1
|
Su X, Luo Y, Wang Y, Qu P, Liu J, Han S, Ma C, Deng S, Liang Q, Qi X, Cheng P, Hou L. A select inhibitor of MORC2 encapsulated by chimeric membranecoated DNA nanocage target alleviation TNBC progression. Mater Today Bio 2025; 31:101497. [PMID: 39906202 PMCID: PMC11791359 DOI: 10.1016/j.mtbio.2025.101497] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2024] [Revised: 01/02/2025] [Accepted: 01/16/2025] [Indexed: 02/06/2025] Open
Abstract
Triple-negative breast cancer (TNBC) is the most malignant type of breast cancer and lacks effective targeted therapeutic drugs, resulting in a high recurrence rate and worse outcome. In this study, bioinformatic analysis and a series of experiments demonstrated that MOCR2 was highly expressed in TNBC and closely associated with poor prognosis, indicating that MOCR2 may be a potential therapeutic target for TNBC. Subsequently, Angoline was identified as an inhibitor of MORC2 protein by high-throughput screening and can significantly kill the TNBC cells by blocking cell cycle and inducing apoptosis. Furthermore, the biomimetic nanodrug delivery system (PMD) was designed by encapsulating tetrahedral DNA nanostructures with biomimetic cell membrane, and it can efficiently evade the phagocytosis of immune system and target TNBC tissue. Additionally, PMD can markedly enhance the killing effect of Angoline on TNBC tumors. Therefore, PMD-enveloped Angoline provide a highly effective targeted therapeutic regimen for TNBC and may improve the outcome for patients with TNBC.
Collapse
Affiliation(s)
- Xiaohan Su
- Breast Surgery, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
- Department of Breast Surgery, Mianyang 404 hospital, Mianyang, China
| | - Yunbo Luo
- Breast Surgery, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
- Department of Breast and Thyroid Surgery, Biological Targeting Laboratory of Breast Cancer, Academician (expert) Workstation, Affiliated Hospital of North Sichuan Medical College, Nanchong, China
| | - Yali Wang
- Department of Breast and Thyroid Surgery, Biological Targeting Laboratory of Breast Cancer, Academician (expert) Workstation, Affiliated Hospital of North Sichuan Medical College, Nanchong, China
| | - Peng Qu
- Department of Laboratory Medicine, Affiliated Hospital of North Sichuan Medical College, Nanchong, China
| | - Jun Liu
- Institute of Cardiovascular Diseases & Department of Cardiology, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Shiqi Han
- Department of Breast and Thyroid Surgery, Biological Targeting Laboratory of Breast Cancer, Academician (expert) Workstation, Affiliated Hospital of North Sichuan Medical College, Nanchong, China
| | - Cui Ma
- Department of Mathematics, Army Medical University, Chongqing, China
| | - Shishan Deng
- Department of Breast and Thyroid Surgery, Biological Targeting Laboratory of Breast Cancer, Academician (expert) Workstation, Affiliated Hospital of North Sichuan Medical College, Nanchong, China
| | - Qi Liang
- Department of Laboratory Medicine, Affiliated Hospital of North Sichuan Medical College, Nanchong, China
| | - Xiaowei Qi
- Department of Breast Surgery, Southwest Hospital, Army Medical University, Chongqing, China
| | - Panke Cheng
- Institute of Cardiovascular Diseases & Department of Cardiology, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
- Ultrasound in Cardiac Electrophysiology and Biomechanics Key Laboratory of Sichuan Province, Chengdu, China
| | - Lingmi Hou
- Breast Surgery, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| |
Collapse
|
2
|
Hashem S, Dougha A, Tufféry P. Ligand-Induced Biased Activation of GPCRs: Recent Advances and New Directions from In Silico Approaches. Molecules 2025; 30:1047. [PMID: 40076272 PMCID: PMC11901715 DOI: 10.3390/molecules30051047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2025] [Revised: 02/21/2025] [Accepted: 02/23/2025] [Indexed: 03/14/2025] Open
Abstract
G-protein coupled receptors (GPCRs) are the largest family of membrane proteins engaged in transducing signals from the extracellular environment into the cell. GPCR-biased signaling occurs when two different ligands, sharing the same binding site, induce distinct signaling pathways. This selective signaling offers significant potential for the design of safer and more effective drugs. Although its molecular mechanism remains elusive, big efforts are made to try to explain this mechanism using a wide range of methods. Recent advances in computational techniques and AI technology have introduced a variety of simulations and machine learning tools that facilitate the modeling of GPCR signal transmission and the analysis of ligand-induced biased signaling. In this review, we present the current state of in silico approaches to elucidate the structural mechanism of GPCR-biased signaling. This includes molecular dynamics simulations that capture the main interactions causing the bias. We also highlight the major contributions and impacts of transmembrane domains, loops, and mutations in mediating biased signaling. Moreover, we discuss the impact of machine learning models on bias prediction and diffusion-based generative AI to design biased ligands. Ultimately, this review addresses the future directions for studying the biased signaling problem through AI approaches.
Collapse
Affiliation(s)
| | | | - Pierre Tufféry
- Unité de Biologie Fonctionnelle et Adaptative, INSERM ERL 1133, CNRS UMR 8251, Université Paris Cité, F-75013 Paris, France; (S.H.); (A.D.)
| |
Collapse
|
3
|
Brahma R, Moon S, Shin JM, Cho KH. AiGPro: a multi-tasks model for profiling of GPCRs for agonist and antagonist. J Cheminform 2025; 17:12. [PMID: 39881398 PMCID: PMC11780767 DOI: 10.1186/s13321-024-00945-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2024] [Accepted: 12/27/2024] [Indexed: 01/31/2025] Open
Abstract
G protein-coupled receptors (GPCRs) play vital roles in various physiological processes, making them attractive drug discovery targets. Meanwhile, deep learning techniques have revolutionized drug discovery by facilitating efficient tools for expediting the identification and optimization of ligands. However, existing models for the GPCRs often focus on single-target or a small subset of GPCRs or employ binary classification, constraining their applicability for high throughput virtual screening. To address these issues, we introduce AiGPro, a novel multitask model designed to predict small molecule agonists (EC50) and antagonists (IC50) across the 231 human GPCRs, making it a first-in-class solution for large-scale GPCR profiling. Leveraging multi-scale context aggregation and bidirectional multi-head cross-attention mechanisms, our approach demonstrates that ensemble models may not be necessary for predicting complex GPCR states and small molecule interactions. Through extensive validation using stratified tenfold cross-validation, AiGPro achieves robust performance with Pearson's correlation coefficient of 0.91, indicating broad generalizability. This breakthrough sets a new standard in the GPCR studies, outperforming previous studies. Moreover, our first-in-class multi-tasking model can predict agonist and antagonist activities across a wide range of GPCRs, offering a comprehensive perspective on ligand bioactivity within this diverse superfamily. To facilitate easy accessibility, we have deployed a web-based platform for model access at https://aicadd.ssu.ac.kr/AiGPro . Scientific Contribution We introduce a deep learning-based multi-task model to generalize the agonist and antagonist bioactivity prediction for GPCRs accurately. The model is implemented on a user-friendly web server to facilitate rapid screening of small-molecule libraries, expediting GPCR-targeted drug discovery. Covering a diverse set of 231 GPCR targets, the platform delivers a robust, scalable solution for advancing GPCR-focused therapeutic development. The proposed framework incorporates an innovative dual-label prediction strategy, enabling the simultaneous classification of molecules as agonists, antagonists, or both. Each prediction is further accompanied by a confidence score, offering a quantitative measure of activity likelihood. This advancement moves beyond conventional models focusing solely on binding affinity, providing a more comprehensive understanding of ligand-receptor interactions. At the core of our model lies the Bi-Directional Multi-Head Cross-Attention (BMCA) module, a novel architecture that captures forward and backward contextual embeddings of protein and ligand features. By leveraging BMCA, the model effectively integrates structural and sequence-level information, ensuring a precise representation of molecular interactions. Results show that this approach is highly accurate in binding affinity predictions and consistent across diverse GPCR families. By unifying agonist and antagonist bioactivity prediction into a single model architecture, we bridge a critical gap in GPCR modeling. This enhances prediction accuracy and accelerates virtual screening workflows, offering a valuable and innovative solution for advancing GPCR-targeted drug discovery.
Collapse
Affiliation(s)
- Rahul Brahma
- School of Systems Biomedical Science, Soongsil University, 369 Sangdo-ro, Dongjak-gu, 06978, Seoul, Republic of Korea
| | - Sunghyun Moon
- School of Systems Biomedical Science, Soongsil University, 369 Sangdo-ro, Dongjak-gu, 06978, Seoul, Republic of Korea
| | - Jae-Min Shin
- AzothBio, Rm. DA724 Hyundai Knowledge Industry Center, Hanam-si, Gyeonggi-do, Republic of Korea.
| | - Kwang-Hwi Cho
- School of Systems Biomedical Science, Soongsil University, 369 Sangdo-ro, Dongjak-gu, 06978, Seoul, Republic of Korea.
| |
Collapse
|
4
|
Sherif ZA, Ogunwobi OO, Ressom HW. Mechanisms and technologies in cancer epigenetics. Front Oncol 2025; 14:1513654. [PMID: 39839798 PMCID: PMC11746123 DOI: 10.3389/fonc.2024.1513654] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2024] [Accepted: 12/04/2024] [Indexed: 01/23/2025] Open
Abstract
Cancer's epigenetic landscape, a labyrinthine tapestry of molecular modifications, has long captivated researchers with its profound influence on gene expression and cellular fate. This review discusses the intricate mechanisms underlying cancer epigenetics, unraveling the complex interplay between DNA methylation, histone modifications, chromatin remodeling, and non-coding RNAs. We navigate through the tumultuous seas of epigenetic dysregulation, exploring how these processes conspire to silence tumor suppressors and unleash oncogenic potential. The narrative pivots to cutting-edge technologies, revolutionizing our ability to decode the epigenome. From the granular insights of single-cell epigenomics to the holistic view offered by multi-omics approaches, we examine how these tools are reshaping our understanding of tumor heterogeneity and evolution. The review also highlights emerging techniques, such as spatial epigenomics and long-read sequencing, which promise to unveil the hidden dimensions of epigenetic regulation. Finally, we probed the transformative potential of CRISPR-based epigenome editing and computational analysis to transmute raw data into biological insights. This study seeks to synthesize a comprehensive yet nuanced understanding of the contemporary landscape and future directions of cancer epigenetic research.
Collapse
Affiliation(s)
- Zaki A. Sherif
- Department of Biochemistry & Molecular Biology, Howard University College of Medicine, Washington, DC, United States
| | - Olorunseun O. Ogunwobi
- Department of Biochemistry & Molecular Biology, Michigan State University, East Lansing, MI, United States
| | - Habtom W. Ressom
- Department of Oncology, Georgetown University Medical Center, Washington, DC, United States
| |
Collapse
|
5
|
Caniceiro AB, Orzeł U, Rosário-Ferreira N, Filipek S, Moreira IS. Leveraging Artificial Intelligence in GPCR Activation Studies: Computational Prediction Methods as Key Drivers of Knowledge. Methods Mol Biol 2025; 2870:183-220. [PMID: 39543036 DOI: 10.1007/978-1-0716-4213-9_10] [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: 11/17/2024]
Abstract
G protein-coupled receptors (GPCRs) are key molecules involved in cellular signaling and are attractive targets for pharmacological intervention. This chapter is designed to explore the range of algorithms used to predict GPCRs' activation states, while also examining the pharmaceutical implications of these predictions. Our primary objective is to show how artificial intelligence (AI) is key in GPCR research to reveal the intricate dynamics of activation and inactivation processes, shedding light on the complex regulatory mechanisms of this vital protein family. We describe several computational strategies that leverage diverse structural data from the Protein Data Bank, molecular dynamic simulations, or ligand-based methods to predict the activation states of GPCRs. We demonstrate how the integration of AI into GPCR research not only enhances our understanding of their dynamic properties but also presents immense potential for driving pharmaceutical research and development, offering promising new avenues in the search for newer, better therapeutic agents.
Collapse
Affiliation(s)
- Ana B Caniceiro
- Department of Life Sciences, University of Coimbra, Coimbra, Portugal
- CNC-UC - Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
| | - Urszula Orzeł
- Department of Life Sciences, University of Coimbra, Coimbra, Portugal
- CNC-UC - Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
- Faculty of Chemistry, Biological and Chemical Research Centre, University of Warsaw, Warsaw, Poland
| | - Nícia Rosário-Ferreira
- CNC-UC - Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
- CIBB - Centre for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal
| | - Sławomir Filipek
- Faculty of Chemistry, Biological and Chemical Research Centre, University of Warsaw, Warsaw, Poland
| | - Irina S Moreira
- Department of Life Sciences, University of Coimbra, Coimbra, Portugal.
- CNC-UC - Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal.
- CIBB - Centre for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal.
| |
Collapse
|
6
|
Galdino GT, Mailhot O, Najmanovich R. Understanding and Predicting Ligand Efficacy in the μ-Opioid Receptor through Quantitative Dynamical Analysis of Complex Structures. J Chem Inf Model 2024; 64:8549-8561. [PMID: 39496284 DOI: 10.1021/acs.jcim.4c00788] [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: 11/06/2024]
Abstract
The μ-opioid receptor (MOR) is a G-protein coupled receptor involved in nociception and the primary target of opioid drugs. Understanding the relationships among the ligand structure, receptor dynamics, and efficacy in activating MOR is crucial for drug discovery and development. Here, we use coarse-grained normal-mode analysis to predict ligand-induced changes in receptor dynamics with the Quantitative Dynamics Activity Relationship (QDAR) DynaSig-ML methodology, training a LASSO regression model on the entropic signatures (ESs) computed from ligand-receptor complexes. We train and validate the methodology using a data set of 179 MOR ligands with experimentally measured efficacies split into strictly chemically different cross-validation sets. By analyzing the coefficients of the ES LASSO model, we identified key residues involved in MOR activation, several of which have mutational data supporting their role in MOR activation. Additionally, we explored a contact-only LASSO model based on ligand-protein interactions. While the model showed predictive power, it failed at predicting efficacy for ligands with low structural similarity to the training set, emphasizing the importance of receptor dynamics for predicting ligand-induced receptor activation. Moreover, the low computational cost of our approach, at 3 CPU s per ligand-receptor complex, opens the door to its application in large-scale virtual screening contexts. Our work contributes to a better understanding of dynamics-function relationships in the μ-opioid receptor and provides a framework for predicting ligand efficacy based on ligand-induced changes in receptor dynamics.
Collapse
Affiliation(s)
- Gabriel T Galdino
- Department of Pharmacology and Physiology Faculty of Medicine, University of Montreal, 2960 Chemin de la Tour, H3T 1J4 Montréal, Quebec, Canada
| | - Olivier Mailhot
- Department of Pharmacology and Physiology Faculty of Medicine, University of Montreal, 2960 Chemin de la Tour, H3T 1J4 Montréal, Quebec, Canada
| | - Rafael Najmanovich
- Department of Pharmacology and Physiology Faculty of Medicine, University of Montreal, Room 3147, Pavillon Paul-G.-Desmarais 2960 Chemin de la Tour, H3T 1J4 Montréal, Quebec, Canada
| |
Collapse
|
7
|
Li Z, Huang R, Xia M, Patterson TA, Hong H. Fingerprinting Interactions between Proteins and Ligands for Facilitating Machine Learning in Drug Discovery. Biomolecules 2024; 14:72. [PMID: 38254672 PMCID: PMC10813698 DOI: 10.3390/biom14010072] [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: 11/02/2023] [Revised: 12/26/2023] [Accepted: 12/28/2023] [Indexed: 01/24/2024] Open
Abstract
Molecular recognition is fundamental in biology, underpinning intricate processes through specific protein-ligand interactions. This understanding is pivotal in drug discovery, yet traditional experimental methods face limitations in exploring the vast chemical space. Computational approaches, notably quantitative structure-activity/property relationship analysis, have gained prominence. Molecular fingerprints encode molecular structures and serve as property profiles, which are essential in drug discovery. While two-dimensional (2D) fingerprints are commonly used, three-dimensional (3D) structural interaction fingerprints offer enhanced structural features specific to target proteins. Machine learning models trained on interaction fingerprints enable precise binding prediction. Recent focus has shifted to structure-based predictive modeling, with machine-learning scoring functions excelling due to feature engineering guided by key interactions. Notably, 3D interaction fingerprints are gaining ground due to their robustness. Various structural interaction fingerprints have been developed and used in drug discovery, each with unique capabilities. This review recapitulates the developed structural interaction fingerprints and provides two case studies to illustrate the power of interaction fingerprint-driven machine learning. The first elucidates structure-activity relationships in β2 adrenoceptor ligands, demonstrating the ability to differentiate agonists and antagonists. The second employs a retrosynthesis-based pre-trained molecular representation to predict protein-ligand dissociation rates, offering insights into binding kinetics. Despite remarkable progress, challenges persist in interpreting complex machine learning models built on 3D fingerprints, emphasizing the need for strategies to make predictions interpretable. Binding site plasticity and induced fit effects pose additional complexities. Interaction fingerprints are promising but require continued research to harness their full potential.
Collapse
Affiliation(s)
- Zoe Li
- National Center for Toxicological Research, US Food and Drug Administration, Jefferson, AR 72079, USA; (Z.L.); (T.A.P.)
| | - Ruili Huang
- National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, MD 20892, USA; (R.H.); (M.X.)
| | - Menghang Xia
- National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, MD 20892, USA; (R.H.); (M.X.)
| | - Tucker A. Patterson
- National Center for Toxicological Research, US Food and Drug Administration, Jefferson, AR 72079, USA; (Z.L.); (T.A.P.)
| | - Huixiao Hong
- National Center for Toxicological Research, US Food and Drug Administration, Jefferson, AR 72079, USA; (Z.L.); (T.A.P.)
| |
Collapse
|
8
|
Goßen J, Ribeiro RP, Bier D, Neumaier B, Carloni P, Giorgetti A, Rossetti G. AI-based identification of therapeutic agents targeting GPCRs: introducing ligand type classifiers and systems biology. Chem Sci 2023; 14:8651-8661. [PMID: 37592985 PMCID: PMC10430665 DOI: 10.1039/d3sc02352d] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2023] [Accepted: 07/20/2023] [Indexed: 08/19/2023] Open
Abstract
Identifying ligands targeting G protein coupled receptors (GPCRs) with novel chemotypes other than the physiological ligands is a challenge for in silico screening campaigns. Here we present an approach that identifies novel chemotype ligands by combining structural data with a random forest agonist/antagonist classifier and a signal-transduction kinetic model. As a test case, we apply this approach to identify novel antagonists of the human adenosine transmembrane receptor type 2A, an attractive target against Parkinson's disease and cancer. The identified antagonists were tested here in a radio ligand binding assay. Among those, we found a promising ligand whose chemotype differs significantly from all so-far reported antagonists, with a binding affinity of 310 ± 23.4 nM. Thus, our protocol emerges as a powerful approach to identify promising ligand candidates with novel chemotypes while preserving antagonistic potential and affinity in the nanomolar range.
Collapse
Affiliation(s)
- Jonas Goßen
- Institute for Computational Biomedicine (INM-9/IAS-5) Forschungszentrum Jülich Wilhelm-Johnen-Straße 52428 Jülich Germany
- Faculty of Mathematics, Computer Science and Natural Sciences RWTH Aachen University Aachen Germany
| | - Rui Pedro Ribeiro
- Institute for Computational Biomedicine (INM-9/IAS-5) Forschungszentrum Jülich Wilhelm-Johnen-Straße 52428 Jülich Germany
| | - Dirk Bier
- Institute of Neuroscience and Medicine, Nuclear Chemistry (INM-5), Forschungszentrum Jülich GmbH Wilhelm-Johnen-Straße 52428 Jülich Germany
| | - Bernd Neumaier
- Institute of Neuroscience and Medicine, Nuclear Chemistry (INM-5), Forschungszentrum Jülich GmbH Wilhelm-Johnen-Straße 52428 Jülich Germany
- Institute of Radiochemistry and Experimental Molecular Imaging, University of Cologne, Faculty of Medicine and University Hospital Cologne Kerpener Straße 62 50937 Cologne Germany
| | - Paolo Carloni
- Institute for Computational Biomedicine (INM-9/IAS-5) Forschungszentrum Jülich Wilhelm-Johnen-Straße 52428 Jülich Germany
- Faculty of Mathematics, Computer Science and Natural Sciences RWTH Aachen University Aachen Germany
- JARA-Institut Molecular Neuroscience and Neuroimaging (INM-11) Forschungszentrum Jülich Wilhelm-Johnen-Straße 52428 Jülich Germany
| | - Alejandro Giorgetti
- Institute for Computational Biomedicine (INM-9/IAS-5) Forschungszentrum Jülich Wilhelm-Johnen-Straße 52428 Jülich Germany
- Department of Biotechnology University of Verona Verona Italy
| | - Giulia Rossetti
- Institute for Computational Biomedicine (INM-9/IAS-5) Forschungszentrum Jülich Wilhelm-Johnen-Straße 52428 Jülich Germany
- Jülich Supercomputing Centre (JSC) Forschungszentrum Jülich Jülich Germany
- Department of Neurology University Hospital Aachen (UKA), RWTH Aachen University Aachen Germany
| |
Collapse
|
9
|
Mollaei P, Barati Farimani A. Activity Map and Transition Pathways of G Protein-Coupled Receptor Revealed by Machine Learning. J Chem Inf Model 2023; 63:2296-2304. [PMID: 37036101 PMCID: PMC10131220 DOI: 10.1021/acs.jcim.3c00032] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Indexed: 04/11/2023]
Abstract
Approximately, one-third of all U.S. Food and Drug Administration approved drugs target G protein-coupled receptors (GPCRs). However, more knowledge of protein structure-activity correlation is required to improve the efficacy of the drugs targeting GPCRs. In this study, we developed a machine learning model to predict the activation state and activity level of the receptors with high prediction accuracy. Furthermore, we applied this model to thousands of molecular dynamics trajectories to correlate residue-level conformational changes of a GPCR to its activity level. Finally, the most probable transition pathway between activation states of a receptor can be identified using the state-activity information. In addition, with this model, we can associate the contribution of each amino acid to the activation process. Using this method, we can design drugs that mainly target principal amino acids driving the transition between activation states of GPCRs. Our advanced method is generalizable to all GPCR classes and provides mechanistic insight into the activation mechanism in the receptors.
Collapse
Affiliation(s)
- Parisa Mollaei
- Department
of Mechanical Engineering, Carnegie Mellon
University, Pittsburgh, Pennsylvania 15213, United States
| | - Amir Barati Farimani
- Department
of Mechanical Engineering, Carnegie Mellon
University, Pittsburgh, Pennsylvania 15213, United States
- Department
of Biomedical Engineering, Carnegie Mellon
University, Pittsburgh, Pennsylvania 15213, United States
- Machine
Learning Department, Carnegie Mellon University, Pittsburgh, Pennsylvania 15213, United States
| |
Collapse
|