1
|
Darsaraee M, Kaveh S, Mani-Varnosfaderani A, Neiband MS. General structure-activity/selectivity relationship patterns for the inhibitors of the chemokine receptors (CCR1/CCR2/CCR4/CCR5) with application for virtual screening of PubChem database. J Biomol Struct Dyn 2024; 42:8781-8799. [PMID: 37599469 DOI: 10.1080/07391102.2023.2248255] [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: 03/16/2023] [Accepted: 08/08/2023] [Indexed: 08/22/2023]
Abstract
CC chemokine receptors (CCRs) form a crucial subfamily of G protein-linked receptors that play a distinct role in the onset and progression of various life-threatening diseases. The main aim of this research is to derive general structure-activity relationship (SAR) patterns to describe the selectivity and activity of CCR inhibitors. To this end, a total of 7332 molecules related to the inhibition of CCR1, CCR2, CCR4, and CCR5 were collected from the Binding Database and analyzed using machine learning techniques. A diverse set of 450 molecular descriptors was calculated for each molecule, and the molecules were classified based on their therapeutic targets and activities. The variable importance in the projection (VIP) approach was used to select discriminatory molecular features, and classification models were developed using supervised Kohonen networks (SKN) and counter-propagation artificial neural networks (CPANN). The reliability and predictability of the models were estimated using 10-fold cross-validation, an external validation set, and an applicability domain approach. We were able to identify different sets of molecular descriptors for discriminating between active and inactive molecules and model the selectivity of inhibitors towards different CCRs. The sensitivities of the predictions for the external test set for the SKN models ranged from 0.827-0.873. Finally, the developed classification models were used to screen approximately 2 million random molecules from the PubChem database, with average values for areas under the receiver operating characteristic curves ranging from 0.78-0.96 for SKN models and 0.75-0.89 for CPANN models.Communicated by Ramaswamy H. Sarma.
Collapse
MESH Headings
- Structure-Activity Relationship
- Humans
- Databases, Chemical
- Receptors, CCR1/antagonists & inhibitors
- Receptors, CCR1/chemistry
- Receptors, CCR1/metabolism
- Receptors, CCR5/chemistry
- Receptors, CCR5/metabolism
- Receptors, CCR/antagonists & inhibitors
- Receptors, CCR/chemistry
- Receptors, CCR/metabolism
- Receptors, CCR2/antagonists & inhibitors
- Receptors, CCR2/chemistry
- Receptors, CCR2/metabolism
- Receptors, Chemokine/antagonists & inhibitors
- Receptors, Chemokine/chemistry
- Receptors, Chemokine/metabolism
- Models, Molecular
- Neural Networks, Computer
Collapse
Affiliation(s)
- M Darsaraee
- Chemometrics and Cheminformatics Laboratory, Department of Analytical Chemistry, Tarbiat Modares University, Tehran, Iran
| | - S Kaveh
- Chemometrics and Cheminformatics Laboratory, Department of Analytical Chemistry, Tarbiat Modares University, Tehran, Iran
| | - A Mani-Varnosfaderani
- Chemometrics and Cheminformatics Laboratory, Department of Analytical Chemistry, Tarbiat Modares University, Tehran, Iran
| | - M S Neiband
- Department of Chemistry, Payame Noor University (PNU), Tehran, Iran
| |
Collapse
|
2
|
Kulsoom K, Ali W, Saba Z, Hussain S, Zahra S, Irshad M, Ramzan MS. Revealing Melatonin's Mysteries: Receptors, Signaling Pathways, and Therapeutics Applications. Horm Metab Res 2024; 56:405-418. [PMID: 38081221 DOI: 10.1055/a-2226-3971] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/21/2024]
Abstract
Melatonin (5-methoxy-acetyl tryptamine) is a sleep-inducing hormone, and the pineal gland produces it in response to the circadian clock of darkness. In the body, MT1 and MT2 receptors are mostly found, having an orthosteric pocket and ligand binding determinants. Melatonin acts by binding on melatonin receptors, intracellular proteins, and orphan nuclear receptors. It inhibits adenyl cyclase and activates phospholipase C, resulting in gene expression and an intracellular alteration environment. Melatonin signaling pathways are also associated with other intracellular signaling pathways, i. e., cAMP/PKA and MAPK/ERK pathways. Relative expression of different proteins depends on the coupling profile of G protein, accounting pharmacology of the melatonin receptor bias system, and mediates action in a Gi-dependent manner. It shows antioxidant, antitumor, antiproliferative, and neuroprotective activity. Different types of melatonin agonists have been synthesized for the treatment of sleeping disorders. Researchers have developed therapeutics that target melatonin signaling, which could benefit a wide range of medical conditions. This review focuses on melatonin receptors, pharmacology, and signaling cascades; it aims to provide basic mechanical aspects of the receptor's pharmacology, melatonin's functions in cancer and neurodegenerative diseases, and any treatments and drugs designed for these diseases. This will allow a basic comparison between the receptors in question, highlighting any parallels and differences that may exist and providing fundamental knowledge about these receptors to future researchers.
Collapse
Affiliation(s)
- Kulsoom Kulsoom
- Department of Biochemistry, Bahauddin Zakariya University, Multan, Pakistan
| | - Wajahat Ali
- School of Basic Medical Sciences, Xi'an Jiaotong University, Xian, China
| | - Zainab Saba
- Department of Optometry, Khwaja Fareed University of Engineering & Information Technology, Rahim Yar Khan, Pakistan
| | - Shabab Hussain
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, Universita degli studi di Messina, Messina, Italy
| | - Samra Zahra
- Department of Biosciences, COMSATS University Islamabad, Pakistan
| | - Maria Irshad
- Department of Medical, Surgical and Health Sciences, University of Trieste, Trieste, Italy
| | - Muhammad Saeed Ramzan
- Department of Pharmacology, The Islamia University of Bahawalpur, Bahawalpur, Pakistan
| |
Collapse
|
3
|
da Rocha MN, da Fonseca AM, Dantas ANM, Dos Santos HS, Marinho ES, Marinho GS. In Silico Study in MPO and Molecular Docking of the Synthetic Drynaran Analogues Against the Chronic Tinnitus: Modulation of the M1 Muscarinic Acetylcholine Receptor. Mol Biotechnol 2024; 66:254-269. [PMID: 37079267 DOI: 10.1007/s12033-023-00748-5] [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: 02/17/2023] [Accepted: 04/03/2023] [Indexed: 04/21/2023]
Abstract
Tinnitus is a syndrome that affects the human auditory system and is characterized by a perception of sounds in the absence of acoustic stimuli, or in total silence. Research indicates that muscarinic acetylcholine receptors (mAChRs), especially the M1 type, have a fundamental role in the alterations of auditory perceptions of tinnitus. Here, a series of computer-aided tools were used, from molecular surface analysis software to services available on the web for estimating pharmacokinetics and pharmacodynamics. The results infer that the low lipophilicity ligands, that is, the 1a-d alkyl furans, present the best pharmacokinetic profile, as compounds with an optimal alignment between permeability and clearance. However, only ligands 1a and 1b have properties that are safe for the central nervous system, the site of cholinergic modulation. These ligands showed similarity with compounds deposited in the European Molecular Biology Laboratory chemical (ChEMBL) database acting on the mAChRs M1 type, the target selected for the molecular docking test. The simulations suggest that the 1 g ligand can form the ligand-receptor complex with the best affinity energy order and that, together with the 1b ligand, they are competitive agonists in relation to the antagonist Tiotropium, in addition to acting in synergism with the drug Bromazepam in the treatment of chronic tinnitus.
Collapse
Affiliation(s)
- Matheus Nunes da Rocha
- Graduate Program in Natural Sciences, Center for Science and Technology, State University of Ceará, Fortaleza, CE, Brazil.
| | - Aluísio Marques da Fonseca
- Institute of Engineering and Sustainable Development, Academic Master in Sociobiodiversity and Sustainable Technologies, University of International Integration of Afro-Brazilian Lusofonia, Acarape, CE, Brazil
| | | | | | - Emmanuel Silva Marinho
- Graduate Program in Natural Sciences, Center for Science and Technology, State University of Ceará, Fortaleza, CE, Brazil
- Group of Theoretical Chemistry and Electrochemistry, State University of Ceará, Limoeiro Do Norte, CE, Brazil
| | - Gabrielle Silva Marinho
- Group of Theoretical Chemistry and Electrochemistry, State University of Ceará, Limoeiro Do Norte, CE, Brazil
| |
Collapse
|
4
|
Burghi V, Paradis JS, Officer A, Adame-Garcia SR, Wu X, Matthees ESF, Barsi-Rhyne B, Ramms DJ, Clubb L, Acosta M, Tamayo P, Bouvier M, Inoue A, von Zastrow M, Hoffmann C, Gutkind JS. Gαs is dispensable for β-arrestin coupling but dictates GRK selectivity and is predominant for gene expression regulation by β2-adrenergic receptor. J Biol Chem 2023; 299:105293. [PMID: 37774973 PMCID: PMC10641165 DOI: 10.1016/j.jbc.2023.105293] [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: 02/15/2023] [Revised: 09/03/2023] [Accepted: 09/14/2023] [Indexed: 10/01/2023] Open
Abstract
β-arrestins play a key role in G protein-coupled receptor (GPCR) internalization, trafficking, and signaling. Whether β-arrestins act independently of G protein-mediated signaling has not been fully elucidated. Studies using genome-editing approaches revealed that whereas G proteins are essential for mitogen-activated protein kinase activation by GPCRs., β-arrestins play a more prominent role in signal compartmentalization. However, in the absence of G proteins, GPCRs may not activate β-arrestins, thereby limiting the ability to distinguish G protein from β-arrestin-mediated signaling events. We used β2-adrenergic receptor (β2AR) and its β2AR-C tail mutant expressed in human embryonic kidney 293 cells wildtype or CRISPR-Cas9 gene edited for Gαs, β-arrestin1/2, or GPCR kinases 2/3/5/6 in combination with arrestin conformational sensors to elucidate the interplay between Gαs and β-arrestins in controlling gene expression. We found that Gαs is not required for β2AR and β-arrestin conformational changes, β-arrestin recruitment, and receptor internalization, but that Gαs dictates the GPCR kinase isoforms involved in β-arrestin recruitment. By RNA-Seq analysis, we found that protein kinase A and mitogen-activated protein kinase gene signatures were activated by stimulation of β2AR in wildtype and β-arrestin1/2-KO cells but absent in Gαs-KO cells. These results were validated by re-expressing Gαs in the corresponding KO cells and silencing β-arrestins in wildtype cells. These findings were extended to cellular systems expressing endogenous levels of β2AR. Overall, our results support that Gs is essential for β2AR-promoted protein kinase A and mitogen-activated protein kinase gene expression signatures, whereas β-arrestins initiate signaling events modulating Gαs-driven nuclear transcriptional activity.
Collapse
Affiliation(s)
- Valeria Burghi
- Moores Cancer Center, University of California San Diego, La Jolla, California, USA; Department of Pharmacology, University of California San Diego, La Jolla, California, USA
| | - Justine S Paradis
- Moores Cancer Center, University of California San Diego, La Jolla, California, USA; Department of Pharmacology, University of California San Diego, La Jolla, California, USA
| | - Adam Officer
- Moores Cancer Center, University of California San Diego, La Jolla, California, USA; Department of Pharmacology, University of California San Diego, La Jolla, California, USA
| | - Sendi Rafael Adame-Garcia
- Moores Cancer Center, University of California San Diego, La Jolla, California, USA; Department of Pharmacology, University of California San Diego, La Jolla, California, USA
| | - Xingyu Wu
- Moores Cancer Center, University of California San Diego, La Jolla, California, USA; Department of Pharmacology, University of California San Diego, La Jolla, California, USA
| | - Edda S F Matthees
- Institut für Molekulare Zellbiologie, CMB - Center for Molecular Biomedicine, Universitätsklinikum Jena, Friedrich-Schiller-Universität Jena, Jena, Germany
| | - Benjamin Barsi-Rhyne
- Department of Pharmaceutical Chemistry, University of California San Francisco, San Francisco, California, USA
| | - Dana J Ramms
- Moores Cancer Center, University of California San Diego, La Jolla, California, USA; Department of Pharmacology, University of California San Diego, La Jolla, California, USA
| | - Lauren Clubb
- Moores Cancer Center, University of California San Diego, La Jolla, California, USA; Department of Pharmacology, University of California San Diego, La Jolla, California, USA
| | - Monica Acosta
- Moores Cancer Center, University of California San Diego, La Jolla, California, USA; Department of Pharmacology, University of California San Diego, La Jolla, California, USA
| | - Pablo Tamayo
- Moores Cancer Center, University of California San Diego, La Jolla, California, USA
| | - Michel Bouvier
- Department of Biochemistry and Molecular Medicine, Institute for Research in Immunology and Cancer, Université de Montréal, Québec, Canada
| | - Asuka Inoue
- Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Miyagi, Japan
| | - Mark von Zastrow
- Department of Psychiatry and Department of Cellular and Molecular Pharmacology, University of California San Francisco, San Francisco, California, USA
| | - Carsten Hoffmann
- Institut für Molekulare Zellbiologie, CMB - Center for Molecular Biomedicine, Universitätsklinikum Jena, Friedrich-Schiller-Universität Jena, Jena, Germany
| | - J Silvio Gutkind
- Moores Cancer Center, University of California San Diego, La Jolla, California, USA; Department of Pharmacology, University of California San Diego, La Jolla, California, USA.
| |
Collapse
|
5
|
Caniceiro AB, Bueschbell B, Schiedel AC, Moreira IS. Class A and C GPCR Dimers in Neurodegenerative Diseases. Curr Neuropharmacol 2022; 20:2081-2141. [PMID: 35339177 PMCID: PMC9886835 DOI: 10.2174/1570159x20666220327221830] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Revised: 02/21/2022] [Accepted: 03/23/2022] [Indexed: 11/22/2022] Open
Abstract
Neurodegenerative diseases affect over 30 million people worldwide with an ascending trend. Most individuals suffering from these irreversible brain damages belong to the elderly population, with onset between 50 and 60 years. Although the pathophysiology of such diseases is partially known, it remains unclear upon which point a disease turns degenerative. Moreover, current therapeutics can treat some of the symptoms but often have severe side effects and become less effective in long-term treatment. For many neurodegenerative diseases, the involvement of G proteincoupled receptors (GPCRs), which are key players of neuronal transmission and plasticity, has become clearer and holds great promise in elucidating their biological mechanism. With this review, we introduce and summarize class A and class C GPCRs, known to form heterodimers or oligomers to increase their signalling repertoire. Additionally, the examples discussed here were shown to display relevant alterations in brain signalling and had already been associated with the pathophysiology of certain neurodegenerative diseases. Lastly, we classified the heterodimers into two categories of crosstalk, positive or negative, for which there is known evidence.
Collapse
Affiliation(s)
- Ana B. Caniceiro
- Center for Neuroscience and Cell Biology, University of Coimbra, 3004-504 Coimbra, Portugal; ,These authors contributed equally to this work.
| | - Beatriz Bueschbell
- PhD Programme in Experimental Biology and Biomedicine, Institute for Interdisciplinary Research (IIIUC), University of Coimbra, Casa Costa Alemão, 3030-789 Coimbra, Portugal; ,These authors contributed equally to this work.
| | - Anke C. Schiedel
- Department of Pharmaceutical & Medicinal Chemistry, Pharmaceutical Institute, University of Bonn, D-53121 Bonn, Germany;
| | - Irina S. Moreira
- University of Coimbra, Department of Life Sciences, Calçada Martim de Freitas, 3000-456 Coimbra, Portugal; ,Center for Neuroscience and Cell Biology, Center for Innovative Biomedicine and Biotechnology, 3004-504 Coimbra, Portugal,Address correspondence to this author at the Center for Neuroscience and Cell Biology, Center for Innovative Biomedicine and Biotechnology, 3004-504 Coimbra, Portugal; E-mail:
| |
Collapse
|
6
|
Screening of the siGPCR library in combination with cisplatin against lung cancers. Sci Rep 2022; 12:17358. [PMID: 36253428 PMCID: PMC9576725 DOI: 10.1038/s41598-022-21063-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Accepted: 09/22/2022] [Indexed: 01/10/2023] Open
Abstract
The screening of siRNAs targeting 390 human G protein-coupled receptors (GPCRs) was multiplexed in combination with cisplatin against lung cancer cells. While the cell viability measure hardly captured the anticancer effect of siGPCRs, the direct cell count revealed the anticancer potential of diverse GPCRs (46 hits with > twofold growth inhibition, p-value < 0.01). In combined treatment with cisplatin, siRNAs against five genes (ADRA2A, F2RL3, NPSR1, NPY and TACR3) enhanced the anti-proliferation efficacy on cancer cells and reduced the self-recovery ability of surviving cells after the removal of the combined treatment. Further on-target validation confirmed that the knockdown of TACR3 expression exhibited anticancer efficacy under both single and combined treatment with cisplatin. Q-omics ( http://qomics.io ) analysis showed that high expression of TACR3 was unfavorable for patient survival, particularly with mutations in GPCR signaling pathways. The present screening data provide a useful resource for GPCR targets and biomarkers for improving the efficacy of cisplatin treatment.
Collapse
|
7
|
Ma X, Guo J, Fu Y, Shen C, Jiang P, Zhang Y, Zhang L, Yu Y, Fan J, Chai R. G protein-coupled receptors in cochlea: Potential therapeutic targets for hearing loss. Front Mol Neurosci 2022; 15:1028125. [PMID: 36311029 PMCID: PMC9596917 DOI: 10.3389/fnmol.2022.1028125] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Accepted: 09/21/2022] [Indexed: 11/29/2022] Open
Abstract
The prevalence of hearing loss-related diseases caused by different factors is increasing worldwide year by year. Currently, however, the patient’s hearing loss has not been effectively improved. Therefore, there is an urgent need to adopt new treatment measures and treatment techniques to help improve the therapeutic effect of hearing loss. G protein-coupled receptors (GPCRs), as crucial cell surface receptors, can widely participate in different physiological and pathological processes, particularly play an essential role in many disease occurrences and be served as promising therapeutic targets. However, no specific drugs on the market have been found to target the GPCRs of the cochlea. Interestingly, many recent studies have demonstrated that GPCRs can participate in various pathogenic process related to hearing loss in the cochlea including heredity, noise, ototoxic drugs, cochlear structure, and so on. In this review, we comprehensively summarize the functions of 53 GPCRs known in the cochlea and their relationships with hearing loss, and highlight the recent advances of new techniques used in cochlear study including cryo-EM, AI, GPCR drug screening, gene therapy vectors, and CRISPR editing technology, as well as discuss in depth the future direction of novel GPCR-based drug development and gene therapy for cochlear hearing loss. Collectively, this review is to facilitate basic and (pre-) clinical research in this area, and provide beneficial help for emerging GPCR-based cochlear therapies.
Collapse
Affiliation(s)
- Xiangyu Ma
- State Key Laboratory of Bioelectronics, Jiangsu Province High-Tech Key Laboratory for Bio-Medical Research, Department of Otolaryngology Head and Neck Surgery, Zhongda Hospital, School of Life Sciences and Technology, Advanced Institute for Life and Health, Southeast University, Nanjing, China
| | - Jiamin Guo
- State Key Laboratory of Bioelectronics, Jiangsu Province High-Tech Key Laboratory for Bio-Medical Research, Department of Otolaryngology Head and Neck Surgery, Zhongda Hospital, School of Life Sciences and Technology, Advanced Institute for Life and Health, Southeast University, Nanjing, China
| | - Yaoyang Fu
- Department of Psychiatry, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Cangsong Shen
- College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, China
| | - Pei Jiang
- State Key Laboratory of Bioelectronics, Jiangsu Province High-Tech Key Laboratory for Bio-Medical Research, Department of Otolaryngology Head and Neck Surgery, Zhongda Hospital, School of Life Sciences and Technology, Advanced Institute for Life and Health, Southeast University, Nanjing, China
| | - Yuan Zhang
- Jiangsu Provincial Key Medical Discipline (Laboratory), Department of Otolaryngology Head and Neck Surgery, Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, China
- Research Institute of Otolaryngology, Nanjing, China
| | - Lei Zhang
- Department of Otorhinolaryngology, Head and Neck Surgery, The Second Hospital of Anhui Medical University, Hefei, China
| | - Yafeng Yu
- First Affiliated Hospital of Soochow University, Soochow, China
- *Correspondence: Yafeng Yu,
| | - Jiangang Fan
- Department of Otolaryngology Head and Neck Surgery, Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu, China
- Jiangang Fan,
| | - Renjie Chai
- State Key Laboratory of Bioelectronics, Jiangsu Province High-Tech Key Laboratory for Bio-Medical Research, Department of Otolaryngology Head and Neck Surgery, Zhongda Hospital, School of Life Sciences and Technology, Advanced Institute for Life and Health, Southeast University, Nanjing, China
- Department of Otolaryngology Head and Neck Surgery, Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu, China
- Co-Innovation Center of Neuroregeneration, Nantong University, Nantong, China
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China
- Beijing Key Laboratory of Neural Regeneration and Repair, Capital Medical University, Beijing, China
- Renjie Chai,
| |
Collapse
|
8
|
Wang Y, Li M, Liang W, Shi X, Fan J, Kong R, Liu Y, Zhang J, Chen T, Lu S. Delineating the activation mechanism and conformational landscape of a class B G protein-coupled receptor glucagon receptor. Comput Struct Biotechnol J 2022; 20:628-639. [PMID: 35140883 PMCID: PMC8801358 DOI: 10.1016/j.csbj.2022.01.015] [Citation(s) in RCA: 34] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 01/17/2022] [Accepted: 01/17/2022] [Indexed: 02/09/2023] Open
Abstract
Class B G protein-coupled receptors (GPCRs) are important targets in the treatment of metabolic syndrome and diabetes. Although multiple structures of class B GPCRs-G protein complexes have been elucidated, the detailed activation mechanism of the receptors remains unclear. Here, we combine Gaussian accelerated molecular dynamics simulations and Markov state models (MSM) to investigate the activation mechanism of a canonical class B GPCR, human glucagon receptor-GCGR, including the negative allosteric modulator-bound inactive state, the agonist glucagon-bound active state, and both glucagon- and Gs-bound fully active state. The free-energy landscapes of GCGR show the conformational ensemble consisting of three activation-associated states: inactive, active, and fully active. The structural analysis indicates the high dynamics of GCGR upon glucagon binding with both active and inactive conformations in the ensemble. Significantly, the H8 and TM6 exhibits distinct features from the inactive to the active states. The additional simulations demonstrate the role of H8 in the recruitment of Gs. Gs binding presents a crucial function of stabilizing the glucagon binding site and MSM highlights the absolute requirement of Gs to help the GCGR reach the fully active state. Together, our results reveal the detailed activation mechanism of GCGR from the view of conformational dynamics.
Collapse
Affiliation(s)
- Ying Wang
- Department of Pathophysiology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University, School of Medicine, Shanghai 200025, China
- Medicinal Chemistry and Bioinformatics Center, Shanghai Jiao Tong University, School of Medicine, Shanghai 200025, China
| | - Mingyu Li
- Department of Pathophysiology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University, School of Medicine, Shanghai 200025, China
| | - Wenqi Liang
- Department of Emergency, Changhai Hospital, Naval Medical University, Shanghai 200433, China
| | - Xinchao Shi
- Department of Pathophysiology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University, School of Medicine, Shanghai 200025, China
| | - Jigang Fan
- Department of Pathophysiology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University, School of Medicine, Shanghai 200025, China
| | - Ren Kong
- Institute of Bioinformatics and Medical Engineering, School of Electrical and Information Engineering, Jiangsu University of Technology, Changzhou 213001, China
| | - Yaqin Liu
- Medicinal Chemistry and Bioinformatics Center, Shanghai Jiao Tong University, School of Medicine, Shanghai 200025, China
| | - Jian Zhang
- Department of Pathophysiology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University, School of Medicine, Shanghai 200025, China
- Medicinal Chemistry and Bioinformatics Center, Shanghai Jiao Tong University, School of Medicine, Shanghai 200025, China
| | - Ting Chen
- Department of Cardiology, Changzheng Hospital, Naval Medical University, Shanghai 200023, China
| | - Shaoyong Lu
- Department of Pathophysiology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University, School of Medicine, Shanghai 200025, China
- Medicinal Chemistry and Bioinformatics Center, Shanghai Jiao Tong University, School of Medicine, Shanghai 200025, China
| |
Collapse
|
9
|
Zhao Q, Chapman A, Huang Y, Ferguson M, McBride S, Kelly M, Weiner M, Li X. Ligand-Directed GPCR Antibody Discovery. Methods Mol Biol 2022; 2394:319-342. [PMID: 35094336 DOI: 10.1007/978-1-0716-1811-0_19] [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] [Indexed: 06/14/2023]
Abstract
Developing affinity reagents recognizing and modulating G-protein coupled receptors (GPCR) function by traditional animal immunization or in vitro screening methods is challenging. Some anti-GPCR antibodies exist on the market, but the success rate of development is still poor compared with antibodies targeting soluble or peripherally anchored proteins. More importantly, most of these antibodies do not modulate GPCR function. The current pipeline for antibody development primarily screens for overall affinity rather than functional epitope recognition. We developed a new strategy utilizing natural ligand affinity to generate a library of antibody variants with an inherent bias toward the active site of the GPCR. Instead of using phage libraries displaying antibodies with random CDR sequences at polymorphism sites observed in natural immune repertoire sequences, we generated focused antibody libraries with a natural ligand encoded within or conjugated to one of the CDRs or the N-terminus. To tailor antibody binding to the active site, we limited the sequence randomization of the antibody in regions holstering the ligand while leaving the ligand-carrying part unaltered in the first round of randomization. With hits from the successful first round, the second round of randomization of the ligand-carrying part was then performed to eliminate the bias of the ligand. Based on our results on three different GPCR targets, the proposed pipeline will enable the rapid generation of functional antibodies (both agonists and antagonists) against high-value targets with poor function epitope exposures including GPCR, channels, transporters as well as cell surface targets whose binding site is heavily masked by glycosylation.
Collapse
Affiliation(s)
- Qi Zhao
- Abcam plc, Branford, CT, USA.
| | | | | | | | | | | | | | | |
Collapse
|
10
|
Michael E, Covic L, Kuliopulos A. Lipopeptide Pepducins as Therapeutic Agents. Methods Mol Biol 2021; 2383:307-333. [PMID: 34766299 DOI: 10.1007/978-1-0716-1752-6_21] [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] [Indexed: 03/29/2023]
Abstract
Pepducins are lipidated peptides that target the intracellular loops of G protein-coupled receptors (GPCRs) in order to modulate transmembrane signaling to internally located effectors. With a wide array of potential activities ranging from partial, biased, or full agonism to antagonism, pepducins represent a versatile class of compounds that can be used to potentially treat diverse human diseases or be employed as novel tools to probe complex mechanisms of receptor activation and signaling in cells and in animals. Here, we describe a number of different pepducins including an advanced compound, PZ-128, that has successfully progressed through phase 2 clinical trials in cardiac patients demonstrating safety and efficacy in suppressing myonecrosis and arterial thrombosis.
Collapse
Affiliation(s)
- Emily Michael
- Center of Hemostasis and Thrombosis Research, Division of Hematology-Oncology, Department of Medicine, Tufts Medical Center, Tufts University School of Medicine, Boston, MA, USA
| | - Lidija Covic
- Center of Hemostasis and Thrombosis Research, Division of Hematology-Oncology, Department of Medicine, Tufts Medical Center, Tufts University School of Medicine, Boston, MA, USA
| | - Athan Kuliopulos
- Center of Hemostasis and Thrombosis Research, Division of Hematology-Oncology, Department of Medicine, Tufts Medical Center, Tufts University School of Medicine, Boston, MA, USA.
| |
Collapse
|
11
|
Sompel K, Elango A, Smith AJ, Tennis MA. Cancer chemoprevention through Frizzled receptors and EMT. Discov Oncol 2021; 12:32. [PMID: 34604862 PMCID: PMC8429367 DOI: 10.1007/s12672-021-00429-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Accepted: 08/31/2021] [Indexed: 12/11/2022] Open
Abstract
Frizzled (FZD) transmembrane receptors are well known for their role in β-catenin signaling and development and now understanding of their role in the context of cancer is growing. FZDs are often associated with the process of epithelial to mesenchymal transition (EMT) through β-catenin, but some also influence EMT through non-canonical pathways. With ten different FZDs, there is a wide range of activity from oncogenic to tumor suppressive depending on the tissue context. Alterations in FZD signaling can occur during development of premalignant lesions, supporting their potential as targets of chemoprevention agents. Agonizing or antagonizing FZD activity may affect EMT, which is a key process in lesion progression often targeted by chemoprevention agents. Recent studies identified a specific FZD as important for activity of an EMT inhibiting chemopreventive agent and other studies have highlighted the previously unrecognized potential for targeting small molecules to FZD receptors. This work demonstrates the value of investigating FZDs in chemoprevention and here we provide a review of FZDs in cancer EMT and their potential as chemoprevention targets.
Collapse
Affiliation(s)
- K. Sompel
- Division of Pulmonary Sciences and Critical Care Medicine, School of Medicine, University of Colorado Anschutz Medical Campus, 12700 E 19th AVE, RC2 Box C272, Aurora, CO 80045 USA
| | - A. Elango
- Division of Pulmonary Sciences and Critical Care Medicine, School of Medicine, University of Colorado Anschutz Medical Campus, 12700 E 19th AVE, RC2 Box C272, Aurora, CO 80045 USA
| | - A. J. Smith
- Division of Pulmonary Sciences and Critical Care Medicine, School of Medicine, University of Colorado Anschutz Medical Campus, 12700 E 19th AVE, RC2 Box C272, Aurora, CO 80045 USA
| | - M. A. Tennis
- Division of Pulmonary Sciences and Critical Care Medicine, School of Medicine, University of Colorado Anschutz Medical Campus, 12700 E 19th AVE, RC2 Box C272, Aurora, CO 80045 USA
| |
Collapse
|
12
|
Zhang S, Li L, Li H. Role of ectopic olfactory receptors in glucose and lipid metabolism. Br J Pharmacol 2021; 178:4792-4807. [PMID: 34411276 DOI: 10.1111/bph.15666] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2020] [Revised: 05/25/2021] [Accepted: 08/11/2021] [Indexed: 12/25/2022] Open
Abstract
The metabolic syndrome has become one of the major public health challenges in the world, and adjusting glucose and lipid levels to their normal values is crucial for treating the metabolic syndrome. Olfactory receptors (ORs) expressed in extra-nasal tissues participate in diverse biological processes, including the regulation of glucose and lipid metabolism. Ectopic ORs can regulate a variety of metabolic events including insulin secretion, glucagon secretion, fatty acid oxidation, lipogenesis and thermogenesis. Understanding the physiological function and deciphering the olfactory recognition code by suitable ligands make ectopic ORs potential targets for the treatment of the metabolic syndrome. In this review, we delineate the roles and mechanisms of ectopic ORs in the regulation of glucose and lipid metabolism, summarize the corresponding natural ligands, and discuss existing problems and the therapeutic potential of targeting ORs in the metabolic syndrome.
Collapse
Affiliation(s)
- Siyu Zhang
- Institute of Pharmacology, Department of Pharmaceutical Sciences, Zhejiang University of Technology, Hangzhou, China
| | - Linghuan Li
- Institute of Pharmacology, Department of Pharmaceutical Sciences, Zhejiang University of Technology, Hangzhou, China
| | - Hanbing Li
- Institute of Pharmacology, Department of Pharmaceutical Sciences, Zhejiang University of Technology, Hangzhou, China.,Section of Endocrinology, School of Medicine, Yale University, New Haven, Connecticut, USA
| |
Collapse
|
13
|
Jabeen A, Vijayram R, Ranganathan S. BIO-GATS: A Tool for Automated GPCR Template Selection Through a Biophysical Approach for Homology Modeling. Front Mol Biosci 2021; 8:617176. [PMID: 33898512 PMCID: PMC8059640 DOI: 10.3389/fmolb.2021.617176] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2020] [Accepted: 02/24/2021] [Indexed: 11/13/2022] Open
Abstract
G protein-coupled receptors (GPCRs) are the largest family of membrane proteins with more than 800 members. GPCRs are involved in numerous physiological functions within the human body and are the target of more than 30% of the United States Food and Drug Administration (FDA) approved drugs. At present, over 400 experimental GPCR structures are available in the Protein Data Bank (PDB) representing 76 unique receptors. The absence of an experimental structure for the majority of GPCRs demand homology models for structure-based drug discovery workflows. The generation of good homology models requires appropriate templates. The commonly used methods for template selection are based on sequence identity. However, there exists low sequence identity among the GPCRs. Sequences with similar patterns of hydrophobic residues are often structural homologs, even with low sequence identity. Extending this, we propose a biophysical approach for template selection based principally on hydrophobicity correspondence between the target and the template. Our approach takes into consideration other relevant parameters, including resolution, similarity within the orthosteric binding pocket of GPCRs, and structure completeness, for template selection. The proposed method was implemented in the form of a free tool called Bio-GATS, to provide the user with easy selection of the appropriate template for a query GPCR sequence. Bio-GATS was successfully validated with recent published benchmarking datasets. An application to an olfactory receptor to select an appropriate template has also been provided as a case study.
Collapse
Affiliation(s)
- Amara Jabeen
- Department of Molecular Sciences, Macquarie University, Sydney, NSW, Australia
| | - Ramya Vijayram
- Department of Biotechnology, Bhupat and Jyoti Mehta School of Biosciences, Indian Institute of Technology Madras, Chennai, India
| | - Shoba Ranganathan
- Department of Molecular Sciences, Macquarie University, Sydney, NSW, Australia
| |
Collapse
|
14
|
Ma W, Wang C, Liu R, Wang N, Lv Y, Dai B, He L. Advances in cell membrane chromatography. J Chromatogr A 2021; 1639:461916. [PMID: 33548663 DOI: 10.1016/j.chroma.2021.461916] [Citation(s) in RCA: 49] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Revised: 01/11/2021] [Accepted: 01/15/2021] [Indexed: 12/14/2022]
Abstract
Cell membrane chromatography (CMC) is a biomimetic chromatographic method based on the ability of membrane receptors to selectively interact with their ligands in vivo. Using membrane receptors as a stationary phase, the CMC method helps in determining the binding characteristics between ligands and membrane receptors and in efficiently identifying specific target components in a complex sample that produce the cellular biological effects of ligands (drugs, antibodies, enzymes, cytokines, etc.). CMC is an analytical tool for revealing characteristics of ligand-receptor interactions, screening and discovering target substances, and accurately controlling the quality of drugs. Since establishment of CMC in the early 1990s, with the rapid development of cell biology, significant progress has been made in the development of high-expression receptors, engineered cell cultures, and standardized preparations, which allowed in vitro immobilization of cell membrane receptors and miniaturization of binding assays. A variety of CMC models have been established using different membrane receptors as a stationary phase, and many new methods have been developed by combining CMC with high-performance liquid chromatography (HPLC)/mass spectrometry or HPLC-IT-TOF technologies. CMC methods have been widely used to study drug-receptor interactions and to screen complex samples for effective or harmful components.
Collapse
Affiliation(s)
- Weina Ma
- School of Pharmacy, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China; Institute of Vascular Materia Medica, Xi'an Jiaotong University, Xi'an, Shaanxi 710116, China
| | - Cheng Wang
- School of Pharmacy, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China; Institute of Vascular Materia Medica, Xi'an Jiaotong University, Xi'an, Shaanxi 710116, China
| | - Rui Liu
- School of Pharmacy, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China; Institute of Vascular Materia Medica, Xi'an Jiaotong University, Xi'an, Shaanxi 710116, China
| | - Nan Wang
- School of Pharmacy, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China; Institute of Vascular Materia Medica, Xi'an Jiaotong University, Xi'an, Shaanxi 710116, China
| | - Yanni Lv
- School of Pharmacy, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China; Institute of Vascular Materia Medica, Xi'an Jiaotong University, Xi'an, Shaanxi 710116, China
| | - Bingling Dai
- School of Pharmacy, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China; Institute of Vascular Materia Medica, Xi'an Jiaotong University, Xi'an, Shaanxi 710116, China
| | - Langchong He
- School of Pharmacy, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China; Institute of Vascular Materia Medica, Xi'an Jiaotong University, Xi'an, Shaanxi 710116, China.
| |
Collapse
|
15
|
Neuropeptide receptor genes GHSR and NMUR1 are candidate epigenetic biomarkers and predictors for surgically treated patients with oropharyngeal cancer. Sci Rep 2020; 10:1007. [PMID: 31974445 PMCID: PMC6978330 DOI: 10.1038/s41598-020-57920-z] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2019] [Accepted: 01/08/2020] [Indexed: 12/20/2022] Open
Abstract
Pathological staging and histological grading systems are useful, but imperfect, predictors of recurrence in head and neck squamous cell carcinoma (HNSCC). Aberrant promoter methylation is the main type of epigenetic modification that plays a role in the inactivation of tumor suppressor genes. To identify new potential prognostic markers, we investigated the promoter methylation status of five neuropeptide receptor genes. The methylation status of the target genes was compared with clinical characteristics in 278 cases; 72 hypopharyngeal cancers, 54 laryngeal cancers, 75 oropharyngeal cancers, and 77 oral cavity cancers were studied. We found that the NTSR1, NTSR2, GHSR, MLNR, and NMUR1 promoters were methylated in 47.8%, 46.8%, 54.3%, 39.2%, and 43.5% of the samples, respectively. GHSR and NMUR1 promoter methylation independently predicted recurrence in HNSCC. In patients with oropharyngeal cancer (n = 75), GHSR and NMUR1 promoter methylation significantly correlates with survival in surgically treated patients. We classified our patients as having a low, intermediate, or high-risk of death based on three factors: HPV status, and GHSR and NMUR1 promoter methylation. The disease-free survival (DFS) rates were 87.1%, 42.7%, and 17.0%, respectively. Combined data analysis of the methylation status of ten-eleven translocation (TET) family genes indicated a trend toward greater methylation indices as the number of TET methylation events increased. In the current study, we presented the relationship between the methylation status of the GHSR and NMUR1 genes and recurrence in HNSCC, specifically in risk classification of oropharyngeal carcinomas cases with HPV status.
Collapse
|