1
|
Mohammed I, Sagurthi SR. Current Approaches and Strategies Applied in First-in-class Drug Discovery. ChemMedChem 2024:e202400639. [PMID: 39648151 DOI: 10.1002/cmdc.202400639] [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: 08/16/2024] [Revised: 11/30/2024] [Accepted: 12/05/2024] [Indexed: 12/10/2024]
Abstract
First-in-class drug discovery (FICDD) offers novel therapies, new biological targets and mechanisms of action (MOAs) toward targeting various diseases and provides opportunities to understand unexplored biology and to target unmet diseases. Current screening approaches followed in FICDD for discovery of hit and lead molecules can be broadly categorized and discussed under phenotypic drug discovery (PDD) and target-based drug discovery (TBDD). Each category has been further classified and described with suitable examples from the literature outlining the current trends in screening approaches applied in small molecule drug discovery (SMDD). Similarly, recent applications of functional genomics, structural biology, artificial intelligence (AI), machine learning (ML), and other such advanced approaches in FICDD have also been highlighted in the article. Further, some of the current medicinal chemistry strategies applied during discovery of hits and optimization studies such as hit-to-lead (HTL) and lead optimization (LO) have been simultaneously overviewed in this article.
Collapse
Affiliation(s)
- Idrees Mohammed
- Drug Design & Molecular Medicine Laboratory, Department of Genetics & Biotechnology, Osmania University, Hyderabad, 500007, Telangana, India
| | - Someswar Rao Sagurthi
- Drug Design & Molecular Medicine Laboratory, Department of Genetics & Biotechnology, Osmania University, Hyderabad, 500007, Telangana, India
- Special Center for Molecular Medicine, Jawaharlal Nehru University, New Delhi, 110067, India
| |
Collapse
|
2
|
Sinha K, Parwez S, Mv S, Yadav A, Siddiqi MI, Banerjee D. Machine learning and biological evaluation-based identification of a potential MMP-9 inhibitor, effective against ovarian cancer cells SKOV3. J Biomol Struct Dyn 2024; 42:6823-6841. [PMID: 37504963 DOI: 10.1080/07391102.2023.2240416] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2023] [Accepted: 07/08/2023] [Indexed: 07/29/2023]
Abstract
MMP-9, also known as gelatinase B, is a zinc-metalloproteinase family protein that plays a key role in the degradation of the extracellular matrix (ECM). The normal function of MMP-9 includes the breakdown of ECM, a process that aids in normal physiological processes such as embryonic development, angiogenesis, etc. Interruptions in these processes due to the over-expression or downregulation of MMP-9 are reported to cause some pathological conditions like neurodegenerative diseases and cancer. In the present study, an integrated approach for ML-based virtual screening of the Maybridge library was carried out and their biological activity was tested in an attempt to identify novel small molecule scaffolds that can inhibit the activity of MMP-9. The top hits were identified and selected for target-based activity against MMP-9 protein using the kit (Biovision K844). Further, MTT assay was performed in various cancer cell lines such as breast (MCF-7, MDA-MB-231), colorectal (HCT119, DL-D-1), cervical (HeLa), lung (A549) and ovarian cancer (SKOV3). Interestingly, one compound viz., RJF02215 exhibited anti-cancer activity selectively in SKOV3. Wound healing assay and colony formation assay performed on SKOV3 cell line in the presence of RJF02215 confirmed that the compound had a significant inhibitory effect on this cell line. Thus, we have identified a novel molecule that can inhibit MMP-9 activity in vitro and inhibits the proliferation of SKOV3 cells. Novel molecules based on the structure of RJF02215 may become a good value addition for the treatment of ovarian cancer by exhibiting selective MMP-9 activity.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Khushboo Sinha
- Cancer Biology Division, CSIR-Central Drug Research Institute, Lucknow, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| | - Shahid Parwez
- Biochemistry and Structural Biology Division, CSIR-Central Drug Research Institute, Lucknow, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| | - Shahana Mv
- Cancer Biology Division, CSIR-Central Drug Research Institute, Lucknow, India
| | - Ananya Yadav
- Cancer Biology Division, CSIR-Central Drug Research Institute, Lucknow, India
| | - Mohammad Imran Siddiqi
- Biochemistry and Structural Biology Division, CSIR-Central Drug Research Institute, Lucknow, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| | - Dibyendu Banerjee
- Cancer Biology Division, CSIR-Central Drug Research Institute, Lucknow, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| |
Collapse
|
3
|
Guidotti IL, Neis A, Martinez DP, Seixas FK, Machado K, Kremer FS. Bambu and its applications in the discovery of active molecules against melanoma. J Mol Graph Model 2023; 124:108564. [PMID: 37453311 DOI: 10.1016/j.jmgm.2023.108564] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Revised: 06/14/2023] [Accepted: 07/10/2023] [Indexed: 07/18/2023]
Abstract
PURPOSE OR OBJECTIVE Melanoma is one of the most dangerous forms of skin cancer and the discovery of novel drugs is an ongoing effort. Quantitative Structure Activity Relationship (QSAR) is a computational method that allows the estimation of the properties of a molecule, including its biological activity. QSAR models have been widely employed in the search for potential drug candidates, but also for agrochemicals and other molecules with applications in different branches of the industry. Here we present Bambu, a simple command line tool to generate QSAR models from high-throughput screening bioassays datasets. METHODS The tool was developed using the Python programming language and relies mainly on RDKit for molecule data manipulation, FLAML for automated machine learning and the PubChem REST API for data retrieval. As a proof-of-concept we have employed the tool to generate QSAR models for melanoma cell growth inhibition based on HTS data and used them to screen libraries of FDA-approved drugs and natural compounds. Additionally, Bambu was compared to QSAR-Co, another automated tool for QSAR model generation. RESULTS based on the developed tool we were able to produce QSAR models and identify a wide variety of molecules with potential melanoma cell growth inhibitors, many of which with anti-tumoral activity already described. The QSAR models are available through the URL http://caramel.ufpel.edu.br, and all data and code used to generate its models are available at Zenodo (https://doi.org/10.5281/zenodo.7495214). Bambu source code is available at GitHub (https://github.com/omixlab/bambu-v2). In the benchmark, Bambu was able to produce models with higher accuracy, recall, F1 and ROC AUC when compared to QSAR-Co for the selected datasets. CONCLUSIONS Bambu is an free and open source tool which facilitates the creation of QSAR models and can be futurely applied in a wide variety of drug discovery projects.
Collapse
Affiliation(s)
- Isadora Leitzke Guidotti
- Centro de Desenvolvimento Tecnológico, Universidade Federal de Pelotas, Pelotas, Rio Grande do Sul, Brazil
| | - Alessandra Neis
- Centro de Desenvolvimento Tecnológico, Universidade Federal de Pelotas, Pelotas, Rio Grande do Sul, Brazil
| | - Daniela Peres Martinez
- Centro de Desenvolvimento Tecnológico, Universidade Federal de Pelotas, Pelotas, Rio Grande do Sul, Brazil
| | - Fabiana Kömmling Seixas
- Centro de Desenvolvimento Tecnológico, Universidade Federal de Pelotas, Pelotas, Rio Grande do Sul, Brazil
| | - Karina Machado
- Centro de Ciências Computacionais, Universidade Federal do Rio Grande, Rio Grande, Rio Grande do Sul, Brazil
| | - Frederico Schmitt Kremer
- Centro de Desenvolvimento Tecnológico, Universidade Federal de Pelotas, Pelotas, Rio Grande do Sul, Brazil.
| |
Collapse
|
4
|
Venkatraman V. FP-MAP: an extensive library of fingerprint-based molecular activity prediction tools. Front Chem 2023; 11:1239467. [PMID: 37649967 PMCID: PMC10462816 DOI: 10.3389/fchem.2023.1239467] [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: 06/13/2023] [Accepted: 07/31/2023] [Indexed: 09/01/2023] Open
Abstract
Discovering new drugs for disease treatment is challenging, requiring a multidisciplinary effort as well as time, and resources. With a view to improving hit discovery and lead compound identification, machine learning (ML) approaches are being increasingly used in the decision-making process. Although a number of ML-based studies have been published, most studies only report fragments of the wider range of bioactivities wherein each model typically focuses on a particular disease. This study introduces FP-MAP, an extensive atlas of fingerprint-based prediction models that covers a diverse range of activities including neglected tropical diseases (caused by viral, bacterial and parasitic pathogens) as well as other targets implicated in diseases such as Alzheimer's. To arrive at the best predictive models, performance of ≈4,000 classification/regression models were evaluated on different bioactivity data sets using 12 different molecular fingerprints. The best performing models that achieved test set AUC values of 0.62-0.99 have been integrated into an easy-to-use graphical user interface that can be downloaded from https://gitlab.com/vishsoft/fpmap.
Collapse
Affiliation(s)
- Vishwesh Venkatraman
- Department of Chemistry, Norwegian University of Science and Technology, Trondheim, Norway
| |
Collapse
|
5
|
Herman D, Kańduła MM, Freitas LGA, van Dongen C, Le Van T, Mesens N, Jaensch S, Gustin E, Micholt L, Lardeau CH, Varsakelis C, Reumers J, Zoffmann S, Will Y, Peeters PJ, Ceulemans H. Leveraging Cell Painting Images to Expand the Applicability Domain and Actively Improve Deep Learning Quantitative Structure-Activity Relationship Models. Chem Res Toxicol 2023. [PMID: 37327474 DOI: 10.1021/acs.chemrestox.2c00404] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/18/2023]
Abstract
The search for chemical hit material is a lengthy and increasingly expensive drug discovery process. To improve it, ligand-based quantitative structure-activity relationship models have been broadly applied to optimize primary and secondary compound properties. Although these models can be deployed as early as the stage of molecule design, they have a limited applicability domain─if the structures of interest differ substantially from the chemical space on which the model was trained, a reliable prediction will not be possible. Image-informed ligand-based models partly solve this shortcoming by focusing on the phenotype of a cell caused by small molecules, rather than on their structure. While this enables chemical diversity expansion, it limits the application to compounds physically available and imaged. Here, we employ an active learning approach to capitalize on both of these methods' strengths and boost the model performance of a mitochondrial toxicity assay (Glu/Gal). Specifically, we used a phenotypic Cell Painting screen to build a chemistry-independent model and adopted the results as the main factor in selecting compounds for experimental testing. With the additional Glu/Gal annotation for selected compounds we were able to dramatically improve the chemistry-informed ligand-based model with respect to the increased recognition of compounds from a 10% broader chemical space.
Collapse
Affiliation(s)
- Dorota Herman
- In-Silico Discovery, Janssen Research & Development, Pharmaceutical Companies of Johnson & Johnson, Beerse B-2340, Belgium
| | - Maciej M Kańduła
- Discovery Technology and Molecular Pharmacology, Janssen Research & Development, Pharmaceutical Companies of Johnson & Johnson, Beerse B-2340, Belgium
| | - Lorena G A Freitas
- In-Silico Discovery, Janssen Research & Development, Pharmaceutical Companies of Johnson & Johnson, Beerse B-2340, Belgium
| | | | - Thanh Le Van
- Discovery Technology and Molecular Pharmacology, Janssen Research & Development, Pharmaceutical Companies of Johnson & Johnson, Beerse B-2340, Belgium
| | - Natalie Mesens
- Predictive, Investigative and Translational Toxicology, PSTS, Janssen Research & Development, Pharmaceutical Companies of Johnson & Johnson, Beerse B-2340, Belgium
| | - Steffen Jaensch
- Discovery Technology and Molecular Pharmacology, Janssen Research & Development, Pharmaceutical Companies of Johnson & Johnson, Beerse B-2340, Belgium
| | - Emmanuel Gustin
- Discovery Technology and Molecular Pharmacology, Janssen Research & Development, Pharmaceutical Companies of Johnson & Johnson, Beerse B-2340, Belgium
| | - Liesbeth Micholt
- Discovery Technology and Molecular Pharmacology, Janssen Research & Development, Pharmaceutical Companies of Johnson & Johnson, Beerse B-2340, Belgium
| | - Charles-Hugues Lardeau
- Discovery Technology and Molecular Pharmacology, Janssen Research & Development, Pharmaceutical Companies of Johnson & Johnson, Beerse B-2340, Belgium
| | - Christos Varsakelis
- In-Silico Discovery, Janssen Research & Development, Pharmaceutical Companies of Johnson & Johnson, Beerse B-2340, Belgium
| | - Joke Reumers
- Discovery Technology and Molecular Pharmacology, Janssen Research & Development, Pharmaceutical Companies of Johnson & Johnson, Beerse B-2340, Belgium
| | - Sannah Zoffmann
- Discovery Technology and Molecular Pharmacology, Janssen Research & Development, Pharmaceutical Companies of Johnson & Johnson, Beerse B-2340, Belgium
| | - Yvonne Will
- Predictive, Investigative and Translational Toxicology, PSTS, Janssen Research & Development, Pharmaceutical Companies of Johnson & Johnson, Beerse B-2340, Belgium
| | - Pieter J Peeters
- Discovery Technology and Molecular Pharmacology, Janssen Research & Development, Pharmaceutical Companies of Johnson & Johnson, Beerse B-2340, Belgium
| | - Hugo Ceulemans
- In-Silico Discovery, Janssen Research & Development, Pharmaceutical Companies of Johnson & Johnson, Beerse B-2340, Belgium
| |
Collapse
|
6
|
Zhang Y, Luo M, Wu P, Wu S, Lee TY, Bai C. Application of Computational Biology and Artificial Intelligence in Drug Design. Int J Mol Sci 2022; 23:13568. [PMID: 36362355 PMCID: PMC9658956 DOI: 10.3390/ijms232113568] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2022] [Revised: 10/29/2022] [Accepted: 11/03/2022] [Indexed: 08/24/2023] Open
Abstract
Traditional drug design requires a great amount of research time and developmental expense. Booming computational approaches, including computational biology, computer-aided drug design, and artificial intelligence, have the potential to expedite the efficiency of drug discovery by minimizing the time and financial cost. In recent years, computational approaches are being widely used to improve the efficacy and effectiveness of drug discovery and pipeline, leading to the approval of plenty of new drugs for marketing. The present review emphasizes on the applications of these indispensable computational approaches in aiding target identification, lead discovery, and lead optimization. Some challenges of using these approaches for drug design are also discussed. Moreover, we propose a methodology for integrating various computational techniques into new drug discovery and design.
Collapse
Affiliation(s)
- Yue Zhang
- School of Life and Health Sciences, School of Medicine, The Chinese University of Hong Kong, Shenzhen 518172, China
- School of Chemistry and Materials Science, University of Science and Technology of China, Hefei 230026, China
- Warshel Institute for Computational Biology, Shenzhen 518172, China
| | - Mengqi Luo
- School of Life and Health Sciences, School of Medicine, The Chinese University of Hong Kong, Shenzhen 518172, China
- South China Hospital, Health Science Center, Shenzhen University, Shenzhen 518116, China
| | - Peng Wu
- School of Biomedical Engineering, Health Science Center, Shenzhen University, Shenzhen 518055, China
| | - Song Wu
- South China Hospital, Health Science Center, Shenzhen University, Shenzhen 518116, China
| | - Tzong-Yi Lee
- School of Life and Health Sciences, School of Medicine, The Chinese University of Hong Kong, Shenzhen 518172, China
- Warshel Institute for Computational Biology, Shenzhen 518172, China
| | - Chen Bai
- School of Life and Health Sciences, School of Medicine, The Chinese University of Hong Kong, Shenzhen 518172, China
- Warshel Institute for Computational Biology, Shenzhen 518172, China
| |
Collapse
|
7
|
Virtual Screening of Drug Proteins Based on the Prediction Classification Model of Imbalanced Data Mining. Processes (Basel) 2022. [DOI: 10.3390/pr10071420] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
We propose a virtual screening method based on imbalanced data mining in this paper, which combines virtual screening techniques with imbalanced data classification methods to improve the traditional virtual screening process. First, in the actual virtual screening process, we apply k-means and smote heuristic oversampling method to deal with imbalanced data. Meanwhile, to enhance the accuracy of the virtual screening process, a particle swarm optimization algorithm is introduced to optimize the parameters of the support vector machine classifier, and the concept of ensemble learning is brought in. The classification technique based on particle swarm optimization, support vector machine and adaptive boosting is used to screen the molecular docking conformation to improve the accuracy of the prediction. Finally, in the experimental construction and analysis section, the proposed method was validated using relevant data from the protein data bank database and PubChem database. The experimental results indicated that the proposed method can effectively improve the accuracy of virus screening and has practical guidance for new drug development. This research regards virtual screening as a problem of imbalanced data classification, which has obvious guiding significance and also provides a certain reference for the problems faced by virtual screening technology.
Collapse
|
8
|
Abstract
Stochastic computing is an emerging scientific field pushed by the need for developing high-performance artificial intelligence systems in hardware to quickly solve complex data processing problems. This is the case of virtual screening, a computational task aimed at searching across huge molecular databases for new drug leads. In this work, we show a classification framework in which molecules are described by an energy-based vector. This vector is then processed by an ultra-fast artificial neural network implemented through FPGA by using stochastic computing techniques. Compared to other previously published virtual screening methods, this proposal provides similar or higher accuracy, while it improves processing speed by about two or three orders of magnitude.
Collapse
|
9
|
Vijayan RSK, Kihlberg J, Cross JB, Poongavanam V. Enhancing preclinical drug discovery with artificial intelligence. Drug Discov Today 2021; 27:967-984. [PMID: 34838731 DOI: 10.1016/j.drudis.2021.11.023] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Revised: 10/15/2021] [Accepted: 11/19/2021] [Indexed: 12/14/2022]
Abstract
Artificial intelligence (AI) is becoming an integral part of drug discovery. It has the potential to deliver across the drug discovery and development value chain, starting from target identification and reaching through clinical development. In this review, we provide an overview of current AI technologies and a glimpse of how AI is reimagining preclinical drug discovery by highlighting examples where AI has made a real impact. Considering the excitement and hyperbole surrounding AI in drug discovery, we aim to present a realistic view by discussing both opportunities and challenges in adopting AI in drug discovery.
Collapse
Affiliation(s)
- R S K Vijayan
- Institute for Applied Cancer Science, MD Anderson Cancer Center, Houston, TX, USA
| | - Jan Kihlberg
- Department of Chemistry-BMC, Uppsala University, Uppsala, Sweden
| | - Jason B Cross
- Institute for Applied Cancer Science, MD Anderson Cancer Center, Houston, TX, USA.
| | | |
Collapse
|
10
|
Sun J, Zhong H, Wang K, Li N, Chen L. Gains from no real PAINS: Where 'Fair Trial Strategy' stands in the development of multi-target ligands. Acta Pharm Sin B 2021; 11:3417-3432. [PMID: 34900527 PMCID: PMC8642439 DOI: 10.1016/j.apsb.2021.02.023] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2020] [Revised: 02/15/2021] [Accepted: 02/25/2021] [Indexed: 12/26/2022] Open
Abstract
Compounds that selectively modulate multiple targets can provide clinical benefits and are an alternative to traditional highly selective agents for unique targets. High-throughput screening (HTS) for multitarget-directed ligands (MTDLs) using approved drugs, and fragment-based drug design has become a regular strategy to achieve an ideal multitarget combination. However, the unexpected presence of pan-assay interference compounds (PAINS) suspects in the development of MTDLs frequently results in nonspecific interactions or other undesirable effects leading to artefacts or false-positive data of biological assays. Publicly available filters can help to identify PAINS suspects; however, these filters cannot comprehensively conclude whether these suspects are "bad" or innocent. Additionally, these in silico approaches may inappropriately label a ligand as PAINS. More than 80% of the initial hits can be identified as PAINS by the filters if appropriate biochemical tests are not used resulting in false positive data that are unacceptable for medicinal chemists in manuscript peer review and future studies. Therefore, extensive offline experiments should be used after online filtering to discriminate "bad" PAINS and avoid incorrect evaluation of good scaffolds. We suggest that the use of "Fair Trial Strategy" to identify interesting molecules in PAINS suspects to provide certain structure‒function insight in MTDL development.
Collapse
Key Words
- AD, Alzheimer disease
- ALARM NMR, a La assay to detect reactive molecules by nuclear magnetic resonance
- Biochemical experiment
- CADD, computer-aided drug design technology
- CoA, coenzyme A
- EGFR, epidermal growth factor receptor
- Fair trial strategy
- GSH, glutathione
- HER2, human epidermal growth factor receptor 2
- HTS, high-throughput screening
- In silico filtering
- LC−MS, liquid chromatography−mass spectrometry
- MTDLs, multitarget-directed ligands
- Multitarget-directed ligands
- PAINS suspects
- PAINS, pan-assay interference compounds
- QSAR, quantitative structure–activity relationship
- ROS, radicals and oxygen reactive species
Collapse
Affiliation(s)
- Jianbo Sun
- State Key Laboratory of Natural Medicines, Department of Natural Medicinal Chemistry, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Hui Zhong
- Department of Pharmacology of Traditional Chinese Medicine, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Kun Wang
- State Key Laboratory of Natural Medicines, Department of Natural Medicinal Chemistry, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Na Li
- State Key Laboratory of Natural Medicines, Department of Natural Medicinal Chemistry, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Li Chen
- State Key Laboratory of Natural Medicines, Department of Natural Medicinal Chemistry, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| |
Collapse
|
11
|
Gupta R, Srivastava D, Sahu M, Tiwari S, Ambasta RK, Kumar P. Artificial intelligence to deep learning: machine intelligence approach for drug discovery. Mol Divers 2021; 25:1315-1360. [PMID: 33844136 PMCID: PMC8040371 DOI: 10.1007/s11030-021-10217-3] [Citation(s) in RCA: 331] [Impact Index Per Article: 82.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Accepted: 03/22/2021] [Indexed: 02/06/2023]
Abstract
Drug designing and development is an important area of research for pharmaceutical companies and chemical scientists. However, low efficacy, off-target delivery, time consumption, and high cost impose a hurdle and challenges that impact drug design and discovery. Further, complex and big data from genomics, proteomics, microarray data, and clinical trials also impose an obstacle in the drug discovery pipeline. Artificial intelligence and machine learning technology play a crucial role in drug discovery and development. In other words, artificial neural networks and deep learning algorithms have modernized the area. Machine learning and deep learning algorithms have been implemented in several drug discovery processes such as peptide synthesis, structure-based virtual screening, ligand-based virtual screening, toxicity prediction, drug monitoring and release, pharmacophore modeling, quantitative structure-activity relationship, drug repositioning, polypharmacology, and physiochemical activity. Evidence from the past strengthens the implementation of artificial intelligence and deep learning in this field. Moreover, novel data mining, curation, and management techniques provided critical support to recently developed modeling algorithms. In summary, artificial intelligence and deep learning advancements provide an excellent opportunity for rational drug design and discovery process, which will eventually impact mankind. The primary concern associated with drug design and development is time consumption and production cost. Further, inefficiency, inaccurate target delivery, and inappropriate dosage are other hurdles that inhibit the process of drug delivery and development. With advancements in technology, computer-aided drug design integrating artificial intelligence algorithms can eliminate the challenges and hurdles of traditional drug design and development. Artificial intelligence is referred to as superset comprising machine learning, whereas machine learning comprises supervised learning, unsupervised learning, and reinforcement learning. Further, deep learning, a subset of machine learning, has been extensively implemented in drug design and development. The artificial neural network, deep neural network, support vector machines, classification and regression, generative adversarial networks, symbolic learning, and meta-learning are examples of the algorithms applied to the drug design and discovery process. Artificial intelligence has been applied to different areas of drug design and development process, such as from peptide synthesis to molecule design, virtual screening to molecular docking, quantitative structure-activity relationship to drug repositioning, protein misfolding to protein-protein interactions, and molecular pathway identification to polypharmacology. Artificial intelligence principles have been applied to the classification of active and inactive, monitoring drug release, pre-clinical and clinical development, primary and secondary drug screening, biomarker development, pharmaceutical manufacturing, bioactivity identification and physiochemical properties, prediction of toxicity, and identification of mode of action.
Collapse
Affiliation(s)
- Rohan Gupta
- Molecular Neuroscience and Functional Genomics Laboratory, Department of Biotechnology, Delhi Technological University (Formerly DCE), Shahbad Daulatpur, Bawana Road, Delhi, 110042, India
| | - Devesh Srivastava
- Molecular Neuroscience and Functional Genomics Laboratory, Department of Biotechnology, Delhi Technological University (Formerly DCE), Shahbad Daulatpur, Bawana Road, Delhi, 110042, India
| | - Mehar Sahu
- Molecular Neuroscience and Functional Genomics Laboratory, Department of Biotechnology, Delhi Technological University (Formerly DCE), Shahbad Daulatpur, Bawana Road, Delhi, 110042, India
| | - Swati Tiwari
- Molecular Neuroscience and Functional Genomics Laboratory, Department of Biotechnology, Delhi Technological University (Formerly DCE), Shahbad Daulatpur, Bawana Road, Delhi, 110042, India
| | - Rashmi K Ambasta
- Molecular Neuroscience and Functional Genomics Laboratory, Department of Biotechnology, Delhi Technological University (Formerly DCE), Shahbad Daulatpur, Bawana Road, Delhi, 110042, India
| | - Pravir Kumar
- Molecular Neuroscience and Functional Genomics Laboratory, Department of Biotechnology, Delhi Technological University (Formerly DCE), Shahbad Daulatpur, Bawana Road, Delhi, 110042, India.
| |
Collapse
|
12
|
Issa NT, Stathias V, Schürer S, Dakshanamurthy S. Machine and deep learning approaches for cancer drug repurposing. Semin Cancer Biol 2021; 68:132-142. [PMID: 31904426 PMCID: PMC7723306 DOI: 10.1016/j.semcancer.2019.12.011] [Citation(s) in RCA: 119] [Impact Index Per Article: 29.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2019] [Revised: 10/31/2019] [Accepted: 12/15/2019] [Indexed: 02/07/2023]
Abstract
Knowledge of the underpinnings of cancer initiation, progression and metastasis has increased exponentially in recent years. Advanced "omics" coupled with machine learning and artificial intelligence (deep learning) methods have helped elucidate targets and pathways critical to those processes that may be amenable to pharmacologic modulation. However, the current anti-cancer therapeutic armamentarium continues to lag behind. As the cost of developing a new drug remains prohibitively expensive, repurposing of existing approved and investigational drugs is sought after given known safety profiles and reduction in the cost barrier. Notably, successes in oncologic drug repurposing have been infrequent. Computational in-silico strategies have been developed to aid in modeling biological processes to find new disease-relevant targets and discovering novel drug-target and drug-phenotype associations. Machine and deep learning methods have especially enabled leaps in those successes. This review will discuss these methods as they pertain to cancer biology as well as immunomodulation for drug repurposing opportunities in oncologic diseases.
Collapse
Affiliation(s)
- Naiem T Issa
- Dr. Phillip Frost Department of Dermatology and Cutaneous Surgery, University of Miami School of Medicine, Miami, FL, USA
| | - Vasileios Stathias
- Department of Molecular and Cellular Pharmacology, University of Miami School of Medicine, Miami, FL, USA
| | - Stephan Schürer
- Department of Molecular and Cellular Pharmacology, University of Miami School of Medicine, Miami, FL, USA
| | - Sivanesan Dakshanamurthy
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC, USA.
| |
Collapse
|
13
|
Wang YL, Wang F, Shi XX, Jia CY, Wu FX, Hao GF, Yang GF. Cloud 3D-QSAR: a web tool for the development of quantitative structure-activity relationship models in drug discovery. Brief Bioinform 2020; 22:5934782. [PMID: 33140820 DOI: 10.1093/bib/bbaa276] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Revised: 09/11/2020] [Accepted: 09/21/2020] [Indexed: 12/22/2022] Open
Abstract
Effective drug discovery contributes to the treatment of numerous diseases but is limited by high costs and long cycles. The Quantitative Structure-Activity Relationship (QSAR) method was introduced to evaluate the activity of a large number of compounds virtually, reducing the time and labor costs required for chemical synthesis and experimental determination. Hence, this method increases the efficiency of drug discovery. To meet the needs of researchers to utilize this technology, numerous QSAR-related web servers, such as Web-4D-QSAR and DPubChem, have been developed in recent years. However, none of the servers mentioned above can perform a complete QSAR modeling and supply activity prediction functions. We introduce Cloud 3D-QSAR by integrating the functions of molecular structure generation, alignment, molecular interaction field (MIF) computing and results analysis to provide a one-stop solution. We rigidly validated this server, and the activity prediction correlation was R2 = 0.934 in 834 test molecules. The sensitivity, specificity and accuracy were 86.9%, 94.5% and 91.5%, respectively, with AUC = 0.981, AUCPR = 0.971. The Cloud 3D-QSAR server may facilitate the development of good QSAR models in drug discovery. Our server is free and now available at http://chemyang.ccnu.edu.cn/ccb/server/cloud3dQSAR/ and http://agroda.gzu.edu.cn:9999/ccb/server/cloud3dQSAR/.
Collapse
Affiliation(s)
- Yu-Liang Wang
- College of Chemistry, Central China Normal University (CCNU)
| | | | | | - Chen-Yang Jia
- College of Chemistry, Central China Normal University (CCNU)
| | | | - Ge-Fei Hao
- Pesticide Design and Bioinformatics in College of Chemistry, CCNU
| | | |
Collapse
|
14
|
Slavov S, Beger RD. Quantitative structure–toxicity relationships in translational toxicology. CURRENT OPINION IN TOXICOLOGY 2020. [DOI: 10.1016/j.cotox.2020.04.002] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
15
|
Selecting machine-learning scoring functions for structure-based virtual screening. DRUG DISCOVERY TODAY. TECHNOLOGIES 2020; 32-33:81-87. [PMID: 33386098 DOI: 10.1016/j.ddtec.2020.09.001] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Revised: 09/02/2020] [Accepted: 09/07/2020] [Indexed: 12/27/2022]
Abstract
Interest in docking technologies has grown parallel to the ever increasing number and diversity of 3D models for macromolecular therapeutic targets. Structure-Based Virtual Screening (SBVS) aims at leveraging these experimental structures to discover the necessary starting points for the drug discovery process. It is now established that Machine Learning (ML) can strongly enhance the predictive accuracy of scoring functions for SBVS by exploiting large datasets from targets, molecules and their associations. However, with greater choice, the question of which ML-based scoring function is the most suitable for prospective use on a given target has gained importance. Here we analyse two approaches to select an existing scoring function for the target along with a third approach consisting in generating a scoring function tailored to the target. These analyses required discussing the limitations of popular SBVS benchmarks, the alternatives to benchmark scoring functions for SBVS and how to generate them or use them using freely-available software.
Collapse
|
16
|
Achary PGR. Applications of Quantitative Structure-Activity Relationships (QSAR) based Virtual Screening in Drug Design: A Review. Mini Rev Med Chem 2020; 20:1375-1388. [DOI: 10.2174/1389557520666200429102334] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2019] [Revised: 11/07/2019] [Accepted: 11/08/2019] [Indexed: 12/18/2022]
Abstract
The scientists, and the researchers around the globe generate tremendous amount of information
everyday; for instance, so far more than 74 million molecules are registered in Chemical
Abstract Services. According to a recent study, at present we have around 1060 molecules, which are
classified as new drug-like molecules. The library of such molecules is now considered as ‘dark chemical
space’ or ‘dark chemistry.’ Now, in order to explore such hidden molecules scientifically, a good
number of live and updated databases (protein, cell, tissues, structure, drugs, etc.) are available today.
The synchronization of the three different sciences: ‘genomics’, proteomics and ‘in-silico simulation’
will revolutionize the process of drug discovery. The screening of a sizable number of drugs like molecules
is a challenge and it must be treated in an efficient manner. Virtual screening (VS) is an important
computational tool in the drug discovery process; however, experimental verification of the
drugs also equally important for the drug development process. The quantitative structure-activity relationship
(QSAR) analysis is one of the machine learning technique, which is extensively used in VS
techniques. QSAR is well-known for its high and fast throughput screening with a satisfactory hit rate.
The QSAR model building involves (i) chemo-genomics data collection from a database or literature
(ii) Calculation of right descriptors from molecular representation (iii) establishing a relationship
(model) between biological activity and the selected descriptors (iv) application of QSAR model to
predict the biological property for the molecules. All the hits obtained by the VS technique needs to be
experimentally verified. The present mini-review highlights: the web-based machine learning tools, the
role of QSAR in VS techniques, successful applications of QSAR based VS leading to the drug discovery
and advantages and challenges of QSAR.
Collapse
Affiliation(s)
- Patnala Ganga Raju Achary
- Department of Chemistry, Faculty of Engineering & Technology (ITER), Siksha ‘O’ Anusandhan, Deemed to be University, Khandagiri Square, Bhubaneswar- 751030, India
| |
Collapse
|
17
|
Gao K, Nguyen DD, Chen J, Wang R, Wei GW. Repositioning of 8565 Existing Drugs for COVID-19. J Phys Chem Lett 2020; 11:5373-5382. [PMID: 32543196 PMCID: PMC7313673 DOI: 10.1021/acs.jpclett.0c01579] [Citation(s) in RCA: 67] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Accepted: 06/16/2020] [Indexed: 05/06/2023]
Abstract
The coronavirus disease 2019 (COVID-19) pandemic caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has infected over 7.1 million people and led to over 0.4 million deaths. Currently, there is no specific anti-SARS-CoV-2 medication. New drug discovery typically takes more than 10 years. Drug repositioning becomes one of the most feasible approaches for combating COVID-19. This work curates the largest available experimental data set for SARS-CoV-2 or SARS-CoV 3CL (main) protease inhibitors. On the basis of this data set, we develop validated machine learning models with relatively low root-mean-square error to screen 1553 FDA-approved drugs as well as another 7012 investigational or off-market drugs in DrugBank. We found that many existing drugs might be potentially potent to SARS-CoV-2. The druggability of many potent SARS-CoV-2 3CL protease inhibitors is analyzed. This work offers a foundation for further experimental studies of COVID-19 drug repositioning.
Collapse
Affiliation(s)
- Kaifu Gao
- Department of Mathematics, Michigan State University, East Lansing, Michigan 48824, United States
| | - Duc Duy Nguyen
- Department of Mathematics, University of Kentucky, Lexington, Kentucky 40506, United States
| | - Jiahui Chen
- Department of Mathematics, Michigan State University, East Lansing, Michigan 48824, United States
| | - Rui Wang
- Department of Mathematics, Michigan State University, East Lansing, Michigan 48824, United States
| | - Guo-Wei Wei
- Department of Mathematics, Michigan State University, East Lansing, Michigan 48824, United States
- Department of Biochemistry and Molecular Biology, Michigan State University, East Lansing, Michigan 48824, United States
- Department of Electrical and Computer Engineering, Michigan State University, East Lansing, Michigan 48824, United States
| |
Collapse
|
18
|
Li H, Sze K, Lu G, Ballester PJ. Machine‐learning scoring functions for structure‐based virtual screening. WILEY INTERDISCIPLINARY REVIEWS-COMPUTATIONAL MOLECULAR SCIENCE 2020. [DOI: 10.1002/wcms.1478] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Affiliation(s)
- Hongjian Li
- Cancer Research Center of Marseille (INSERM U1068, Institut Paoli‐Calmettes, Aix‐Marseille Université UM105, CNRS UMR7258) Marseille France
- CUHK‐SDU Joint Laboratory on Reproductive Genetics, School of Biomedical Sciences Chinese University of Hong Kong Shatin Hong Kong
| | - Kam‐Heung Sze
- CUHK‐SDU Joint Laboratory on Reproductive Genetics, School of Biomedical Sciences Chinese University of Hong Kong Shatin Hong Kong
| | - Gang Lu
- CUHK‐SDU Joint Laboratory on Reproductive Genetics, School of Biomedical Sciences Chinese University of Hong Kong Shatin Hong Kong
| | - Pedro J. Ballester
- Cancer Research Center of Marseille (INSERM U1068, Institut Paoli‐Calmettes, Aix‐Marseille Université UM105, CNRS UMR7258) Marseille France
| |
Collapse
|
19
|
Singh N, Chaput L, Villoutreix BO. Virtual screening web servers: designing chemical probes and drug candidates in the cyberspace. Brief Bioinform 2020; 22:1790-1818. [PMID: 32187356 PMCID: PMC7986591 DOI: 10.1093/bib/bbaa034] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
The interplay between life sciences and advancing technology drives a continuous cycle of chemical data growth; these data are most often stored in open or partially open databases. In parallel, many different types of algorithms are being developed to manipulate these chemical objects and associated bioactivity data. Virtual screening methods are among the most popular computational approaches in pharmaceutical research. Today, user-friendly web-based tools are available to help scientists perform virtual screening experiments. This article provides an overview of internet resources enabling and supporting chemical biology and early drug discovery with a main emphasis on web servers dedicated to virtual ligand screening and small-molecule docking. This survey first introduces some key concepts and then presents recent and easily accessible virtual screening and related target-fishing tools as well as briefly discusses case studies enabled by some of these web services. Notwithstanding further improvements, already available web-based tools not only contribute to the design of bioactive molecules and assist drug repositioning but also help to generate new ideas and explore different hypotheses in a timely fashion while contributing to teaching in the field of drug development.
Collapse
Affiliation(s)
- Natesh Singh
- Univ. Lille, Inserm, Institut Pasteur de Lille, U1177 Drugs and Molecules for Living Systems, F-59000 Lille, France
| | - Ludovic Chaput
- Univ. Lille, Inserm, Institut Pasteur de Lille, U1177 Drugs and Molecules for Living Systems, F-59000 Lille, France
| | - Bruno O Villoutreix
- Univ. Lille, Inserm, Institut Pasteur de Lille, U1177 Drugs and Molecules for Living Systems, F-59000 Lille, France
| |
Collapse
|
20
|
Pirzada RH, Javaid N, Choi S. The Roles of the NLRP3 Inflammasome in Neurodegenerative and Metabolic Diseases and in Relevant Advanced Therapeutic Interventions. Genes (Basel) 2020; 11:E131. [PMID: 32012695 PMCID: PMC7074480 DOI: 10.3390/genes11020131] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2019] [Revised: 01/22/2020] [Accepted: 01/22/2020] [Indexed: 02/07/2023] Open
Abstract
Inflammasomes are intracellular multiprotein complexes in the cytoplasm that regulate inflammation activation in the innate immune system in response to pathogens and to host self-derived molecules. Recent advances greatly improved our understanding of the activation of nucleotide-binding oligomerization domain-like receptor (NLR) family pyrin domain containing 3 (NLRP3) inflammasomes at the molecular level. The NLRP3 belongs to the subfamily of NLRP which activates caspase 1, thus causing the production of proinflammatory cytokines (interleukin 1β and interleukin 18) and pyroptosis. This inflammasome is involved in multiple neurodegenerative and metabolic disorders including Alzheimer's disease, multiple sclerosis, type 2 diabetes mellitus, and gout. Therefore, therapeutic targeting to the NLRP3 inflammasome complex is a promising way to treat these diseases. Recent research advances paved the way toward drug research and development using a variety of machine learning-based and artificial intelligence-based approaches. These state-of-the-art approaches will lead to the discovery of better drugs after the training of such a system.
Collapse
Affiliation(s)
| | | | - Sangdun Choi
- Department of Molecular Science and Technology, Ajou University, Suwon 16499, Korea; (R.H.P.); (N.J.)
| |
Collapse
|
21
|
Maltarollo VG. Classification of Staphylococcus Aureus FabI Inhibitors by Machine Learning Techniques. ACTA ACUST UNITED AC 2019. [DOI: 10.4018/ijqspr.2019100101] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Enoyl-acyl carrier protein reductase (FabI) is a key enzyme in the fatty acid metabolism of gram-positive bacteria and is considered a potential target for new antibacterial drugs development. Indeed, triclosan is a widely employed antibacterial and AFN-1252 is currently under phase-II clinical trials, both are known as FabI inhibitors. Nowadays, there is an urgent need for new drug discovery due to increasing antibacterial resistance. In the present study, classification models using machine learning techniques were generated to distinguish SaFabI inhibitors from non-inhibitors successfully (e.g., Mathews correlation coefficient values equal to 0.837 and 0.789 calculated with internal and external validations). The interpretation of a selected model indicates that larger compounds, number of N atoms and the distance between central amide and naphthyridinone ring are important to biological activity, corroborating previous studies. Therefore, these obtained information and generated models can be useful for design/discovery of novel bioactive ligands as potential antibacterial agents.
Collapse
|
22
|
Vukovic K, Gadaleta D, Benfenati E. Methodology of aiQSAR: a group-specific approach to QSAR modelling. J Cheminform 2019; 11:27. [PMID: 30945010 PMCID: PMC6446381 DOI: 10.1186/s13321-019-0350-y] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2019] [Accepted: 03/25/2019] [Indexed: 12/26/2022] Open
Abstract
Background Several QSAR methodology developments have shown promise in recent years. These include the consensus approach to generate the final prediction of a model, utilizing new, advanced machine learning algorithms and streamlining, standardization and automation of various QSAR steps. One approach that seems under-explored is at-the-runtime generation of local models specific to individual compounds. This approach was quite likely limited by the computational requirements, but with current increases in processing power and the widespread availability of cluster-computing infrastructure, this limitation is no longer that severe. Results We propose a new QSAR methodology: aiQSAR, whose aim is to generate endpoint predictions directly from the input dataset by building an array of local models generated at-the-runtime and specific for each compound in the dataset. The local group of each compound is selected on the basis of fingerprint similarities and the final prediction is calculated by integrating the results of a number of autonomous mathematical models. The method is applicable to regression, binary classification and multi-class classification and was tested on one dataset for each endpoint type: bioconcentration factor (BCF) for regression, Ames test for binary classification and Environmental Protection Agency (EPA) acute rat oral toxicity ranking for multi-class classification. As part of this method, the applicability domain of each prediction is assessed through the applicability domain measure, calculated on the basis of the fingerprint similarities in each local group of compounds. Conclusions We outline the methodology for a new QSAR-based predictive tool whose advantages are automation, group-specific approach to modelling and simplicity of execution. Our aim now will be to develop this method into a stand-alone software tool. We hope that eventual adoption of our tool would make QSAR modelling more accessible and transparent. Our methodology could be used as an initial modelling step, to predict new compounds by simply loading the training dataset as an input. Predictions could then be further evaluated and refined either by other tools or through optimization of aiQSAR parameters. Electronic supplementary material The online version of this article (10.1186/s13321-019-0350-y) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Kristijan Vukovic
- Istituto di Ricerche Farmacologiche Mario Negri-IRCCS, Via Mario Negri 2, 20156, Milan, Italy. .,Jozef Stefan International Postgraduate School, Jamova cesta 39, 1000, Ljubljana, Slovenia.
| | - Domenico Gadaleta
- Istituto di Ricerche Farmacologiche Mario Negri-IRCCS, Via Mario Negri 2, 20156, Milan, Italy
| | - Emilio Benfenati
- Istituto di Ricerche Farmacologiche Mario Negri-IRCCS, Via Mario Negri 2, 20156, Milan, Italy
| |
Collapse
|
23
|
Niu C, Jiang M, Li N, Cao J, Hou M, Ni DA, Chu Z. Integrated bioinformatics analysis of As, Au, Cd, Pb and Cu heavy metal responsive marker genes through Arabidopsis thaliana GEO datasets. PeerJ 2019; 7:e6495. [PMID: 30918749 PMCID: PMC6428040 DOI: 10.7717/peerj.6495] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2018] [Accepted: 01/19/2019] [Indexed: 01/11/2023] Open
Abstract
BACKGROUND Current environmental pollution factors, particularly the distribution and diffusion of heavy metals in soil and water, are a high risk to local environments and humans. Despite striking advances in methods to detect contaminants by a variety of chemical and physical solutions, these methods have inherent limitations such as small dimensions and very low coverage. Therefore, identifying novel contaminant biomarkers are urgently needed. METHODS To better track heavy metal contaminations in soil and water, integrated bioinformatics analysis to identify biomarkers of relevant heavy metal, such as As, Cd, Pb and Cu, is a suitable method for long-term and large-scale surveys of such heavy metal pollutants. Subsequently, the accuracy and stability of the results screened were experimentally validated by quantitative PCR experiment. RESULTS We obtained 168 differentially expressed genes (DEGs) which contained 59 up-regulated genes and 109 down-regulated genes through comparative bioinformatics analyses. Subsequently, the gene ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichments of these DEGs were performed, respectively. GO analyses found that these DEGs were mainly related to responses to chemicals, responses to stimulus, responses to stress, responses to abiotic stimulus, and so on. KEGG pathway analyses of DEGs were mainly involved in the protein degradation process and other biologic process, such as the phenylpropanoid biosynthesis pathways and nitrogen metabolism. Moreover, we also speculated that nine candidate core biomarker genes (namely, NILR1, PGPS1, WRKY33, BCS1, AR781, CYP81D8, NR1, EAP1 and MYB15) might be tightly correlated with the response or transport of heavy metals. Finally, experimental results displayed that these genes had the same expression trend response to different stresses as mentioned above (Cd, Pb and Cu) and no mentioned above (Zn and Cr). CONCLUSION In general, the identified biomarker genes could help us understand the potential molecular mechanisms or signaling pathways responsive to heavy metal stress in plants, and could be applied as marker genes to track heavy metal pollution in soil and water through detecting their expression in plants growing in those environments.
Collapse
Affiliation(s)
- Chao Niu
- School of Ecological Technology and Engineering, Shanghai Institute of Technology, Shanghai, Shanghai, China
- Shanghai Key Laboratory of Plant Functional Genomics and Resources, Shanghai Chenshan Botanical Garden, Shanghai, Shanghai, China
- Shanghai Chenshan Plant Science Research Center, Chinese Academy of Sciences, Shanghai, Shanghai, China
| | - Min Jiang
- Shanghai Key Laboratory of Plant Functional Genomics and Resources, Shanghai Chenshan Botanical Garden, Shanghai, Shanghai, China
- Shanghai Chenshan Plant Science Research Center, Chinese Academy of Sciences, Shanghai, Shanghai, China
| | - Na Li
- Shanghai Key Laboratory of Plant Functional Genomics and Resources, Shanghai Chenshan Botanical Garden, Shanghai, Shanghai, China
- Shanghai Chenshan Plant Science Research Center, Chinese Academy of Sciences, Shanghai, Shanghai, China
- College of Life Sciences, Shanghai Normal University, Shanghai, Shanghai, China
| | - Jianguo Cao
- College of Life Sciences, Shanghai Normal University, Shanghai, Shanghai, China
| | - Meifang Hou
- School of Ecological Technology and Engineering, Shanghai Institute of Technology, Shanghai, Shanghai, China
| | - Di-an Ni
- School of Ecological Technology and Engineering, Shanghai Institute of Technology, Shanghai, Shanghai, China
| | - Zhaoqing Chu
- Shanghai Key Laboratory of Plant Functional Genomics and Resources, Shanghai Chenshan Botanical Garden, Shanghai, Shanghai, China
- Shanghai Chenshan Plant Science Research Center, Chinese Academy of Sciences, Shanghai, Shanghai, China
| |
Collapse
|