1
|
Zhu R, Wu C, Zha J, Lu S, Zhang J. Decoding allosteric landscapes: computational methodologies for enzyme modulation and drug discovery. RSC Chem Biol 2025; 6:539-554. [PMID: 39981029 PMCID: PMC11836628 DOI: 10.1039/d4cb00282b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2024] [Accepted: 02/14/2025] [Indexed: 02/22/2025] Open
Abstract
Allosteric regulation is a fundamental mechanism in enzyme function, enabling dynamic modulation of activity through ligand binding at sites distal to the active site. Allosteric modulators have gained significant attention due to their unique advantages, including enhanced specificity, reduced off-target effects, and the potential for synergistic interaction with orthosteric agents. However, the inherent complexity of allosteric mechanisms has posed challenges to the systematic discovery and design of allosteric modulators. This review discusses recent advancements in computational methodologies for identifying and characterizing allosteric sites in enzymes, emphasizing techniques such as molecular dynamics (MD) simulations, enhanced sampling methods, normal mode analysis (NMA), evolutionary conservation analysis, and machine learning (ML) approaches. Advanced tools like PASSer, AlloReverse, and AlphaFold have further enhanced the understanding of allosteric mechanisms and facilitated the design of selective allosteric modulators. Case studies on enzymes such as Sirtuin 6 (SIRT6) and MAPK/ERK kinase (MEK) demonstrate the practical applications of these approaches in drug discovery. By integrating computational predictions with experimental validation, this review highlights the transformative potential of computational strategies in advancing allosteric drug discovery, offering innovative opportunities to regulate enzyme activity for therapeutic benefits.
Collapse
Affiliation(s)
- Ruidi Zhu
- Medicinal Chemistry and Bioinformatics Center, Shanghai Jiao Tong University, School of Medicine Shanghai 200025 China
| | - Chengwei Wu
- Medicinal Chemistry and Bioinformatics Center, Shanghai Jiao Tong University, School of Medicine Shanghai 200025 China
| | - Jinyin Zha
- Medicinal Chemistry and Bioinformatics Center, Shanghai Jiao Tong University, School of Medicine Shanghai 200025 China
| | - Shaoyong Lu
- Medicinal Chemistry and Bioinformatics Center, Shanghai Jiao Tong University, School of Medicine Shanghai 200025 China
- College of Pharmacy, Ningxia Medical University Yinchuan Ningxia Hui Autonomous Region 750004 China
- State Key Laboratory of Oncogenes and Related Genes, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University, School of Medicine Shanghai 200025 China
| | - Jian Zhang
- Medicinal Chemistry and Bioinformatics Center, Shanghai Jiao Tong University, School of Medicine Shanghai 200025 China
- College of Pharmacy, Ningxia Medical University Yinchuan Ningxia Hui Autonomous Region 750004 China
- State Key Laboratory of Oncogenes and Related Genes, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University, School of Medicine Shanghai 200025 China
| |
Collapse
|
2
|
Chum S, Naveira Montalvo A, Hassoun S. Computational analysis of the gut microbiota-mediated drug metabolism. Comput Struct Biotechnol J 2025; 27:1472-1481. [PMID: 40248646 PMCID: PMC12005296 DOI: 10.1016/j.csbj.2025.03.016] [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: 12/19/2024] [Revised: 02/28/2025] [Accepted: 03/08/2025] [Indexed: 04/19/2025] Open
Abstract
The gut microbiota, an extensive ecosystem harboring trillions of bacteria, plays a pivotal role in human health and disease, influencing diverse conditions from obesity to cancer. Among the microbiota's myriad functions, the capacity to metabolize drugs remains relatively unexplored despite its potential implications for drug efficacy and toxicity. Experimental methods are resource-intensive, prompting the need for innovative computational approaches. We present a computational analysis, termed MDM, aimed at predicting gut microbiota-mediated drug metabolism. This computational analysis incorporates data from diverse sources, e.g., UHGG, MagMD, MASI, KEGG, and RetroRules. An existing tool, PROXIMAL2, is used iteratively over all drug candidates from experimental databases queried against biotransformation rules from RetroRules to predict potential drug metabolites along with the enzyme commission number responsible for that biotransformation. These potential metabolites are then categorized into gut MDM metabolites by cross referencing UHGG. The analysis' efficacy is validated by its coverage on each of the experimental databases in the gut microbial context, being able to recall up to 74 % of experimental data and producing a list of potential metabolites, of which an average of about 65 % are relevant to the gut microbial context. Moreover, explorations into ranking metabolites, iterative applications to account for multi-step metabolic pathways, and potential applications in experimental studies showcase its versatility and potential impact beyond raw predictions. Overall, this study presents a promising computational framework for further research and applications gut MDM, drug development and human health.
Collapse
Affiliation(s)
| | | | - Soha Hassoun
- Department of Computer Science, Tufts University, Medford, MA 02155, United States
| |
Collapse
|
3
|
Martin M, Bittremieux W, Hassoun S. Molecular Structure Discovery for Untargeted Metabolomics Using Biotransformation Rules and Global Molecular Networking. Anal Chem 2025; 97:3213-3219. [PMID: 39903752 PMCID: PMC11841678 DOI: 10.1021/acs.analchem.4c01565] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2024] [Revised: 12/18/2024] [Accepted: 12/22/2024] [Indexed: 02/06/2025]
Abstract
Although untargeted mass spectrometry-based metabolomics is crucial for understanding life's molecular underpinnings, its effectiveness is hampered by low annotation rates of the generated tandem mass spectra. To address this issue, we introduce a novel data-driven approach, Biotransformation-based Annotation Method (BAM), that leverages molecular structural similarities inherent in biochemical reactions. BAM operates by applying biotransformation rules to known "anchor" molecules, which exhibit high spectral similarity to unknown spectra, thereby hypothesizing and ranking potential structures for the corresponding "suspect" molecule. BAM's effectiveness is demonstrated by its success in annotating query spectra in a global molecular network comprising hundreds of millions of spectra. BAM was able to assign correct molecular structures to 24.2% of examined anchor-suspect cases, thereby demonstrating remarkable advancement in metabolite annotation.
Collapse
Affiliation(s)
- Margaret
R. Martin
- Department
of Computer Science, Tufts University, Medford, Massachusetts 02155, United States
| | - Wout Bittremieux
- Department
of Computer Science, University of Antwerp, 2020 Antwerp, Belgium
| | - Soha Hassoun
- Department
of Computer Science, Tufts University, Medford, Massachusetts 02155, United States
- Department
of Chemical and Biological Engineering, Tufts University, Medford, Massachusetts 02155, United States
| |
Collapse
|
4
|
Biziukova NY, Rudik AV, Dmitriev AV, Tarasova OA, Filimonov DA, Poroikov VV. XenoMet: A Corpus of Texts to Extract Data on Metabolites of Xenobiotics. ACS OMEGA 2025; 10:2459-2471. [PMID: 39895765 PMCID: PMC11780559 DOI: 10.1021/acsomega.4c05723] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Revised: 10/25/2024] [Accepted: 11/21/2024] [Indexed: 02/04/2025]
Abstract
Understanding the biotransformation of xenobiotics in the human body is critical for a comprehensive assessment of drug effects since pharmacologically active drug metabolites may exhibit a range of biological effects that often differ from those of the original pharmaceutical agent. Studies of the biotransformation mechanisms of xenobiotics have resulted in numerous publications. Extracting information about the parent compounds (substrates) and their metabolites from the texts allows retrieval of information on their biological activities, molecular mechanisms of action, and toxicity. Manual curation of the names of xenobiotics, their metabolites, and biotransformation reactions in the text is a challenging task due to the large number of publications related to studies of pharmaceutical agents metabolism. Our aim is to create an annotated corpus of texts that can be used for automated extraction of the names of xenobiotics, including pharmaceutical agents that undergo biotransformation and their metabolites. Prior to manual annotation of the corpus, semiautomatic annotation was carried out based on the earlier developed rule-based method for parent compounds and their metabolites extraction. To create XenoMet, we automatically extracted relevant texts from PubMed using a query based on MeSH terms. The names of biotransformation reactions were recognized by using an in-house-developed dictionary. Then, we manually verified the extracted data by correcting errors in the named entity annotation and identified the associations between substrates and metabolites. We tested the applicability of XenoMet for the reconstruction of a metabolic tree and for the automated extraction of the chemical names of substrates, metabolites, and reactions of biotransformation. Classification of the named entities of metabolites, substrates, and biotransformation reactions by a conditional random fields approach using XenoMet as the training set provides an F1-score of 0.79.
Collapse
Affiliation(s)
- Nadezhda Yu. Biziukova
- Institute of Biomedical
Chemistry, 10-8, Pogodinskaya
Str., Moscow 119121, Russian Federation
| | - Anastasia V. Rudik
- Institute of Biomedical
Chemistry, 10-8, Pogodinskaya
Str., Moscow 119121, Russian Federation
| | - Alexander V. Dmitriev
- Institute of Biomedical
Chemistry, 10-8, Pogodinskaya
Str., Moscow 119121, Russian Federation
| | - Olga A. Tarasova
- Institute of Biomedical
Chemistry, 10-8, Pogodinskaya
Str., Moscow 119121, Russian Federation
| | - Dmitry A. Filimonov
- Institute of Biomedical
Chemistry, 10-8, Pogodinskaya
Str., Moscow 119121, Russian Federation
| | - Vladimir V. Poroikov
- Institute of Biomedical
Chemistry, 10-8, Pogodinskaya
Str., Moscow 119121, Russian Federation
| |
Collapse
|
5
|
Qian W, Wang X, Huang Y, Kang Y, Pan P, Hsieh CY, Hou T. Deep Learning-Driven Insights into Enzyme-Substrate Interaction Discovery. J Chem Inf Model 2025; 65:187-200. [PMID: 39721977 DOI: 10.1021/acs.jcim.4c01801] [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: 12/28/2024]
Abstract
Enzymes are ubiquitous catalysts with enormous application potential in biomedicine, green chemistry, and biotechnology. However, accurately predicting whether a molecule serves as a substrate for a specific enzyme, especially for novel entities, remains a significant challenge. Compared with traditional experimental methods, computational approaches are much more resource-efficient and time-saving, but they often compromise on accuracy. To address this, we introduce the molecule-enzyme interaction (MEI) model, a novel machine learning framework designed to predict the probability that a given molecule is a substrate for a specified enzyme with high accuracy. Utilizing a comprehensive data set that encapsulates extensive information on enzymatic reactions and enzyme sequences, the MEI model seamlessly combines atomic environmental data with amino acid sequence features through an advanced attention mechanism within a hierarchical neural network. Empirical evaluations have confirmed that the MEI model outperforms the current state-of-the-art model by at least 6.7% in prediction accuracy and 8.5% in AUROC, underscoring its enhanced predictive capabilities. Additionally, the MEI model demonstrates remarkable generalization across data sets of varying qualities and sizes. This adaptability is further evidenced by its successful application in diverse areas, such as predicting interactions within the CYP450 enzyme family and achieving an outstanding accuracy of 90.5% in predicting the enzymatic breakdown of complex plastics within environmental applications. These examples illustrate the model's ability to effectively transfer knowledge from coarsely annotated enzyme databases to smaller, high-precision data sets, robustly modeling both sparse and high-quality databases. We believe that this versatility firmly establishes the MEI model as a foundational tool in enzyme research with immense potential to extend beyond its original scope.
Collapse
Affiliation(s)
- Wenjia Qian
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, Zhejiang, China
| | - Xiaorui Wang
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, Zhejiang, China
| | - Yuansheng Huang
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, Zhejiang, China
| | - Yu Kang
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, Zhejiang, China
| | - Peichen Pan
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, Zhejiang, China
| | - Chang-Yu Hsieh
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, Zhejiang, China
| | - Tingjun Hou
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, Zhejiang, China
| |
Collapse
|
6
|
Yang H, Liu J, Chen K, Cong S, Cai S, Li Y, Jia Z, Wu H, Lou T, Wei Z, Yang X, Xiao H. D-CyPre: a machine learning-based tool for accurate prediction of human CYP450 enzyme metabolic sites. PeerJ Comput Sci 2024; 10:e2040. [PMID: 38855237 PMCID: PMC11157575 DOI: 10.7717/peerj-cs.2040] [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: 11/17/2023] [Accepted: 04/15/2024] [Indexed: 06/11/2024]
Abstract
The advancement of graph neural networks (GNNs) has made it possible to accurately predict metabolic sites. Despite the combination of GNNs with XGBOOST showing impressive performance, this technology has not yet been applied in the realm of metabolic site prediction. Previous metabolic site prediction tools focused on bonds and atoms, regardless of the overall molecular skeleton. This study introduces a novel tool, named D-CyPre, that amalgamates atom, bond, and molecular skeleton information via two directed message-passing neural networks (D-MPNN) to predict the metabolic sites of the nine cytochrome P450 enzymes using XGBOOST. In D-CyPre Precision Mode, the model produces fewer, but more accurate results (Jaccard score: 0.497, F1: 0.660, and precision: 0.737 in the test set). In D-CyPre Recall Mode, the model produces less accurate, but more comprehensive results (Jaccard score: 0.506, F1: 0.669, and recall: 0.720 in the test set). In the test set of 68 reactants, D-CyPre outperformed BioTransformer on all isoenzymes and CyProduct on most isoenzymes (5/9). For the subtypes where D-CyPre outperformed CyProducts, the Jaccard score and F1 scores increased by 24% and 16% in Precision Mode (4/9) and 19% and 12% in Recall Mode (5/9), respectively, relative to the second-best CyProduct. Overall, D-CyPre provides more accurate prediction results for human CYP450 enzyme metabolic sites.
Collapse
Affiliation(s)
- Haolan Yang
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
- Beijing University of Chinese Medicine, Research Center of Chinese Medicine Analysis and Transformation, Beijing, China
| | - Jie Liu
- Beijing University of Chinese Medicine, Research Center of Chinese Medicine Analysis and Transformation, Beijing, China
| | - Kui Chen
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
- Beijing University of Chinese Medicine, Research Center of Chinese Medicine Analysis and Transformation, Beijing, China
| | - Shiyu Cong
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
- Beijing University of Chinese Medicine, Research Center of Chinese Medicine Analysis and Transformation, Beijing, China
| | - Shengnan Cai
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
- Beijing University of Chinese Medicine, Research Center of Chinese Medicine Analysis and Transformation, Beijing, China
| | - Yueting Li
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
- Beijing University of Chinese Medicine, Research Center of Chinese Medicine Analysis and Transformation, Beijing, China
| | - Zhixin Jia
- Beijing University of Chinese Medicine, Research Center of Chinese Medicine Analysis and Transformation, Beijing, China
| | - Hao Wu
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
- Beijing University of Chinese Medicine, Research Center of Chinese Medicine Analysis and Transformation, Beijing, China
| | - Tianyu Lou
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
- Beijing University of Chinese Medicine, Research Center of Chinese Medicine Analysis and Transformation, Beijing, China
| | - Zuying Wei
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
- Beijing University of Chinese Medicine, Research Center of Chinese Medicine Analysis and Transformation, Beijing, China
| | - Xiaoqin Yang
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
- Beijing University of Chinese Medicine, Research Center of Chinese Medicine Analysis and Transformation, Beijing, China
| | - Hongbin Xiao
- Beijing University of Chinese Medicine, Research Center of Chinese Medicine Analysis and Transformation, Beijing, China
| |
Collapse
|
7
|
Martin MR, Bittremieux W, Hassoun S. Molecular structure discovery for untargeted metabolomics using biotransformation rules and global molecular networking. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.02.04.578795. [PMID: 38370723 PMCID: PMC10871291 DOI: 10.1101/2024.02.04.578795] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/20/2024]
Abstract
Although untargeted mass spectrometry-based metabolomics is crucial for understanding life's molecular underpinnings, its effectiveness is hampered by low annotation rates of the generated tandem mass spectra. To address this issue, we introduce a novel data-driven approach, Biotransformation-based Annotation Method (BAM), that leverages molecular structural similarities inherent in biochemical reactions. BAM operates by applying biotransformation rules to known 'anchor' molecules, which exhibit high spectral similarity to unknown spectra, thereby hypothesizing and ranking potential structures for the corresponding 'suspect' molecule. BAM's effectiveness is demonstrated by its success in annotating suspect spectra in a global molecular network comprising hundreds of millions of spectra. BAM was able to assign correct molecular structures to 24.2 % of examined anchor-suspect cases, thereby demonstrating remarkable advancement in metabolite annotation.
Collapse
|
8
|
Dudas B, Miteva MA. Computational and artificial intelligence-based approaches for drug metabolism and transport prediction. Trends Pharmacol Sci 2024; 45:39-55. [PMID: 38072723 DOI: 10.1016/j.tips.2023.11.001] [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: 08/02/2023] [Revised: 11/09/2023] [Accepted: 11/09/2023] [Indexed: 01/07/2024]
Abstract
Drug metabolism and transport, orchestrated by drug-metabolizing enzymes (DMEs) and drug transporters (DTs), are implicated in drug-drug interactions (DDIs) and adverse drug reactions (ADRs). Reliable and precise predictions of DDIs and ADRs are critical in the early stages of drug development to reduce the rate of drug candidate failure. A variety of experimental and computational technologies have been developed to predict DDIs and ADRs. Recent artificial intelligence (AI) approaches offer new opportunities for better predicting and understanding the complex processes related to drug metabolism and transport. We summarize the role of major DMEs and DTs, and provide an overview of current progress in computational approaches for the prediction of drug metabolism, transport, and DDIs, with an emphasis on AI including machine learning (ML) and deep learning (DL) modeling.
Collapse
Affiliation(s)
- Balint Dudas
- Université Paris Cité, CNRS UMR 8038 CiTCoM, Inserm U1268 MCTR, Paris, France
| | - Maria A Miteva
- Université Paris Cité, CNRS UMR 8038 CiTCoM, Inserm U1268 MCTR, Paris, France.
| |
Collapse
|