Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
1.
Mol Syst Biol ; 19(4): e11127, 2023 04 12.
Article in English | MEDLINE | ID: mdl-36856068

ABSTRACT

Cancers represent complex autonomous systems, displaying self-sufficiency in growth signaling. Autonomous growth is fueled by a cancer cell's ability to "secrete-and-sense" growth factors (GFs): a poorly understood phenomenon. Using an integrated computational and experimental approach, here we dissect the impact of a feedback-coupled GTPase circuit within the secretory pathway that imparts secretion-coupled autonomy. The circuit is assembled when the Ras-superfamily monomeric GTPase Arf1, and the heterotrimeric GTPase Giαßγ and their corresponding GAPs and GEFs are coupled by GIV/Girdin, a protein that is known to fuel aggressive traits in diverse cancers. One forward and two key negative feedback loops within the circuit create closed-loop control, allow the two GTPases to coregulate each other, and convert the expected switch-like behavior of Arf1-dependent secretion into an unexpected dose-response alignment behavior of sensing and secretion. Such behavior translates into cell survival that is self-sustained by stimulus-proportionate secretion. Proteomic studies and protein-protein interaction network analyses pinpoint GFs (e.g., the epidermal GF) as key stimuli for such self-sustenance. Findings highlight how the enhanced coupling of two biological switches in cancer cells is critical for multiscale feedback control to achieve secretion-coupled autonomy of growth factors.


Subject(s)
Eukaryotic Cells , Proteomics , Signal Transduction , GTP Phosphohydrolases
2.
FASEB J ; 36(2): e22132, 2022 02.
Article in English | MEDLINE | ID: mdl-34986275

ABSTRACT

Ponesimod is a sphingosine 1-phosphate (S1P) receptor (S1PR) modulator that was recently approved for treating relapsing forms of multiple sclerosis (MS). Three other FDA-approved S1PR modulators for MS-fingolimod, siponimod, and ozanimod-share peripheral immunological effects via common S1P1 interactions, yet ponesimod may access distinct central nervous system (CNS) mechanisms through its selectivity for the S1P1 receptor. Here, ponesimod was examined for S1PR internalization and binding, human astrocyte signaling and single-cell RNA-seq (scRNA-seq) gene expression, and in vivo using murine cuprizone-mediated demyelination. Studies confirmed ponesimod's selectivity for S1P1 without comparable engagement to the other S1PR subtypes (S1P2,3,4,5 ). Ponesimod showed pharmacological properties of acute agonism followed by chronic functional antagonism of S1P1 . A major locus of S1P1 expression in the CNS is on astrocytes, and scRNA-seq of primary human astrocytes exposed to ponesimod identified a gene ontology relationship of reduced neuroinflammation and reduction in known astrocyte disease-related genes including those of immediate early astrocytes that have been strongly associated with disease progression in MS animal models. Remarkably, ponesimod prevented cuprizone-induced demyelination selectively in the cingulum, but not in the corpus callosum. These data support the CNS activities of ponesimod through S1P1 , including protective, and likely selective, effects against demyelination in a major connection pathway of the brain, the limbic fibers of the cingulum, lesions of which have been associated with several neurologic impairments including MS fatigue.


Subject(s)
Astrocytes/metabolism , Central Nervous System/drug effects , Neuroinflammatory Diseases/drug therapy , Neuroinflammatory Diseases/metabolism , Protective Agents/pharmacology , Sphingosine-1-Phosphate Receptors/metabolism , Thiazoles/pharmacology , Animals , Astrocytes/drug effects , Cell Line, Tumor , Cells, Cultured , Central Nervous System/metabolism , Humans , Male , Mice , Mice, Inbred C57BL , Signal Transduction/drug effects
3.
PLoS Biol ; 18(4): e3000656, 2020 04.
Article in English | MEDLINE | ID: mdl-32271748

ABSTRACT

Chemokines and their receptors are orchestrators of cell migration in humans. Because dysregulation of the receptor-chemokine system leads to inflammation and cancer, both chemokines and receptors are highly sought therapeutic targets. Yet one of the barriers for their therapeutic targeting is the limited understanding of the structural principles behind receptor-chemokine recognition and selectivity. The existing structures do not include CXC subfamily complexes and lack information about the receptor distal N-termini, despite the importance of the latter in signaling, regulation, and bias. Here, we report the discovery of the geometry of the complex between full-length CXCR4, a prototypical CXC receptor and driver of cancer metastasis, and its endogenous ligand CXCL12. By comprehensive disulfide cross-linking, we establish the existence and the structure of a novel interface between the CXCR4 distal N-terminus and CXCL12 ß1-strand, while also recapitulating earlier findings from nuclear magnetic resonance, modeling and crystallography of homologous receptors. A cross-linking-informed high-resolution model of the CXCR4-CXCL12 complex pinpoints the interaction determinants and reveals the occupancy of the receptor major subpocket by the CXCL12 proximal N terminus. This newly found positioning of the chemokine proximal N-terminus provides a structural explanation of CXC receptor-chemokine selectivity against other subfamilies. Our findings challenge the traditional two-site understanding of receptor-chemokine recognition, suggest the possibility of new affinity and signaling determinants, and fill a critical void on the structural map of an important class of therapeutic targets. These results will aid the rational design of selective chemokine-receptor targeting small molecules and biologics with novel pharmacology.


Subject(s)
Chemokine CXCL12/chemistry , Chemokine CXCL12/metabolism , Receptors, CXCR4/chemistry , Receptors, CXCR4/metabolism , Animals , Binding Sites , Blotting, Western , Chemokine CXCL12/genetics , Cysteine/chemistry , Cysteine/genetics , Disulfides/chemistry , Flow Cytometry , HEK293 Cells , Humans , Insecta/cytology , Models, Molecular , Mutation , Protein Conformation , Protein Interaction Domains and Motifs , Receptors, CXCR4/genetics , beta-Arrestins/metabolism
4.
Proc Natl Acad Sci U S A ; 117(46): 28763-28774, 2020 11 17.
Article in English | MEDLINE | ID: mdl-33139573

ABSTRACT

The molecular mechanisms by which receptor tyrosine kinases (RTKs) and heterotrimeric G proteins, two major signaling hubs in eukaryotes, independently relay signals across the plasma membrane have been extensively characterized. How these hubs cross-talk has been a long-standing question, but answers remain elusive. Using linear ion-trap mass spectrometry in combination with biochemical, cellular, and computational approaches, we unravel a mechanism of activation of heterotrimeric G proteins by RTKs and chart the key steps that mediate such activation. Upon growth factor stimulation, the guanine-nucleotide exchange modulator dissociates Gαi•ßγ trimers, scaffolds monomeric Gαi with RTKs, and facilitates the phosphorylation on two tyrosines located within the interdomain cleft of Gαi. Phosphorylation triggers the activation of Gαi and inhibits second messengers (cAMP). Tumor-associated mutants reveal how constitutive activation of this pathway impacts cell's decision to "go" vs. "grow." These insights define a tyrosine-based G protein signaling paradigm and reveal its importance in eukaryotes.


Subject(s)
GTP-Binding Protein alpha Subunits/metabolism , Heterotrimeric GTP-Binding Proteins/metabolism , Receptor Protein-Tyrosine Kinases/metabolism , Animals , COS Cells , Chlorocebus aethiops , ErbB Receptors/metabolism , HEK293 Cells , HeLa Cells , Heterotrimeric GTP-Binding Proteins/physiology , Humans , Phosphorylation , Receptor Protein-Tyrosine Kinases/physiology , Signal Transduction , Tyrosine/metabolism
5.
J Biol Chem ; 296: 100493, 2021.
Article in English | MEDLINE | ID: mdl-33675748

ABSTRACT

PDZ domains are one of the most abundant protein domains in eukaryotes and are frequently found on junction-localized scaffold proteins. Various signaling molecules bind to PDZ proteins via PDZ-binding motifs (PBM) and fine-tune cellular signaling. However, how such interaction affects protein function is difficult to predict and must be solved empirically. Here we describe a long isoform of the guanine nucleotide exchange factor GIV/Girdin (CCDC88A) that we named GIV-L, which is conserved throughout evolution, from invertebrates to vertebrates, and contains a PBM. Unlike GIV, which lacks PBM and is cytosolic, GIV-L localizes onto cell junctions and has a PDZ interactome (as shown through annotating Human Cell Map and BioID-proximity labeling studies), which impacts GIV-L's ability to bind and activate trimeric G-protein, Gαi, through its guanine-nucleotide exchange modulator (GEM) module. This GEM module is found exclusively in vertebrates. We propose that the two functional modules in GIV may have evolved sequentially: the ability to bind PDZ proteins via the PBM evolved earlier in invertebrates, whereas G-protein binding and activation may have evolved later only among vertebrates. Phenotypic studies in Caco-2 cells revealed that GIV and GIV-L may have antagonistic effects on cell growth, proliferation (cell cycle), and survival. Immunohistochemical analysis in human colon tissues showed that GIV expression increases with a concomitant decrease in GIV-L during cancer initiation. Taken together, these findings reveal how regulation in GIV/CCDC88A transcript helps to achieve protein modularity, which allows the protein to play opposing roles either as a tumor suppressor (GIV-L) or as an oncogene (GIV).


Subject(s)
Colonic Neoplasms/metabolism , GTP-Binding Protein alpha Subunits, Gi-Go/metabolism , Guanine Nucleotide Exchange Factors/metabolism , Microfilament Proteins/metabolism , Vesicular Transport Proteins/metabolism , Animals , Cell Line , Cell Line, Tumor/physiology , Cell Proliferation , Colonic Neoplasms/genetics , Colonic Neoplasms/pathology , Humans , Microfilament Proteins/chemistry , PDZ Domains , Phosphorylation , Protein Binding , Protein Isoforms , Protein Transport , Signal Transduction , Vesicular Transport Proteins/chemistry , Zebrafish
6.
Proc Natl Acad Sci U S A ; 116(33): 16394-16403, 2019 08 13.
Article in English | MEDLINE | ID: mdl-31363053

ABSTRACT

Heterotrimeric G proteins are key molecular switches that control cell behavior. The canonical activation of G proteins by agonist-occupied G protein-coupled receptors (GPCRs) has recently been elucidated from the structural perspective. In contrast, the structural basis for GPCR-independent G protein activation by a novel family of guanine-nucleotide exchange modulators (GEMs) remains unknown. Here, we present a 2.0-Å crystal structure of Gαi in complex with the GEM motif of GIV/Girdin. Nucleotide exchange assays, molecular dynamics simulations, and hydrogen-deuterium exchange experiments demonstrate that GEM binding to the conformational switch II causes structural changes that allosterically propagate to the hydrophobic core of the Gαi GTPase domain. Rearrangement of the hydrophobic core appears to be a common mechanism by which GPCRs and GEMs activate G proteins, although with different efficiency. Atomic-level insights presented here will aid structure-based efforts to selectively target the noncanonical G protein activation.


Subject(s)
GTP-Binding Protein alpha Subunits, Gi-Go/chemistry , Heterotrimeric GTP-Binding Proteins/chemistry , Microfilament Proteins/chemistry , Receptors, G-Protein-Coupled/chemistry , Vesicular Transport Proteins/chemistry , Allosteric Regulation/genetics , Crystallography, X-Ray , GTP-Binding Protein alpha Subunits, Gi-Go/genetics , Guanine Nucleotide Exchange Factors/chemistry , Guanine Nucleotide Exchange Factors/genetics , HeLa Cells , Heterotrimeric GTP-Binding Proteins/genetics , Humans , Hydrophobic and Hydrophilic Interactions , Microfilament Proteins/genetics , Molecular Dynamics Simulation , Protein Binding/genetics , Protein Conformation , Receptors, G-Protein-Coupled/genetics , Signal Transduction/genetics , Vesicular Transport Proteins/genetics
7.
Nat Chem Biol ; 13(2): 235-242, 2017 02.
Article in English | MEDLINE | ID: mdl-27992882

ABSTRACT

Understanding the pharmacological similarity of G protein-coupled receptors (GPCRs) is paramount for predicting ligand off-target effects, drug repurposing, and ligand discovery for orphan receptors. Phylogenetic relationships do not always correctly capture pharmacological similarity. Previous family-wide attempts to define pharmacological relationships were based on three-dimensional structures and/or known receptor-ligand pairings, both unavailable for orphan GPCRs. Here, we present GPCR-CoINPocket, a novel contact-informed neighboring pocket metric of GPCR binding-site similarity that is informed by patterns of ligand-residue interactions observed in crystallographically characterized GPCRs. GPCR-CoINPocket is applicable to receptors with unknown structure or ligands and accurately captures known pharmacological relationships between GPCRs, even those undetected by phylogeny. When applied to orphan receptor GPR37L1, GPCR-CoINPocket identified its pharmacological neighbors, and transfer of their pharmacology aided in discovery of the first surrogate ligands for this orphan with a 30% success rate. Although primarily designed for GPCRs, the method is easily transferable to other protein families.


Subject(s)
Drug Discovery , Ligands , Receptors, G-Protein-Coupled/antagonists & inhibitors , HEK293 Cells , Humans , Molecular Structure
10.
J Comput Aided Mol Des ; 27(4): 305-19, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23625023

ABSTRACT

The α1-adrenoceptors (α1-ARs), in particular the α1A-AR subtype, are current therapeutic targets of choice for the treatment of urogenital conditions, such as benign prostatic hyperplasia (BPH). Due to the similarity between the transmembrane domains of the α1-AR subtypes, and the serotonin receptor subtype 1A (5-HT1A-R), currently used α1-AR subtype-selective drugs to treat BPH display considerable off-target affinity for the 5-HT1A-R, leading to side effects. We describe the construction and validation of pharmacophores for 5-HT1A-R agonists and antagonists. Through the structural diversity of the training sets used in their development, these pharmacophores define the properties of a compound needed to bind to 5-HT1A receptors. Using these and previously published pharmacophores in virtual screening and profiling, we have identified unique chemical compounds (hits) that fit the requirements to bind to our target, the α1A-AR, selectively over the off-target, the 5-HT1A-R. Selected hits have been obtained and their affinities for α1A-AR, α1B-AR and 5-HT1A-R determined in radioligand binding assays, using membrane preparations which contain human receptors expressed individually. Three of the tested hits demonstrate statistically significant selectivity for α1A-AR over 5-HT1A-R. All seven tested hits bind to α1A-AR, with two compounds displaying K i values below 1 µM, and a further two K i values of around 10 µM. The insights and knowledge gained through the development of the new 5-HT1A-R pharmacophores will greatly aid in the design and synthesis of derivatives of our lead compound, and allow the generation of more efficacious and selective ligands.


Subject(s)
Drug Design , Receptor, Serotonin, 5-HT1A/metabolism , Serotonin 5-HT1 Receptor Antagonists/chemistry , Serotonin 5-HT1 Receptor Antagonists/pharmacology , Animals , COS Cells , Chlorocebus aethiops , Humans , Models, Molecular , Radioligand Assay , Recombinant Proteins/metabolism , Serotonin 5-HT1 Receptor Agonists/chemistry , Serotonin 5-HT1 Receptor Agonists/pharmacology
11.
Protein Expr Purif ; 82(1): 55-60, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22133714

ABSTRACT

Desensitization of agonist-activated G protein-coupled receptors (GPCRs) requires phosphorylation followed by the binding of arrestin, a ~48 kDa soluble protein. While crystal structures for the inactive, 'basal' state of various arrestins are available, the conformation of 'activated' arrestin adopted upon interaction with activated GPCRs remains unknown. As a first step towards applying high-resolution structural methods to study arrestin conformation and dynamics, we have utilized the subtilisin prodomain/Profinity eXact™ fusion-tag system for the high-level bacterial expression and one-step purification of wild-type visual arrestin (arrestin 1) as well as a mutant form (R175E) of the protein that binds to non-phosphorylated, light-activated rhodopsin (Rho∗). The results show that both prodomain/Profinity eXact™ fusion-tagged wild-type and R175E arrestins can be expressed to levels approaching 2-3 mg/l in Luria-Bertani media, and that the processed, tag-free mature forms can be purified to near homogeneity using a Bio-Scale™ Mini Profinity eXact™ cartridge on the Profinia™ purification system. Functional analysis of R175E arrestin generated using this approach shows that it binds to non-phosphorylated rhodopsin in a light-dependent manner. These findings should facilitate the structure determination of this 'constitutively activated' state of arrestin 1 as well as the monitoring of conformational changes upon interaction with Rho∗.


Subject(s)
Arrestin/isolation & purification , Arrestin/metabolism , Animals , Arrestin/chemistry , Arrestin/genetics , Cattle , Cloning, Molecular , Escherichia coli/genetics , Gene Expression , Genetic Vectors , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/isolation & purification , Recombinant Fusion Proteins/metabolism , Rhodopsin/metabolism , Subtilisin/chemistry , Subtilisin/genetics , Subtilisin/isolation & purification
12.
Sci Signal ; 13(640)2020 07 14.
Article in English | MEDLINE | ID: mdl-32665413

ABSTRACT

Because of their prominent roles in development, cancer, and HIV, the chemokine receptor CXCR4 and its ligand CXCL12 have been the subject of numerous structural and functional studies, but the determinants of ligand binding, selectivity, and signaling are still poorly understood. Here, building on our latest structural model, we used a systematic mutagenesis strategy to dissect the functional anatomy of the CXCR4-CXCL12 complex. Key charge swap mutagenesis experiments provided evidence for pairwise interactions between oppositely charged residues in the receptor and chemokine, confirming the accuracy of the predicted orientation of the chemokine relative to the receptor and providing insight into ligand selectivity. Progressive deletion of N-terminal residues revealed an unexpected contribution of the receptor N terminus to chemokine signaling. This finding challenges a longstanding "two-site" hypothesis about the essential features of the receptor-chemokine interaction in which the N terminus contributes only to binding affinity. Our results suggest that although the interaction of the chemokine N terminus with the receptor-binding pocket is the key driver of signaling, the signaling amplitude depends on the extent to which the receptor N terminus binds the chemokine. Together with systematic characterization of other epitopes, these data enable us to propose an experimentally consistent structural model for how CXCL12 binds CXCR4 and initiates signal transmission through the receptor transmembrane domain.


Subject(s)
Chemokine CXCL12/chemistry , Models, Molecular , Multiprotein Complexes/chemistry , Receptors, CXCR4/chemistry , Animals , CHO Cells , Chemokine CXCL12/genetics , Chemokine CXCL12/metabolism , Cricetulus , HEK293 Cells , Humans , Multiprotein Complexes/genetics , Multiprotein Complexes/metabolism , Mutagenesis, Site-Directed , Protein Structure, Quaternary , Receptors, CXCR4/genetics , Receptors, CXCR4/metabolism
13.
Sci Rep ; 10(1): 19995, 2020 11 17.
Article in English | MEDLINE | ID: mdl-33203955

ABSTRACT

GPR37L1 is an orphan G protein-coupled receptor expressed exclusively in the brain and linked to seizures, neuroprotection and cardiovascular disease. Based upon the observation that fragments of the GPR37L1 N-terminus are found in human cerebrospinal fluid, we hypothesized that GPR37L1 was subject to post-translational modification. Heterologous expression of GPR37L1-eYFP in either HEK293 or U87 glioblastoma cells yielded two cell surface species of approximately equivalent abundance, the larger of which is N-glycosylated at Asn105. The smaller species is produced by matrix metalloprotease/ADAM-mediated proteolysis (shown by the use of pharmacological inhibitors) and has a molecular weight identical to that of a mutant lacking the entire N-terminus, Δ122 GPR37L1. Serial truncation of the N-terminus prevented GPR37L1 expression except when the entire N-terminus was removed, narrowing the predicted site of N-terminal proteolysis to residues 105-122. Using yeast expressing different G protein chimeras, we found that wild type GPR37L1, but not Δ122 GPR37L1, coupled constitutively to Gpa1/Gαs and Gpa1/Gα16 chimeras, in contrast to previous studies. We tested the peptides identified in cerebrospinal fluid as well as their putative newly-generated N-terminal 'tethered' counterparts in both wild type and Δ122 GPR37L1 Gpa1/Gαs strains but saw no effect, suggesting that GPR37L1 does not signal in a manner akin to the protease-activated receptor family. We also saw no evidence of receptor activation or regulation by the reported GPR37L1 ligand, prosaptide/TX14A. Finally, the proteolytically processed species predominated both in vivo and ex vivo in organotypic cerebellar slice preparations, suggesting that GPR37L1 is rapidly processed to a signaling-inactive form. Our data indicate that the function of GPR37L1 in vivo is tightly regulated by metalloprotease-dependent N-terminal cleavage.


Subject(s)
Matrix Metalloproteinases/metabolism , Receptors, G-Protein-Coupled/metabolism , Animals , Cell Line, Tumor , Cerebellum/metabolism , Female , GTP-Binding Proteins/metabolism , HEK293 Cells , Humans , Male , Mice , Mice, Inbred C57BL , Nerve Growth Factors/metabolism , Peptide Hydrolases/metabolism , Proteolysis , Rats , Rats, Sprague-Dawley , Signal Transduction/physiology
14.
Photochem Photobiol ; 85(2): 431-6, 2009.
Article in English | MEDLINE | ID: mdl-19222798

ABSTRACT

To better understand the mechanism by which the activating signal is transmitted from the receptor-interacting regions on the G protein alpha-subunit (G(alpha)) to the guanine nucleotide-binding pocket, we generated and characterized mutant forms of G(alpha) with alterations in switch II (Trp-207-->Phe) and the carboxyl-terminus (Phe-350-->Ala). Previously reported bacterial expression methods for the high-level production of a uniformly isotope-labeled G(talpha)/G(i1alpha) chimera, ChiT, were successfully used to isolate milligram quantities of (15)N-labeled mutant protein. NMR analysis showed that while the GDP/Mg(2+)-bound state of both mutants shared an overall conformation similar to that of the GDP/Mg(2+)-bound state of ChiT, formation of the "transition/activated" state in the presence of aluminum fluoride (AlF(4) (-)) revealed distinct differences between the wild-type and mutant G(alpha) subunits, particularly in the response of the (1)HN, (15)N cross-peak for the Trp-254 indole in the Trp-207-->Phe mutant and the (1)HN, (15)N cross-peak for Ala-350 in the Phe-350-->Ala mutant. Consistent with the NMR data, the F350-->Ala mutant showed an increase in intrinsic fluorescence that was similar to G(talpha) and ChiT upon formation of the "transition/activated" state in the presence of AlF(4) (-), whereas the intrinsic fluorescence of the Trp-207-->Phe mutant decreased. These results show that the substitution of key amino acid positions in G(alpha) can effect structural changes that may compromise receptor interactions and GDP/GTP exchange.


Subject(s)
GTP-Binding Protein alpha Subunits/chemistry , GTP-Binding Protein alpha Subunits/metabolism , Point Mutation/genetics , Aluminum Compounds , Fluorescent Dyes/chemistry , Fluorides , GTP-Binding Protein alpha Subunits/genetics , GTP-Binding Protein alpha Subunits/isolation & purification , Gene Expression , Models, Molecular , Nuclear Magnetic Resonance, Biomolecular , Protein Multimerization , Protein Structure, Quaternary , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Tryptophan/chemistry
15.
Biol Sex Differ ; 9(1): 14, 2018 04 06.
Article in English | MEDLINE | ID: mdl-29625592

ABSTRACT

BACKGROUND: Over 100 mammalian G protein-coupled receptors are yet to be matched with endogenous ligands; these so-called orphans are prospective drug targets for the treatment of disease. GPR37L1 is one such orphan, abundant in the brain and detectable as mRNA in the heart and kidney. GPR37L1 ablation was reported to cause hypertension and left ventricular hypertrophy, and thus, we sought to further define the role of GPR37L1 in blood pressure homeostasis. METHODS: We investigated the cardiovascular effects of GPR37L1 using wild-type (GPR37L1wt/wt) and null (GPR37L1KO/KO) mice established on a C57BL/6J background, both under baseline conditions and during AngII infusion. We profiled GPR37L1 tissue expression, examining the endogenous receptor by immunoblotting and a ß-galactosidase reporter mouse by immunohistochemistry. RESULTS: GPR37L1 protein was abundant in the brain but not detectable in the heart and kidney. We measured blood pressure in GPR37L1wt/wt and GPR37L1KO/KO mice and found that deletion of GPR37L1 causes a female-specific increase in systolic, diastolic, and mean arterial pressures. When challenged with short-term AngII infusion, only male GPR37L1KO/KO mice developed exacerbated left ventricular hypertrophy and evidence of heart failure, while the female GPR37L1KO/KO mice were protected from cardiac fibrosis. CONCLUSIONS: Despite its absence in the heart and kidney, GPR37L1 regulates baseline blood pressure in female mice and is crucial for cardiovascular compensatory responses in males. The expression of GPR37L1 in the brain, yet absence from peripheral cardiovascular tissues, suggests this orphan receptor is a hitherto unknown contributor to central cardiovascular control.


Subject(s)
Blood Pressure , Receptors, G-Protein-Coupled/physiology , Animals , Brain/metabolism , Female , Fibrosis , Kidney/metabolism , Male , Mice, Inbred C57BL , Mice, Knockout , Myocardium/metabolism , Myocardium/pathology , Sex Characteristics
16.
Cell Signal ; 33: 1-9, 2017 05.
Article in English | MEDLINE | ID: mdl-28188824

ABSTRACT

G protein-coupled receptors (GPCRs) are a vast family of membrane-traversing proteins, essential to the ability of eukaryotic life to detect, and mount an intracellular response to, a diverse range of extracellular stimuli. GPCRs have evolved with archetypal features including an extracellular N-terminus and intracellular C-terminus that flank a transmembrane structure of seven sequential helices joined by intracellular and extracellular loops. These structural domains contribute to the ability of a GPCR to be correctly synthesised and inserted into the cell membrane, to interact with its cognate ligand(s) and to couple with signal-transducing heterotrimeric G proteins, allowing the activated receptor to selectively modulate a number of signalling cascades. Whilst well known for its importance in receptor translation and trafficking, the GPCR N-terminus is underexplored as a participant in receptor signalling. This review aims to discuss and integrate recent advances in knowledge of the vital roles of the GPCR N-terminus in receptor signalling.


Subject(s)
Receptors, G-Protein-Coupled/chemistry , Receptors, G-Protein-Coupled/metabolism , Signal Transduction , Animals , Cell Adhesion , Humans , Models, Molecular , Peptide Hydrolases/metabolism
17.
PeerJ ; 5: e3765, 2017.
Article in English | MEDLINE | ID: mdl-28948099

ABSTRACT

BACKGROUND: The rapid increase in antibiotic resistance by various bacterial pathogens underlies the significance of developing new therapies and exploring different drug targets. A fraction of bacterial pathogens abbreviated as ESKAPE by the European Center for Disease Prevention and Control have been considered a major threat due to the rise in nosocomial infections. Here, we compared putative drug binding pockets of twelve essential and mostly conserved metabolic enzymes in numerous bacterial pathogens including those of the ESKAPE group and Mycobacterium tuberculosis. The comparative analysis will provide guidelines for the likelihood of transferability of the inhibitors from one species to another. METHODS: Nine bacterial species including six ESKAPE pathogens, Mycobacterium tuberculosis along with Mycobacterium smegmatis and Eschershia coli, two non-pathogenic bacteria, have been selected for drug binding pocket analysis of twelve essential enzymes. The amino acid sequences were obtained from Uniprot, aligned using ICM v3.8-4a and matched against the Pocketome encyclopedia. We used known co-crystal structures of selected target enzyme orthologs to evaluate the location of their active sites and binding pockets and to calculate a matrix of pairwise sequence identities across each target enzyme across the different species. This was used to generate sequence maps. RESULTS: High sequence identity of enzyme binding pockets, derived from experimentally determined co-crystallized structures, was observed among various species. Comparison at both full sequence level and for drug binding pockets of key metabolic enzymes showed that binding pockets are highly conserved (sequence similarity up to 100%) among various ESKAPE pathogens as well as Mycobacterium tuberculosis. Enzymes orthologs having conserved binding sites may have potential to interact with inhibitors in similar way and might be helpful for design of similar class of inhibitors for a particular species. The derived pocket alignments and distance-based maps provide guidelines for drug discovery and repurposing. In addition they also provide recommendations for the relevant model bacteria that may be used for initial drug testing. DISCUSSION: Comparing ligand binding sites through sequence identity calculation could be an effective approach to identify conserved orthologs as drug binding pockets have shown higher level of conservation among various species. By using this approach we could avoid the problems associated with full sequence comparison. We identified essential metabolic enzymes among ESKAPE pathogens that share high sequence identity in their putative drug binding pockets (up to 100%), of which known inhibitors can potentially antagonize these identical pockets in the various species in a similar manner.

18.
Structure ; 10(4): 515-25, 2002 Apr.
Article in English | MEDLINE | ID: mdl-11937056

ABSTRACT

Components from the extracellular surface of CCR5 interact with certain macrophage-tropic strains of human immunodeficiency virus type 1 (HIV-1) to mediate viral fusion and entry. To mimic these viral interacting site(s), the amino-terminal and extracellular loop segments of CCR5 were linked in tandem to form concatenated polypeptides, or grafted onto a seven-transmembrane bacteriorhodopsin scaffold to generate several chimeras. The chimera studies identified specific regions in CCR5 that confer HIV-1 coreceptor function, structural rearrangements in the transmembrane region that may modulate this activity, and a role for the extracellular surface in folding and assembly. Methods developed here may be applicable to the dissection of functional domains from other seven-transmembrane receptors and form a basis for future structural studies.


Subject(s)
Bacteriorhodopsins/chemistry , HIV-1/metabolism , Receptors, CCR5/chemistry , Receptors, HIV/metabolism , Amino Acid Sequence , Animals , Bacteriorhodopsins/genetics , Bacteriorhodopsins/metabolism , COS Cells , Cattle , Genes, Reporter , HIV-1/chemistry , Humans , Molecular Sequence Data , Protein Structure, Secondary , Receptors, CCR5/genetics , Receptors, CCR5/metabolism , Receptors, HIV/chemistry , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Spectrum Analysis
19.
Br J Pharmacol ; 173(20): 2934-51, 2016 10.
Article in English | MEDLINE | ID: mdl-26837045

ABSTRACT

GPCRs are the most successful pharmaceutical targets in history. Nevertheless, the pharmacology of many GPCRs remains inaccessible as their endogenous or exogenous modulators have not been discovered. Tools that explore the physiological functions and pharmacological potential of these 'orphan' GPCRs, whether they are endogenous and/or surrogate ligands, are therefore of paramount importance. Rates of receptor deorphanization determined by traditional reverse pharmacology methods have slowed, indicating a need for the development of more sophisticated and efficient ligand screening approaches. Here, we discuss the use of structure-based ligand discovery approaches to identify small molecule modulators for exploring the function of orphan GPCRs. These studies have been buoyed by the growing number of GPCR crystal structures solved in the past decade, providing a broad range of template structures for homology modelling of orphans. This review discusses the methods used to establish the appropriate signalling assays to test orphan receptor activity and provides current examples of structure-based methods used to identify ligands of orphan GPCRs. Linked Articles This article is part of a themed section on Molecular Pharmacology of G Protein-Coupled Receptors. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v173.20/issuetoc.


Subject(s)
Receptors, G-Protein-Coupled/chemistry , Animals , Humans , Ligands , Models, Biological , Receptors, G-Protein-Coupled/agonists , Receptors, G-Protein-Coupled/antagonists & inhibitors
20.
Sci Signal ; 9(423): ra36, 2016 Apr 12.
Article in English | MEDLINE | ID: mdl-27072655

ABSTRACT

Little is known about the pharmacology or physiology of GPR37L1, a G protein (heterotrimeric guanine nucleotide-binding protein)-coupled receptor that is abundant in the cerebellum. Mice deficient in this receptor exhibit precocious cerebellar development and hypertension. We showed that GPR37L1 coupled to the G protein Gα(s) when heterologously expressed in cultured cells in the absence of any added ligand, whereas a mutant receptor that lacked the amino terminus was inactive. Conversely, inhibition of ADAMs (a disintegrin and metalloproteases) enhanced receptor activity, indicating that the presence of the amino terminus is necessary for GPR37L1 signaling. Metalloprotease-dependent processing of GPR37L1 was evident in rodent cerebellum, where we detected predominantly the cleaved, inactive form. However, comparison of the accumulation of cAMP (adenosine 3',5'-monophosphate) in response to phosphodiesterase inhibition in cerebellar slice preparations from wild-type and GPR37L1-null mice showed that some constitutive signaling remained in the wild-type mice. In reporter assays of Gα(s) or Gα(i) signaling, the synthetic, prosaposin-derived peptide prosaptide (TX14A) did not increase GPR37L1 activity. Our data indicate that GPR37L1 may be a constitutively active receptor, or perhaps its ligand is present under the conditions that we used for analysis, and that the activity of this receptor is instead controlled by signals that regulate metalloprotease activity in the tissue.


Subject(s)
Metalloproteases/metabolism , Mutant Proteins/metabolism , Receptors, G-Protein-Coupled/metabolism , Signal Transduction , ADAM Proteins/metabolism , Amino Acid Sequence , Animals , Female , GTP-Binding Protein alpha Subunits, Gs/metabolism , HEK293 Cells , Humans , Immunoblotting , Male , Mice, Inbred C57BL , Mice, Knockout , Microscopy, Confocal , Mutant Proteins/genetics , Proteolysis , Rats, Sprague-Dawley , Receptors, G-Protein-Coupled/genetics , Sequence Homology, Amino Acid
SELECTION OF CITATIONS
SEARCH DETAIL