Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 53
Filter
1.
Nat Commun ; 10(1): 5490, 2019 12 02.
Article in English | MEDLINE | ID: mdl-31792216

ABSTRACT

Field cancerization is a premalignant process marked by clones of oncogenic mutations spreading through the epithelium. The timescales of intestinal field cancerization can be variable and the mechanisms driving the rapid spread of oncogenic clones are unknown. Here we use a Cancer rainbow (Crainbow) modelling system for fluorescently barcoding somatic mutations and directly visualizing the clonal expansion and spread of oncogenes. Crainbow shows that mutations of ß-catenin (Ctnnb1) within the intestinal stem cell results in widespread expansion of oncogenes during perinatal development but not in adults. In contrast, mutations that extrinsically disrupt the stem cell microenvironment can spread in adult intestine without delay. We observe the rapid spread of premalignant clones in Crainbow mice expressing oncogenic Rspondin-3 (RSPO3), which occurs by increasing crypt fission and inhibiting crypt fixation. Crainbow modelling provides insight into how somatic mutations rapidly spread and a plausible mechanism for predetermining the intratumor heterogeneity found in colon cancers.


Subject(s)
Colonic Neoplasms/genetics , Disease Models, Animal , Neoplastic Stem Cells/cytology , Animals , Carcinogenesis , Cell Proliferation , Colonic Neoplasms/metabolism , Colonic Neoplasms/physiopathology , Humans , Mice , Mutation , Neoplastic Stem Cells/metabolism , Oncogenes , Thrombospondins/genetics , Thrombospondins/metabolism
2.
ACS Pharmacol Transl Sci ; 2(4): 230-246, 2019 Aug 09.
Article in English | MEDLINE | ID: mdl-32259059

ABSTRACT

G-protein-coupled receptors (GPCRs) can bias signaling through distinct biochemical pathways that originate from G-protein/receptor and ß-arrestin/receptor complexes. Receptor conformations supporting ß-arrestin engagement depend on multiple receptor determinants. Using ghrelin receptor GHR1a, we demonstrate by bioluminescence resonance energy transfer and fluorescence microscopy a critical role for its second intracellular loop 2 (ICL2) domain in stabilizing ß-arrestin/GHSR1a core interactions and determining receptor trafficking fate. We validate our findings in ICL2 gain- and loss-of-function experiments assessing ß-arrestin and ubiquitin-dependent internalization of the CC chemokine receptor, CCR1. Like all CC and CXC subfamily chemokine receptors, CCR1 lacks a critical proline residue found in the ICL2 consensus domain of rhodopsin-family GPCRs. Our study indicates that ICL2, C-tail determinants, and the orthosteric binding pocket that regulates ß-arrestin/receptor complex stability are sufficient to encode a broad repertoire of the trafficking fates observed for rhodopsin-family GPCRs, suggesting they provide the essential elements for regulating a large fraction of ß-arrestin signaling bias.

3.
Bioorg Med Chem ; 25(16): 4355-4367, 2017 08 15.
Article in English | MEDLINE | ID: mdl-28673732

ABSTRACT

GPR55, a G protein-coupled receptor, is an attractive target to alleviate inflammatory and neuropathic pain and treat osteoporosis and cancer. Identifying a potent and selective ligand will aid to further establish the specific physiological roles and pharmacology of the receptor. Towards this goal, a targeted library of 22 compounds was synthesized in a modular fashion to obtain structure-activity relationship information. The general route consisted of coupling a variety of p-aminophenyl sulfonamides to isothiocyanates to form acylthioureas. For the synthesis of a known naphthyl ethyl alcohol motif, route modification led to a shorter and more efficient process. The 22 analogues were analyzed for their ability to serve as agonists at GPR55 and valuable information for both ends of the molecule was ascertained.


Subject(s)
Drug Design , Receptors, G-Protein-Coupled/agonists , Thiourea/pharmacology , Dose-Response Relationship, Drug , Humans , Molecular Structure , Receptors, Cannabinoid , Structure-Activity Relationship , Thiourea/analogs & derivatives , Thiourea/chemical synthesis
4.
FASEB J ; 31(6): 2507-2519, 2017 06.
Article in English | MEDLINE | ID: mdl-28242772

ABSTRACT

Apelin signaling plays an important role during embryo development and regulates angiogenesis, cardiovascular activity, and energy metabolism in adulthood. Overexpression and hyperactivity of this signaling pathway is observed in various pathologic states, such as cardiovascular diseases and cancer, which highlights the importance of inhibiting apelin receptor (APJ); therefore, we developed a cell-based screening assay that uses fluorescence microscopy to identify APJ antagonists. This approach led us to identify the U.S. Food and Drug Administration-approved compound protamine-already used clinically after cardiac surgery-as an agent to bind to heparin and thereby reverse its anticlotting activity. Protamine displays a 390-nM affinity for APJ and behaves as a full antagonist with regard to G protein and ß-arrestin-dependent intracellular signaling. Ex vivo and in vivo, protamine abolishes well-known apelin effects, such as angiogenesis, glucose tolerance, and vasodilatation. Remarkably, protamine antagonist activity is fully reversed by heparin treatment both in vitro and in vivo Thus, our results demonstrate a new pharmacologic property of protamine-blockade of APJ-that could explain some adverse effects observed in protamine-treated patients. Moreover, our data reveal that the established antiangiogenic activity of protamine would rely on APJ antagonism.-Le Gonidec, S., Chaves-Almagro, C., Bai, Y., Kang, H. J., Smith, A., Wanecq, E., Huang, X.-P., Prats, H., Knibiehler, B., Roth, B. L., Barak, L. S., Caron, M. G., Valet, P., Audigier, Y., Masri, B. Protamine is an antagonist of apelin receptor, and its activity is reversed by heparin.


Subject(s)
Heparin/pharmacology , Protamines/pharmacology , Receptors, G-Protein-Coupled/antagonists & inhibitors , Animals , Apelin Receptors , Cell Line, Tumor , Female , Gene Expression Regulation/drug effects , Gene Expression Regulation/physiology , HEK293 Cells , Humans , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/metabolism
5.
J Biol Chem ; 292(17): 7208-7222, 2017 04 28.
Article in English | MEDLINE | ID: mdl-28275053

ABSTRACT

The leucine-rich G protein-coupled receptor-5 (LGR5) is expressed in adult tissue stem cells of many epithelia, and its overexpression is negatively correlated with cancer prognosis. LGR5 potentiates WNT/ß-catenin signaling through its unique constitutive internalization property that clears negative regulators of the WNT-receptor complex from the membrane. However, both the mechanism and physiological relevance of LGR5 internalization are unclear. Therefore, a natural product library was screened to discover LGR5 internalization inhibitors and gain mechanistic insight into LGR5 internalization. The plant lignan justicidin B blocked the constitutive internalization of LGR5. Justicidin B is structurally similar to more potent vacuolar-type H+-ATPase inhibitors, which all inhibited LGR5 internalization by blocking clathrin-mediated endocytosis. We then tested the physiological relevance of LGR5 internalization blockade in vivo A LGR5-rainbow (LBOW) mouse line was engineered to express three different LGR5 isoforms along with unique fluorescent protein lineage reporters in the same mouse. In this manner, the effects of each isoform on cell fate can be simultaneously assessed through simple fluorescent imaging for each lineage reporter. LBOW mice express three different forms of LGR5, a wild-type form that constitutively internalizes and two mutant forms whose internalization properties have been compromised by genetic perturbations within the carboxyl-terminal tail. LBOW was activated in the intestinal epithelium, and a year-long lineage-tracing course revealed that genetic blockade of LGR5 internalization diminished cell fitness. Together these data provide proof-of-concept genetic evidence that blocking the clathrin-mediated endocytosis of LGR5 could be used to pharmacologically control cell behavior.


Subject(s)
Clathrin/chemistry , Endocytosis , Leucine/chemistry , Receptors, G-Protein-Coupled/chemistry , Adenosine Triphosphatases/chemistry , Animals , Cell Line, Tumor , Cell Lineage , Cell Proliferation , Dioxolanes/chemistry , Epithelium/metabolism , Female , Homeostasis , Humans , Lignans/chemistry , Mice , Mice, Inbred C57BL , Protein Isoforms , Rats , Stem Cells/cytology , Stochastic Processes , Wnt Signaling Pathway
6.
Bioorg Med Chem ; 25(6): 1804-1816, 2017 03 15.
Article in English | MEDLINE | ID: mdl-28233680

ABSTRACT

The Wnt signaling pathway plays a key role in organ and tissue homeostasis, and when dysregulated, can become a major underlying mechanism of disease, particularly cancer. We reported previously that the anthelmintic drug Niclosamide inhibits Wnt/ß-catenin signaling and suppresses colon cancer cell growth in vitro and in vivo. To define Niclosamide's mechanism of Wnt/ß-catenin inhibition, and to improve its selectivity and pharmacokinetic properties as an anticancer treatment, we designed a novel class of benzimidazole inhibitors of Wnt/ß-catenin signaling based on SAR studies of the Niclosamide salicylanilide chemotype. Niclosamide has multiple biological activities. To address selectivity in our design, we interrogated a protonophore SAR model and used the principle of conformational restriction to identify novel Wnt/ß-catenin inhibitors with less effect on ATP cellular homeostasis. These studies led to the identification of 4-chloro-2-(5-(trifluoromethyl)-1H-benzo[d]imidazol-2-yl) phenol (4) and related derivatives with greater selectivity for Wnt/ß-catenin signaling inhibition vs. differential effects on cellular ATP homeostasis. This is the first report that the Wnt signaling inhibitory activity of Niclosamide can be translated into a new chemical class and to show that its effects on ATP homeostasis can be separated from its inhibitory effects on Wnt signaling. These compounds could be useful tools to elucidate the mechanism of Niclosamide's inhibition of Wnt signaling, and aid the discovery of inhibitors with improved pharmacologic properties to treat cancer and diseases in which Niclosamide has important biological activity.


Subject(s)
Adenosine Triphosphate/metabolism , Benzimidazoles/pharmacology , Niclosamide/pharmacology , Signal Transduction/drug effects , Wnt Proteins/metabolism , beta Catenin/metabolism , Benzimidazoles/chemistry , Cell Line, Tumor , HEK293 Cells , Homeostasis , Humans , Niclosamide/chemistry , Structure-Activity Relationship
7.
ACS Chem Biol ; 12(3): 702-714, 2017 03 17.
Article in English | MEDLINE | ID: mdl-28045487

ABSTRACT

Nicotinic acetylcholine receptors regulate the nicotine dependence encountered with cigarette smoking, and this has stimulated a search for drugs binding the responsible receptor subtypes. Studies link a gene cluster encoding for α3ß4α5-D398N nicotinic acetylcholine receptors to lung cancer risk as well as link a second mutation in this cluster to an increased risk for nicotine dependence. However, there are currently no recognized drugs for discriminating α3ß4α5 signaling. In this study, we describe the development of homogeneous HEK-293 cell clones of α3ß4 and α3ß4α5 receptors appropriate for drug screening and characterizing biochemical and pharmacological properties of incorporated α5 subunits. Clones were assessed for plasma membrane expression of the individual receptor subunits by mass spectrometry and immunochemistry, and their calcium flux was measured in the presence of a library of kinase inhibitors and a focused library of acetylcholine receptor ligands. We demonstrated an incorporation of two α3 subunits in approximately 98% of plasma membrane receptor pentamers, indicating a 2/3 subunit expression ratio of α3 to ß4 alone or to coexpressed ß4 and α5. With prolonged nicotine exposure, the plasma membrane expression of receptors with and without incorporated α5 increased. Whereas α5 subunit expression decreased the cell calcium response to nicotine and reduced plasma membrane receptor number, it partially protected receptors from nicotine mediated desensitization. Hit compounds from both libraries suggest the α5 and α5-D398N subunits allosterically modify the behavior of nicotine at the parent α3ß4 nicotinic acetylcholine receptor. These studies identify pharmacological tools from two distinct classes of drugs, antagonists and modifiers that are α5 and α5-D398N subtype selective that provide a means to characterize the role of the CHRNA5/A3/B4 gene cluster in smoking and cancer.


Subject(s)
Receptors, Nicotinic/metabolism , Allosteric Regulation , Calcium/metabolism , Electrophoresis, Polyacrylamide Gel , HEK293 Cells , Humans , Ligands
8.
ACS Chem Biol ; 11(7): 1880-90, 2016 07 15.
Article in English | MEDLINE | ID: mdl-27119457

ABSTRACT

Pharmacological treatment for methamphetamine addiction will provide important societal benefits. Neurotensin receptor NTR1 and dopamine receptor distributions coincide in brain areas regulating methamphetamine-associated reward, and neurotensin peptides produce behaviors opposing psychostimulants. Therefore, undesirable methamphetamine-associated activities should be treatable with druggable NTR1 agonists, but no such FDA-approved therapeutics exist. We address this limitation with proof-of-concept data for ML314, a small-molecule, brain penetrant, ß-arrestin biased, NTR1 agonist. ML314 attenuates amphetamine-like hyperlocomotion in dopamine transporter knockout mice, and in C57BL/6J mice it attenuates methamphetamine-induced hyperlocomotion, potentiates the psychostimulant inhibitory effects of a ghrelin antagonist, and reduces methamphetamine-associated conditioned place preference. In rats, ML314 blocks methamphetamine self-administration. ML314 acts as an allosteric enhancer of endogenous neurotensin, unmasking stoichiometric numbers of hidden NTR1 binding sites in transfected-cell membranes or mouse striatal membranes, while additionally supporting NTR1 endocytosis in cells in the absence of NT peptide. These results indicate ML314 is a viable, preclinical lead for methamphetamine abuse treatment and support an allosteric model of G protein-coupled receptor signaling.


Subject(s)
Amphetamine-Related Disorders/metabolism , Methamphetamine/adverse effects , Piperazines/metabolism , Quinazolines/metabolism , Receptors, Neurotensin/metabolism , Allosteric Regulation , Animals , Dopamine Plasma Membrane Transport Proteins/genetics , Ligands , Locomotion/drug effects , Mice , Mice, Inbred C57BL , Mice, Knockout
9.
Bioorg Med Chem Lett ; 26(7): 1827-1830, 2016 Apr 01.
Article in English | MEDLINE | ID: mdl-26916440

ABSTRACT

A series of 1,3,4-oxadiazol-2-ones was synthesized and tested for activity as antagonists at GPR55 in cellular beta-arrestin redistribution assays. The synthesis was designed to be modular in nature so that a sufficient number of analogues could be rapidly accessed to explore initial structure-activity relationships. The design of analogues was guided by the docking of potential compounds into a model of the inactive form of GPR55. The results of the assays were used to learn more about the binding pocket of GPR55. With this oxadiazolone scaffold, it was determined that modification of the aryl group adjacent to the oxadiazolone ring was often detrimental and that the distal cyclopropane was beneficial for activity. These results will guide further exploration of this receptor.


Subject(s)
Drug Design , Oxadiazoles/chemistry , Oxadiazoles/pharmacology , Piperidines/chemistry , Piperidines/pharmacology , Receptors, G-Protein-Coupled/antagonists & inhibitors , Animals , Arrestins/metabolism , CHO Cells , Cricetulus , Humans , Molecular Docking Simulation , Oxadiazoles/chemical synthesis , Piperidines/chemical synthesis , Receptors, Cannabinoid , Receptors, G-Protein-Coupled/metabolism , Structure-Activity Relationship , beta-Arrestins
10.
BMC Biol ; 13: 107, 2015 Dec 17.
Article in English | MEDLINE | ID: mdl-26678094

ABSTRACT

BACKGROUND: Membrane proteins regulate a diversity of physiological processes and are the most successful class of targets in drug discovery. However, the number of targets adequately explored in chemical space and the limited resources available for screening are significant problems shared by drug-discovery centers and small laboratories. Therefore, a low-cost and universally applicable screen for membrane protein trafficking was developed. RESULTS: This high-throughput screen (HTS), termed IRFAP-HTS, utilizes the recently described MarsCy1-fluorogen activating protein and the near-infrared and membrane impermeant fluorogen SCi1. The cell surface expression of MarsCy1 epitope-tagged receptors can be visualized by simple addition of SCi1. User-friendly, rapid, and quantitative detection occurs on a standard infrared western-blotting scanner. The reliability and robustness of IRFAP-HTS was validated by confirming human vasopressin-2 receptor and dopamine receptor-2 trafficking in response to agonist or antagonist. The IRFAP-HTS screen was deployed against the leucine-rich G protein-coupled receptor-5 (Lgr5). Lgr5 is expressed in stem cells, modulates Wnt/ß-catenin signaling, and is therefore a promising drug target. However, small molecule modulators have yet to be reported. The constitutive internalization of Lgr5 appears to be one primary mode through which its function is regulated. Therefore, IRFAP-HTS was utilized to screen 11,258 FDA-approved and drug-like small molecules for those that antagonize Lgr5 internalization. Glucocorticoids were found to potently increase Lgr5 expression at the plasma membrane. CONCLUSION: The IRFAP-HTS platform provides a versatile solution for screening more targets with fewer resources. Using only a standard western-blotting scanner, we were able to screen 5,000 compounds per hour in a robust and quantitative assay. Multi-purposing standardly available laboratory equipment eliminates the need for idiosyncratic and more expensive high-content imaging systems. The modular and user-friendly IRFAP-HTS is a significant departure from current screening platforms. Small laboratories will have unprecedented access to a robust and reliable screening platform and will no longer be limited by the esoteric nature of assay development, data acquisition, and post-screening analysis. The discovery of glucocorticoids as modulators for Lgr5 trafficking confirms that IRFAP-HTS can accelerate drug-discovery and drug-repurposing for even the most obscure targets.


Subject(s)
Drug Discovery/methods , High-Throughput Screening Assays/methods , Membrane Proteins/metabolism , Drug Discovery/economics , HEK293 Cells , High-Throughput Screening Assays/economics , Humans , Protein Transport , Reproducibility of Results
11.
Bioorg Med Chem ; 23(17): 5829-38, 2015 Sep 01.
Article in English | MEDLINE | ID: mdl-26272032

ABSTRACT

The Wnt signaling pathway plays a key role in regulation of organ development and tissue homeostasis. Dysregulated Wnt activity is one of the major underlying mechanisms responsible for many diseases including cancer. We previously reported the FDA-approved anthelmintic drug Niclosamide inhibits Wnt/ß-catenin signaling and suppresses colon cancer cell growth in vitro and in vivo. Niclosamide is a multi-functional drug that possesses important biological activity in addition to inhibition of Wnt/ß-catenin signaling. Here, we studied the SAR of Wnt signaling inhibition in the anilide and salicylamide region of Niclosamide. We found that the 4'-nitro substituent can be effectively replaced by trifluoromethyl or chlorine and that the potency of inhibition was dependent on the substitution pattern in the anilide ring. Non-anilide, N-methyl amides and reverse amide derivatives lost significant potency, while acylated salicylamide derivatives inhibited signaling with potency similar to non-acyl derivatives. Niclosamide's low systemic exposure when dosed orally may hinder its use to treat systemic disease. To overcome this limitation we identified an acyl derivative of Niclosamide, DK-520 (compound 32), that significantly increased both the plasma concentration and the duration of exposure of Niclosamide when dosed orally. The studies herein provide a medicinal chemical foundation to improve the pharmacokinetic exposure of Niclosamide and Wnt-signaling inhibitors based on the Niclosamide chemotype. The identification of novel derivatives of Niclosamide that metabolize to Niclosamide and increase its drug exposure may provide important research tools for in vivo studies and provide drug candidates for treating cancers with dysregulated Wnt signaling including drug-resistant cancers. Moreover, since Niclosamide is a multi-functional drug, new research tools such as DK520 could directly result in novel treatments against bacterial and viral infection, lupus, and metabolic diseases such as type II diabetes, NASH and NAFLD.


Subject(s)
Niclosamide/therapeutic use , Wnt Proteins/antagonists & inhibitors , Wnt Signaling Pathway/drug effects , beta Catenin/metabolism , Cell Line, Tumor , Cell Proliferation , Humans , Structure-Activity Relationship
12.
Breast Cancer Res ; 17: 20, 2015 Feb 15.
Article in English | MEDLINE | ID: mdl-25849870

ABSTRACT

INTRODUCTION: Human epidermal growth factor receptor HER3 has been implicated in promoting the aggressiveness and metastatic potential of breast cancer. Upregulation of HER3 has been found to be a major mechanism underlying drug resistance to EGFR and HER2 tyrosine kinase inhibitors and to endocrine therapy in the treatment of breast cancer. Thus, agents that reduce HER3 expression at the plasma membrane may synergize with current therapies and offer a novel therapeutic strategy to improve treatment. METHODS: We devised an image-based screening platform using membrane localized HER3-YFP to identify small molecules that promote HER3 internalization and degradation. In vitro and in vivo tumor models were used to characterize the signaling effects of perhexiline, an anti-anginal drug, identified by the screening platform. RESULTS: We found perhexiline, an anti-anginal drug, selectively internalized HER3, decreased HER3 expression, and subsequently inhibited signaling downstream of HER3. Consistent with these results, perhexiline inhibited breast cancer cell proliferation in vitro and tumor growth in vivo. CONCLUSIONS: This is the first demonstration that HER3 can be targeted with small molecules by eliminating it from the cell membrane. The novel approach used here led to the discovery that perhexiline ablates HER3 expression, and offers an opportunity to identify HER3 ablation modulators as innovative therapeutics to improve survival in breast cancer patients.


Subject(s)
Antineoplastic Agents/pharmacology , Breast Neoplasms/metabolism , Perhexiline/pharmacology , Receptor, ErbB-3/metabolism , Animals , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Membrane/metabolism , Cell Proliferation/drug effects , Disease Models, Animal , Female , Humans , Mice , Neuregulins/metabolism , Neuregulins/pharmacology , Protein Transport/drug effects , Proteolysis/drug effects , Receptor, ErbB-3/genetics , Signal Transduction/drug effects , Tumor Burden/drug effects , Ubiquitination/drug effects , Xenograft Model Antitumor Assays
13.
J Cell Sci ; 128(6): 1230-40, 2015 Mar 15.
Article in English | MEDLINE | ID: mdl-25653388

ABSTRACT

Embryonic development and adult tissue homeostasis require precise information exchange between cells and their microenvironment to coordinate cell behavior. A specialized class of ultra-long actin-rich filopodia, termed cytonemes, provides one mechanism for this spatiotemporal regulation of extracellular cues. We provide here a mechanism whereby the stem-cell marker Lgr5, and its family member Lgr4, promote the formation of cytonemes. Lgr4- and Lgr5-induced cytonemes exceed lengths of 80 µm, are generated through stabilization of nascent filopodia from an underlying lamellipodial-like network and functionally provide a pipeline for the transit of signaling effectors. As proof-of-principle, we demonstrate that Lgr5-induced cytonemes act as conduits for cell signaling by demonstrating that the actin motor and filopodial cargo carrier protein myosin X (Myo10) and the G-protein-coupled receptor (GPCR) signaling effector ß-arrestin-2 (Arrb2) transit into cytonemes. This work delineates a biological function for Lgr4 and Lgr5 and provides the rationale to fully investigate Lgr4 and Lgr5 function and cytonemes in mammalian stem cell and cancer stem cell behavior.


Subject(s)
Actins/metabolism , Cell Membrane/metabolism , Cell Surface Extensions/metabolism , Receptors, G-Protein-Coupled/metabolism , Stem Cells/metabolism , Adult , Arrestins/metabolism , Biological Transport , Blotting, Western , HEK293 Cells , Humans , Immunoprecipitation , Pseudopodia/physiology , Signal Transduction , Stem Cells/cytology , beta-Arrestin 2 , beta-Arrestins
14.
J Biol Chem ; 289(48): 33442-55, 2014 Nov 28.
Article in English | MEDLINE | ID: mdl-25261469

ABSTRACT

The G protein-coupled ghrelin receptor GHSR1a is a potential pharmacological target for treating obesity and addiction because of the critical role ghrelin plays in energy homeostasis and dopamine-dependent reward. GHSR1a enhances growth hormone release, appetite, and dopamine signaling through G(q/11), G(i/o), and G(12/13) as well as ß-arrestin-based scaffolds. However, the contribution of individual G protein and ß-arrestin pathways to the diverse physiological responses mediated by ghrelin remains unknown. To characterize whether a signaling bias occurs for GHSR1a, we investigated ghrelin signaling in a number of cell-based assays, including Ca(2+) mobilization, serum response factor response element, stress fiber formation, ERK1/2 phosphorylation, and ß-arrestin translocation, utilizing intracellular second loop and C-tail mutants of GHSR1a. We observed that GHSR1a and ß-arrestin rapidly form metastable plasma membrane complexes following exposure to an agonist, but replacement of the GHSR1a C-tail by the tail of the vasopressin 2 receptor greatly stabilizes them, producing complexes observable on the plasma membrane and also in endocytic vesicles. Mutations of the contiguous conserved amino acids Pro-148 and Leu-149 in the GHSR1a intracellular second loop generate receptors with a strong bias to G protein and ß-arrestin, respectively, supporting a role for conformation-dependent signaling bias in the wild-type receptor. Our results demonstrate more balance in GHSR1a-mediated ERK signaling from G proteins and ß-arrestin but uncover an important role for ß-arrestin in RhoA activation and stress fiber formation. These findings suggest an avenue for modulating drug abuse-associated changes in synaptic plasticity via GHSR1a and indicate the development of GHSR1a-biased ligands as a promising strategy for selectively targeting downstream signaling events.


Subject(s)
Arrestin/metabolism , GTP-Binding Proteins/metabolism , MAP Kinase Signaling System/physiology , Receptors, Ghrelin/metabolism , Arrestin/genetics , GTP-Binding Proteins/genetics , HEK293 Cells , Humans , Mitogen-Activated Protein Kinase 1/genetics , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/genetics , Mitogen-Activated Protein Kinase 3/metabolism , Neuronal Plasticity/physiology , Protein Stability , Protein Structure, Secondary , Protein Transport/physiology , Receptors, Ghrelin/genetics , Receptors, Vasopressin/genetics , Receptors, Vasopressin/metabolism
15.
Proc Natl Acad Sci U S A ; 111(35): E3641-9, 2014 Sep 02.
Article in English | MEDLINE | ID: mdl-25136113

ABSTRACT

The pseudostratified airway epithelium of the lung contains a balanced proportion of multiciliated and secretory luminal cells that are maintained and regenerated by a population of basal stem cells. However, little is known about how these processes are modulated in vivo, and about the potential role of cytokine signaling between stem and progenitor cells and their niche. Using a clonal 3D organoid assay, we found that IL-6 stimulated, and Stat3 inhibitors reduced, the generation of ciliated vs. secretory cells from basal cells. Gain-of-function and loss-of-function studies with cultured mouse and human basal cells suggest that IL-6/Stat3 signaling promotes ciliogenesis at multiple levels, including increases in multicilin gene and forkhead box protein J1 expression and inhibition of the Notch pathway. To test the role of IL-6 in vivo genetically, we followed the regeneration of mouse tracheal epithelium after ablation of luminal cells by inhaled SO2. Stat3 is activated in basal cells and their daughters early in the repair process, correlating with an increase in Il-6 expression in platelet-derived growth factor receptor alpha(+) mesenchymal cells in the stroma. Conditional deletion in basal cells of suppressor of cytokine signaling 3, encoding a negative regulator of the Stat3 pathway, results in an increase in multiciliated cells at the expense of secretory and basal cells. By contrast, Il-6 null mice regenerate fewer ciliated cells and an increased number of secretory cells after injury. The results support a model in which IL-6, produced in the reparative niche, functions to enhance the differentiation of basal cells, and thereby acts as a "friend" to promote airway repair rather than a "foe."


Subject(s)
Interleukin-6/metabolism , Respiratory Mucosa/cytology , STAT3 Transcription Factor/metabolism , Animals , Bronchi/cytology , Cell Differentiation/physiology , Cilia/physiology , Disease Models, Animal , Epithelial Cells/cytology , Epithelial Cells/physiology , Green Fluorescent Proteins/genetics , Humans , Interleukin-6/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Phosphorylation/physiology , Primary Cell Culture , Regeneration/physiology , Respiratory Mucosa/physiology , STAT3 Transcription Factor/genetics , Signal Transduction/physiology , Stem Cells/cytology , Stem Cells/physiology , Trachea/cytology
16.
Biochemistry ; 52(52): 9456-69, 2013 Dec 31.
Article in English | MEDLINE | ID: mdl-24274581

ABSTRACT

GPR55 is a class A G protein-coupled receptor (GPCR) that has been implicated in inflammatory pain, neuropathic pain, metabolic disorder, bone development, and cancer. Initially deorphanized as a cannabinoid receptor, GPR55 has been shown to be activated by non-cannabinoid ligands such as l-α-lysophosphatidylinositol (LPI). While there is a growing body of evidence of physiological and pathophysiological roles for GPR55, the paucity of specific antagonists has limited its study. In collaboration with the Molecular Libraries Probe Production Centers Network initiative, we identified a series of GPR55 antagonists using a ß-arrestin, high-throughput, high-content screen of ~300000 compounds. This screen yielded novel, GPR55 antagonist chemotypes with IC50 values in the range of 0.16-2.72 µM [Heynen-Genel, S., et al. (2010) Screening for Selective Ligands for GPR55: Antagonists (ML191, ML192, ML193) (Bookshelf ID NBK66153; PMID entry 22091481)]. Importantly, many of the GPR55 antagonists were completely selective, with no agonism or antagonism against GPR35, CB1, or CB2 up to 20 µM. Using a model of the GPR55 inactive state, we studied the binding of an antagonist series that emerged from this screen. These studies suggest that GPR55 antagonists possess a head region that occupies a horizontal binding pocket extending into the extracellular loop region, a central ligand portion that fits vertically in the receptor binding pocket and terminates with a pendant aromatic or heterocyclic ring that juts out. Both the region that extends extracellularly and the pendant ring are features associated with antagonism. Taken together, our results provide a set of design rules for the development of second-generation GPR55 selective antagonists.


Subject(s)
Drug Evaluation, Preclinical , Receptors, G-Protein-Coupled/antagonists & inhibitors , Receptors, G-Protein-Coupled/chemistry , Binding Sites , Humans , Inhibitory Concentration 50 , Ligands , Models, Molecular , Protein Binding , Receptors, Cannabinoid , Receptors, G-Protein-Coupled/metabolism
17.
Biochemistry ; 52(32): 5403-14, 2013 Aug 13.
Article in English | MEDLINE | ID: mdl-23865508

ABSTRACT

ß-Arrestins regulate G protein-coupled receptor signaling as competitive inhibitors and protein adaptors. Low molecular weight biased ligands that bind receptors and discriminate between the G protein dependent arm and ß-arrestin, clathrin-associated arm of receptor signaling are considered therapeutically valuable as a result of this distinctive pharmacological behavior. Other than receptor agonists, compounds that activate ß-arrestins are not available. We show that within minutes of exposure to the cationic triphenylmethane dyes malachite green and brilliant green, tissue culture cells recruit ß-arrestins to clathrin scaffolds in a receptor-activation independent manner. In the presence of these compounds, G protein signaling is inhibited, ERK and GSK3ß signaling are preserved, and the recruitment of the beta2-adaptin, AP2 adaptor complex to clathrin as well as transferrin internalization is reduced. Moreover, malachite green binds ß-arrestin2-GFP coated immunotrap beads relative to GFP only coated beads. Triphenylmethane dyes are FDA approved for topical use on newborns as components of triple-dye preparations and are not approved but used effectively as aqueous antibiotics in fish husbandry. As possible carcinogens, their chronic ingestion in food preparations, particularly through farmed fish, is discouraged in the U.S. and Europe. Our results indicate triphenylmethane dyes as a result of novel pharmacology may have additional roles as ß-arrestin/clathrin pathway signaling modulators in both pharmacology research and clinical therapy.


Subject(s)
Arrestins/metabolism , Quaternary Ammonium Compounds/metabolism , Rosaniline Dyes/metabolism , Cell Line , Cell Line, Tumor , Coloring Agents , Endocytosis , GTP-Binding Proteins/metabolism , HEK293 Cells , Humans , Quaternary Ammonium Compounds/chemistry , Receptors, Neurotensin/chemistry , Receptors, Neurotensin/metabolism , Rosaniline Dyes/chemistry , Signal Transduction , beta-Arrestins
18.
Bioorg Med Chem Lett ; 23(7): 2187-91, 2013 Apr 01.
Article in English | MEDLINE | ID: mdl-23453073

ABSTRACT

The Wnt signal transduction pathway is dysregulated in many highly prevalent diseases, including cancer. Unfortunately, drug discovery efforts have been hampered by the paucity of targets and drug-like lead molecules amenable to drug discovery. Recently, we reported the FDA-approved anthelmintic drug Niclosamide inhibits Wnt/ß-catenin signaling by a unique mechanism, though the target responsible remains unknown. We interrogated the mechanism and structure-activity relationships to understand drivers of potency and to assist target identification efforts. We found inhibition of Wnt signaling by Niclosamide appears unique among the structurally-related anthelmintic agents tested and found the potency and functional response was dependent on small changes in the chemical structure of Niclosamide. Overall, these findings support efforts to identify the target of Niclosamide inhibition of Wnt/ß-catenin signaling and the discovery of potent and selective modulators to treat human disease.


Subject(s)
Niclosamide/pharmacology , Small Molecule Libraries/pharmacology , Wnt Signaling Pathway/drug effects , beta Catenin/antagonists & inhibitors , Cell Line, Tumor , Dose-Response Relationship, Drug , HEK293 Cells , Humans , Molecular Structure , Niclosamide/chemistry , Small Molecule Libraries/chemistry , Structure-Activity Relationship , beta Catenin/metabolism
19.
Gastroenterology ; 144(4): 771-80, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23313268

ABSTRACT

BACKGROUND & AIMS: Glucose is absorbed into intestine cells via the sodium glucose transporter 1 (SGLT-1) and glucose transporter 2 (GLUT2); various peptides and hormones control this process. Apelin is a peptide that regulates glucose homeostasis and is produced by proximal digestive cells; we studied whether glucose modulates apelin secretion by enterocytes and the effects of apelin on intestinal glucose absorption. METHODS: We characterized glucose-related luminal apelin secretion in vivo and ex vivo by mass spectroscopy and immunologic techniques. The effects of apelin on (14)C-labeled glucose transport were determined in jejunal loops and in mice following apelin gavage. We determined levels of GLUT2 and SGLT-1 proteins and phosphorylation of AMPKα2 by immunoblotting. The net effect of apelin on intestinal glucose transepithelial transport was determined in mice. RESULTS: Glucose stimulated luminal secretion of the pyroglutaminated apelin-13 isoform ([Pyr-1]-apelin-13) in the small intestine of mice. Apelin increased specific glucose flux through the gastric epithelial barrier in jejunal loops and in vivo following oral glucose administration. Conversely, pharmacologic apelin blockade in the intestine reduced the increased glycemia that occurs following oral glucose administration. Apelin activity was associated with phosphorylation of AMPKα2 and a rapid increase of the GLUT2/SGLT-1 protein ratio in the brush border membrane. CONCLUSIONS: Glucose amplifies its own transport from the intestinal lumen to the bloodstream by increasing luminal apelin secretion. In the lumen, active apelin regulates carbohydrate flux through enterocytes by promoting AMPKα2 phosphorylation and modifying the ratio of SGLT-1:GLUT2. The glucose-apelin cycle might be pharmacologically handled to regulate glucose absorption and assess better control of glucose homeostasis.


Subject(s)
Carbohydrates/pharmacokinetics , Glucose/metabolism , Intercellular Signaling Peptides and Proteins/metabolism , Intestinal Absorption/drug effects , Intestinal Absorption/physiology , Analysis of Variance , Animals , Biological Transport/drug effects , Biological Transport/physiology , Blotting, Western , Chromatography, Liquid/methods , Disease Models, Animal , Glucose/pharmacology , Glucose Transporter Type 2/metabolism , Immunohistochemistry , Male , Mass Spectrometry , Mice , Mice, Inbred C57BL , Microscopy, Confocal , Random Allocation , Reference Values , Sodium-Glucose Transporter 1/metabolism
20.
PLoS One ; 8(12): e84476, 2013.
Article in English | MEDLINE | ID: mdl-24386388

ABSTRACT

Lgr5 is a membrane protein related to G protein-coupled receptors (GPCR)s whose expression identifies stem cells in multiple tissues and is strongly correlated with cancer. Despite the recent identification of endogenous ligands for Lgr5, its mode of signaling remains enigmatic. The ability to couple to G proteins and ßarrestins are classical molecular behaviors of GPCRs that have yet to be observed for Lgr5. Therefore, the goal of this study was to determine if Lgr5 can engage a classical GPCR behavior and elucidate the molecular determinants of this process. Structural analysis of Lgr5 revealed several motifs consistent with its ability to recruit ßarr2. Among them, a "SSS" serine cluster located at amino acid position 873-875 within the C-terminal tail (C-tail), is in a region consistent with other GPCRs that bind ßarr2 with high-affinity. To test its functionality, a ligand-independent ßarr2 translocation assay was implemented. We show that Lgr5 recruits ßarr2 and that the "SSS" amino acids (873-875) are absolutely critical to this process. We also demonstrate that for full efficacy, this cluster requires other Lgr5 C-tail serines that were previously shown to be important for constitutive and ßarr2 independent internalization of Lgr5. These data are proof of principle that a classical GPCR behavior can be manifested by Lgr5. The existence of alternative ligands or missing effectors of Lgr5 that scaffold this classical GPCR behavior and the downstream signaling pathways engaged should be considered. Characterizing Lgr5 signaling will be invaluable for assessing its role in tissue maintenance, repair, and disease.


Subject(s)
Arrestins/metabolism , Receptors, G-Protein-Coupled/metabolism , Signal Transduction/physiology , Amino Acid Motifs , Arrestins/genetics , HEK293 Cells , Humans , Protein Structure, Tertiary , Receptors, G-Protein-Coupled/genetics , beta-Arrestin 2 , beta-Arrestins
SELECTION OF CITATIONS
SEARCH DETAIL
...