OnePot 2D formats. We found that the OnePot 2D format provided ∼10× throughput, achieved ∼1.6× protein coverage and involves more straightforward data analysis. We also compared SPROX with the current "gold-standard" stability proteomics technique TPP in the OnePot 2D format. The protein coverage of TPP is ∼1.5 fold of SPROX; however, SPROX offers protein domain-level information, identifies comparable numbers of kinase hits, has higher signal (R value), and requires ∼3× less MS time. Unique SPROX hits encompass higher-molecular-weight proteins, compared to the unique TPP hits, and include atypical kinases. We also discuss hit stratification and prioritization strategies to promote the efficiency of hit followup.


Subject(s)
Protein Kinase Inhibitors/pharmacology , Protein Kinases/analysis , Proteome/analysis , Proteomics/methods , Staurosporine/pharmacology , Humans , K562 Cells , Protein Kinases/metabolism , Proteome/metabolism
4.
Org Biomol Chem ; 18(19): 3669-3673, 2020 05 20.
Article in English | MEDLINE | ID: mdl-32373883

ABSTRACT

A C-H functionalization strategy for the expedient access to photoreactive chemical probes of commonly found heterocyclic fragments or drug molecules of pharmaceutical relevance is described. A series of aryl glyoxylic acid reagents featuring pendant alkyne or azide clickable handles have been developed for application in the radical-mediated appendage of benzoyl fragments onto simple heteroaromatic fragments, as well as more complex drug-like compounds. This unprecedented strategy of chemical probe synthesis allows for direct access to photoreactive chemical probes without any requirement of fragment pre-functionalization or significant synthetic re-evaluation.

5.
Elife ; 82019 10 03.
Article in English | MEDLINE | ID: mdl-31580259

ABSTRACT

Alanine-serine-cysteine transporter 2 (ASCT2, SLC1A5) is the primary transporter of glutamine in cancer cells and regulates the mTORC1 signaling pathway. The SLC1A5 function involves finely tuned orchestration of two domain movements that include the substrate-binding transport domain and the scaffold domain. Here, we present cryo-EM structures of human SLC1A5 and its complex with the substrate, L-glutamine in an outward-facing conformation. These structures reveal insights into the conformation of the critical ECL2a loop which connects the two domains, thus allowing rigid body movement of the transport domain throughout the transport cycle. Furthermore, the structures provide new insights into substrate recognition, which involves conformational changes in the HP2 loop. A putative cholesterol binding site was observed near the domain interface in the outward-facing state. Comparison with the previously determined inward-facing structure of SCL1A5 provides a basis for a more integrated understanding of substrate recognition and transport mechanism in the SLC1 family.


Subject(s)
Amino Acid Transport System ASC/chemistry , Amino Acid Transport System ASC/metabolism , Glutamine/chemistry , Glutamine/metabolism , Minor Histocompatibility Antigens/chemistry , Minor Histocompatibility Antigens/metabolism , Cryoelectron Microscopy , Humans , Protein Binding , Protein Conformation
6.
J Mol Biol ; 430(21): 4102-4118, 2018 10 19.
Article in English | MEDLINE | ID: mdl-30120952

ABSTRACT

Photoreceptors of the squid Loligo pealei contain a G-protein-coupled receptor (GPCR) signaling system that activates phospholipase C in response to light. Analogous to the mammalian visual system, signaling of the photoactivated GPCR rhodopsin is terminated by binding of squid arrestin (sArr). sArr forms a light-dependent, high-affinity complex with squid rhodopsin, which does not require prior receptor phosphorylation for interaction. This is at odds with classical mammalian GPCR desensitization where an agonist-bound phosphorylated receptor is needed to break stabilizing constraints within arrestins, the so-called "three-element interaction" and "polar core" network, before a stable receptor-arrestin complex can be established. Biophysical and mass spectrometric analysis of the squid rhodopsin-arrestin complex indicates that in contrast to mammalian arrestins, the sArr C-tail is not involved in a stable three-element interaction. We determined the crystal structure of C-terminally truncated sArr that adopts a basal conformation common to arrestins and is stabilized by a series of weak but novel polar core interactions. Unlike mammalian arrestin-1, deletion of the sArr C-tail does not influence kinetic properties of complex formation of sArr with the receptor. Hydrogen-deuterium exchange studies revealed the footprint of the light-activated rhodopsin on sArr. Furthermore, double electron-electron resonance spectroscopy experiments provide evidence that receptor-bound sArr adopts a conformation different from the one known for arrestin-1 and molecular dynamics simulations reveal the residues that account for the weak three-element interaction. Insights gleaned from studying this system add to our general understanding of GPCR-arrestin interaction.


Subject(s)
Arrestin/chemistry , Arrestin/metabolism , Decapodiformes/metabolism , Protein Interaction Domains and Motifs , Rhodopsin/chemistry , Rhodopsin/metabolism , Animals , Kinetics , Models, Molecular , Molecular Dynamics Simulation , Phosphorylation , Protein Binding , Protein Conformation , Spectrum Analysis , Structure-Activity Relationship
7.
Proc Natl Acad Sci U S A ; 115(31): E7285-E7292, 2018 07 31.
Article in English | MEDLINE | ID: mdl-30012605

ABSTRACT

Proteolysis targeting chimeras (PROTACs) are heterobifunctional small molecules that simultaneously bind to a target protein and an E3 ligase, thereby leading to ubiquitination and subsequent degradation of the target. They present an exciting opportunity to modulate proteins in a manner independent of enzymatic or signaling activity. As such, they have recently emerged as an attractive mechanism to explore previously "undruggable" targets. Despite this interest, fundamental questions remain regarding the parameters most critical for achieving potency and selectivity. Here we employ a series of biochemical and cellular techniques to investigate requirements for efficient knockdown of Bruton's tyrosine kinase (BTK), a nonreceptor tyrosine kinase essential for B cell maturation. Members of an 11-compound PROTAC library were investigated for their ability to form binary and ternary complexes with BTK and cereblon (CRBN, an E3 ligase component). Results were extended to measure effects on BTK-CRBN cooperative interactions as well as in vitro and in vivo BTK degradation. Our data show that alleviation of steric clashes between BTK and CRBN by modulating PROTAC linker length within this chemical series allows potent BTK degradation in the absence of thermodynamic cooperativity.


Subject(s)
Protein-Tyrosine Kinases/metabolism , Proteolysis , Ubiquitin-Protein Ligases/metabolism , Ubiquitination , Agammaglobulinaemia Tyrosine Kinase , Animals , Cells, Cultured , Ligands , Polyubiquitin/metabolism , Rats , Thermodynamics
8.
Sci Rep ; 8(1): 4664, 2018 03 16.
Article in English | MEDLINE | ID: mdl-29549268

ABSTRACT

Cystic Fibrosis (CF) is caused by mutations in the Cystic Fibrosis Transmembrane Conductance Regulator (CFTR). Mutations associated with CF cause loss-of-function in CFTR leading to salt imbalance in epithelial tissues. Kalydeco (also called VX-770 or ivacaftor) was approved for CF treatment in 2012 but little is known regarding the compound's interactions with CFTR including the site of binding or mechanisms of action. In this study we use hydrogen/deuterium exchange (HDX) coupled with mass spectrometry to assess the conformational dynamics of a thermostabilized form of CFTR in apo and ligand-bound states. We observe HDX protection at a known binding site for AMPPNP and significant protection for several regions of CFTR in the presence of Kalydeco. The ligand-induced changes of CFTR in the presence of Kalydeco suggest a potential binding site.


Subject(s)
Adenosine Diphosphate/metabolism , Adenylyl Imidodiphosphate/metabolism , Aminophenols/pharmacology , Cystic Fibrosis Transmembrane Conductance Regulator/chemistry , Cystic Fibrosis Transmembrane Conductance Regulator/metabolism , Quinolones/pharmacology , Aminophenols/chemistry , Binding Sites , Cryoelectron Microscopy , Deuterium Exchange Measurement , Humans , Models, Molecular , Protein Binding , Protein Conformation/drug effects , Protein Stability , Quinolones/chemistry , Thermodynamics
9.
Methods Mol Biol ; 1732: 29-55, 2018.
Article in English | MEDLINE | ID: mdl-29480467

ABSTRACT

Protein-ligand interactions can be evaluated by a number of different biophysical methods. Here we describe some of the experimental methods that we have used to generate AMPK protein reagents and characterize its interactions with direct synthetic activators. Recombinant heterotrimeric AMPK complexes were generated using standard molecular biology methods by expression either in insect cells via infection with three different viruses or more routinely in Escherichia coli with a tricistronic expression vector. Hydrogen/deuterium exchange (HDX) coupled with mass spectrometry was used to probe protein conformational changes and potential binding sites of activators on AMPK. X-ray crystallographic studies were carried out on crystals of AMPK with bound ligands to reveal detailed molecular interactions formed by AMPK activators at near-atomic resolution. In order to gain insights into the mechanism of enzyme activation and to probe the effects of AMPK activators on kinetic parameters such as Michaelis-Menten constant (K m ) or maximal reaction velocity (V max), we performed classical enzyme kinetic studies using radioactive 33P-ATP-based filter assay. Equilibrium dissociation constants (K D ) and on and off rates of ligand binding were obtained by application of surface plasmon resonance (SPR) technique.


Subject(s)
AMP-Activated Protein Kinases/chemistry , Deuterium Exchange Measurement/methods , Enzyme Activators/chemistry , Surface Plasmon Resonance/methods , AMP-Activated Protein Kinases/isolation & purification , Animals , Binding Sites , Crystallography, X-Ray , Deuterium Exchange Measurement/instrumentation , Enzyme Activation , Enzyme Assays/instrumentation , Enzyme Assays/methods , Kinetics , Ligands , Mass Spectrometry/instrumentation , Mass Spectrometry/methods , Molecular Docking Simulation , Protein Binding , Protein Structure, Quaternary , Recombinant Proteins/chemistry , Recombinant Proteins/isolation & purification , Sf9 Cells , Surface Plasmon Resonance/instrumentation
10.
Methods Mol Biol ; 1513: 11-22, 2017.
Article in English | MEDLINE | ID: mdl-27807827

ABSTRACT

A powerful interplay exists between the recognition of gene families, sensitive techniques in proteomics, and the interrogation of protein function using chemical probes. The most prominent methods, such as affinity capture, activity-based protein profiling and photoaffinity labeling, are extensively reviewed in the literature. Here we briefly review additional methods developed in the past 15 years. These include "stability proteomics" methods such as proteomically analyzed cellular thermal shift assays and the use of chemical oxidation as a probe of structure, the use of multiple bead-linked kinase inhibitors to analyze inhibitor specificities, and advances in the use of proteolysis-targeting chimeras for selective protein elimination.


Subject(s)
Antineoplastic Agents/pharmacology , Drug Discovery/methods , Neoplasm Proteins/genetics , Protein Kinase Inhibitors/pharmacology , Protein Kinases/genetics , Proteomics/methods , Biological Assay , Gene Expression , Humans , K562 Cells , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/metabolism , Oxidation-Reduction , Protein Denaturation/drug effects , Protein Kinases/metabolism , Protein Stability/drug effects , Thermodynamics
11.
Nat Commun ; 7: 13042, 2016 10 11.
Article in English | MEDLINE | ID: mdl-27727204

ABSTRACT

Inhibition of ß-secretase BACE1 is considered one of the most promising approaches for treating Alzheimer's disease. Several structurally distinct BACE1 inhibitors have been withdrawn from development after inducing ocular toxicity in animal models, but the target mediating this toxicity has not been identified. Here we use a clickable photoaffinity probe to identify cathepsin D (CatD) as a principal off-target of BACE1 inhibitors in human cells. We find that several BACE1 inhibitors blocked CatD activity in cells with much greater potency than that displayed in cell-free assays with purified protein. Through a series of exploratory toxicology studies, we show that quantifying CatD target engagement in cells with the probe is predictive of ocular toxicity in vivo. Taken together, our findings designate off-target inhibition of CatD as a principal driver of ocular toxicity for BACE1 inhibitors and more generally underscore the power of chemical proteomics for discerning mechanisms of drug action.


Subject(s)
Amyloid Precursor Protein Secretases/antagonists & inhibitors , Cathepsin D/metabolism , Enzyme Inhibitors/toxicity , Eye/pathology , Proteomics/methods , Toxicity Tests , Amyloid Precursor Protein Secretases/metabolism , Animals , Cell Line , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Eye/drug effects , Humans , Inhibitory Concentration 50 , Mass Spectrometry , Mice, Knockout , Molecular Probes/chemical synthesis , Molecular Probes/chemistry , Peptides/metabolism , Protein Binding , Rats, Wistar , Retinal Pigment Epithelium/drug effects , Retinal Pigment Epithelium/pathology , Staining and Labeling
12.
J Proteome Res ; 14(11): 4571-80, 2015 Nov 06.
Article in English | MEDLINE | ID: mdl-26398198

ABSTRACT

Liver receptor homologue-1 (LRH1) is an orphan nuclear receptor that has been shown to play a role in the transcriptional regulation of pathways involved in cancer. Elucidating the components of the LRH1 transcriptional complex to better understand endogenous regulation of the receptor as well as its role in cancer remains a high priority. A sub-cellular enrichment strategy coupled with proteomic approaches was employed to identify putative LRH1 co-regulators. Nuclear fractionation protocol was essential for detection of LRH1 peptides by mass spectrometry (MS), with most peptides being observed in the insoluble fraction (receptor bound to DNA). SERBP1 and ILF3 were identified as LRH1 interacting partners by both Western blot and MS/MS analysis. Receptor knockdown by siRNA showed an increase in SERBP1 expression, while ILF3 expression was unchanged. In contrast, receptor overexpression decreased only SERBP1 mRNA levels. Consistent with these data, in a promoter:reporter assay, binding of LRH1 to the promoter region of SERBP1 resulted in a decrease in the expression level of the reporter gene, subsequently inhibiting transcription. Given the receptor's role in cancer progression, the study here elucidates additional transcriptional machinery involved in LRH1 signaling and potentially provides new targets for therapeutics development.


Subject(s)
Gene Expression Regulation , Peptides/analysis , RNA-Binding Proteins/metabolism , Receptors, Cytoplasmic and Nuclear/metabolism , Transcription, Genetic , Amino Acid Sequence , Cell Line, Tumor , Cell Nucleus/metabolism , Chemical Fractionation , HEK293 Cells , Hepatocytes/cytology , Hepatocytes/metabolism , Humans , Molecular Sequence Annotation , Molecular Sequence Data , Nuclear Factor 90 Proteins/genetics , Nuclear Factor 90 Proteins/metabolism , Peptides/genetics , Peptides/metabolism , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha , Plasmids/chemistry , Plasmids/metabolism , Promoter Regions, Genetic , Protein-Arginine N-Methyltransferases/genetics , Protein-Arginine N-Methyltransferases/metabolism , Proteolysis , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , RNA-Binding Proteins/antagonists & inhibitors , RNA-Binding Proteins/genetics , Receptors, Cytoplasmic and Nuclear/antagonists & inhibitors , Receptors, Cytoplasmic and Nuclear/genetics , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Repressor Proteins/genetics , Repressor Proteins/metabolism , Signal Transduction , Transcription Factors/genetics , Transcription Factors/metabolism , Transfection
13.
J Am Soc Mass Spectrom ; 26(12): 2133-40, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26265041

ABSTRACT

The goal in proteomics to identify all peptides in a complex mixture has been largely addressed using various LC MS/MS approaches, such as data dependent acquisition, SRM/MRM, and data independent acquisition instrumentation. Despite these developments, many peptides remain unsequenced, often due to low abundance, poor fragmentation patterns, or data analysis difficulties. Many of the unidentified peptides exhibit strong evidence in high resolution MS(1) data and are frequently post-translationally modified, playing a significant role in biological processes. Proteomics Workbench (PWB) software was developed to automate the detection and visualization of all possible peptides in MS(1) data, reveal candidate peptides not initially identified, and build inclusion lists for subsequent MS(2) analysis to uncover new identifications. We used this software on existing data on the autophagy regulating kinase Ulk1 as a proof of concept for this method, as we had already manually identified a number of phosphorylation sites Dorsey, F. C. et al (J. Proteome. Res. 8(11), 5253-5263 (2009)). PWB found all previously identified sites of phosphorylation. The software has been made freely available at http://www.proteomicsworkbench.com . Graphical Abstract ᅟ.


Subject(s)
Intracellular Signaling Peptides and Proteins/chemistry , Protein Processing, Post-Translational , Protein Serine-Threonine Kinases/chemistry , Software , Tandem Mass Spectrometry/methods , Amino Acid Sequence , Autophagy-Related Protein-1 Homolog , HEK293 Cells , Humans , Molecular Sequence Data
14.
Nat Commun ; 6: 7859, 2015 Jul 31.
Article in English | MEDLINE | ID: mdl-26227798

ABSTRACT

Class B G protein-coupled receptors are composed of an extracellular domain (ECD) and a seven-transmembrane (7TM) domain, and their signalling is regulated by peptide hormones. Using a hybrid structural biology approach together with the ECD and 7TM domain crystal structures of the glucagon receptor (GCGR), we examine the relationship between full-length receptor conformation and peptide ligand binding. Molecular dynamics (MD) and disulfide crosslinking studies suggest that apo-GCGR can adopt both an open and closed conformation associated with extensive contacts between the ECD and 7TM domain. The electron microscopy (EM) map of the full-length GCGR shows how a monoclonal antibody stabilizes the ECD and 7TM domain in an elongated conformation. Hydrogen/deuterium exchange (HDX) studies and MD simulations indicate that an open conformation is also stabilized by peptide ligand binding. The combined studies reveal the open/closed states of GCGR and suggest that glucagon binds to GCGR by a conformational selection mechanism.


Subject(s)
Glucagon/metabolism , Molecular Dynamics Simulation , Receptors, Glucagon/metabolism , Animals , Chromatography, Liquid , Deuterium Exchange Measurement , Disulfides/chemistry , Disulfides/metabolism , Humans , Ligands , Microscopy, Electron , Protein Binding , Protein Structure, Secondary , Protein Structure, Tertiary , Receptors, Glucagon/chemistry , Receptors, Glucagon/ultrastructure , Sf9 Cells , Tandem Mass Spectrometry
15.
Nature ; 523(7562): 561-7, 2015 Jul 30.
Article in English | MEDLINE | ID: mdl-26200343

ABSTRACT

G-protein-coupled receptors (GPCRs) signal primarily through G proteins or arrestins. Arrestin binding to GPCRs blocks G protein interaction and redirects signalling to numerous G-protein-independent pathways. Here we report the crystal structure of a constitutively active form of human rhodopsin bound to a pre-activated form of the mouse visual arrestin, determined by serial femtosecond X-ray laser crystallography. Together with extensive biochemical and mutagenesis data, the structure reveals an overall architecture of the rhodopsin-arrestin assembly in which rhodopsin uses distinct structural elements, including transmembrane helix 7 and helix 8, to recruit arrestin. Correspondingly, arrestin adopts the pre-activated conformation, with a ∼20° rotation between the amino and carboxy domains, which opens up a cleft in arrestin to accommodate a short helix formed by the second intracellular loop of rhodopsin. This structure provides a basis for understanding GPCR-mediated arrestin-biased signalling and demonstrates the power of X-ray lasers for advancing the frontiers of structural biology.


Subject(s)
Arrestin/chemistry , Arrestin/metabolism , Rhodopsin/chemistry , Rhodopsin/metabolism , Animals , Binding Sites , Crystallography, X-Ray , Disulfides/chemistry , Disulfides/metabolism , Humans , Lasers , Mice , Models, Molecular , Multiprotein Complexes/biosynthesis , Multiprotein Complexes/chemistry , Multiprotein Complexes/metabolism , Protein Binding , Reproducibility of Results , Signal Transduction , X-Rays
16.
J Proteome Res ; 14(5): 2287-97, 2015 May 01.
Article in English | MEDLINE | ID: mdl-25825992

ABSTRACT

Current methods for the large-scale characterization of disease states generally rely on the analysis of gene and/or protein expression levels. These existing methods fail to detect proteins with disease-related functions and unaltered expression levels. Here we describe the large-scale use of thermodynamic measurements of protein folding and stability for the characterization of disease states. Using the Stable Isotope Labeling with Amino Acids in Cell Culture and Stability of Proteins from Rates of Oxidation (SILAC-SPROX) technique, we assayed ∼800 proteins for protein folding and stability changes in three different cell culture models of breast cancer including the MCF-10A, MCF-7, and MDA-MB-231 cell lines. The thermodynamic stability profiles generated here created distinct molecular markers to differentiate the three cell lines, and a significant fraction (∼45%) of the differentially stabilized proteins did not have altered expression levels. Thus, the differential thermodynamic profiling strategy reported here created novel molecular signatures of breast cancer and provided additional insight into the molecular basis of the disease. Our results establish the utility of protein folding and stability measurements for the study of disease processes, and they suggest that such measurements may be useful for biomarker discovery in disease.


Subject(s)
Biomarkers, Tumor/isolation & purification , Breast Neoplasms/genetics , Neoplasm Proteins/isolation & purification , Proteome/isolation & purification , Amino Acids/metabolism , Biomarkers, Tumor/chemistry , Biomarkers, Tumor/genetics , Breast Neoplasms/diagnosis , Breast Neoplasms/pathology , Cell Line, Tumor , Female , Gene Expression , Humans , Isotope Labeling , Molecular Sequence Annotation , Neoplasm Proteins/chemistry , Neoplasm Proteins/genetics , Oxidation-Reduction , Protein Folding , Protein Stability , Proteome/chemistry , Proteome/genetics , Proteomics/methods , Thermodynamics
17.
Mol Pharmacol ; 87(2): 296-304, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25473120

ABSTRACT

The orphan nuclear receptor liver receptor homolog 1 (LRH-1; NR5A2) is a potent regulator of cholesterol metabolism and bile acid homeostasis. Recently, LRH-1 has been shown to play an important role in intestinal inflammation and in the progression of estrogen receptor positive and negative breast cancers and pancreatic cancer. Structural studies have revealed that LRH-1 can bind phospholipids and the dietary phospholipid dilauroylphosphatidylcholine activates LRH-1 activity in rodents. Here we characterize the activity of a novel synthetic nonphospholipid small molecule repressor of LRH-1, SR1848 (6-[4-(3-chlorophenyl)piperazin-1-yl]-3-cyclohexyl-1H-pyrimidine-2,4-dione). In cotransfection studies, SR1848 reduced LRH-1-dependent expression of a reporter gene and in cells that endogenously express LRH-1 dose dependently reduced the expression of cyclin-D1 and -E1, resulting in inhibition of cell proliferation. The cellular effects of SR1848 treatment are recapitulated after transfection of cells with small-interfering RNA targeting LRH-1. Immunocytochemistry analysis shows that SR1848 induces rapid translocation of nuclear LRH-1 to the cytoplasm. Combined, these results suggest that SR1848 is a functional repressor of LRH-1 that impacts expression of genes involved in proliferation in LRH-1-expressing cancers. Thus, SR1848 represents a novel chemical scaffold for the development of therapies targeting malignancies driven by LRH-1.


Subject(s)
Cell Proliferation/physiology , Pyrimidines/metabolism , Receptors, Cytoplasmic and Nuclear/antagonists & inhibitors , Receptors, Cytoplasmic and Nuclear/metabolism , Animals , Cell Proliferation/drug effects , HEK293 Cells , Hep G2 Cells , Humans , Mice , Mice, Inbred C57BL , Pyrimidines/chemistry , Pyrimidines/pharmacology
18.
J Proteome Res ; 13(12): 5801-11, 2014 Dec 05.
Article in English | MEDLINE | ID: mdl-25285362

ABSTRACT

The genes encoding many viral proteins such as HIV-1 envelope glycoprotein gp120 have a tendency for codons that are poorly used by the human genome. Why these codons are frequently present in the HIV genome is not known. The presence of these codons limits expression of HIV-1 gp120 for biochemical studies. The poor codons are replaced by synonymous codons that are frequently present in the highly expressed human genes to overexpress this protein. Whether this codon optimization affects functional properties of gp120 such as its N-linked glycosylation is unknown. We applied a bottom-up mass-spectrometry-based workflow for the direct measurement of deglycosylated and unglycosylated peptides with putative N-linked glycosylation sites, that is, NxS/T motifs. Using this mass-spectrometry approach in combination with ELISA, it is found that codon optimization significantly reduces the frequency with which the dolichol pyrophosphate-linked oligosaccharide is added by the catalytic subunits of oligosaccharide transferase complex to the glycosylation sites. This reduction affects binding of glycan-dependent broadly neutralizing antibodies. These data are essential for biochemical studies of gp120 and successful development of a vaccine against HIV-1. Furthermore, they demonstrate a mass-spectrometry approach for studying the site-specific N-linked glycosylation efficiency of glycoproteins.


Subject(s)
Enzyme-Linked Immunosorbent Assay/methods , HIV Envelope Protein gp120/metabolism , Oligosaccharides/metabolism , Tandem Mass Spectrometry/methods , Amino Acid Sequence , Binding Sites/genetics , Codon/genetics , Electrophoresis, Polyacrylamide Gel , Glycoproteins/genetics , Glycoproteins/metabolism , Glycosylation , HEK293 Cells , HIV Envelope Protein gp120/genetics , Humans , Molecular Sequence Data , Mutation , Peptides/analysis , Peptides/metabolism , Proteomics/methods
19.
PLoS One ; 9(9): e105683, 2014.
Article in English | MEDLINE | ID: mdl-25180755

ABSTRACT

Activation of the glucagon-like peptide-1 receptor (GLP-1R) in pancreatic ß-cells potentiates insulin production and is a current therapeutic target for the treatment of type 2 diabetes mellitus (T2DM). Like other class B G protein-coupled receptors (GPCRs), the GLP-1R contains an N-terminal extracellular ligand binding domain. N-terminal truncations on the peptide agonist generate antagonists capable of binding to the extracellular domain, but not capable of activating full length receptor. The main objective of this study was to use Hydrogen/deuterium exchange (HDX) to identify how the amide hydrogen bonding network of peptide ligands and the extracellular domain of GLP-1R (nGLP-1R) were altered by binding interactions and to then use this platform to validate direct binding events for putative GLP-1R small molecule ligands. The HDX studies presented here for two glucagon-like peptide-1 receptor (GLP-1R) peptide ligands indicates that the antagonist exendin-4[9-39] is significantly destabilized in the presence of nonionic detergents as compared to the agonist exendin-4. Furthermore, HDX can detect stabilization of exendin-4 and exendin-4[9-39] hydrogen bonding networks at the N-terminal helix [Val19 to Lys27] upon binding to the N-terminal extracellular domain of GLP-1R (nGLP-1R). In addition we show hydrogen bonding network stabilization on nGLP-1R in response to ligand binding, and validate direct binding events with the extracellular domain of the receptor for putative GLP-1R small molecule ligands.


Subject(s)
Receptors, Glucagon/chemistry , Receptors, Glucagon/metabolism , Amino Acid Sequence , Crystallography, X-Ray , Deuterium Exchange Measurement , Exenatide , Glucagon-Like Peptide-1 Receptor , Ligands , Molecular Sequence Data , Peptides/chemistry , Peptides/metabolism , Protein Structure, Tertiary , Small Molecule Libraries/chemistry , Small Molecule Libraries/metabolism , Venoms/chemistry , Venoms/metabolism
20.
ACS Chem Biol ; 9(3): 649-55, 2014 Mar 21.
Article in English | MEDLINE | ID: mdl-24358983

ABSTRACT

Protein arginine methyltransferase 1 (PRMT1)-dependent methylation contributes to the onset and progression of numerous diseases (e.g., cancer, heart disease, ALS); however, the regulatory mechanisms that control PRMT1 activity are relatively unexplored. We therefore set out to decipher how phosphorylation regulates PRMT1 activity. Curated mass spectrometry data identified Tyr291, a residue adjacent to the conserved THW loop, as being phosphorylated. Natural and unnatural amino acid mutagenesis, including the incorporation of p-carboxymethyl-l-phenylalanine (pCmF) as a phosphotyrosine mimic, were used to show that Tyr291 phosphorylation alters the substrate specificity of PRMT1. Additionally, p-benzoyl-l-phenylalanine (pBpF) was incorporated at the Tyr291 position, and cross-linking experiments with K562 cell extracts identified several proteins (e.g., hnRNPA1 and hnRNP H3) that bind specifically to this site. Moreover, we also demonstrate that Tyr291 phosphorylation impairs PRMT1's ability to bind and methylate both proteins. In total, these studies demonstrate that Tyr291 phosphorylation alters both PRMT1 substrate specificity and protein-protein interactions.


Subject(s)
Mutagenesis, Site-Directed , Phenylalanine/analogs & derivatives , Protein-Arginine N-Methyltransferases/genetics , Protein-Arginine N-Methyltransferases/metabolism , Repressor Proteins/genetics , Repressor Proteins/metabolism , Tyrosine/genetics , Amino Acid Substitution , Benzophenones/metabolism , Cross-Linking Reagents/chemistry , Histones/metabolism , Humans , Immunoprecipitation , K562 Cells , Methylation , Phenylalanine/genetics , Phenylalanine/metabolism , Phosphorylation , Protein Processing, Post-Translational , Substrate Specificity , Tyrosine/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...