1
|
Cruz A, Warshel A. Unraveling GPCRs Allosteric Modulation. Cannabinoid 1 Receptor as a Case Study. Proteins 2025; 93:763-785. [PMID: 39584635 PMCID: PMC11879764 DOI: 10.1002/prot.26762] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Revised: 10/14/2024] [Accepted: 10/18/2024] [Indexed: 11/26/2024]
Abstract
G-protein-coupled receptors (GPCRs) constitute one of the most prominent families of integral membrane receptor proteins that mediate most transmembrane signaling processes. Malfunction of these signal transduction processes is one of the underlying causes of many human pathologies (Parkinson's, Huntington's, heart diseases, etc), provoking that GPCRs are the largest family of druggable proteins. However, these receptors have been targeted traditionally by orthosteric ligands, which usually causes side effects due to the simultaneous targeting of homologous receptor subtypes. Allosteric modulation offers a promising alternative approach to circumvent this problematic and, thus, comprehending its details is a most important task. Here we use the Cannabinoid type-1 receptor (CB1R) in trying to shed light on this issue, focusing on positive allosteric modulation. This is done by using the protein-dipole Langevin-dipole (PDLD) within the linear response approximation (LRA) framework (PDLD/S-2000) along with our coarse-grained (CG) model of membrane proteins to evaluate the dissociation constants (K Bs) and cooperativity factors (αs) for a diverse series of CB1R positive allosteric modulators belonging to the 2-phenylindole structural class, considering CP55940 as an agonist. The agreement with the experimental data evinces that significantly populated allosteric modulator:CB1R and allosteric modulator:CP55940:CB1R complexes have been identified and characterized successfully. Analyzing them, it has been determined that CB1R positive allosteric modulation lies in an outwards displacement of transmembrane α helix (TM) 4 extracellular end and in the regulation of the range of motion of a compound TM7 movement for binary and ternary complexes, respectively. In this respect, we achieved a better comprehension of the molecular architecture of CB1R positive allosteric site, identifying Lys1923.28 and Gly1943.30 as key residues regarding electrostatic interactions inside this cavity, and to rationalize (at both structural and molecular level) the exhibited stereoselectivity in relation to positive allosteric modulation activity by considered CB1R allosteric modulators. Additionally, putative/postulated allosteric binding sites have been screened successfully, identifying the real CB1R positive allosteric site, and most structure-activity relationship (SAR) studies of CB1R 2-phenylindole allosteric modulators have been rationalized. All these findings point out towards the predictive value of the methodology used in the current work, which can be applied to other biophysical systems of interest. The results presented in this study contribute significantly to understand GPCRs allosteric modulation and, hopefully, will encourage a more thorough exploration of the topic.
Collapse
Affiliation(s)
- Alejandro Cruz
- Department of Chemistry, University of Southern California, Los Angeles, California, 90089-1062, United States
| | - Arieh Warshel
- Department of Chemistry, University of Southern California, Los Angeles, California, 90089-1062, United States
| |
Collapse
|
2
|
Armstrong Suthahar SS, Nettersheim FS, Alimadadi A, Wang E, Billitti M, Resto-Trujillo N, Roy P, Hedrick CC, Ley K, Orecchioni M. Olfr2-positive macrophages originate from monocytes proliferate in situ and present a pro-inflammatory foamy-like phenotype. Cardiovasc Res 2024; 120:1577-1589. [PMID: 39229899 DOI: 10.1093/cvr/cvae153] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Revised: 04/25/2024] [Accepted: 06/13/2024] [Indexed: 09/05/2024] Open
Abstract
AIMS Olfactory receptor 2 (Olfr2) has been identified in a minimum of 30% of vascular macrophages, and its depletion was shown to reduce atherosclerosis progression. Mononuclear phagocytes, including monocytes and macrophages within the vessel wall, are major players in atherosclerosis. Single-cell RNA sequencing studies revealed that atherosclerotic artery walls encompass several monocytes and vascular macrophages, defining at least nine distinct subsets potentially serving diverse functions in disease progression. This study investigates the functional phenotype and ontogeny of Olfr2-expressing vascular macrophages in atherosclerosis. METHODS AND RESULTS Olfr2+ macrophages rapidly increase in Apoe-/- mice's aorta when fed a Western diet (WD). Mass cytometry showed that Olfr2+ cells are clustered within the CD64 high population and enriched for CD11c and Ccr2 markers. Olfr2+ macrophages express many pro-inflammatory cytokines, including Il1b, Il6, Il12, and Il23, and chemokines, including Ccl5, Cx3cl1, Cxcl9, and Ccl22. By extracting differentially expressed genes from bulk RNA sequencing (RNA-seq) of Olfr2+ vs. Olfr2- macrophages, we defined a signature that significantly mapped to single-cell data of plaque myeloid cells, including monocytes, subendothelial MacAir, and Trem2Gpnmb foamy macrophages. By adoptive transfer experiments, we identified that Olfr2 competent monocytes from CD45.1Apoe-/-Olfr2+/+ mice transferred into CD45.2Apoe-/-Olfr2-/- recipient mice fed WD for 12 weeks, accumulate in the atherosclerotic aorta wall already at 72 h, and differentiate in macrophages. Olfr2+ macrophages showed significantly increased BrdU incorporation compared to Olfr2- macrophages. Flow cytometry confirmed that at least 50% of aortic Olfr2+ macrophages are positive for BODIPY staining and have increased expression of both tumour necrosis factor and interleukin 6 compared to Olfr2- macrophages. Gene set enrichment analysis of the Olfr2+ macrophage signature revealed a similar enrichment pattern in human atherosclerotic plaques, particularly within foamy/TREM2hi-Mφ and monocytes. CONCLUSIONS In summary, we conclude that Olfr2+ macrophages in the aorta originate from monocytes and can accumulate at the early stages of disease progression. These cells can undergo differentiation into MacAir and Trem2Gpnmb foamy macrophages, exhibiting proliferative and pro-inflammatory potentials. This dynamic behaviour positions them as key influencers in shaping the myeloid landscape within the atherosclerotic plaque.
Collapse
Affiliation(s)
| | - Felix Sebastian Nettersheim
- Division of Inflammation Biology, La Jolla Institute for Immunology, 9420 Athena Cir, La Jolla, CA 92037, USA
| | - Ahmad Alimadadi
- Division of Inflammation Biology, La Jolla Institute for Immunology, 9420 Athena Cir, La Jolla, CA 92037, USA
- Immunology Center of Georgia, Augusta University, 1410 Laney Walker Blvd, Augusta, GA 30912, USA
| | - Erpei Wang
- Division of Inflammation Biology, La Jolla Institute for Immunology, 9420 Athena Cir, La Jolla, CA 92037, USA
| | - Monica Billitti
- Division of Inflammation Biology, La Jolla Institute for Immunology, 9420 Athena Cir, La Jolla, CA 92037, USA
| | - Natalya Resto-Trujillo
- Immunology Center of Georgia, Augusta University, 1410 Laney Walker Blvd, Augusta, GA 30912, USA
| | - Payel Roy
- Division of Inflammation Biology, La Jolla Institute for Immunology, 9420 Athena Cir, La Jolla, CA 92037, USA
- Immunology Center of Georgia, Augusta University, 1410 Laney Walker Blvd, Augusta, GA 30912, USA
| | - Catherine C Hedrick
- Immunology Center of Georgia, Augusta University, 1410 Laney Walker Blvd, Augusta, GA 30912, USA
- Department of Medicine, Augusta University, 1120 15th St BA 8412, Augusta, GA 30912, USA
| | - Klaus Ley
- Immunology Center of Georgia, Augusta University, 1410 Laney Walker Blvd, Augusta, GA 30912, USA
- Department of Physiology, Augusta University, 1462 Laney Walker Blvd, Augusta, GA 30912, USA
| | - Marco Orecchioni
- Immunology Center of Georgia, Augusta University, 1410 Laney Walker Blvd, Augusta, GA 30912, USA
- Department of Pharmacology & Toxicology, Augusta University, 1459 Laney Walker Blvd, Augusta, GA 30901, USA
| |
Collapse
|
3
|
Justus CR, Marie MA, Sanderlin EJ, Yang LV. The Roles of Proton-Sensing G-Protein-Coupled Receptors in Inflammation and Cancer. Genes (Basel) 2024; 15:1151. [PMID: 39336742 PMCID: PMC11431078 DOI: 10.3390/genes15091151] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Revised: 08/27/2024] [Accepted: 08/27/2024] [Indexed: 09/30/2024] Open
Abstract
The precise regulation of pH homeostasis is crucial for normal physiology. However, in tissue microenvironments, it can be impacted by pathological conditions such as inflammation and cancer. Due to the overproduction and accumulation of acids (protons), the extracellular pH is characteristically more acidic in inflamed tissues and tumors in comparison to normal tissues. A family of proton-sensing G-protein-coupled receptors (GPCRs) has been identified as molecular sensors for cells responding to acidic tissue microenvironments. Herein, we review the current research progress pertaining to these proton-sensing GPCRs, including GPR4, GPR65 (TDAG8), and GPR68 (OGR1), in inflammation and cancer. Growing evidence suggests that GPR4 and GPR68 are mainly pro-inflammatory, whereas GPR65 is primarily anti-inflammatory, in various inflammatory disorders. Both anti- and pro-tumorigenic effects have been reported for this family of receptors. Moreover, antagonists and agonists targeting proton-sensing GPCRs have been developed and evaluated in preclinical models. Further research is warranted to better understand the roles of these proton-sensing GPCRs in pathophysiology and is required in order to exploit them as potential therapeutic targets for disease treatment.
Collapse
Affiliation(s)
- Calvin R Justus
- Department of Internal Medicine, Brody School of Medicine, East Carolina University, Greenville, NC 27834, USA
| | - Mona A Marie
- Department of Internal Medicine, Brody School of Medicine, East Carolina University, Greenville, NC 27834, USA
| | - Edward J Sanderlin
- Department of Internal Medicine, Brody School of Medicine, East Carolina University, Greenville, NC 27834, USA
| | - Li V Yang
- Department of Internal Medicine, Brody School of Medicine, East Carolina University, Greenville, NC 27834, USA
| |
Collapse
|
4
|
Gaiser BI, Danielsen M, Xu X, Røpke Jørgensen K, Fronik P, Märcher-Rørsted E, Wróbel TM, Liu X, Mosolff Mathiesen J, Sejer Pedersen D. Bitopic Ligands Support the Presence of a Metastable Binding Site at the β 2 Adrenergic Receptor. J Med Chem 2024; 67:11053-11068. [PMID: 38952152 DOI: 10.1021/acs.jmedchem.4c00578] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/03/2024]
Abstract
Metastable binding sites (MBS) have been observed in a multitude of molecular dynamics simulations and can be considered low affinity allosteric binding sites (ABS) that function as stepping stones as the ligand moves toward the orthosteric binding site (OBS). Herein, we show that MBS can be utilized as ABS in ligand design, resulting in ligands with improved binding kinetics. Four homobivalent bitopic ligands (1-4) were designed by molecular docking of (S)-alprenolol ((S)-ALP) in the cocrystal structure of the β2 adrenergic receptor (β2AR) bound to the antagonist ALP. Ligand 4 displayed a potency and affinity similar to (S)-ALP, but with a >4-fold increase in residence time. The proposed binding mode was confirmed by X-ray crystallography of ligand 4 in complex with the β2AR. This ligand design principle can find applications beyond the β2AR and G protein-coupled receptors (GPCRs) as a general approach for improving the pharmacological profile of orthosteric ligands by targeting the OBS and an MBS simultaneously.
Collapse
Affiliation(s)
- Birgit Isabel Gaiser
- Department of Drug Design and Pharmacology, University of Copenhagen, Jagtvej 162, 2100 Copenhagen, Denmark
| | - Mia Danielsen
- Department of Drug Design and Pharmacology, University of Copenhagen, Jagtvej 162, 2100 Copenhagen, Denmark
| | - Xinyu Xu
- State Key Laboratory of Membrane Biology, Tsinghua-Peking Center for Life Sciences, Beijing Frontier Research Center for Biological Structure, School of Pharmaceutical Sciences, Tsinghua University, Beijing 100084 ,China
| | - Kira Røpke Jørgensen
- Department of Drug Design and Pharmacology, University of Copenhagen, Jagtvej 162, 2100 Copenhagen, Denmark
| | - Philipp Fronik
- Department of Drug Design and Pharmacology, University of Copenhagen, Jagtvej 162, 2100 Copenhagen, Denmark
| | - Emil Märcher-Rørsted
- Department of Drug Design and Pharmacology, University of Copenhagen, Jagtvej 162, 2100 Copenhagen, Denmark
| | - Tomasz M Wróbel
- Department of Drug Design and Pharmacology, University of Copenhagen, Jagtvej 162, 2100 Copenhagen, Denmark
- Department of Synthesis and Chemical Technology of Pharmaceutical Substances, Medical University of Lublin, Chodźki 4a, 20093 Lublin, Poland
| | - Xiangyu Liu
- State Key Laboratory of Membrane Biology, Tsinghua-Peking Center for Life Sciences, Beijing Frontier Research Center for Biological Structure, School of Pharmaceutical Sciences, Tsinghua University, Beijing 100084 ,China
| | - Jesper Mosolff Mathiesen
- Department of Drug Design and Pharmacology, University of Copenhagen, Jagtvej 162, 2100 Copenhagen, Denmark
| | - Daniel Sejer Pedersen
- Department of Drug Design and Pharmacology, University of Copenhagen, Jagtvej 162, 2100 Copenhagen, Denmark
| |
Collapse
|
5
|
Abrams KL, Ward DA, Sabiniewicz A, Hummel T. Olfaction evaluation in dogs with sudden acquired retinal degeneration syndrome. Vet Ophthalmol 2024; 27:127-138. [PMID: 37399129 DOI: 10.1111/vop.13121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Revised: 06/05/2023] [Accepted: 06/09/2023] [Indexed: 07/05/2023]
Abstract
PURPOSE To evaluate olfaction in dogs with sudden acquired retinal degeneration syndrome (SARDS) compared with sighted dogs and blind dogs without SARDS as control groups. ANIMALS STUDIED Forty client-owned dogs. PROCEDURE Olfactory threshold testing was performed on three groups: SARDS, sighted, and blind/non-SARDS using eugenol as the test odorant. The olfactory threshold was determined when subjects indicated the detection of a specific eugenol concentration with behavioral responses. Olfactory threshold, age, body weight, and environmental room factors were evaluated. RESULTS Sixteen dogs with SARDS, 12 sighted dogs, and 12 blind/non-SARDS dogs demonstrated mean olfactory threshold pen numbers of 2.8 (SD = 1.4), 13.8 (SD = 1.4), and 13.4 (SD = 1.1), respectively, which correspond to actual mean concentrations of 0.017 g/mL, 1.7 × 10-13 g/mL and 4.26 × 10-13 g/mL, respectively. Dogs with SARDS had significantly poorer olfactory threshold scores compared with the two control groups (p < .001), with no difference between the control groups (p = .5). Age, weight, and room environment did not differ between the three groups. CONCLUSIONS Dogs with SARDS have severely decreased olfaction capabilities compared with sighted dogs and blind/non-SARDS dogs. This finding supports the suspicion that SARDS is a systemic disease causing blindness, endocrinopathy, and hyposmia. Since the molecular pathways are similar in photoreceptors, olfactory receptors, and steroidogenesis with all using G-protein coupled receptors in the cell membrane, the cause of SARDS may exist at the G-protein associated interactions with intracellular cyclic nucleotides. Further investigations into G-protein coupled receptors pathway and canine olfactory receptor genes in SARDS patients may be valuable in revealing the cause of SARDS.
Collapse
Affiliation(s)
- Kenneth L Abrams
- Veterinary Ophthalmology Services, North Kingstown, Rhode Island, USA
| | - Daniel A Ward
- Department of Small Animal Clinical Sciences, College of Veterinary Medicine, University of Tennessee, Knoxville, Tennessee, USA
| | - Agnieszka Sabiniewicz
- Department of Otorhinolaryngology, Smell and Taste Clinic, Technische Universität Dresden, Dresden, Germany
- Institute of Psychology, University of Wrocław, Wrocław, Poland
| | - Thomas Hummel
- Department of Otorhinolaryngology, Smell and Taste Clinic, Technische Universität Dresden, Dresden, Germany
| |
Collapse
|
6
|
Gu X, Liu J, Yu Y, Xiao P, Ding Y. MFD-GDrug: multimodal feature fusion-based deep learning for GPCR-drug interaction prediction. Methods 2024; 223:75-82. [PMID: 38286333 DOI: 10.1016/j.ymeth.2024.01.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2023] [Revised: 01/14/2024] [Accepted: 01/26/2024] [Indexed: 01/31/2024] Open
Abstract
The accurate identification of drug-protein interactions (DPIs) is crucial in drug development, especially concerning G protein-coupled receptors (GPCRs), which are vital targets in drug discovery. However, experimental validation of GPCR-drug pairings is costly, prompting the need for accurate predictive methods. To address this, we propose MFD-GDrug, a multimodal deep learning model. Leveraging the ESM pretrained model, we extract protein features and employ a CNN for protein feature representation. For drugs, we integrated multimodal features of drug molecular structures, including three-dimensional features derived from Mol2vec and the topological information of drug graph structures extracted through Graph Convolutional Neural Networks (GCN). By combining structural characterizations and pretrained embeddings, our model effectively captures GPCR-drug interactions. Our tests on leading GPCR-drug interaction datasets show that MFD-GDrug outperforms other methods, demonstrating superior predictive accuracy.
Collapse
Affiliation(s)
- Xingyue Gu
- State Key Laboratory of Digital Medical Engineering, School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, China
| | - Junkai Liu
- School of Electronic and Information Engineering, Suzhou University of Science and Technology, Suzhou 215009, China
| | - Yue Yu
- School of Computer and Artificial Intelligence, Zhengzhou University, Zhengzhou 450001, China
| | - Pengfeng Xiao
- State Key Laboratory of Digital Medical Engineering, School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, China.
| | - Yijie Ding
- Yangtze Delta Region Institute (Quzhou), University of Electronic Science and Technology of China, Quzhou, Zhejiang 324003, China; Institute of Fundamental and Frontier Sciences, University of Electronic Science and Technology of China, Chengdu 611730, China.
| |
Collapse
|
7
|
Yahiro I, Barnuevo KDE, Sato O, Mohapatra S, Toyoda A, Itoh T, Ohno K, Matsuyama M, Chakraborty T, Ohta K. Modeling the SDF-1/CXCR4 protein using advanced artificial intelligence and antagonist screening for Japanese anchovy. Front Physiol 2024; 15:1349119. [PMID: 38370015 PMCID: PMC10869568 DOI: 10.3389/fphys.2024.1349119] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Accepted: 01/16/2024] [Indexed: 02/20/2024] Open
Abstract
SDF-1/CXCR4 chemokine signaling are indispensable for cell migration, especially the Primordial Germ Cell (PGC) migration towards the gonadal ridge during early development. We earlier found that this signaling is largely conserved in the Japanese anchovy (Engraulis japonicus, EJ), and a mere treatment of CXCR4 antagonist, AMD3100, leads to germ cell depletion and thereafter gonad sterilization. However, the effect of AMD3100 was limited. So, in this research, we scouted for CXCR4 antagonist with higher potency by employing advanced artificial intelligence deep learning-based computer simulations. Three potential candidates, AMD3465, WZ811, and LY2510924, were selected and in vivo validation was conducted using Japanese anchovy embryos. We found that seven transmembrane motif of EJ CXCR4a and EJ CXCR4b were extremely similar with human homolog while the CXCR4 chemokine receptor N terminal (PF12109, essential for SDF-1 binding) was missing in EJ CXCR4b. 3D protein analysis and cavity search predicted the cavity in EJ CXCR4a to be five times larger (6,307 ų) than that in EJ CXCR4b (1,241 ų). Docking analysis demonstrated lower binding energy of AMD3100 and AMD3465 to EJ CXCR4a (Vina score -9.6) and EJ CXCR4b (Vina score -8.8), respectively. Furthermore, we observed significant PGC mismigration in microinjected AMD3465 treated groups at 10, 100 and 1 × 105 nM concentration in 48 h post fertilized embryos. The other three antagonists showed various degrees of PGC dispersion, but no significant effect compared to their solvent control at tested concentrations was observed. Cumulatively, our results suggests that AMD3645 might be a better candidate for abnormal PGC migration in Japanese anchovy and warrants further investigation.
Collapse
Affiliation(s)
- Issei Yahiro
- Laboratory of Marine Biology, Faculty of Agriculture, Kyushu University, Fukuoka, Japan
| | | | - Oga Sato
- Laboratory of Marine Biology, Faculty of Agriculture, Kyushu University, Fukuoka, Japan
| | - Sipra Mohapatra
- Laboratory of Marine Biology, Faculty of Agriculture, Kyushu University, Fukuoka, Japan
- Aqua-Bioresource Innovation Center, Kyushu University, Saga, Japan
| | - Atsushi Toyoda
- Advanced Genomics Center, National Institute of Genetics, Shizuoka, Japan
| | - Takehiko Itoh
- School and Graduate School of Bioscience and Biotechnology, Tokyo Institute of Technology, Kanagawa, Japan
| | - Kaoru Ohno
- National Institute for Basic Biology (NIBB), Aichi, Japan
| | | | - Tapas Chakraborty
- Laboratory of Marine Biology, Faculty of Agriculture, Kyushu University, Fukuoka, Japan
- Aqua-Bioresource Innovation Center, Kyushu University, Saga, Japan
| | - Kohei Ohta
- Laboratory of Marine Biology, Faculty of Agriculture, Kyushu University, Fukuoka, Japan
- Aqua-Bioresource Innovation Center, Kyushu University, Saga, Japan
| |
Collapse
|
8
|
Ullrich M, Brandt F, Löser R, Pietzsch J, Wodtke R. Comparative Saturation Binding Analysis of 64Cu-Labeled Somatostatin Analogues Using Cell Homogenates and Intact Cells. ACS OMEGA 2023; 8:24003-24009. [PMID: 37426243 PMCID: PMC10324063 DOI: 10.1021/acsomega.3c02755] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/21/2023] [Accepted: 06/09/2023] [Indexed: 07/11/2023]
Abstract
The development of novel ligands for G-protein-coupled receptors (GPCRs) typically entails the characterization of their binding affinity, which is often performed with radioligands in a competition or saturation binding assay format. Since GPCRs are transmembrane proteins, receptor samples for binding assays are prepared from tissue sections, cell membranes, cell homogenates, or intact cells. As part of our investigations on modulating the pharmacokinetics of radiolabeled peptides for improved theranostic targeting of neuroendocrine tumors with a high abundance of the somatostatin receptor sub-type 2 (SST2), we characterized a series of 64Cu-labeled [Tyr3]octreotate (TATE) derivatives in vitro in saturation binding assays. Herein, we report on the SST2 binding parameters measured toward intact mouse pheochromocytoma cells and corresponding cell homogenates and discuss the observed differences taking the physiology of SST2 and GPCRs in general into account. Furthermore, we point out method-specific advantages and limitations.
Collapse
Affiliation(s)
- Martin Ullrich
- Helmholtz-Zentrum
Dresden-Rossendorf, Institute of Radiopharmaceutical Cancer Research, Bautzner Landstraße 400, Dresden 01328, Germany
| | - Florian Brandt
- Helmholtz-Zentrum
Dresden-Rossendorf, Institute of Radiopharmaceutical Cancer Research, Bautzner Landstraße 400, Dresden 01328, Germany
- School
of Science, Faculty of Chemistry and Food Chemistry, Technische Universität Dresden, Mommsenstraße 4, Dresden 01069, Germany
| | - Reik Löser
- Helmholtz-Zentrum
Dresden-Rossendorf, Institute of Radiopharmaceutical Cancer Research, Bautzner Landstraße 400, Dresden 01328, Germany
- School
of Science, Faculty of Chemistry and Food Chemistry, Technische Universität Dresden, Mommsenstraße 4, Dresden 01069, Germany
| | - Jens Pietzsch
- Helmholtz-Zentrum
Dresden-Rossendorf, Institute of Radiopharmaceutical Cancer Research, Bautzner Landstraße 400, Dresden 01328, Germany
- School
of Science, Faculty of Chemistry and Food Chemistry, Technische Universität Dresden, Mommsenstraße 4, Dresden 01069, Germany
| | - Robert Wodtke
- Helmholtz-Zentrum
Dresden-Rossendorf, Institute of Radiopharmaceutical Cancer Research, Bautzner Landstraße 400, Dresden 01328, Germany
| |
Collapse
|
9
|
Armando I, Cuevas S, Fan C, Kumar M, Izzi Z, Jose PA, Konkalmatt PR. G Protein-Coupled Receptor 37L1 Modulates Epigenetic Changes in Human Renal Proximal Tubule Cells. Int J Mol Sci 2022; 23:ijms232214456. [PMID: 36430934 PMCID: PMC9698582 DOI: 10.3390/ijms232214456] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Revised: 10/19/2022] [Accepted: 10/31/2022] [Indexed: 11/23/2022] Open
Abstract
Renal luminal sodium transport is essential for physiological blood pressure control, and abnormalities in this process are strongly implicated in the pathogenesis of essential hypertension. Renal G protein-coupled receptors (GPCRs) are critical for the regulation of the reabsorption of essential nutrients, ions, and water from the glomerular filtrate. Recently, we showed that GPCR 37L1 (GPR37L1) is expressed on the apical membrane of renal proximal tubules (RPT) and regulates luminal sodium transport and blood pressure by modulating the function of the sodium proton exchanger 3 (NHE3). However, little is known about GPR37L1 intracellular signaling. Here, we show that GPR37L1 is localized to the nuclear membrane, in addition to the plasma membrane in human RPT cells. Furthermore, GPR37L1 signals via the PI3K/AKT/mTOR pathway to decrease the expression of DNA (cytosine-5)-methyltransferase 1 (DNMT1) and enhance NHE3 transcription. Overall, we demonstrate the direct role of a nuclear membrane GPCR in the regulation of renal sodium through epigenetic gene regulation.
Collapse
|
10
|
Muhl L, Mocci G, Pietilä R, Liu J, He L, Genové G, Leptidis S, Gustafsson S, Buyandelger B, Raschperger E, Hansson EM, Björkegren JL, Vanlandewijck M, Lendahl U, Betsholtz C. A single-cell transcriptomic inventory of murine smooth muscle cells. Dev Cell 2022; 57:2426-2443.e6. [DOI: 10.1016/j.devcel.2022.09.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Revised: 09/12/2022] [Accepted: 09/27/2022] [Indexed: 11/28/2022]
|
11
|
de Bartolomeis A, Vellucci L, Barone A, Manchia M, De Luca V, Iasevoli F, Correll CU. Clozapine's multiple cellular mechanisms: What do we know after more than fifty years? A systematic review and critical assessment of translational mechanisms relevant for innovative strategies in treatment-resistant schizophrenia. Pharmacol Ther 2022; 236:108236. [PMID: 35764175 DOI: 10.1016/j.pharmthera.2022.108236] [Citation(s) in RCA: 45] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2022] [Revised: 06/21/2022] [Accepted: 06/21/2022] [Indexed: 12/21/2022]
Abstract
Almost fifty years after its first introduction into clinical care, clozapine remains the only evidence-based pharmacological option for treatment-resistant schizophrenia (TRS), which affects approximately 30% of patients with schizophrenia. Despite the long-time experience with clozapine, the specific mechanism of action (MOA) responsible for its superior efficacy among antipsychotics is still elusive, both at the receptor and intracellular signaling level. This systematic review is aimed at critically assessing the role and specific relevance of clozapine's multimodal actions, dissecting those mechanisms that under a translational perspective could shed light on molecular targets worth to be considered for further innovative antipsychotic development. In vivo and in vitro preclinical findings, supported by innovative techniques and methods, together with pharmacogenomic and in vivo functional studies, point to multiple and possibly overlapping MOAs. To better explore this crucial issue, the specific affinity for 5-HT2R, D1R, α2c, and muscarinic receptors, the relatively low occupancy at dopamine D2R, the interaction with receptor dimers, as well as the potential confounder effects resulting in biased ligand action, and lastly, the role of the moiety responsible for lipophilic and alkaline features of clozapine are highlighted. Finally, the role of transcription and protein changes at the synaptic level, and the possibility that clozapine can directly impact synaptic architecture are addressed. Although clozapine's exact MOAs that contribute to its unique efficacy and some of its severe adverse effects have not been fully understood, relevant information can be gleaned from recent mechanistic understandings that may help design much needed additional therapeutic strategies for TRS.
Collapse
Affiliation(s)
- Andrea de Bartolomeis
- Section of Psychiatry, Laboratory of Translational and Molecular Psychiatry and Unit of Treatment Resistant Psychosis, Department of Neuroscience, Reproductive Science and Dentistry, University Medical School of Naples "Federico II", Naples, Italy.
| | - Licia Vellucci
- Section of Psychiatry, Laboratory of Translational and Molecular Psychiatry and Unit of Treatment Resistant Psychosis, Department of Neuroscience, Reproductive Science and Dentistry, University Medical School of Naples "Federico II", Naples, Italy
| | - Annarita Barone
- Section of Psychiatry, Laboratory of Translational and Molecular Psychiatry and Unit of Treatment Resistant Psychosis, Department of Neuroscience, Reproductive Science and Dentistry, University Medical School of Naples "Federico II", Naples, Italy
| | - Mirko Manchia
- Section of Psychiatry, Department of Medical Sciences and Public Health, University of Cagliari, Cagliari, Italy; Department of Pharmacology, Dalhousie University, Halifax, Nova Scotia, Canada
| | | | - Felice Iasevoli
- Section of Psychiatry, Laboratory of Translational and Molecular Psychiatry and Unit of Treatment Resistant Psychosis, Department of Neuroscience, Reproductive Science and Dentistry, University Medical School of Naples "Federico II", Naples, Italy
| | - Christoph U Correll
- The Zucker Hillside Hospital, Department of Psychiatry, Northwell Health, Glen Oaks, NY, USA; Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Department of Psychiatry and Molecular Medicine, Hempstead, NY, USA; Charité Universitätsmedizin Berlin, Department of Child and Adolescent Psychiatry, Berlin, Germany
| |
Collapse
|
12
|
Bandzerewicz A, Gadomska-Gajadhur A. Into the Tissues: Extracellular Matrix and Its Artificial Substitutes: Cell Signalling Mechanisms. Cells 2022; 11:914. [PMID: 35269536 PMCID: PMC8909573 DOI: 10.3390/cells11050914] [Citation(s) in RCA: 50] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Revised: 03/02/2022] [Accepted: 03/04/2022] [Indexed: 02/06/2023] Open
Abstract
The existence of orderly structures, such as tissues and organs is made possible by cell adhesion, i.e., the process by which cells attach to neighbouring cells and a supporting substance in the form of the extracellular matrix. The extracellular matrix is a three-dimensional structure composed of collagens, elastin, and various proteoglycans and glycoproteins. It is a storehouse for multiple signalling factors. Cells are informed of their correct connection to the matrix via receptors. Tissue disruption often prevents the natural reconstitution of the matrix. The use of appropriate implants is then required. This review is a compilation of crucial information on the structural and functional features of the extracellular matrix and the complex mechanisms of cell-cell connectivity. The possibilities of regenerating damaged tissues using an artificial matrix substitute are described, detailing the host response to the implant. An important issue is the surface properties of such an implant and the possibilities of their modification.
Collapse
|
13
|
Abstract
Membrane proteins, particularly those that are α-helical, such as transporters and G-protein-coupled receptors (GPCRs), have significant biological relevance. However, their expression and purification pose difficulties because of their poor water solubilities, which impedes progress in this field. The QTY method, a code-based protein-engineering approach, was recently developed to produce soluble transmembrane proteins. Here, we describe a comprehensive Web server built for QTY design and its relevance for in silico analyses. Typically, the simple design model is expected to require only 2 to 4 min of computer time, and the library design model requires 2 to 5 h, depending on the target protein size and the number of transmembrane helices. Detailed protocols for using the server with both the simple design and library design modules are provided. Methods for experiments following the QTY design are also included to facilitate the implementation of this approach. The design pipeline was further evaluated using microbial transmembrane proteins and structural alignment between the designed proteins and their origins by employing AlphaFold2. The results reveal that mutants generated by the developed pipeline were highly identical to their origins in terms of three-dimensional (3D) structures. In summary, the utilization of our Web server and associated protocols will enable QTY-based protein engineering to be implemented in a convenient, fast, accurate, and rational manner. The Protein Solubilizing Server (PSS) is publicly available at http://pss.sjtu.edu.cn. IMPORTANCE Water-soluble expression and purification are of considerable importance for protein identification and characterization. However, there has been a lack of an effective method for water-soluble expression of membrane proteins, which has severely hampered their studies. Here, an enabling comprehensive Web server, PSS, was developed for designing water-soluble mutants of α-helical membrane proteins, based on QTY design, a code-based protein-engineering approach. With microbial transmembrane proteins and GPCRs as examples, we systematically evaluated the server and demonstrated its successful performance. PSS is readily available for worldwide users as a Web-based tool, rendering QTY-based protein engineering convenient, efficient, accurate, and rational.
Collapse
|
14
|
Roy N, Parhar I. Habenula orphan G-protein coupled receptors in the pathophysiology of fear and anxiety. Neurosci Biobehav Rev 2021; 132:870-883. [PMID: 34801259 DOI: 10.1016/j.neubiorev.2021.11.008] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Revised: 11/02/2021] [Accepted: 11/08/2021] [Indexed: 10/19/2022]
Abstract
The phasic emotion, fear, and the tonic emotion, anxiety, have been conventionally inspected in clinical frameworks to epitomize memory acquisition, storage, and retrieval. However, inappropriate expression of learned fear in a safe environment and its resistance to suppression is a cardinal feature of various fear-related disorders. A significant body of literature suggests the involvement of extra-amygdala circuitry in fear disorders. Consistent with this view, the present review underlies incentives for the association between the habenula and fear memory. G protein-coupled receptors (GPCRs) are important to understand the molecular mechanisms central to fear learning due to their neuromodulatory role. The efficacy of a pharmacological strategy aimed at exploiting habenular-GPCR desensitization machinery can serve as a therapeutic target combating the pathophysiology of fear disorders. Originating from this milieu, the conserved nature of orphan GPCRs in the brain, with some having the highest expression in the habenula can lead to recent endeavors in understanding its functionality in fear circuitry.
Collapse
Affiliation(s)
- Nisa Roy
- Brain Research Institute, Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Bandar Sunway, Selangor, Malaysia.
| | - Ishwar Parhar
- Brain Research Institute, Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Bandar Sunway, Selangor, Malaysia.
| |
Collapse
|
15
|
Ramasamy M, Damaj MB, Vargas-Bautista C, Mora V, Liu J, Padilla CS, Irigoyen S, Saini T, Sahoo N, DaSilva JA, Mandadi KK. A Sugarcane G-Protein-Coupled Receptor, ShGPCR1, Confers Tolerance to Multiple Abiotic Stresses. FRONTIERS IN PLANT SCIENCE 2021; 12:745891. [PMID: 35295863 PMCID: PMC8919185 DOI: 10.3389/fpls.2021.745891] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Accepted: 10/14/2021] [Indexed: 06/14/2023]
Abstract
Sugarcane (Saccharum spp.) is a prominent source of sugar and serves as bioenergy/biomass feedstock globally. Multiple biotic and abiotic stresses, including drought, salinity, and cold, adversely affect sugarcane yield. G-protein-coupled receptors (GPCRs) are components of G-protein-mediated signaling affecting plant growth, development, and stress responses. Here, we identified a GPCR-like protein (ShGPCR1) from sugarcane and energy cane (Saccharum spp. hybrids) and characterized its function in conferring tolerance to multiple abiotic stresses. ShGPCR1 protein sequence contained nine predicted transmembrane (TM) domains connected by four extracellular and four intracellular loops, which could interact with various ligands and heterotrimeric G proteins in the cells. ShGPCR1 sequence displayed other signature features of a GPCR, such as a putative guanidine triphosphate (GTP)-binding domain, as well as multiple myristoylation and protein phosphorylation sites, presumably important for its biochemical function. Expression of ShGPCR1 was upregulated by drought, salinity, and cold stresses. Subcellular imaging and calcium (Ca2+) measurements revealed that ShGPCR1 predominantly localized to the plasma membrane and enhanced intracellular Ca2+ levels in response to GTP, respectively. Furthermore, constitutive overexpression of ShGPCR1 in sugarcane conferred tolerance to the three stressors. The stress-tolerance phenotype of the transgenic lines corresponded with activation of multiple drought-, salinity-, and cold-stress marker genes, such as Saccharum spp. LATE EMBRYOGENESIS ABUNDANT, DEHYDRIN, DROUGHT RESPONSIVE 4, GALACTINOL SYNTHASE, ETHYLENE RESPONSIVE FACTOR 3, SALT OVERLY SENSITIVE 1, VACUOLAR Na+/H+ ANTIPORTER 1, NAM/ATAF1/2/CUC2, COLD RESPONSIVE FACTOR 2, and ALCOHOL DEHYDROGENASE 3. We suggest that ShGPCR1 plays a key role in conferring tolerance to multiple abiotic stresses, and the engineered lines may be useful to enhance sugarcane production in marginal environments with fewer resources.
Collapse
Affiliation(s)
- Manikandan Ramasamy
- Texas A&M AgriLife Research and Extension Center, Weslaco, TX, United States
| | - Mona B. Damaj
- Texas A&M AgriLife Research and Extension Center, Weslaco, TX, United States
| | | | - Victoria Mora
- Texas A&M AgriLife Research and Extension Center, Weslaco, TX, United States
| | - Jiaxing Liu
- Texas A&M AgriLife Research and Extension Center, Weslaco, TX, United States
| | - Carmen S. Padilla
- Texas A&M AgriLife Research and Extension Center, Weslaco, TX, United States
| | - Sonia Irigoyen
- Texas A&M AgriLife Research and Extension Center, Weslaco, TX, United States
| | - Tripti Saini
- Department of Biology, University of Texas Rio Grande Valley, Edinburg, TX, United States
| | - Nirakar Sahoo
- Department of Biology, University of Texas Rio Grande Valley, Edinburg, TX, United States
| | - Jorge A. DaSilva
- Texas A&M AgriLife Research and Extension Center, Weslaco, TX, United States
- Department of Soil and Crop Sciences, Texas A&M University, College Station, TX, United States
| | - Kranthi K. Mandadi
- Texas A&M AgriLife Research and Extension Center, Weslaco, TX, United States
- Department of Plant Pathology and Microbiology, Texas A&M University, College Station, TX, United States
| |
Collapse
|
16
|
Höring C, Conrad M, Söldner CA, Wang J, Sticht H, Strasser A, Miao Y. Specific Engineered G Protein Coupling to Histamine Receptors Revealed from Cellular Assay Experiments and Accelerated Molecular Dynamics Simulations. Int J Mol Sci 2021; 22:10047. [PMID: 34576210 PMCID: PMC8467750 DOI: 10.3390/ijms221810047] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Revised: 09/15/2021] [Accepted: 09/15/2021] [Indexed: 01/29/2023] Open
Abstract
G protein-coupled receptors (GPCRs) are targets of extracellular stimuli and hence occupy a key position in drug discovery. By specific and not yet fully elucidated coupling profiles with α subunits of distinct G protein families, they regulate cellular responses. The histamine H2 and H4 receptors (H2R and H4R) are prominent members of Gs- and Gi-coupled GPCRs. Nevertheless, promiscuous G protein and selective Gi signaling have been reported for the H2R and H4R, respectively, the molecular mechanism of which remained unclear. Using a combination of cellular experimental assays and Gaussian accelerated molecular dynamics (GaMD) simulations, we investigated the coupling profiles of the H2R and H4R to engineered mini-G proteins (mG). We obtained coupling profiles of the mGs, mGsi, or mGsq proteins to the H2R and H4R from the mini-G protein recruitment assays using HEK293T cells. Compared to H2R-mGs expressing cells, histamine responses were weaker (pEC50, Emax) for H2R-mGsi and -mGsq. By contrast, the H4R selectively bound to mGsi. Similarly, in all-atom GaMD simulations, we observed a preferential binding of H2R to mGs and H4R to mGsi revealed by the structural flexibility and free energy landscapes of the complexes. Although the mG α5 helices were consistently located within the HR binding cavity, alternative binding orientations were detected in the complexes. Due to the specific residue interactions, all mG α5 helices of the H2R complexes adopted the Gs-like orientation toward the receptor transmembrane (TM) 6 domain, whereas in H4R complexes, only mGsi was in the Gi-like orientation toward TM2, which was in agreement with Gs- and Gi-coupled GPCRs structures resolved by X-ray/cryo-EM. These cellular and molecular insights support (patho)physiological profiles of the histamine receptors, especially the hitherto little studied H2R function in the brain, as well as of the pharmacological potential of H4R selective drugs.
Collapse
Affiliation(s)
- Carina Höring
- Institute of Pharmacy, Faculty of Chemistry and Pharmacy, University of Regensburg, 93040 Regensburg, Germany
| | - Marcus Conrad
- Bioinformatik, Institut für Biochemie, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Fahrstraße 17, 91054 Erlangen, Germany
| | - Christian A Söldner
- Bioinformatik, Institut für Biochemie, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Fahrstraße 17, 91054 Erlangen, Germany
| | - Jinan Wang
- Department of Computational Biology and Molecular Biosciences, University of Kansas, Lawrence, KS 66047, USA
| | - Heinrich Sticht
- Bioinformatik, Institut für Biochemie, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Fahrstraße 17, 91054 Erlangen, Germany
- Erlangen National High Performance Computing Center (NHR@FAU), Friedrich-Alexander-University Erlangen-Nürnberg (FAU), 91058 Erlangen, Germany
| | - Andrea Strasser
- Institute of Pharmacy, Faculty of Chemistry and Pharmacy, University of Regensburg, 93040 Regensburg, Germany
| | - Yinglong Miao
- Department of Computational Biology and Molecular Biosciences, University of Kansas, Lawrence, KS 66047, USA
| |
Collapse
|
17
|
Qiu W, Lv Z, Xiao X, Shao S, Lin H. EMCBOW-GPCR: A method for identifying G-protein coupled receptors based on word embedding and wordbooks. Comput Struct Biotechnol J 2021; 19:4961-4969. [PMID: 34527200 PMCID: PMC8437786 DOI: 10.1016/j.csbj.2021.08.044] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Revised: 08/07/2021] [Accepted: 08/27/2021] [Indexed: 11/15/2022] Open
Abstract
An computational method was developed to identify G-protein coupled receptors. Three word-embedding models and a bag-of-words model are used to extract original features. A high accuracy was achieved by using fusion information. A powerful tool was established.
G Protein-Coupled Receptors (GPCRs) are one of the largest membrane protein receptor family in human, which are also important targets for many drugs. Thence, it’s of great significance to judge whether a protein is a GPCR or not. However, identifying GPCRs by experimental methods is very expensive and time-consuming. As more and more GPCR primary sequences are accumulated, it’s feasible to develop a computational model to predict GPCRs precisely and quickly. In this paper, a novel method called EMCBOW-GPCR has been proposed to improve the accuracy of identifying GPCRs based on natural language processing (NLP). For representing GPCRs, three word-embedding models and a bag-of-words model are used to extract original features. Then, the original features are thrown into a Deep-learning algorithm to extract features further and reduce the dimension. Finally, the obtained features are fed into Extreme Gradient Boosting. As shown with the results comparison, the overall prediction metrics of EMCBOW-GPCR are higher than the state of the arts. In order to be convenient for more researchers to use EMCBOW-GPCR, the method and source code have been opened in github, which are available at https://github.com/454170054/EMCBOW-GPCR, and a user-friendly web-server for EMCBOW-GPCR has been established at http://www.jci-bioinfo.cn/emcbowgpcr.
Collapse
Affiliation(s)
- Wangren Qiu
- School of Information Engineering, Jingdezhen Ceramic Institute, Jingdezhen, China
| | - Zhe Lv
- School of Information Engineering, Jingdezhen Ceramic Institute, Jingdezhen, China
| | - Xuan Xiao
- School of Information Engineering, Jingdezhen Ceramic Institute, Jingdezhen, China
| | - Shuai Shao
- School of Information Engineering, Jingdezhen Ceramic Institute, Jingdezhen, China
| | - Hao Lin
- Center for Informational Biology, University of Electronic Science and Technology of China, Chengdu 610054, China
| |
Collapse
|
18
|
Liu N, Li T, Wang Y, Liu S. G-Protein Coupled Receptors (GPCRs) in Insects-A Potential Target for New Insecticide Development. Molecules 2021; 26:2993. [PMID: 34069969 PMCID: PMC8157829 DOI: 10.3390/molecules26102993] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2021] [Revised: 05/10/2021] [Accepted: 05/12/2021] [Indexed: 02/07/2023] Open
Abstract
G-protein coupled receptors (GPCRs) play important roles in cell biology and insects' physiological processes, toxicological response and the development of insecticide resistance. New information on genome sequences, proteomic and transcriptome analysis and expression patterns of GPCRs in organs such as the central nervous system in different organisms has shown the importance of these signaling regulatory GPCRs and their impact on vital cell functions. Our growing understanding of the role played by GPCRs at the cellular, genome, transcriptome and tissue levels is now being utilized to develop new targets that will sidestep many of the problems currently hindering human disease control and insect pest management. This article reviews recent work on the expression and function of GPCRs in insects, focusing on the molecular complexes governing the insect physiology and development of insecticide resistance and examining the genome information for GPCRs in two medically important insects, mosquitoes and house flies, and their orthologs in the model insect species Drosophila melanogaster. The tissue specific distribution and expression of the insect GPCRs is discussed, along with fresh insights into practical aspects of insect physiology and toxicology that could be fundamental for efforts to develop new, more effective, strategies for pest control and resistance management.
Collapse
Affiliation(s)
- Nannan Liu
- Department of Entomology and Plant Pathology, Auburn University, Auburn, AL 36849, USA; (T.L.); (Y.W.)
| | - Ting Li
- Department of Entomology and Plant Pathology, Auburn University, Auburn, AL 36849, USA; (T.L.); (Y.W.)
| | - Yifan Wang
- Department of Entomology and Plant Pathology, Auburn University, Auburn, AL 36849, USA; (T.L.); (Y.W.)
| | - Shikai Liu
- College of Fisheries, Ocean University of China, Qingdao 266100, China;
| |
Collapse
|
19
|
Liu N, Wang Y, Li T, Feng X. G-Protein Coupled Receptors (GPCRs): Signaling Pathways, Characterization, and Functions in Insect Physiology and Toxicology. Int J Mol Sci 2021; 22:ijms22105260. [PMID: 34067660 PMCID: PMC8156084 DOI: 10.3390/ijms22105260] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Revised: 05/12/2021] [Accepted: 05/13/2021] [Indexed: 02/06/2023] Open
Abstract
G-protein-coupled receptors (GPCRs) are known to play central roles in the physiology of many organisms. Members of this seven α-helical transmembrane protein family transduce the extracellular signals and regulate intracellular second messengers through coupling to heterotrimeric G-proteins, adenylate cyclase, cAMPs, and protein kinases. As a result of the critical function of GPCRs in cell physiology and biochemistry, they not only play important roles in cell biology and the medicines used to treat a wide range of human diseases but also in insects’ physiological functions. Recent studies have revealed the expression and function of GPCRs in insecticide resistance, improving our understanding of the molecular complexes governing the development of insecticide resistance. This article focuses on the review of G-protein coupled receptor (GPCR) signaling pathways in insect physiology, including insects’ reproduction, growth and development, stress responses, feeding, behaviors, and other physiological processes. Hormones and polypeptides that are involved in insect GPCR regulatory pathways are reviewed. The review also gives a brief introduction of GPCR pathways in organisms in general. At the end of the review, it provides the recent studies on the function of GPCRs in the development of insecticide resistance, focusing in particular on our current knowledge of the expression and function of GPCRs and their downstream regulation pathways and their roles in insecticide resistance and the regulation of resistance P450 gene expression. The latest insights into the exciting technological advances and new techniques for gene expression and functional characterization of the GPCRs in insects are provided.
Collapse
Affiliation(s)
- Nannan Liu
- Department of Entomology and Plant Pathology, Auburn University, Auburn, AL 36849, USA; (Y.W.); (T.L.)
- Correspondence: ; Tel.: +1-334-844-5076
| | - Yifan Wang
- Department of Entomology and Plant Pathology, Auburn University, Auburn, AL 36849, USA; (Y.W.); (T.L.)
| | - Ting Li
- Department of Entomology and Plant Pathology, Auburn University, Auburn, AL 36849, USA; (Y.W.); (T.L.)
| | - Xuechun Feng
- Department of Biology Sciences, University of California, San Diego, CA 92093, USA;
| |
Collapse
|
20
|
Qiu W, Lv Z, Hong Y, Jia J, Xiao X. BOW-GBDT: A GBDT Classifier Combining With Artificial Neural Network for Identifying GPCR-Drug Interaction Based on Wordbook Learning From Sequences. Front Cell Dev Biol 2021; 8:623858. [PMID: 33598456 PMCID: PMC7882597 DOI: 10.3389/fcell.2020.623858] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Accepted: 12/15/2020] [Indexed: 12/28/2022] Open
Abstract
Background: As a class of membrane protein receptors, G protein-coupled receptors (GPCRs) are very important for cells to complete normal life function and have been proven to be a major drug target for widespread clinical application. Hence, it is of great significance to find GPCR targets that interact with drugs in the process of drug development. However, identifying the interaction of the GPCR–drug pairs by experimental methods is very expensive and time-consuming on a large scale. As more and more database about GPCR–drug pairs are opened, it is viable to develop machine learning models to accurately predict whether there is an interaction existing in a GPCR–drug pair. Methods: In this paper, the proposed model aims to improve the accuracy of predicting the interactions of GPCR–drug pairs. For GPCRs, the work extracts protein sequence features based on a novel bag-of-words (BOW) model improved with weighted Silhouette Coefficient and has been confirmed that it can extract more pattern information and limit the dimension of feature. For drug molecules, discrete wavelet transform (DWT) is used to extract features from the original molecular fingerprints. Subsequently, the above-mentioned two types of features are contacted, and SMOTE algorithm is selected to balance the training dataset. Then, artificial neural network is used to extract features further. Finally, a gradient boosting decision tree (GBDT) model is trained with the selected features. In this paper, the proposed model is named as BOW-GBDT. Results: D92M and Check390 are selected for testing BOW-GBDT. D92M is used for a cross-validation dataset which contains 635 interactive GPCR–drug pairs and 1,225 non-interactive pairs. Check390 is used for an independent test dataset which consists of 130 interactive GPCR–drug pairs and 260 non-interactive GPCR–drug pairs, and each element in Check390 cannot be found in D92M. According to the results, the proposed model has a better performance in generation ability compared with the existing machine learning models. Conclusion: The proposed predictor improves the accuracy of the interactions of GPCR–drug pairs. In order to facilitate more researchers to use the BOW-GBDT, the predictor has been settled into a brand-new server, which is available at http://www.jci-bioinfo.cn/bowgbdt.
Collapse
Affiliation(s)
- Wangren Qiu
- School of Information Engineering, Jingdezhen Ceramic Institute, Jingdezhen, China
| | - Zhe Lv
- School of Information Engineering, Jingdezhen Ceramic Institute, Jingdezhen, China
| | - Yaoqiu Hong
- School of Information Engineering, Jingdezhen University, Jingdezhen, China
| | - Jianhua Jia
- School of Information Engineering, Jingdezhen Ceramic Institute, Jingdezhen, China
| | - Xuan Xiao
- School of Information Engineering, Jingdezhen Ceramic Institute, Jingdezhen, China
| |
Collapse
|
21
|
Yang D, Zhou Q, Labroska V, Qin S, Darbalaei S, Wu Y, Yuliantie E, Xie L, Tao H, Cheng J, Liu Q, Zhao S, Shui W, Jiang Y, Wang MW. G protein-coupled receptors: structure- and function-based drug discovery. Signal Transduct Target Ther 2021; 6:7. [PMID: 33414387 PMCID: PMC7790836 DOI: 10.1038/s41392-020-00435-w] [Citation(s) in RCA: 331] [Impact Index Per Article: 82.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2020] [Revised: 11/30/2020] [Accepted: 12/05/2020] [Indexed: 02/08/2023] Open
Abstract
As one of the most successful therapeutic target families, G protein-coupled receptors (GPCRs) have experienced a transformation from random ligand screening to knowledge-driven drug design. We are eye-witnessing tremendous progresses made recently in the understanding of their structure-function relationships that facilitated drug development at an unprecedented pace. This article intends to provide a comprehensive overview of this important field to a broader readership that shares some common interests in drug discovery.
Collapse
Affiliation(s)
- Dehua Yang
- The National Center for Drug Screening, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 201203, Shanghai, China.,The CAS Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 201203, Shanghai, China
| | - Qingtong Zhou
- School of Basic Medical Sciences, Fudan University, 200032, Shanghai, China
| | - Viktorija Labroska
- The National Center for Drug Screening, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 201203, Shanghai, China.,University of Chinese Academy of Sciences, 100049, Beijing, China
| | - Shanshan Qin
- iHuman Institute, ShanghaiTech University, 201210, Shanghai, China
| | - Sanaz Darbalaei
- The National Center for Drug Screening, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 201203, Shanghai, China.,University of Chinese Academy of Sciences, 100049, Beijing, China
| | - Yiran Wu
- iHuman Institute, ShanghaiTech University, 201210, Shanghai, China
| | - Elita Yuliantie
- The National Center for Drug Screening, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 201203, Shanghai, China.,University of Chinese Academy of Sciences, 100049, Beijing, China
| | - Linshan Xie
- iHuman Institute, ShanghaiTech University, 201210, Shanghai, China.,School of Life Science and Technology, ShanghaiTech University, 201210, Shanghai, China
| | - Houchao Tao
- iHuman Institute, ShanghaiTech University, 201210, Shanghai, China
| | - Jianjun Cheng
- iHuman Institute, ShanghaiTech University, 201210, Shanghai, China
| | - Qing Liu
- The National Center for Drug Screening, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 201203, Shanghai, China.,The CAS Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 201203, Shanghai, China
| | - Suwen Zhao
- iHuman Institute, ShanghaiTech University, 201210, Shanghai, China.,School of Life Science and Technology, ShanghaiTech University, 201210, Shanghai, China
| | - Wenqing Shui
- iHuman Institute, ShanghaiTech University, 201210, Shanghai, China. .,School of Life Science and Technology, ShanghaiTech University, 201210, Shanghai, China.
| | - Yi Jiang
- The CAS Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 201203, Shanghai, China.
| | - Ming-Wei Wang
- The National Center for Drug Screening, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 201203, Shanghai, China. .,The CAS Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 201203, Shanghai, China. .,School of Basic Medical Sciences, Fudan University, 200032, Shanghai, China. .,University of Chinese Academy of Sciences, 100049, Beijing, China. .,School of Life Science and Technology, ShanghaiTech University, 201210, Shanghai, China. .,School of Pharmacy, Fudan University, 201203, Shanghai, China.
| |
Collapse
|
22
|
Calderón-Zamora L, Canizalez-Román A, León-Sicairos N, Aguilera-Mendez A, Huang F, Hong E, Villafaña S. Changes in expression of orphan receptors GPR99 and GPR107 during the development and establishment of hypertension in spontaneously hypertensive rats. J Recept Signal Transduct Res 2020; 41:558-565. [PMID: 33121311 DOI: 10.1080/10799893.2020.1835959] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
Hypertension is a disease, which in spite of existing treatments continues to have high morbidity and mortality, which suggests that there are other mechanisms involved in this pathology. In this sense, the orphan receptors are G protein-coupled receptor associated with various pathologies such as GPR99 which has been linked to mice develop left ventricular hypertrophy induced by blood pressure overload while GPR107 with patients with idiopathic pulmonary arterial hypertension. For this reason, the aim of this work was to study if the expression of the orphan receptors GPR99 and GPR107 are modified by arterial hypertension. Male SHR and WKY rats of 6-8 and 10-12 weeks old were used. The weight, systolic blood pressure and heart rate were measured, as well as the mRNA of the receptors GPR99 and GPR107 in the aorta, kidney, heart and brain by RT-PCR, also was realized an in silico analysis to predict which G protein could be coupled the orphan receptor GPR107. Our results showed that receptors GPR99 and GPR107 are expressed in the analyzed tissues and their expression profile tends to change at different ages and with the development of hypertension, for the other hand, the bioinformatics analysis for GPR107 showed that is coupled to Gi protein. Therefore, we do not rule out that GPR99 and GPR107 could be involved in the pathophysiology of hypertension and could be used as targets therapeutic in hypertension.
Collapse
Affiliation(s)
| | | | - Nidia León-Sicairos
- CIASaP, Facultad de Medicina, Universidad Autónoma de Sinaloa, Culiacán, México
| | - Asdrubal Aguilera-Mendez
- Instituto de Investigaciones Químico Biológicas, Universidad Michoacana de San Nicolás Hidalgo, Morelia, México
| | - Fengyang Huang
- Laboratorio de Investigación de Farmacología, Hospital Infantil de México Federico Gómez (HIMFG), Ciudad de México, México
| | | | - Santiago Villafaña
- Laboratorio de Farmacología Molecular, Sección de Estudios de Posgrado e Investigación, Escuela Superior de Medicina, Instituto Politécnico Nacional, Ciudad de México, México
| |
Collapse
|
23
|
Cytosolic and intra-organellar Ca2+ oscillations: mechanisms and function. CURRENT OPINION IN PHYSIOLOGY 2020. [DOI: 10.1016/j.cophys.2020.08.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
|
24
|
Wang P, Huang X, Qiu W, Xiao X. Identifying GPCR-drug interaction based on wordbook learning from sequences. BMC Bioinformatics 2020; 21:150. [PMID: 32312232 PMCID: PMC7171867 DOI: 10.1186/s12859-020-3488-8] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2019] [Accepted: 04/13/2020] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND G protein-coupled receptors (GPCRs) mediate a variety of important physiological functions, are closely related to many diseases, and constitute the most important target family of modern drugs. Therefore, the research of GPCR analysis and GPCR ligand screening is the hotspot of new drug development. Accurately identifying the GPCR-drug interaction is one of the key steps for designing GPCR-targeted drugs. However, it is prohibitively expensive to experimentally ascertain the interaction of GPCR-drug pairs on a large scale. Therefore, it is of great significance to predict the interaction of GPCR-drug pairs directly from the molecular sequences. With the accumulation of known GPCR-drug interaction data, it is feasible to develop sequence-based machine learning models for query GPCR-drug pairs. RESULTS In this paper, a new sequence-based method is proposed to identify GPCR-drug interactions. For GPCRs, we use a novel bag-of-words (BoW) model to extract sequence features, which can extract more pattern information from low-order to high-order and limit the feature space dimension. For drug molecules, we use discrete Fourier transform (DFT) to extract higher-order pattern information from the original molecular fingerprints. The feature vectors of two kinds of molecules are concatenated and input into a simple prediction engine distance-weighted K-nearest-neighbor (DWKNN). This basic method is easy to be enhanced through ensemble learning. Through testing on recently constructed GPCR-drug interaction datasets, it is found that the proposed methods are better than the existing sequence-based machine learning methods in generalization ability, even an unconventional method in which the prediction performance was further improved by post-processing procedure (PPP). CONCLUSIONS The proposed methods are effective for GPCR-drug interaction prediction, and may also be potential methods for other target-drug interaction prediction, or protein-protein interaction prediction. In addition, the new proposed feature extraction method for GPCR sequences is the modified version of the traditional BoW model and may be useful to solve problems of protein classification or attribute prediction. The source code of the proposed methods is freely available for academic research at https://github.com/wp3751/GPCR-Drug-Interaction.
Collapse
Affiliation(s)
- Pu Wang
- Computer School, Hubei University of Arts and Science, Xiangyang, 441053 China
| | - Xiaotong Huang
- Computer School, Hubei University of Arts and Science, Xiangyang, 441053 China
| | - Wangren Qiu
- Computer Department, Jingdezhen Ceramic Institute, Jingdezhen, 333403 China
| | - Xuan Xiao
- Computer Department, Jingdezhen Ceramic Institute, Jingdezhen, 333403 China
| |
Collapse
|
25
|
Khan S, Nisar A, Yuan J, Luo X, Dou X, Liu F, Zhao X, Li J, Ahmad H, Mehmood SA, Feng X. A Whole Genome Re-Sequencing Based GWA Analysis Reveals Candidate Genes Associated with Ivermectin Resistance in Haemonchus contortus. Genes (Basel) 2020; 11:E367. [PMID: 32231078 PMCID: PMC7230667 DOI: 10.3390/genes11040367] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2020] [Revised: 03/11/2020] [Accepted: 03/26/2020] [Indexed: 11/23/2022] Open
Abstract
The most important and broad-spectrum drug used to control the parasitic worms to date is ivermectin (IVM). Resistance against IVM has emerged in parasites, and preserving its efficacy is now becoming a serious issue. The parasitic nematode Haemonchus contortus (Rudolphi, 1803) is economically an important parasite of small ruminants across the globe, which has a successful track record in IVM resistance. There are growing evidences regarding the multigenic nature of IVM resistance, and although some genes have been proposed as candidates of IVM resistance using lower magnification of genome, the genetic basis of IVM resistance still remains poorly resolved. Using the full magnification of genome, we herein applied a population genomics approach to characterize genome-wide signatures of selection among pooled worms from two susceptible and six ivermectin-resistant isolates of H. contortus, and revealed candidate genes under selection in relation to IVM resistance. These candidates also included a previously known IVM-resistance-associated candidate gene HCON_00148840, glc-3. Finally, an RNA-interference-based functional validation assay revealed the HCON_00143950 as IVM-tolerance-associated gene in H. contortus. The possible role of this gene in IVM resistance could be detoxification of xenobiotic in phase I of xenobiotic metabolism. The results of this study further enhance our understanding on the IVM resistance and continue to provide further evidence in favor of multigenic nature of IVM resistance.
Collapse
Affiliation(s)
- Sawar Khan
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Key Laboratory of Animal Parasitology, Ministry of Agriculture of China, Shanghai 200241, China
| | - Ayesha Nisar
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Key Laboratory of Animal Parasitology, Ministry of Agriculture of China, Shanghai 200241, China
| | - Jianqi Yuan
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Key Laboratory of Animal Parasitology, Ministry of Agriculture of China, Shanghai 200241, China
| | - Xiaoping Luo
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Key Laboratory of Animal Parasitology, Ministry of Agriculture of China, Shanghai 200241, China
- Veterinary Research Institute, Inner Mongolia Academy of Agricultural and Animal Husbandry Sciences, Hohhot 010031, China
| | - Xueqin Dou
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Key Laboratory of Animal Parasitology, Ministry of Agriculture of China, Shanghai 200241, China
| | - Fei Liu
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Key Laboratory of Animal Parasitology, Ministry of Agriculture of China, Shanghai 200241, China
| | - Xiaochao Zhao
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Key Laboratory of Animal Parasitology, Ministry of Agriculture of China, Shanghai 200241, China
| | - Junyan Li
- Veterinary Research Institute, Inner Mongolia Academy of Agricultural and Animal Husbandry Sciences, Hohhot 010031, China
| | - Habib Ahmad
- Department of Genetics, Hazara University, Mansehra 21300, Pakistan
| | | | - Xingang Feng
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Key Laboratory of Animal Parasitology, Ministry of Agriculture of China, Shanghai 200241, China
| |
Collapse
|
26
|
Franco R, Castelló J, Canela EI. The Kinetic Component in Drug Discovery: Using the Most Basic Pharmacological Concepts to Advance in Selecting Drugs to Combat CNS Diseases. Curr Neuropharmacol 2020; 18:250-257. [PMID: 31573886 PMCID: PMC7327946 DOI: 10.2174/1570159x17666191001144309] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2019] [Revised: 07/02/2019] [Accepted: 09/24/2019] [Indexed: 01/14/2023] Open
Abstract
To reach the central nervous system (CNS), drugs must cross the brain-blood barrier and have appropriate pharmacokinetic/dynamic properties. However, in early drug discovery steps, the selection of lead compounds, for example, those targeting G-protein-coupled receptors (GPCRs), is made according to i) affinity, which is calculated in in vitro equilibrium conditions, and ii) potency, a signal transduction-related parameter, usually quantified at a fixed time-point in a heterologous expression system. This paper argues that kinetics must be considered in the early steps of lead compound selection. While affinity calculation requires the establishment of a ligand-receptor equilibrium, the signal transduction starts as soon as the receptor senses the agonist. Taking cAMP production as an example, the in vitro-measured cytoplasmic levels of this cyclic nucleotide do not depend on equilibrium dissociation constant, KD. Signaling occurs far from the equilibrium and correlates more with the binding rate (kon) than with KD. Furthermore, residence time, a parameter to consider in lead optimization, may significantly vary from in vitro to in vivo conditions. The results are discussed from the perspective of dopaminergic neurotransmission and dopaminereceptor- based drug discovery.
Collapse
Affiliation(s)
- Rafael Franco
- Department of Biochemistry and Molecular Biomedicine, Molecular Neurobiology Laboratory, Biology School, University of Barcelona, Barcelona, Spain
- Centro de Investigación en Red, Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain
| | - Josema Castelló
- Department of Biochemistry and Molecular Biomedicine, Molecular Neurobiology Laboratory, Biology School, University of Barcelona, Barcelona, Spain
| | - Enric I. Canela
- Department of Biochemistry and Molecular Biomedicine, Molecular Neurobiology Laboratory, Biology School, University of Barcelona, Barcelona, Spain
- Centro de Investigación en Red, Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain
| |
Collapse
|
27
|
Fazli shojai E, Najafi M, Charkhpour M. Evaluating the Effects of Chronic Administration of Natural Honey on the Development of Dependence on Morphine in the Male Rats. PHARMACEUTICAL SCIENCES 2019. [DOI: 10.15171/ps.2019.35] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
Background: According to the previous studies, the exact mechanism of dependence on opioids and withdrawal syndrome has not been fully understood but one of the most important mechanisms is the increase of pro-inflammatory cytokines in CNS. On the other way, previous studies showed that natural honey (NHO) has anti-inflammatory properties. This study was aimed to evaluate the effects of chronic administration of natural honey on the development of morphine dependence in male rats. Methods: Honey was prepared from Tarom Oliya region in Zanjan province. Experiments were performed on male Wistar rats weighing 225-275 g, randomly divided into 6 groups (n=8). The study groups included morphine group, the three doses of morphine plus honey group (at doses of 200,400 and 800 mg/kg, i.p.), the morphine plus vehicle group, and the saline group. The subcutaneous injections of additive doses of morphine were used for 9 days to create morphine dependency. On the 9th day, one hour after the morning dose of morphine, naloxone (4 mg/kg, i.p.) was injected, and symptoms of withdrawal syndrome were assessed for 60 minutes. Then, blood samples were taken to measure TNF-α. One-way ANOVA and Tukey tests were used to compare the results. P- Value of <0.05 was considered as statistically significant. Results: The results of this study showed that intraperitoneal injection of honey at 3 doses (200, 400 and 800 mg/kg with p <0.001) could significantly decrease the total score of the symptoms compared to the morphine-vehicle control group. Natural honey (NHO) could significantly decrease TNF-α at dose of 400 mg/kg. Conclusion: The results indicated that chronic administration of NHO had beneficial effects in reducing symptoms of morphine withdrawal syndrome, and this effect is probably due to the anti-inflammatory effect caused by the polyphenolic compounds in honey.<br />
Collapse
Affiliation(s)
- Elham Fazli shojai
- Student Research Committee, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Moslem Najafi
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mohammad Charkhpour
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
28
|
CCR5: Established paradigms and new frontiers for a 'celebrity' chemokine receptor. Cytokine 2019; 109:81-93. [PMID: 29903576 DOI: 10.1016/j.cyto.2018.02.018] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2017] [Revised: 02/14/2018] [Accepted: 02/16/2018] [Indexed: 01/04/2023]
Abstract
Because of the level of attention it received due to its role as the principal HIV coreceptor, CCR5 has been described as a 'celebrity' chemokine receptor. Here we describe the development of CCR5 inhibitory strategies that have been developed for HIV therapy and which are now additionally being considered for use in HIV prevention and cure. The wealth of CCR5-related tools that have been developed during the intensive investigation of CCR5 as an HIV drug target can now be turned towards the study of CCR5 as a model chemokine receptor. We also summarize what is currently known about the cell biology and pharmacology of CCR5, providing an update on new areas of investigation that have emerged in recent research. Finally, we discuss the potential of CCR5 as a drug target for diseases other than HIV, discussing the evidence linking CCR5 and its natural chemokine ligands with inflammatory diseases, particularly neuroinflammation, and certain cancers. These pathologies may provide new uses for the strategies for CCR5 blockade originally developed to combat HIV/AIDS.
Collapse
|
29
|
A 2A Receptor Homodimer-Disrupting Sequence Efficiently Delivered by a Protease-Resistant, Cyclic CPP Vector. Int J Mol Sci 2019; 20:ijms20194937. [PMID: 31590403 PMCID: PMC6801510 DOI: 10.3390/ijms20194937] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2019] [Revised: 09/26/2019] [Accepted: 09/30/2019] [Indexed: 12/14/2022] Open
Abstract
G-protein-coupled receptors associate into dimers/oligomers whose function is not well understood. One approach to investigate this issue is to challenge oligomerization by peptides replicating transmembrane domains and to study their effect on receptor functionality. The disruptor peptides are typically delivered by means of cell-penetrating vectors such as the human immunodeficiency virus (HIV) transcription trans-activation protein Tat. In this paper we report a cyclic, Tat-like peptide that significantly improves its linear analogue in targeting interreceptor sequences in the transmembrane space. The same cyclic Tat-like vector fused to a transmembrane region not involved in receptor oligomerization was totally ineffective. Besides higher efficacy, the cyclic version has enhanced proteolytic stability, as shown by trypsin digestion experiments.
Collapse
|
30
|
Pace JR, Jog R, Burgess DJ, Hadden MK. Formulation and evaluation of itraconazole liposomes for Hedgehog pathway inhibition. J Liposome Res 2019; 30:305-311. [PMID: 31576768 DOI: 10.1080/08982104.2019.1668011] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
Itraconazole (ITZ) is an FDA-approved antifungal agent that has recently been explored for novel biological properties. In particular, ITZ was identified as a potent inhibitor of the hedgehog (Hh) pathway, a cell signalling pathway that has been linked to a variety of cancers and accounts for ∼25% of paediatric medulloblastoma (MB) cases. To date, there is not a targeted therapeutic option for paediatric MB, resulting in long-term side effects such as hormone deficiency, organ damage and secondary cancers. A primary obstacle for developing targeted therapy for brain ailments is the presence of the blood-brain barrier (BBB), which protects the brain from potentially harmful substances. Due to its size and hydrophobicity, ITZ does not penetrate the BBB. Alternatively, liposomes are being increasingly used within the clinic to increase drug bioavailability, target specificity and BBB permeability. With this in mind, we have successfully developed ITZ-containing liposomes with an optimal size for BBB penetration (<100 nm) and encapsulation efficiency (∼95%) by utilizing a continuous manufacturing approach-turbulent coaxial jet in co-flow. Our preliminary in vitro data demonstrate that these liposomes inhibit the Hh pathway, albeit at a reduced level in comparison to free ITZ. (196/250 words).
Collapse
Affiliation(s)
- Jennifer R Pace
- Department of Pharmaceutical Sciences, University of Connecticut, Storrs, CT, USA
| | - Rajan Jog
- Department of Pharmaceutical Sciences, University of Connecticut, Storrs, CT, USA
| | - Diane J Burgess
- Department of Pharmaceutical Sciences, University of Connecticut, Storrs, CT, USA
| | - M Kyle Hadden
- Department of Pharmaceutical Sciences, University of Connecticut, Storrs, CT, USA
| |
Collapse
|
31
|
Gaiser BI, Danielsen M, Marcher-Rørsted E, Røpke Jørgensen K, Wróbel TM, Frykman M, Johansson H, Bräuner-Osborne H, Gloriam DE, Mathiesen JM, Sejer Pedersen D. Probing the Existence of a Metastable Binding Site at the β 2-Adrenergic Receptor with Homobivalent Bitopic Ligands. J Med Chem 2019; 62:7806-7839. [PMID: 31298548 DOI: 10.1021/acs.jmedchem.9b00595] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Herein, we report the development of bitopic ligands aimed at targeting the orthosteric binding site (OBS) and a metastable binding site (MBS) within the same receptor unit. Previous molecular dynamics studies on ligand binding to the β2-adrenergic receptor (β2AR) suggested that ligands pause at transient, less-conserved MBSs. We envisioned that MBSs can be regarded as allosteric binding sites and targeted by homobivalent bitopic ligands linking two identical pharmacophores. Such ligands were designed based on docking of the antagonist (S)-alprenolol into the OBS and an MBS and synthesized. Pharmacological characterization revealed ligands with similar potency and affinity, slightly increased β2/β1AR-selectivity, and/or substantially slower β2AR off-rates compared to (S)-alprenolol. Truncated bitopic ligands suggested the major contribution of the metastable pharmacophore to be a hydrophobic interaction with the β2AR, while the linkers alone decreased the potency of the orthosteric fragment. Altogether, the study underlines the potential of targeting MBSs for improving the pharmacological profiles of ligands.
Collapse
Affiliation(s)
- Birgit I Gaiser
- Department of Drug Design and Pharmacology , University of Copenhagen , Jagtvej 162 , 2100 Copenhagen , Denmark
| | - Mia Danielsen
- Department of Drug Design and Pharmacology , University of Copenhagen , Jagtvej 162 , 2100 Copenhagen , Denmark
| | - Emil Marcher-Rørsted
- Department of Drug Design and Pharmacology , University of Copenhagen , Jagtvej 162 , 2100 Copenhagen , Denmark
| | - Kira Røpke Jørgensen
- Department of Drug Design and Pharmacology , University of Copenhagen , Jagtvej 162 , 2100 Copenhagen , Denmark
| | - Tomasz M Wróbel
- Department of Drug Design and Pharmacology , University of Copenhagen , Jagtvej 162 , 2100 Copenhagen , Denmark.,Department of Synthesis and Chemical Technology of Pharmaceutical Substances, Faculty of Pharmacy , Medical University of Lublin , 4A Chodźki 20093 Lublin , Poland
| | - Mikael Frykman
- Department of Drug Design and Pharmacology , University of Copenhagen , Jagtvej 162 , 2100 Copenhagen , Denmark
| | - Henrik Johansson
- Department of Drug Design and Pharmacology , University of Copenhagen , Jagtvej 162 , 2100 Copenhagen , Denmark
| | - Hans Bräuner-Osborne
- Department of Drug Design and Pharmacology , University of Copenhagen , Jagtvej 162 , 2100 Copenhagen , Denmark
| | - David E Gloriam
- Department of Drug Design and Pharmacology , University of Copenhagen , Jagtvej 162 , 2100 Copenhagen , Denmark
| | - Jesper Mosolff Mathiesen
- Department of Drug Design and Pharmacology , University of Copenhagen , Jagtvej 162 , 2100 Copenhagen , Denmark
| | - Daniel Sejer Pedersen
- Department of Drug Design and Pharmacology , University of Copenhagen , Jagtvej 162 , 2100 Copenhagen , Denmark
| |
Collapse
|
32
|
Yaginuma K, Aoki W, Miura N, Ohtani Y, Aburaya S, Kogawa M, Nishikawa Y, Hosokawa M, Takeyama H, Ueda M. High-throughput identification of peptide agonists against GPCRs by co-culture of mammalian reporter cells and peptide-secreting yeast cells using droplet microfluidics. Sci Rep 2019; 9:10920. [PMID: 31358824 PMCID: PMC6662714 DOI: 10.1038/s41598-019-47388-x] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2019] [Accepted: 07/15/2019] [Indexed: 12/25/2022] Open
Abstract
Since G-protein coupled receptors (GPCRs) are linked to various diseases, screening of functional ligands against GPCRs is vital for drug discovery. In the present study, we developed a high-throughput functional cell-based assay by combining human culture cells producing a GPCR, yeast cells secreting randomized peptide ligands, and a droplet microfluidic device. We constructed a reporter human cell line that emits fluorescence in response to the activation of human glucagon-like peptide-1 receptor (hGLP1R). We then constructed a yeast library secreting an agonist of hGLP1R or randomized peptide ligands. We demonstrated that high-throughput identification of functional ligands against hGLP1R could be performed by co-culturing the reporter cells and the yeast cells in droplets. We identified functional ligands, one of which had higher activity than that of an original sequence. The result suggests that our system could facilitate the discovery of functional peptide ligands of GPCRs.
Collapse
Affiliation(s)
- Kenshi Yaginuma
- Division of Applied Life Sciences, Graduate School of Agriculture, Kyoto University, Sakyo-ku, Kyoto, 606-8502, Japan
| | - Wataru Aoki
- Division of Applied Life Sciences, Graduate School of Agriculture, Kyoto University, Sakyo-ku, Kyoto, 606-8502, Japan.,JST, CREST, 7 Goban-cho, Chiyoda-ku, Tokyo, 102-0076, Japan.,JST, PRESTO, 7 Goban-cho, Chiyoda-ku, Tokyo, 102-0076, Japan
| | - Natsuko Miura
- Graduate School of Life and Environmental Sciences, Osaka Prefecture University, 1-1 Gakuen-cho, Naka-ku, Sakai, Osaka, 599-8531, Japan
| | - Yuta Ohtani
- Division of Applied Life Sciences, Graduate School of Agriculture, Kyoto University, Sakyo-ku, Kyoto, 606-8502, Japan
| | - Shunsuke Aburaya
- Division of Applied Life Sciences, Graduate School of Agriculture, Kyoto University, Sakyo-ku, Kyoto, 606-8502, Japan.,Japan Society for the Promotion of Science, 5-3-1 Kojimachi, Chiyoda-ku, Tokyo, 102-0083, Japan
| | - Masato Kogawa
- Department of Life Science & Medical Bioscience, School of Advanced Science and Engineering, Waseda University, Shinjuku-ku, Tokyo, 169-8555, Japan.,Computational Bio Big-Data Open Innovation Laboratory, AIST-Waseda University, Shinjuku-ku, Tokyo, 169-0072, Japan
| | - Yohei Nishikawa
- Department of Life Science & Medical Bioscience, School of Advanced Science and Engineering, Waseda University, Shinjuku-ku, Tokyo, 169-8555, Japan.,Computational Bio Big-Data Open Innovation Laboratory, AIST-Waseda University, Shinjuku-ku, Tokyo, 169-0072, Japan
| | - Masahito Hosokawa
- JST, PRESTO, 7 Goban-cho, Chiyoda-ku, Tokyo, 102-0076, Japan.,Institute for Advanced Research of Biosystem Dynamics, Waseda Research Institute for Science and Engineering, Waseda University, Shinjuku-ku, Tokyo, 169-8555, Japan
| | - Haruko Takeyama
- Department of Life Science & Medical Bioscience, School of Advanced Science and Engineering, Waseda University, Shinjuku-ku, Tokyo, 169-8555, Japan.,Computational Bio Big-Data Open Innovation Laboratory, AIST-Waseda University, Shinjuku-ku, Tokyo, 169-0072, Japan.,Institute for Advanced Research of Biosystem Dynamics, Waseda Research Institute for Science and Engineering, Waseda University, Shinjuku-ku, Tokyo, 169-8555, Japan
| | - Mitsuyoshi Ueda
- Division of Applied Life Sciences, Graduate School of Agriculture, Kyoto University, Sakyo-ku, Kyoto, 606-8502, Japan. .,JST, CREST, 7 Goban-cho, Chiyoda-ku, Tokyo, 102-0076, Japan.
| |
Collapse
|
33
|
Syu GD, Wang SC, Ma G, Liu S, Pearce D, Prakash A, Henson B, Weng LC, Ghosh D, Ramos P, Eichinger D, Pino I, Dong X, Xiao J, Wang S, Tao N, Kim KS, Desai PJ, Zhu H. Development and application of a high-content virion display human GPCR array. Nat Commun 2019; 10:1997. [PMID: 31040288 PMCID: PMC6491619 DOI: 10.1038/s41467-019-09938-9] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2018] [Accepted: 04/05/2019] [Indexed: 12/21/2022] Open
Abstract
Human G protein-coupled receptors (GPCRs) respond to various ligands and stimuli. However, GPCRs rely on membrane for proper folding, making their biochemical properties difficult to study. By displaying GPCRs in viral envelopes, we fabricated a Virion Display (VirD) array containing 315 non-olfactory human GPCRs for functional characterization. Using this array, we found that 10 of 20 anti-GPCR mAbs were ultra-specific. We further demonstrated that those failed in the mAb assays could recognize their canonical ligands, suggesting proper folding. Next, using two peptide ligands on the VirD-GPCR array, we identified expected interactions and novel interactions. Finally, we screened the array with group B Streptococcus, a major cause of neonatal meningitis, and demonstrated that inhibition of a newly identified target, CysLTR1, reduced bacterial penetration both in vitro and in vivo. We believe that the VirD-GPCR array holds great potential for high-throughput screening for small molecule drugs, affinity reagents, and ligand deorphanization.
Collapse
Affiliation(s)
- Guan-Da Syu
- Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
- Center for High-Throughput Biology, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
- Viral Oncology Program, Department of Oncology, The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, 21231, USA
| | - Shih-Chin Wang
- Department of Biophysics and Biophysical Chemistry, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Guangzhong Ma
- Biodesign Center for Bioelectronics and Biosensors, Arizona State University, Tempe, AZ, 85287, USA
| | - Shuang Liu
- Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
- Center for High-Throughput Biology, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Donna Pearce
- Division of Paediatric Infectious Diseases, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA
| | - Atish Prakash
- Division of Paediatric Infectious Diseases, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA
| | - Brandon Henson
- Viral Oncology Program, Department of Oncology, The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, 21231, USA
| | - Lien-Chun Weng
- Viral Oncology Program, Department of Oncology, The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, 21231, USA
| | - Devlina Ghosh
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Pedro Ramos
- CDI Laboratories, Inc., Mayaguez, Puerto Rico, 00682, USA
| | | | - Ignacio Pino
- CDI Laboratories, Inc., Mayaguez, Puerto Rico, 00682, USA
| | - Xinzhong Dong
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Howard Hughes Medical Institute, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Jie Xiao
- Department of Biophysics and Biophysical Chemistry, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Shaopeng Wang
- Biodesign Center for Bioelectronics and Biosensors, Arizona State University, Tempe, AZ, 85287, USA
| | - Nongjian Tao
- Biodesign Center for Bioelectronics and Biosensors, Arizona State University, Tempe, AZ, 85287, USA
- School of Electrical, Computer and Energy Engineering, Arizona State University, Tempe, AZ, 85287, USA
| | - Kwang Sik Kim
- Division of Paediatric Infectious Diseases, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA.
| | - Prashant J Desai
- Viral Oncology Program, Department of Oncology, The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, 21231, USA.
| | - Heng Zhu
- Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA.
- Center for High-Throughput Biology, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA.
- Viral Oncology Program, Department of Oncology, The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, 21231, USA.
| |
Collapse
|
34
|
Rustler K, Pockes S, König B. Light-Switchable Antagonists for the Histamine H 1 Receptor at the Isolated Guinea Pig Ileum. ChemMedChem 2019; 14:636-644. [PMID: 30628180 DOI: 10.1002/cmdc.201800815] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2018] [Indexed: 12/13/2022]
Abstract
The histamine H1 G protein-coupled receptor (GPCR) plays an important role in allergy and inflammation. Existing drugs that address the H1 receptor differ in their chemical structure, pharmacology, and side effects. Light-controllable spatial and temporal activity regulation of photochromic H1 ligands may contribute to a better mechanistic understanding and the development of improved correlations between ligand structure and pharmacologic effects. We report photochromic H1 receptor ligands, which were investigated in an organ-pharmacological assay. Initially, five photochromic azobenzene derivatives of reported dual H1 -H4 receptor antagonists were designed, synthesized, photochemically characterized, and organ-pharmacologically tested on the isolated guinea pig ileum. Among them, one compound [trans-19: (Z)-1-(4-chlorophenyl)-1-(4-methylpiperazin-1-yl)-N-(4-((E)-phenyldiazenyl)phenyl)methanimine] retained the antagonistic activity of its non-photochromic lead, and trans-cis isomerization by irradiation induced a fourfold difference in the pharmacological response. Further structural optimization resulted in two bathochromically shifted derivatives of 19 [NO2 -substituted 35 {(Z)-1-(4-chlorophenyl)-1-(4-methylpiperazin-1-yl)-N-(4-((E)-(4-nitrophenyl)diazenyl)phenyl)methanimine} and SO3 - -substituted 41 {4-((E)-(4-(((Z)-(4-chlorophenyl)(4-methylpiperazin-1-yl)methylene)amino)phenyl)diazenyl)benzenesulfonate}], which do not require the use of UV light for photoisomerization and which also have improved solubility and show reduced tissue impairment. The trans isomers of both compounds showed a remarkable increase in antagonistic activity relative to their lead trans-19; furthermore, a 46-fold difference in activity on the isolated guinea pig ileum was observed between trans- and cis-35.
Collapse
Affiliation(s)
- Karin Rustler
- Institute of Organic Chemistry, Faculty of Chemistry and Pharmacy, University of Regensburg, Universitätsstraße 31, 93053, Regensburg, Germany
| | - Steffen Pockes
- Institute of Pharmacy, Faculty of Chemistry and Pharmacy, University of Regensburg, Universitätsstraße 31, 93053, Regensburg, Germany
| | - Burkhard König
- Institute of Organic Chemistry, Faculty of Chemistry and Pharmacy, University of Regensburg, Universitätsstraße 31, 93053, Regensburg, Germany
| |
Collapse
|
35
|
Affiliation(s)
- Oliver Plettenburg
- Institute of Medicinal ChemistryHelmholtz Zentrum München Ingolstädter Landstr. 1 D-85764 Neuherberg Germany
- Institute of Organic Chemistry and Centre of Biomolecular Drug Research (BMWZ)Leibniz Universität Hannover Schneiderberg 1b D-30167 Hannover Germany
| |
Collapse
|
36
|
Wold EA, Chen J, Cunningham KA, Zhou J. Allosteric Modulation of Class A GPCRs: Targets, Agents, and Emerging Concepts. J Med Chem 2019; 62:88-127. [PMID: 30106578 PMCID: PMC6556150 DOI: 10.1021/acs.jmedchem.8b00875] [Citation(s) in RCA: 108] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
G-protein-coupled receptors (GPCRs) have been tractable drug targets for decades with over one-third of currently marketed drugs targeting GPCRs. Of these, the class A GPCR superfamily is highly represented, and continued drug discovery for this family of receptors may provide novel therapeutics for a vast range of diseases. GPCR allosteric modulation is an innovative targeting approach that broadens the available small molecule toolbox and is proving to be a viable drug discovery strategy, as evidenced by recent FDA approvals and clinical trials. Numerous class A GPCR allosteric modulators have been discovered recently, and emerging trends such as the availability of GPCR crystal structures, diverse functional assays, and structure-based computational approaches are improving optimization and development. This Perspective provides an update on allosterically targeted class A GPCRs and their disease indications and the medicinal chemistry approaches toward novel allosteric modulators and highlights emerging trends and opportunities in the field.
Collapse
Affiliation(s)
- Eric A. Wold
- Department of Pharmacology and Toxicology, Chemical Biology Program, University of Texas Medical Branch, Galveston, Texas 77555, United States
- Department of Pharmacology and Toxicology, Center for Addiction Research, University of Texas Medical Branch, Galveston, Texas 77555, United States
| | - Jianping Chen
- Department of Pharmacology and Toxicology, Chemical Biology Program, University of Texas Medical Branch, Galveston, Texas 77555, United States
- Department of Pharmacology and Toxicology, Center for Addiction Research, University of Texas Medical Branch, Galveston, Texas 77555, United States
| | - Kathryn A. Cunningham
- Department of Pharmacology and Toxicology, Chemical Biology Program, University of Texas Medical Branch, Galveston, Texas 77555, United States
- Department of Pharmacology and Toxicology, Center for Addiction Research, University of Texas Medical Branch, Galveston, Texas 77555, United States
| | - Jia Zhou
- Department of Pharmacology and Toxicology, Chemical Biology Program, University of Texas Medical Branch, Galveston, Texas 77555, United States
- Department of Pharmacology and Toxicology, Center for Addiction Research, University of Texas Medical Branch, Galveston, Texas 77555, United States
| |
Collapse
|
37
|
Wu Y, Tong J, Ding K, Zhou Q, Zhao S. GPCR Allosteric Modulator Discovery. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1163:225-251. [PMID: 31707706 DOI: 10.1007/978-981-13-8719-7_10] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
G protein-coupled receptors (GPCRs) influence virtually every aspect of human physiology; about one-third of all marketed drugs target members of this family. GPCR allosteric ligands hold the promise of improved subtype selectivity, spatiotemporal sensitivity, and possible biased property over typical orthosteric ligands. However, only a small number of GPCR allosteric ligands have been approved as drugs or in clinical trials since the discovery process is very challenging. The rapid development of GPCR structural biology leads to the discovery of several allosteric sites and sheds light on understanding the mechanism of GPCR allosteric ligands, which is critical for discovering novel therapeutics. This book chapter summarized different GPCR allosteric modulating mechanisms and discussed validated mechanisms based on allosteric modulator-GPCR complex structures.
Collapse
Affiliation(s)
- Yiran Wu
- iHuman Institute, ShanghaiTech University, Shanghai, China
| | - Jiahui Tong
- iHuman Institute, ShanghaiTech University, Shanghai, China.,School of Life Science and Technology, ShanghaiTech University, Shanghai, China
| | - Kang Ding
- iHuman Institute, ShanghaiTech University, Shanghai, China
| | - Qingtong Zhou
- iHuman Institute, ShanghaiTech University, Shanghai, China
| | - Suwen Zhao
- iHuman Institute, ShanghaiTech University, Shanghai, China. .,School of Life Science and Technology, ShanghaiTech University, Shanghai, China.
| |
Collapse
|
38
|
Gates C, Backos DS, Reigan P, Kang HJ, Koerner C, Mirzaei J, Natale NR. Isoxazolo[3,4-d]pyridazinones positively modulate the metabotropic glutamate subtypes 2 and 4. Bioorg Med Chem 2018; 26:4797-4803. [PMID: 30143366 PMCID: PMC6675577 DOI: 10.1016/j.bmc.2018.08.012] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2018] [Revised: 08/01/2018] [Accepted: 08/09/2018] [Indexed: 11/29/2022]
Abstract
Isoxazolo[3,4-d] pyridazinones ([3,4-d]s) are selective positive modulators of the metabotropic glutamate receptors (mGluRs) subtypes 2 and 4, with no functional cross reactivity at mGluR1a, mGLuR5 or mGluR8. Modest binding for two of the [3,4-d]s is observed at the allosteric fenobam mGluR5 site, but not sufficient to translate into a functional effect. The structure activity relationship (SAR) for mGluR2 and mGluR4 are distinct: the compounds which select for mGluR2 all contain fluorine on the N-6 aryl group. Furthermore, the [3,4-d]s in this study showed no significant binding at inhibitory GABAA, nor excitatory NMDA receptors, and previously we had disclosed that they lack significant activity at the System Xc-Antiporter. A homology model based on Conn's mGluR1 crystal structure was examined, and suggested explanations for a preference for allosteric over orthosteric binding, subtype selectivity, and suggested avenues for optimization of efficacy as a reasonable working hypothesis.
Collapse
Affiliation(s)
- Christina Gates
- Department of Biomedical and Pharmaceutical Sciences, University of Montana, Missoula, MT 59812, United States
| | - Donald S Backos
- Skaggs School of Pharmacy and Pharmaceutical Sciences, Anschutz Medical Campus, University of Colorado Denver, Aurora, CO 80045, United States
| | - Philip Reigan
- Skaggs School of Pharmacy and Pharmaceutical Sciences, Anschutz Medical Campus, University of Colorado Denver, Aurora, CO 80045, United States
| | - Hye Jin Kang
- Psychoactive Drug Screening Program, University of North Carolina at Chapel Hill, Department of Pharmacology, School of Medicine, 2113 Genetics Medicine Building, 120 Mason Farm Road, Chapel Hill, NC 27599, United States
| | - Chris Koerner
- Department of Biomedical and Pharmaceutical Sciences, University of Montana, Missoula, MT 59812, United States
| | - Joseph Mirzaei
- Department of Biomedical and Pharmaceutical Sciences, University of Montana, Missoula, MT 59812, United States
| | - N R Natale
- Department of Biomedical and Pharmaceutical Sciences, University of Montana, Missoula, MT 59812, United States; Skaggs School of Pharmacy and Pharmaceutical Sciences, Anschutz Medical Campus, University of Colorado Denver, Aurora, CO 80045, United States.
| |
Collapse
|
39
|
Li T, Liu N. The Function of G-Protein-Coupled Receptor-Regulatory Cascade in Southern House Mosquitoes (Diptera: Culicidae). JOURNAL OF MEDICAL ENTOMOLOGY 2018; 55:862-870. [PMID: 29608693 PMCID: PMC6025232 DOI: 10.1093/jme/tjy022] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/24/2017] [Indexed: 05/11/2023]
Abstract
G-protein-coupled receptors (GPCRs) are a large family of seven-transmembrane domain proteins that exist in plants and animals, playing critical physiological functions through intracellular cascades. Previous studies revealed an important regulation pathway of GPCR/Guanine nucleotide-binding protein (G-protein)/Adenylyl Cyclase (AC)/cAMP-dependent protein kinase A (PKA) in the insecticide resistance and regulation of resistance-related P450 gene expression in highly resistant southern house mosquitoes, Culex quinquefasciatus Say (Diptera: Culicidae). However, the function of this regulation pathway in field-collected and laboratory-susceptible mosquitoes is still unknown. In the current study, we characterized the function of each effector (GPCR, G-protein, AC, and PKA) in this GPCR intracellular pathway in both field-collected and laboratory Cx. quinquefasciatus strains, showing that knockdown of the expression of each effector gene can cause 1) decreased expression of their downstream respective genes and 2) increased sensitivity of the mosquitoes to permethrin insecticide. These results, together with our previous findings, strongly suggest the universal function of the GPCR-regulation cascade in the mosquito's sensitivity to insecticides and its regulation of resistance development through P450-mediated detoxification.
Collapse
Affiliation(s)
- Ting Li
- Department of Entomology and Plant Pathology, Funchess Hall, Auburn University, Auburn, AL
| | - Nannan Liu
- Department of Entomology and Plant Pathology, Funchess Hall, Auburn University, Auburn, AL
- Corresponding author, e-mail:
| |
Collapse
|
40
|
A Novel G-Protein-Coupled Receptors Gene from Upland Cotton Enhances Salt Stress Tolerance in Transgenic Arabidopsis. Genes (Basel) 2018; 9:genes9040209. [PMID: 29649144 PMCID: PMC5924551 DOI: 10.3390/genes9040209] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2018] [Revised: 03/28/2018] [Accepted: 04/03/2018] [Indexed: 11/17/2022] Open
Abstract
Plants have developed a number of survival strategies which are significant for enhancing their adaptation to various biotic and abiotic stress factors. At the transcriptome level, G-protein-coupled receptors (GPCRs) are of great significance, enabling the plants to detect a wide range of endogenous and exogenous signals which are employed by the plants in regulating various responses in development and adaptation. In this research work, we carried out genome-wide analysis of target of Myb1 (TOM1), a member of the GPCR gene family. The functional role of TOM1 in salt stress tolerance was studied using a transgenic Arabidopsis plants over-expressing the gene. By the use of the functional domain PF06454, we obtained 16 TOM genes members in Gossypium hirsutum, 9 in Gossypium arboreum, and 11 in Gossypium raimondii. The genes had varying physiochemical properties, and it is significant to note that all the grand average of hydropathy (GRAVY) values were less than one, indicating that all are hydrophobic in nature. In all the genes analysed here, both the exonic and intronic regions were found. The expression level of Gh_A07G0747 (GhTOM) was significantly high in the transgenic lines as compared to the wild type; a similar trend in expression was observed in all the salt-related genes tested in this study. The study in epidermal cells confirmed the localization of the protein coded by the gene TOM1 in the plasma membrane. Analysis of anti-oxidant enzymes showed higher concentrations of antioxidants in transgenic lines and relatively lower levels of oxidant substances such as H₂O₂. The low malondialdehyde (MDA) level in transgenic lines indicated that the transgenic lines had relatively low level of oxidative damage compared to the wild types. The results obtained indicate that Gh_A07G0747 (GhTOM) can be a putative target gene for enhancing salt stress tolerance in plants and could be exploited in the future for the development of salt stress-tolerant cotton cultivars.
Collapse
|
41
|
Ok HG, Kim SY, Lee SJ, Kim TK, Huh BK, Kim KH. Can oliceridine (TRV130), an ideal novel µ receptor G protein pathway selective (µ-GPS) modulator, provide analgesia without opioid-related adverse reactions? Korean J Pain 2018; 31:73-79. [PMID: 29686804 PMCID: PMC5904350 DOI: 10.3344/kjp.2018.31.2.73] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2018] [Revised: 03/06/2018] [Accepted: 03/08/2018] [Indexed: 01/19/2023] Open
Abstract
All drugs have both favorable therapeutic and untoward adverse effects. Conventional opioid analgesics possess both analgesia and adverse reactions, such as nausea, vomiting, and respiratory depression. The opioid ligand binds to µ opioid receptor and non-selectively activates two intracellular signaling pathways: the G protein pathway induce analgesia, while the β-arrestin pathway is responsible for the opioid-related adverse reactions. An ideal opioid should activate the G protein pathway while deactivating the β-arrestin pathway. Oliceridine (TRV130) has a novel characteristic mechanism on the action of the µ receptor G protein pathway selective (µ-GPS) modulation. Even though adverse reactions (ADRs) are significantly attenuated, while the analgesic effect is augmented, the some residual ADRs persist. Consequently, a G protein biased µ opioid ligand, oliceridine, improves the therapeutic index owing to increased analgesia with decreased adverse events. This review article provides a brief history, mechanism of action, pharmacokinetics, pharmacodynamics, and ADRs of oliceridine.
Collapse
Affiliation(s)
- Hwoe Gyeong Ok
- Department of Anesthesia and Pain Medicine, School of Medicine, Pusan National University, Yangsan, Korea
| | - Su Young Kim
- Department of Anesthesia and Pain Medicine, School of Medicine, Pusan National University, Yangsan, Korea
| | - Su Jung Lee
- Department of Anesthesia and Pain Medicine, School of Medicine, Pusan National University, Yangsan, Korea
| | - Tae Kyun Kim
- Department of Anesthesia and Pain Medicine, School of Medicine, Pusan National University, Yangsan, Korea
| | - Billy K Huh
- Department of Pain Medicine, The University of Texas M.D. Anderson Cancer Center, Houston, TX, USA
| | - Kyung Hoon Kim
- Department of Anesthesia and Pain Medicine, School of Medicine, Pusan National University, Yangsan, Korea
| |
Collapse
|
42
|
Singh SS, Jois SD. Homo- and Heterodimerization of Proteins in Cell Signaling: Inhibition and Drug Design. ADVANCES IN PROTEIN CHEMISTRY AND STRUCTURAL BIOLOGY 2018; 111:1-59. [PMID: 29459028 DOI: 10.1016/bs.apcsb.2017.08.003] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Protein dimerization controls many physiological processes in the body. Proteins form homo-, hetero-, or oligomerization in the cellular environment to regulate the cellular processes. Any deregulation of these processes may result in a disease state. Protein-protein interactions (PPIs) can be inhibited by antibodies, small molecules, or peptides, and inhibition of PPI has therapeutic value. PPI drug discovery research has steadily increased in the last decade, and a few PPI inhibitors have already reached the pharmaceutical market. Several PPI inhibitors are in clinical trials. With advancements in structural and molecular biology methods, several methods are now available to study protein homo- and heterodimerization and their inhibition by drug-like molecules. Recently developed methods to study PPI such as proximity ligation assay and enzyme-fragment complementation assay that detect the PPI in the cellular environment are described with examples. At present, the methods used to design PPI inhibitors can be classified into three major groups: (1) structure-based drug design, (2) high-throughput screening, and (3) fragment-based drug design. In this chapter, we have described some of the experimental methods to study PPIs and their inhibition. Examples of homo- and heterodimers of proteins, their structural and functional aspects, and some of the inhibitors that have clinical importance are discussed. The design of PPI inhibitors of epidermal growth factor receptor heterodimers and CD2-CD58 is discussed in detail.
Collapse
Affiliation(s)
- Sitanshu S Singh
- Basic Pharmaceutical Sciences, School of Pharmacy, University of Louisiana at Monroe, Monroe, LA, United States
| | - Seetharama D Jois
- Basic Pharmaceutical Sciences, School of Pharmacy, University of Louisiana at Monroe, Monroe, LA, United States.
| |
Collapse
|
43
|
Tu J, Li JJ, Song LT, Zhai HL, Wang J, Zhang XY. Molecular modeling study on resistance of WT/D473H SMO to antagonists LDE-225 and LEQ-506. Pharmacol Res 2017; 129:491-499. [PMID: 29175550 DOI: 10.1016/j.phrs.2017.11.025] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/17/2017] [Revised: 11/17/2017] [Accepted: 11/20/2017] [Indexed: 01/28/2023]
Abstract
The smoothened (SMO) receptor, an essential signal transducer in the Hedgehog pathway, was targeted with antagonists to suppress the tumor. It is interesting that SMO D473H mutation confers resistance on inhibitor LDE-225 rather than LEQ-506. In this paper, the binding modes of them against the wild type and mutant SMO receptors were identified to gain insights into the resistant and non-resistant factors, based on a comprehensive protocol involving molecular docking, molecular dynamic simulations, free energy calculation and decomposition. A comparison of resistant LDE-225 and non-resistant LEQ-506 indicates that the volume of the binding cavity decreases seriously in the mutant complex with resistant LDE-225. In addition, the D473H mutation disrupts the hydrogen bond network with residues R400 and Q477, which results in the TM6 conformation inward. Owing to the absence of the hydrogen bond, residues R400 and Q477 make weak contributions to LDE-225. However, the D473H mutation along with TM6 conformational change has no effect on non-resistant LEQ-506. Finally, the resistance ascribes to adverse interaction between the greater polarity of mutant residue H473 and the nonpolar phenmethyl of LDE-225. The elaborate insights into structural and energetic mechanism of drug resistance provide an effective strategy to design rationally non-resistant antagonists.
Collapse
Affiliation(s)
- Jing Tu
- College of Chemistry & Chemical Engineering, Lanzhou University, Lanzhou, 730000, PR China
| | - Jiao Jiao Li
- College of Chemistry & Chemical Engineering, Lanzhou University, Lanzhou, 730000, PR China
| | - Li Ting Song
- College of Chemistry & Chemical Engineering, Lanzhou University, Lanzhou, 730000, PR China
| | - Hong Lin Zhai
- College of Chemistry & Chemical Engineering, Lanzhou University, Lanzhou, 730000, PR China.
| | - Juan Wang
- College of Chemistry & Chemical Engineering, Lanzhou University, Lanzhou, 730000, PR China
| | - Xiao Yun Zhang
- College of Chemistry & Chemical Engineering, Lanzhou University, Lanzhou, 730000, PR China
| |
Collapse
|
44
|
Heterodimerization of the prostaglandin E2 receptor EP2 and the calcitonin receptor CTR. PLoS One 2017; 12:e0187711. [PMID: 29095955 PMCID: PMC5667882 DOI: 10.1371/journal.pone.0187711] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2017] [Accepted: 10/24/2017] [Indexed: 12/14/2022] Open
Abstract
G protein-coupled receptors (GPCRs) have been found to form heterodimers and modulate or fine-tune the functions of GPCRs. However, the involvement of GPCR heterodimerization and its functional consequences in gonadal tissues, including granulosa cells, have been poorly investigated, mainly due to the lack of efficient method for identification of novel GPCR heterodimers. In this paper, we identified a novel GPCR heterodimer between prostaglandin E2 (PGE2) receptor 2 (EP2) and calcitonin (CT) receptor (CTR). High-resolution liquid chromatography (LC)-tandem mass spectrometry (MS/MS) of protease-digested EP2-coimmunoprecipitates detected protein fragments of CTR in an ovarian granulosa cell line, OV3121. Western blotting of EP2- and CTR-coimmunoprecipitates detected a specific band for EP2-CTR heterodimer. Specific heterodimerization between EP2 and CTR was also observed by fluorescence resonance energy transfer analysis in HEK293MSR cells expressing cyan- and yellow-fluorescent protein-fused EP2 and CTR, respectively. Collectively, these results provided evidence for heterodimerization between EP2 and CTR. Moreover, Ca2+ mobilization by CT was approximately 40% less potent in HEK293MSR cells expressing an EP2-CTR heterodimer, whereas cAMP production by EP2 or CT was not significantly altered compared with cells expressing EP2- or CTR alone. These functional analyses verified that CTR-mediated Ca2+ mobilization is specifically decreased via heterodimerization with EP2. Altogether, the present study suggests that a novel GPCR heterodimer, EP2-CTR, is involved in some functional regulation, and paves the way for investigation of novel biological roles of CTR and EP2 in various tissues.
Collapse
|
45
|
Venuti A, Pastori C, Lopalco L. The Role of Natural Antibodies to CC Chemokine Receptor 5 in HIV Infection. Front Immunol 2017; 8:1358. [PMID: 29163468 PMCID: PMC5670346 DOI: 10.3389/fimmu.2017.01358] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2017] [Accepted: 10/03/2017] [Indexed: 12/22/2022] Open
Abstract
The CC chemokine receptor 5 (CCR5) is responsible for immune and inflammatory responses by mediation of chemotactic activity in leukocytes, although it is expressed on different cell types. It has been shown to act as co-receptor for the human and simian immunodeficiency viruses (HIV-1, HIV-2, and SIV). Natural reactive antibodies (Abs) recognizing first loop (ECL1) of CCR5 have been detected in several pools of immunoglobulins from healthy donors and from several cohorts of either HIV-exposed but uninfected subjects (ESN) or HIV-infected individuals who control disease progression (LTNP) as well. The reason of development of anti-CCR5 Abs in the absence of autoimmune disease is still unknown; however, the presence of these Abs specific for CCR5 or for other immune receptors and mediators probably is related to homeostasis maintenance. The majority of anti-CCR5 Abs is directed to HIV binding site (N-terminus and ECL2) of the receptor. Conversely, it is well known that ECL1 of CCR5 does not bind HIV; thus, the anti-CCR5 Abs directed to ECL1 elicit a long-lasting internalization of CCR5 but not interfere with HIV binding directly; these Abs block HIV infection in either epithelial cells or CD4+ T lymphocytes and the mechanism differs from those ones described for all other CCR5-specific ligands. The Ab-mediated CCR5 internalization allows the formation of a stable signalosome by interaction of CCR5, β-arrestin2 and ERK1 proteins. The signalosome degradation and the subsequent de novo proteins synthesis determine the CCR5 reappearance on the cell membrane with a very long-lasting kinetics (8 days). The use of monoclonal Abs to CCR5 with particular characteristics and mode of action may represent a novel mode to fight viral infection in either vaccinal or therapeutic strategies.
Collapse
Affiliation(s)
- Assunta Venuti
- Division of Immunology, Transplantation and Infectious Diseases, DIBIT - San Raffaele Scientific Institute, Milan, Italy
| | - Claudia Pastori
- Division of Immunology, Transplantation and Infectious Diseases, DIBIT - San Raffaele Scientific Institute, Milan, Italy
| | - Lucia Lopalco
- Division of Immunology, Transplantation and Infectious Diseases, DIBIT - San Raffaele Scientific Institute, Milan, Italy
| |
Collapse
|
46
|
Gacasan SB, Baker DL, Parrill AL. G protein-coupled receptors: the evolution of structural insight. AIMS BIOPHYSICS 2017; 4:491-527. [PMID: 29951585 DOI: 10.3934/biophy.2017.3.491] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
G protein-coupled receptors (GPCR) comprise a diverse superfamily of over 800 proteins that have gained relevance as biological targets for pharmaceutical drug design. Although these receptors have been investigated for decades, three-dimensional structures of GPCR have only recently become available. In this review, we focus on the technological advancements that have facilitated efforts to gain insights into GPCR structure. Progress in these efforts began with the initial crystal structure determination of rhodopsin (PDB: 1F88) in 2000 and has continued to the most recently published structure of the A1AR (PDB: 5UEN) in 2017. Numerous experimental developments over the past two decades have opened the door for widespread GPCR structural characterization. These efforts have resulted in the determination of three-dimensional structures for over 40 individual GPCR family members. Herein we present a comprehensive list and comparative analysis of over 180 individual GPCR structures. This includes a summary of different GPCR functional states crystallized with agonists, dual agonists, partial agonists, inverse agonists, antagonists, and allosteric modulators.
Collapse
Affiliation(s)
- Samantha B Gacasan
- Department of Chemistry, University of Memphis, 3744 Walker Ave, Memphis, TN 38152, USA
| | - Daniel L Baker
- Department of Chemistry, University of Memphis, 3744 Walker Ave, Memphis, TN 38152, USA
| | - Abby L Parrill
- Department of Chemistry, University of Memphis, 3744 Walker Ave, Memphis, TN 38152, USA
| |
Collapse
|
47
|
Calderón-Zamora L, Ruiz-Hernandez A, Romero-Nava R, León-Sicairos N, Canizalez-Román A, Hong E, Huang F, Villafaña S. Possible involvement of orphan receptors GPR88 and GPR124 in the development of hypertension in spontaneously hypertensive rat. Clin Exp Hypertens 2017; 39:513-519. [PMID: 28678544 DOI: 10.1080/10641963.2016.1273949] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Hypertension (HBP) is a chronic disease characterized by increased blood pressure, which despite several treatments maintains a high morbi-mortality, which suggests that there are other mechanisms involved in this pathology, within which the orphan receptors could be candidates for the treatment of the HBP; these receptors are called orphan receptors because their ligand is unknown. These receptors have been suggested to participate in some pathologies because they are associated with various systems such as GPR88, which has been linked to the dopaminergic system, and GPR124 with angiogenesis, suggesting that these receptors could take part in HBP. Hence, the aim of this work was to study the expression of orphan receptors GPR88 and GPR124 in various tissues of normotensive and hypertensive rats. We used Wistar Kyoto (WKY) and spontaneously hypertensive rat (SHR) of 6-8 and 10-12 weeks of age and we determined systolic blood pressure (SBP), heart rate, as well as mRNA of GPR88 and GPR124 receptors by reverse transcription polymerase chain reaction (RT-PCR) in the aorta, heart, kidney, and brain. Our results showed that GPR88 and GPR124 were expressed in all analyzed tissues, but their expression is dependent on the age and development of HBP because their expression tends to be modified as HBP is established. Therefore, we conclude that GPR88 and GPR124 receptors may be involved in the development or maintenance of high blood pressure.
Collapse
Affiliation(s)
- L Calderón-Zamora
- a Laboratorio de Señalización Intracelular, Sección de Posgrado , Escuela Superior de Medicina del Instituto Politécnico Nacional Ciudad de México , México
| | - A Ruiz-Hernandez
- a Laboratorio de Señalización Intracelular, Sección de Posgrado , Escuela Superior de Medicina del Instituto Politécnico Nacional Ciudad de México , México
| | - R Romero-Nava
- a Laboratorio de Señalización Intracelular, Sección de Posgrado , Escuela Superior de Medicina del Instituto Politécnico Nacional Ciudad de México , México
| | - N León-Sicairos
- b CIASaP, Facultad de Medicina , Universidad Autónoma de Sinaloa Culiacán , Sinaloa , México
| | - A Canizalez-Román
- b CIASaP, Facultad de Medicina , Universidad Autónoma de Sinaloa Culiacán , Sinaloa , México
| | - E Hong
- c Departamento de Farmacobiología , Centro de Investigación y de Estudios Avanzados , Ciudad de México , México
| | - F Huang
- d Departamento de Farmacología y Toxicología , Hospital Infantil de México Federico Gómez (HIMFG) , Ciudad de México , México
| | - S Villafaña
- a Laboratorio de Señalización Intracelular, Sección de Posgrado , Escuela Superior de Medicina del Instituto Politécnico Nacional Ciudad de México , México
| |
Collapse
|
48
|
Abstract
Previously, drugs were developed focusing on target affinity and selectivity. However, it is becoming evident that the drug-target residence time, related to the off-rate, is an important parameter for successful drug development. The residence time influences both the on-rate and overall effectiveness of drugs. Furthermore, ligand binding is now appreciated to be a multistep process because metastable and/or intermediate binding sites in the extracellular region have been identified. In this review, we summarize experimental ligand-binding data for G-protein-coupled receptors (GPCRs), and their binding pathways, analyzed by molecular dynamics (MD). The kinetics of drug binding to GPCRs are complex and depend on several factors, including charge distribution on the receptor surface, ligand-receptor interactions in the binding channel and the binding site, or solvation.
Collapse
Affiliation(s)
- Andrea Strasser
- Department of Pharmaceutical/Medicinal Chemistry II, University of Regensburg, Regensburg, Germany.
| | | | - Roland Seifert
- Institute of Pharmacology, Hannover Medical School, Hannover, Germany
| |
Collapse
|
49
|
Sasaki T, Mita M, Ikari N, Kuboyama A, Hashimoto S, Kaneko T, Ishiguro M, Shimizu M, Inoue J, Sato R. Identification of key amino acid residues in the hTGR5-nomilin interaction and construction of its binding model. PLoS One 2017; 12:e0179226. [PMID: 28594916 PMCID: PMC5464637 DOI: 10.1371/journal.pone.0179226] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2017] [Accepted: 05/25/2017] [Indexed: 12/12/2022] Open
Abstract
TGR5, a member of the G protein-coupled receptor (GPCR) family, is activated by bile acids. Because TGR5 promotes energy expenditure and improves glucose homeostasis, it is recognized as a key target in treating metabolic diseases. We previously showed that nomilin, a citrus limonoid, activates TGR5 and confers anti-obesity and anti-hyperglycemic effects in mice. Information on the TGR5–nomilin interaction regarding molecular structure, however, has not been reported. In the present study, we found that human TGR5 (hTGR5) shows higher nomilin responsiveness than does mouse TGR5 (mTGR5). Using mouse–human chimeric TGR5, we also found that three amino acid residues (Q77ECL1, R80ECL1, and Y893.29) are important in the hTGR5–nomilin interaction. Based on these results, an hTGR5–nomilin binding model was constructed using in silico docking simulation, demonstrating that four hydrophilic hydrogen-bonding interactions occur between nomilin and hTGR5. The binding mode of hTGR5–nomilin is vastly different from those of other TGR5 agonists previously reported, suggesting that TGR5 forms various binding patterns depending on the type of agonist. Our study promotes a better understanding of the structure of TGR5, and it may be useful in developing and screening new TGR5 agonists.
Collapse
Affiliation(s)
- Takashi Sasaki
- Food Biochemistry Laboratory, Department of Applied Biological Chemistry, Graduate School of Agricultural and Life Sciences, University of Tokyo, Tokyo, Japan
| | - Moeko Mita
- Food Biochemistry Laboratory, Department of Applied Biological Chemistry, Graduate School of Agricultural and Life Sciences, University of Tokyo, Tokyo, Japan
| | - Naho Ikari
- Food Biochemistry Laboratory, Department of Applied Biological Chemistry, Graduate School of Agricultural and Life Sciences, University of Tokyo, Tokyo, Japan
| | - Ayane Kuboyama
- Food Biochemistry Laboratory, Department of Applied Biological Chemistry, Graduate School of Agricultural and Life Sciences, University of Tokyo, Tokyo, Japan
| | - Shuzo Hashimoto
- Food Biochemistry Laboratory, Department of Applied Biological Chemistry, Graduate School of Agricultural and Life Sciences, University of Tokyo, Tokyo, Japan
| | - Tatsuya Kaneko
- Food Biochemistry Laboratory, Department of Applied Biological Chemistry, Graduate School of Agricultural and Life Sciences, University of Tokyo, Tokyo, Japan
| | - Masaji Ishiguro
- Department of Applied Life Sciences, Niigata University of Pharmacy and Applied Life Sciences, Higashijima, Akiha-ku, Niigata, Japan
| | - Makoto Shimizu
- Food Biochemistry Laboratory, Department of Applied Biological Chemistry, Graduate School of Agricultural and Life Sciences, University of Tokyo, Tokyo, Japan
| | - Jun Inoue
- Food Biochemistry Laboratory, Department of Applied Biological Chemistry, Graduate School of Agricultural and Life Sciences, University of Tokyo, Tokyo, Japan
| | - Ryuichiro Sato
- Food Biochemistry Laboratory, Department of Applied Biological Chemistry, Graduate School of Agricultural and Life Sciences, University of Tokyo, Tokyo, Japan
- Nutri-Life Science Laboratory, Department of Applied Biological Chemistry, Graduate School of Agricultural and Life Sciences, University of Tokyo, Tokyo, Japan
- AMED-CREST, Japan Agency for Medical Research and Development, Chiyoda-ku, Tokyo, Japan
- * E-mail:
| |
Collapse
|
50
|
Yamamoto K, Suzuki T, Imamura R, Nagano T, Okabe T, Miyachi H. Synthesis of both enantiomers of 1,2,3,4-tetrahydroisoquinoline derivative IPPAM-1 and enantio-dependency of its positive allosteric modulation of prostacyclin receptor. Bioorg Med Chem Lett 2017; 27:2567-2570. [PMID: 28462839 DOI: 10.1016/j.bmcl.2017.03.083] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2017] [Revised: 03/24/2017] [Accepted: 03/27/2017] [Indexed: 02/02/2023]
Abstract
We present a practical synthesis of both enantiomers of 1,2,3,4-tetrahydroisoquinoline derivative IPPAM-1 (1), which is a positive allosteric modulator (PAM) of prostacyclin receptor (IP) and a candidate for treatment of pulmonary arterial hypertension without the side effects caused by IP agonists. Assay of cAMP production by CHO-K1 cells stably expressing human IP clearly demonstrated that the IPPAM activity resides exclusively on the R-form of 1.
Collapse
Affiliation(s)
- Kohki Yamamoto
- Graduate School of Pharmaceutical Sciences, Okayama University, 1-1-1, Tsushima-Naka, Kita-ku, Okayama 700-8530, Japan
| | - Toshifumi Suzuki
- Graduate School of Pharmaceutical Sciences, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Riyo Imamura
- Drug Discovery Initiative, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Tetsuo Nagano
- Drug Discovery Initiative, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Takayoshi Okabe
- Drug Discovery Initiative, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Hiroyuki Miyachi
- Lead Exploration Unit, Drug Discovery Initiative, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan.
| |
Collapse
|