1
|
Kauffman J, Cuevas J, Feiner J, Metzger M, Shetye G, Wan B, Qader M, Nguyen D, Nugent A, Hossain A, Franzblau S, Umesiri FE. Discovery of ultra short β-peptoids with selective activity against drug-resistant Mycobacterium tuberculosis. Eur J Med Chem 2025; 290:117531. [PMID: 40147341 DOI: 10.1016/j.ejmech.2025.117531] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2024] [Revised: 01/27/2025] [Accepted: 03/18/2025] [Indexed: 03/29/2025]
Abstract
There is an urgent need to develop new anti-tuberculosis (anti-TB) drugs to tackle drug-resistant strains of Mycobacterium tuberculosis (M.tb). Whereas antimicrobial peptides (AMPs) have received attention because of their antibacterial properties, oligo-N-substituted glycines (peptoids) are now seen as favorable alternatives to AMPs because they are more stable and less vulnerable to protease degradation, less expensive to produce, and better suited to potential pharmaceutical adoption and development. In this work, therefore, we designed, synthesized, and screened 22 new α- and β-peptoids against drug susceptible M. tb strain H37Rv using the Microplate Alamar Blue assay (MABA) to evaluate minimum inhibitory concentration (MIC). Eight compounds (JC5, MM2, MM5, MM9, MM10, MM11, JF11, and JF13) had MICs of less than 10 μg/ml, the most potent of which were JC5 and MM2, with MICs of 1.48 μg/ml and 2.97 μg/ml, respectively. JC5 and MM2 also retained potency against strains mono-resistant to isoniazid and rifampin, and against five of the global M. tb clade representatives. Furthermore, peptoids JC5 and MM2 showed minimum bactericidal concentration (MBC) of 3.02 μg/ml and 5.48 μg/ml respectively. Intracellular activity by luminescence showed a macrophage EC90 of less than 10 μg/ml for both JC5 and MM2. In addition, both compounds showed remarkable narrow spectrum activity. Selectivity with respect to Vero cells was modest but sufficient to consider these classes of alpha and beta-peptoids as good leads for further development of anti-TB drugs.
Collapse
Affiliation(s)
- John Kauffman
- Chemistry Department, Wheaton College, 501 College Ave, Wheaton, IL 60187, United States
| | - Jake Cuevas
- Chemistry Department, Wheaton College, 501 College Ave, Wheaton, IL 60187, United States
| | - Janaya Feiner
- Chemistry Department, Wheaton College, 501 College Ave, Wheaton, IL 60187, United States
| | - Margaret Metzger
- Chemistry Department, Wheaton College, 501 College Ave, Wheaton, IL 60187, United States
| | - Gauri Shetye
- Institute for Tuberculosis Research, Department of Pharmaceutical Sciences, University of Illinois Chicago, 833 South Wood Street, Chicago, IL 60612, United States
| | - Baojie Wan
- Institute for Tuberculosis Research, Department of Pharmaceutical Sciences, University of Illinois Chicago, 833 South Wood Street, Chicago, IL 60612, United States
| | - Mallique Qader
- Institute for Tuberculosis Research, Department of Pharmaceutical Sciences, University of Illinois Chicago, 833 South Wood Street, Chicago, IL 60612, United States
| | - Duc Nguyen
- Institute for Tuberculosis Research, Department of Pharmaceutical Sciences, University of Illinois Chicago, 833 South Wood Street, Chicago, IL 60612, United States
| | - Angela Nugent
- Institute for Tuberculosis Research, Department of Pharmaceutical Sciences, University of Illinois Chicago, 833 South Wood Street, Chicago, IL 60612, United States
| | - Akil Hossain
- Institute for Tuberculosis Research, Department of Pharmaceutical Sciences, University of Illinois Chicago, 833 South Wood Street, Chicago, IL 60612, United States
| | - Scott Franzblau
- Institute for Tuberculosis Research, Department of Pharmaceutical Sciences, University of Illinois Chicago, 833 South Wood Street, Chicago, IL 60612, United States
| | - Francis E Umesiri
- Chemistry Department, Wheaton College, 501 College Ave, Wheaton, IL 60187, United States.
| |
Collapse
|
2
|
Marhöfer RJ, Noack S, Selzer PM. Antiparasitics discovery: from genotype to phenotype to compounds. Trends Parasitol 2025:S1471-4922(25)00101-1. [PMID: 40345885 DOI: 10.1016/j.pt.2025.04.007] [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: 03/06/2025] [Revised: 04/10/2025] [Accepted: 04/11/2025] [Indexed: 05/11/2025]
Abstract
For decades, the discovery of antiparasitics was dominated by whole-organism screening of intact parasite organisms or surrogate parasite models, such as Caenorhabitis elegans, using in vivo animal models or in vitro parasite assays, the latter also known as phenotypic screening. Molecular target-based screening played only a minor role, if at all. While publications using phenotypic screening are abundant in the literature, publications of successful, marketed, antiparasitic drugs discovered using the molecular target-based approach are scarce. This approach, therefore, is often perceived as less relevant for antiparasitic drug discovery than the two other approaches. However, antiparasitics belonging, for example, to the isoxazolines, bispyrazoles, depsipeptides or praziquantel (PZQ) derivatives, imposingly demonstrate the value of this approach, when wisely used in a cooperative manner with phenotypic screening.
Collapse
Affiliation(s)
- Richard J Marhöfer
- Boehringer Ingelheim Animal Health, Binger Str 173, 55216 Ingelheim am Rhein, Germany
| | - Sandra Noack
- Boehringer Ingelheim Animal Health, Binger Str 173, 55216 Ingelheim am Rhein, Germany
| | - Paul M Selzer
- Boehringer Ingelheim Animal Health, Binger Str 173, 55216 Ingelheim am Rhein, Germany.
| |
Collapse
|
3
|
Trachtenberg A, Akabayov B. From Patterns to Pills: How Informatics Is Shaping Medicinal Chemistry. Pharmaceutics 2025; 17:612. [PMID: 40430903 PMCID: PMC12114992 DOI: 10.3390/pharmaceutics17050612] [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: 03/20/2025] [Revised: 04/25/2025] [Accepted: 05/03/2025] [Indexed: 05/29/2025] Open
Abstract
In today's information-driven era, machine learning is revolutionizing medicinal chemistry, offering a paradigm shift from traditional, intuition-based, and often bias-prone methods to the prediction of chemical properties without prior knowledge of the basic principles governing drug function. This perspective highlights the growing importance of informatics in shaping the field of medicinal chemistry, particularly through the concept of the "informacophore". The informacophore refers to the minimal chemical structure, combined with computed molecular descriptors, fingerprints, and machine-learned representations of its structure, that are essential for a molecule to exhibit biological activity. Similar to a skeleton key unlocking multiple locks, the informacophore points to the molecular features that trigger biological responses. By identifying and optimizing informacophores through in-depth analysis of ultra-large datasets of potential lead compounds and automating standard parts in the development process, there will be a significant reduction in biased intuitive decisions, which may lead to systemic errors and a parallel acceleration of drug discovery processes.
Collapse
Affiliation(s)
| | - Barak Akabayov
- Department of Chemistry and Data Science Research Center, Ben-Gurion University of the Negev, Beer-Sheva 8410501, Israel;
| |
Collapse
|
4
|
Niu J, Adam J, Skurk T, Seissler J, Dong Q, Efiong E, Gieger C, Peters A, Sharma S, Grallert H. Machine learning approach on plasma proteomics identifies signatures associated with obesity in the KORA FF4 cohort. Diabetes Obes Metab 2025; 27:2626-2636. [PMID: 40017018 PMCID: PMC11965001 DOI: 10.1111/dom.16264] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/21/2024] [Revised: 01/24/2025] [Accepted: 02/02/2025] [Indexed: 03/01/2025]
Abstract
AIMS This study investigated the role of plasma proteins in obesity to identify predictive biomarkers and explore underlying biological mechanisms. METHODS In the Cooperative Health Research in the Region of Augsburg (KORA) FF4 study, 809 proteins were measured in 2045 individuals (564 obese and 1481 non-obese). Multivariate logistic regression adjusted for confounders (basic and full models) was used to identify obesity-associated proteins. Priority-Lasso was applied for feature selection, followed by machine learning models (support vector machine [SVM], random forest [RF], k-nearest neighbour [KNN] and adaptive boosting [Adaboost]) for prediction. Correlation and enrichment analyses were performed to elucidate relationships between protein biomarkers, obesity risk factors and perturbed pathways. Mendelian randomisation (MR) assessed causal links between proteins and obesity. RESULTS A total of 16 proteins were identified as significantly associated with obesity through multivariable logistic regression in the basic model and subsequent Priority-Lasso analysis. Enrichment analyses highlighted immune response, lipid metabolism and inflammation regulation were linked to obesity. Machine learning models demonstrated robust predictive performance with area under the curves (AUC) of 0.820 (SVM), 0.805 (RF), 0.791 (KNN) and 0.819 (Adaboost). All 16 proteins correlated with obesity-related risk factors such as blood pressure and lipid levels. MR analysis identified AFM, CRP and CFH as causal and potentially modifiable proteins. CONCLUSIONS The protein signatures identified in our study showed promising predictive potential for obesity. These findings warrant further investigation to evaluate their clinical applicability, offering insights into obesity prevention and treatment strategies.
Collapse
Affiliation(s)
- Jiefei Niu
- Research Unit of Molecular EpidemiologyHelmholtz Zentrum MünchenNeuherbergGermany
- Institute of EpidemiologyHelmholtz Zentrum MünchenNeuherbergGermany
- Faculty of MedicineLudwig‐Maximilians‐University MünchenMunichGermany
| | - Jonathan Adam
- Research Unit of Molecular EpidemiologyHelmholtz Zentrum MünchenNeuherbergGermany
- Institute of EpidemiologyHelmholtz Zentrum MünchenNeuherbergGermany
- German Center for Diabetes Research (DZD)NeuherbergGermany
| | - Thomas Skurk
- School of MedicineTechnical University of MunichMunichGermany
- ZIEL Institute for Food & Health, Core Facility Human StudiesTechnical University of MunichFreisingGermany
| | - Jochen Seissler
- German Center for Diabetes Research (DZD)NeuherbergGermany
- Medizinische Klinik und Poliklinik IV, Klinikum der Ludwig‐Maximilians‐Universität, and Clinical Cooperation Group DiabetesLudwig‐Maximilians‐Universität München, and Helmholtz Zentrum MünchenMunichGermany
| | - Qiuling Dong
- Research Unit of Molecular EpidemiologyHelmholtz Zentrum MünchenNeuherbergGermany
- Institute of EpidemiologyHelmholtz Zentrum MünchenNeuherbergGermany
- Faculty of MedicineLudwig‐Maximilians‐University MünchenMunichGermany
| | - Esienanwan Efiong
- Research Unit of Molecular EpidemiologyHelmholtz Zentrum MünchenNeuherbergGermany
- Institute of EpidemiologyHelmholtz Zentrum MünchenNeuherbergGermany
- Faculty of Pharmaceutical, Biomedical and Veterinary Sciences, Department of Pharmaceutical Sciences, Campus Drie EikenUniversiteitsplein 1AntwerpBelgium
- Department of Biochemistry, Faculty of ScienceFederal University of LafiaLafiaNigeria
| | - Christian Gieger
- Research Unit of Molecular EpidemiologyHelmholtz Zentrum MünchenNeuherbergGermany
- Institute of EpidemiologyHelmholtz Zentrum MünchenNeuherbergGermany
- German Center for Diabetes Research (DZD)NeuherbergGermany
| | - Annette Peters
- Institute of EpidemiologyHelmholtz Zentrum MünchenNeuherbergGermany
- German Center for Diabetes Research (DZD)NeuherbergGermany
- Chair of Epidemiology, Institute for Medical Information Processing, Biometry, and Epidemiology (IBE), Faculty of MedicineLudwig‐Maximilians‐University MünchenMunichGermany
| | - Sapna Sharma
- Research Unit of Molecular EpidemiologyHelmholtz Zentrum MünchenNeuherbergGermany
- Institute of EpidemiologyHelmholtz Zentrum MünchenNeuherbergGermany
- German Center for Diabetes Research (DZD)NeuherbergGermany
| | - Harald Grallert
- Research Unit of Molecular EpidemiologyHelmholtz Zentrum MünchenNeuherbergGermany
- Institute of EpidemiologyHelmholtz Zentrum MünchenNeuherbergGermany
- German Center for Diabetes Research (DZD)NeuherbergGermany
| |
Collapse
|
5
|
Yang D, Li J, Mak WY, Zheng A, Zhu X, He Q, Wang Y, Xiang X. PBPK Modeling: Empowering Drug Development and Precision Dosing in China. CPT Pharmacometrics Syst Pharmacol 2025; 14:828-839. [PMID: 39967056 PMCID: PMC12072232 DOI: 10.1002/psp4.70004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2024] [Revised: 01/05/2025] [Accepted: 01/28/2025] [Indexed: 02/20/2025] Open
Abstract
Physiologically based pharmacokinetic (PBPK) modeling, a cornerstone of model-informed drug development and model-informed precision dosing, simulates drug disposition in the human body by integrating physiological, biochemical, and physicochemical parameters. While PBPK modeling has advanced globally since the 1970s, China's adoption of this technology has followed a distinctive path, characterized by accelerated growth over the past 2 decades. This review provides a comprehensive analysis of China's contributions to PBPK modeling, addressing knowledge gaps in publication trends, application domains, and platform preferences. A systematic literature search yielded 266 original PBPK research articles from PubMed up to August 08, 2024. The analysis revealed that drug disposition and drug-drug interaction studies constitute the largest proportion of PBPK analyses in China. Chinese universities and hospitals emerge as the leading contributors to PBPK research among institutions in China. Although established commercial PBPK platform such as GastroPlus and Simcyp remain popular within the Chinese pharmaceutical industry, open-source platforms like PK-Sim are gaining significant traction in PBPK applications across China. This review underscores the transformative potential of PBPK modeling in drug development within China, offering valuable insights into future directions and challenges in the field.
Collapse
Affiliation(s)
- Dongsheng Yang
- Department of Clinical Pharmacy and Pharmacy Administration, School of PharmacyFudan UniversityShanghaiChina
| | - Jian Li
- Center for Drug Evaluation, National Medical Products AdministrationBeijingChina
| | - Wen Yao Mak
- Department of Clinical Pharmacy and Pharmacy Administration, School of PharmacyFudan UniversityShanghaiChina
| | - Aole Zheng
- Department of Clinical Pharmacy and Pharmacy Administration, School of PharmacyFudan UniversityShanghaiChina
| | - Xiao Zhu
- Department of Clinical Pharmacy and Pharmacy Administration, School of PharmacyFudan UniversityShanghaiChina
| | - Qingfeng He
- Department of Clinical Pharmacy and Pharmacy Administration, School of PharmacyFudan UniversityShanghaiChina
| | - Yuzhu Wang
- Center for Drug Evaluation, National Medical Products AdministrationBeijingChina
| | - Xiaoqiang Xiang
- Department of Clinical Pharmacy and Pharmacy Administration, School of PharmacyFudan UniversityShanghaiChina
- Quzhou Fudan InstituteQuzhouChina
- National Key Laboratory of Advanced Drug Formulations for Overcoming Delivery BarriersShanghaiChina
| |
Collapse
|
6
|
Calzetta L, Pistocchini E, Gholamalishahi S, Grugni L, Cazzola M, Rogliani P. Novel drug discovery strategies for chronic obstructive pulmonary disease: the latest developments. Expert Opin Drug Discov 2025; 20:683-692. [PMID: 40223433 DOI: 10.1080/17460441.2025.2490251] [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: 11/27/2024] [Accepted: 04/03/2025] [Indexed: 04/15/2025]
Abstract
INTRODUCTION The journey from initial drug discovery to approval for respiratory diseases typically spans approximately 10.4 years and cost over $2.8 billion. This intricate process involves five stages: target identification, therapeutic molecule discovery, preclinical testing, clinical trials, and regulatory approval. AREAS COVERED This review examines novel drug discovery strategies for chronic obstructive pulmonary disease (COPD), focusing on advanced in vitro models that replicate human lung conditions for accurate drug testing according to the following search string: discovery AND strategy AND COPD. It explores targeted molecular therapies, structure-based drug design, and drug repurposing approaches facilitated by computational analysis. The significance of personalized medicine in tailoring treatments for diverse COPDs is emphasized, highlighting the complexity of the disease and the necessity of these innovative methodologies to improve therapeutic outcomes. EXPERT OPINION COPD remains a challenging area, with a significant unmet medical need. Despite previous efforts, few effective therapies exist. Innovative in vitro models, targeted molecular therapies, and drug repurposing strategies are showing promise. Emphasizing advanced preclinical models and repurposing existing drugs could transform treatment paradigms, promoting more effective therapies for complex diseases like COPD. These innovations hold potential for enhancing drug discovery efficiency, leading to personalized and precision medicine approaches.
Collapse
Affiliation(s)
- Luigino Calzetta
- Respiratory Disease and Lung Function Unit, Department of Medicine and Surgery, University of Parma, Parma, Italy
| | - Elena Pistocchini
- Unit of Respiratory Medicine, Department of Experimental Medicine, University of Rome "Tor Vergata", Rome, Italy
| | - Shima Gholamalishahi
- Unit of Respiratory Medicine, Department of Experimental Medicine, University of Rome "Tor Vergata", Rome, Italy
| | - Lucia Grugni
- Unit of Respiratory Medicine, Department of Experimental Medicine, University of Rome "Tor Vergata", Rome, Italy
| | - Mario Cazzola
- Unit of Respiratory Medicine, Department of Experimental Medicine, University of Rome "Tor Vergata", Rome, Italy
| | - Paola Rogliani
- Unit of Respiratory Medicine, Department of Experimental Medicine, University of Rome "Tor Vergata", Rome, Italy
| |
Collapse
|
7
|
Takács G, Balogh GT, Kiss R. A data-driven journey using results from target-based drug discovery for target deconvolution in phenotypic screening. RSC Med Chem 2025:d4md01051e. [PMID: 40352671 PMCID: PMC12062751 DOI: 10.1039/d4md01051e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2024] [Accepted: 04/12/2025] [Indexed: 05/14/2025] Open
Abstract
In drug discovery, various approaches exist to find compounds that alter the different states in living organisms. There are two fundamental discovery strategies regarding the mechanism of action: target-based and phenotypic methods. Both have strengths and weaknesses in assay development, target selection, target validation and structure optimization. While phenotypic screening can identify chemical starting points with the desired phenotype, it is typically difficult to carry out efficient, structure-based optimization without confirming the mechanism of action of such hits. It is therefore critical to uncover the targets behind the phenotype. Target deconvolution is typically carried out by a set of highly selective compounds, where each ligand is associated with a particular target. Hits of such a high-selectivity set can provide valuable information on the phenotype's underlying targets and may also enable novel target-based therapeutic strategies. Consequently, there is a continuously high demand for novel highly-selective tool compounds for target deconvolution. In this work, the ChEMBL database, comprising over 20 million bioactivity data, was mined to identify the most selective novel ligands for a diverse set of targets. A novel method for the automated selection of such high-selectivity ligands is presented. Using these high-selectivity compounds in phenotypic screening campaigns can provide a valuable preliminary direction during target deconvolution. 87 representative compounds were purchased and screened against 60 cancer cell lines. Several compounds were found to possess selective inhibition of cell growth of a few distinct cell lines. The phenotypic assay results, along with the nanomolar activities of individual proteins obtained from the ChEMBL database suggest some novel mechanisms of action for anti-cancer drug discovery.
Collapse
Affiliation(s)
- Gergely Takács
- Department of Chemical and Environmental Process Engineering, Faculty of Chemical Technology and Biotechnology, Budapest University of Technology and Economics Műegyetem Rakpart 3 Budapest 1111 Hungary
- Mcule.com Kft Bartók Béla út 105-113 Budapest 1115 Hungary
| | - György T Balogh
- Department of Chemical and Environmental Process Engineering, Faculty of Chemical Technology and Biotechnology, Budapest University of Technology and Economics Műegyetem Rakpart 3 Budapest 1111 Hungary
- University Pharmacy Department of Pharmacy Administration, Semmelweis University 7-9 Hőgyes Street 1092 Budapest Hungary
| | - Róbert Kiss
- Mcule.com Kft Bartók Béla út 105-113 Budapest 1115 Hungary
| |
Collapse
|
8
|
Fujimoto Y, Mizuno K, Nakamura Y, Arai M, Kotoku N. Synthesis and Evaluation of Antitumor and Anti-Angiogenesis Activity of Pyrone- or Pyridone-Embedded Analogs of Cortistatin A. Mar Drugs 2025; 23:179. [PMID: 40278300 PMCID: PMC12029069 DOI: 10.3390/md23040179] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2025] [Revised: 04/18/2025] [Accepted: 04/18/2025] [Indexed: 04/26/2025] Open
Abstract
Simplified analogs of cortistatin A were synthesized and biologically evaluated to develop novel antitumor substances that target angiogenesis. To analyze the effect of substituents at positions corresponding to C-2 and/or C-4 of the A-ring, various pyrone- or pyridone-embedded analogs were designed and synthesized. Among the prepared analogs, the pyridone analog 19 bearing a methyl group at C-2 and a hydroxyl group at C-4 showed potent and selective growth inhibitory activity against human umbilical vein endothelial cells (HUVECs, IC50 = 0.001 µM, selective index over that against human epidermoid carcinoma KB3-1 cells = 6400), exceeding those of natural products. The analog 19 of oral administration exhibited excellent in vivo antitumor activity in mice subcutaneously inoculated with sarcoma S180 cells.
Collapse
Affiliation(s)
- Yuri Fujimoto
- College of Pharmaceutical Sciences, Ritsumeikan University, 1-1-1 Noji-higashi, Kusatsu 525-8577, Shiga, Japan; (Y.F.); (Y.N.)
| | - Kanako Mizuno
- Graduate School of Pharmaceutical Sciences, Osaka University, 1-6 Yamadaoka, Suita 565-0871, Osaka, Japan; (K.M.); (M.A.)
| | - Yuta Nakamura
- College of Pharmaceutical Sciences, Ritsumeikan University, 1-1-1 Noji-higashi, Kusatsu 525-8577, Shiga, Japan; (Y.F.); (Y.N.)
| | - Masayoshi Arai
- Graduate School of Pharmaceutical Sciences, Osaka University, 1-6 Yamadaoka, Suita 565-0871, Osaka, Japan; (K.M.); (M.A.)
| | - Naoyuki Kotoku
- College of Pharmaceutical Sciences, Ritsumeikan University, 1-1-1 Noji-higashi, Kusatsu 525-8577, Shiga, Japan; (Y.F.); (Y.N.)
- Graduate School of Pharmaceutical Sciences, Osaka University, 1-6 Yamadaoka, Suita 565-0871, Osaka, Japan; (K.M.); (M.A.)
| |
Collapse
|
9
|
Wong F, Omori S, Li A, Krishnan A, Lach RS, Rufo J, Wilson MZ, Collins JJ. An explainable deep learning platform for molecular discovery. Nat Protoc 2025; 20:1020-1056. [PMID: 39653800 DOI: 10.1038/s41596-024-01084-x] [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: 03/11/2024] [Accepted: 09/26/2024] [Indexed: 04/10/2025]
Abstract
Deep learning approaches have been increasingly applied to the discovery of novel chemical compounds. These predictive approaches can accurately model compounds and increase true discovery rates, but they are typically black box in nature and do not generate specific chemical insights. Explainable deep learning aims to 'open up' the black box by providing generalizable and human-understandable reasoning for model predictions. These explanations can augment molecular discovery by identifying structural classes of compounds with desired activity in lieu of lone compounds. Additionally, these explanations can guide hypothesis generation and make searching large chemical spaces more efficient. Here we present an explainable deep learning platform that enables vast chemical spaces to be mined and the chemical substructures underlying predicted activity to be identified. The platform relies on Chemprop, a software package implementing graph neural networks as a deep learning model architecture. In contrast to similar approaches, graph neural networks have been shown to be state of the art for molecular property prediction. Focusing on discovering structural classes of antibiotics, this protocol provides guidelines for experimental data generation, model implementation and model explainability and evaluation. This protocol does not require coding proficiency or specialized hardware, and it can be executed in as little as 1-2 weeks, starting from data generation and ending in the testing of model predictions. The platform can be broadly applied to discover structural classes of other small molecules, including anticancer, antiviral and senolytic drugs, as well as to discover structural classes of inorganic molecules with desired physical and chemical properties.
Collapse
Affiliation(s)
- Felix Wong
- Infectious Disease and Microbiome Program, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Institute for Medical Engineering and Science and Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
- Integrated Biosciences, Inc., Redwood City, CA, USA
| | - Satotaka Omori
- Infectious Disease and Microbiome Program, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Integrated Biosciences, Inc., Redwood City, CA, USA
| | - Alicia Li
- Integrated Biosciences, Inc., Redwood City, CA, USA
| | - Aarti Krishnan
- Infectious Disease and Microbiome Program, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Institute for Medical Engineering and Science and Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Ryan S Lach
- Integrated Biosciences, Inc., Redwood City, CA, USA
| | - Joseph Rufo
- Center for BioEngineering, University of California Santa Barbara, Santa Barbara, CA, USA
- Biomolecular Science and Engineering Program, University of California Santa Barbara, Santa Barbara, CA, USA
- Department of Molecular, Cellular, and Developmental Biology, University of California Santa Barbara, Santa Barbara, CA, USA
- Neuroscience Research Institute, University of California Santa Barbara, Santa Barbara, CA, USA
| | - Maxwell Z Wilson
- Integrated Biosciences, Inc., Redwood City, CA, USA
- Center for BioEngineering, University of California Santa Barbara, Santa Barbara, CA, USA
- Biomolecular Science and Engineering Program, University of California Santa Barbara, Santa Barbara, CA, USA
- Department of Molecular, Cellular, and Developmental Biology, University of California Santa Barbara, Santa Barbara, CA, USA
- Neuroscience Research Institute, University of California Santa Barbara, Santa Barbara, CA, USA
| | - James J Collins
- Infectious Disease and Microbiome Program, Broad Institute of MIT and Harvard, Cambridge, MA, USA.
- Institute for Medical Engineering and Science and Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA.
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, USA.
| |
Collapse
|
10
|
Ji RL, Tao YX. Biased signaling in drug discovery and precision medicine. Pharmacol Ther 2025; 268:108804. [PMID: 39904401 DOI: 10.1016/j.pharmthera.2025.108804] [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: 10/28/2024] [Revised: 01/10/2025] [Accepted: 01/21/2025] [Indexed: 02/06/2025]
Abstract
Receptors are crucial for converting chemical and environmental signals into cellular responses, making them prime targets in drug discovery, with about 70% of drugs targeting these receptors. Biased signaling, or functional selectivity, has revolutionized drug development by enabling precise modulation of receptor signaling pathways. This concept is more firmly established in G protein-coupled receptor and has now been applied to other receptor types, including ion channels, receptor tyrosine kinases, and nuclear receptors. Advances in structural biology have further refined our understanding of biased signaling. This targeted approach enhances therapeutic efficacy and potentially reduces side effects. Numerous biased drugs have been developed and approved as therapeutics to treat various diseases, demonstrating their significant therapeutic potential. This review provides a comprehensive overview of biased signaling in drug discovery and disease treatment, highlighting recent advancements and exploring the therapeutic potential of these innovative modulators across various diseases.
Collapse
Affiliation(s)
- Ren-Lei Ji
- Department of Anatomy, Physiology and Pharmacology, College of Veterinary Medicine, Auburn University, Auburn, AL 36849, United States.
| | - Ya-Xiong Tao
- Department of Anatomy, Physiology and Pharmacology, College of Veterinary Medicine, Auburn University, Auburn, AL 36849, United States.
| |
Collapse
|
11
|
Ye Q, Zeng Y, Jiang L, Kang Y, Pan P, Chen J, Deng Y, Zhao H, He S, Hou T, Hsieh C. A Knowledge-Guided Graph Learning Approach Bridging Phenotype- and Target-Based Drug Discovery. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2412402. [PMID: 40047372 PMCID: PMC12021103 DOI: 10.1002/advs.202412402] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/05/2024] [Revised: 01/24/2025] [Indexed: 04/26/2025]
Abstract
Discovering therapeutic molecules requires the integration of both phenotype-based drug discovery (PDD) and target-based drug discovery (TDD). However, this integration remains challenging due to the inherent heterogeneity, noise, and bias present in biomedical data. In this study, Knowledge-Guided Drug Relational Predictor (KGDRP), a graph representation learning approach is developed that effectively integrates multimodal biomedical data, including network data containing biological system information, gene expression data, and sequence data that incorporates chemical molecular structures, all within a heterogeneous graph (HG) structure. By incorporating biomedical HG (BioHG) into a heterogeneous graph neural network (HGNN)-based architecture, KGDRP exhibits a remarkable 12% improvement compared to previous methods in real-world screening scenarios. Notably, the biology-informed representation, derived from KGDRP, significantly enhance target prioritization by 26% in drug target discovery. Furthermore, zero-shot evaluation on COVID-19 exhibited a notably higher success rate in identifying diverse potential drugs. The utilization of BioHG facilitates a unique KGDRP-based analysis of cell-target-drug interactions, thereby enabling the elucidation of drug mechanisms. Overall, KGDRP provides a robust infrastructure for the seamlessly integration of multimodal data and biomedical networks, effectively accelerating PDD, guiding therapeutic target discovery, and ultimately expediting therapeutic molecule discovery.
Collapse
Affiliation(s)
- Qing Ye
- College of Control Science and EngineeringZhejiang UniversityHangzhouZhejiang310027China
- College of Pharmaceutical SciencesZhejiang UniversityHangzhouZhejiang310058China
| | - Yundian Zeng
- College of Control Science and EngineeringZhejiang UniversityHangzhouZhejiang310027China
- College of Pharmaceutical SciencesZhejiang UniversityHangzhouZhejiang310058China
| | - Linlong Jiang
- College of Pharmaceutical SciencesZhejiang UniversityHangzhouZhejiang310058China
| | - Yu Kang
- College of Pharmaceutical SciencesZhejiang UniversityHangzhouZhejiang310058China
| | - Peichen Pan
- College of Pharmaceutical SciencesZhejiang UniversityHangzhouZhejiang310058China
| | - Jiming Chen
- College of Control Science and EngineeringZhejiang UniversityHangzhouZhejiang310027China
| | - Yafeng Deng
- CarbonSilicon AI Technology Co., LtdHangzhouZhejiang310018China
| | - Haitao Zhao
- Center for Intelligent and Biomimetic SystemsShenzhen Institutes of Advanced TechnologyChinese Academy of SciencesShenzhenGuangdong440305China
| | - Shibo He
- College of Control Science and EngineeringZhejiang UniversityHangzhouZhejiang310027China
| | - Tingjun Hou
- College of Pharmaceutical SciencesZhejiang UniversityHangzhouZhejiang310058China
| | - Chang‐Yu Hsieh
- College of Pharmaceutical SciencesZhejiang UniversityHangzhouZhejiang310058China
| |
Collapse
|
12
|
Ölander M, Rea Vázquez D, Meier K, Singh A, Silva de Sousa A, Puértolas-Balint F, Milivojevic M, Mooij L, Fredlund J, Calpe Bosch E, Rayón Díaz M, Lundgren M, van der Wal K, Zhu S, Mateus A, Schroeder BO, Lohman JR, Sixt BS. A multi-strategy antimicrobial discovery approach reveals new ways to treat Chlamydia. PLoS Biol 2025; 23:e3003123. [PMID: 40299795 PMCID: PMC12040169 DOI: 10.1371/journal.pbio.3003123] [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: 02/21/2025] [Accepted: 03/19/2025] [Indexed: 05/01/2025] Open
Abstract
While the excessive use of broad-spectrum antibiotics is a major driver of the global antibiotic resistance crisis, more selective therapies remain unavailable for the majority of bacterial pathogens. This includes the obligate intracellular bacterial pathogens of the genus Chlamydia, which cause millions of urogenital, ocular, and respiratory infections each year. Conducting a comprehensive search of the chemical space for novel antichlamydial activities, we identified over 60 compounds that are chemically diverse, structurally distinct from known antibiotics, non-toxic to human cells, and highly potent in preventing the growth of Chlamydia trachomatis in cell cultures. Some blocked C. trachomatis development reversibly, while others eradicated both established and persistent infections in a bactericidal manner. The top molecules displayed compelling selectivity, yet broad activity against diverse Chlamydia strains and species, including both urogenital and ocular serovars of C. trachomatis, as well as Chlamydia muridarum and Chlamydia caviae. Some compounds also displayed synergies with clinically used antibiotics. Critically, we found the most potent antichlamydial compound to inhibit fatty acid biosynthesis via covalent binding to the active site of Chlamydia FabH, identifying a new mechanism of FabH inhibition and highlighting a possible way to selectively treat Chlamydia infections.
Collapse
Affiliation(s)
- Magnus Ölander
- Department of Molecular Biology, Umeå University, Umeå, Sweden
- The Laboratory for Molecular Infection Medicine Sweden (MIMS), Umeå University, Umeå, Sweden
- Umeå Centre for Microbial Research (UCMR), Umeå University, Umeå, Sweden
| | - Daniel Rea Vázquez
- Department of Molecular Biology, Umeå University, Umeå, Sweden
- The Laboratory for Molecular Infection Medicine Sweden (MIMS), Umeå University, Umeå, Sweden
- Umeå Centre for Microbial Research (UCMR), Umeå University, Umeå, Sweden
| | - Karsten Meier
- Department of Molecular Biology, Umeå University, Umeå, Sweden
- The Laboratory for Molecular Infection Medicine Sweden (MIMS), Umeå University, Umeå, Sweden
- Umeå Centre for Microbial Research (UCMR), Umeå University, Umeå, Sweden
| | - Aakriti Singh
- Department of Molecular Biology, Umeå University, Umeå, Sweden
- The Laboratory for Molecular Infection Medicine Sweden (MIMS), Umeå University, Umeå, Sweden
- Umeå Centre for Microbial Research (UCMR), Umeå University, Umeå, Sweden
| | - Amanda Silva de Sousa
- Department of Biochemistry and Molecular Biology, Michigan State University, East Lansing, Michigan, United States of America
| | - Fabiola Puértolas-Balint
- Department of Molecular Biology, Umeå University, Umeå, Sweden
- The Laboratory for Molecular Infection Medicine Sweden (MIMS), Umeå University, Umeå, Sweden
- Umeå Centre for Microbial Research (UCMR), Umeå University, Umeå, Sweden
| | - Milica Milivojevic
- Department of Molecular Biology, Umeå University, Umeå, Sweden
- The Laboratory for Molecular Infection Medicine Sweden (MIMS), Umeå University, Umeå, Sweden
- Umeå Centre for Microbial Research (UCMR), Umeå University, Umeå, Sweden
| | - Lieke Mooij
- Department of Molecular Biology, Umeå University, Umeå, Sweden
- The Laboratory for Molecular Infection Medicine Sweden (MIMS), Umeå University, Umeå, Sweden
- Umeå Centre for Microbial Research (UCMR), Umeå University, Umeå, Sweden
| | - Johanna Fredlund
- Department of Molecular Biology, Umeå University, Umeå, Sweden
- The Laboratory for Molecular Infection Medicine Sweden (MIMS), Umeå University, Umeå, Sweden
- Umeå Centre for Microbial Research (UCMR), Umeå University, Umeå, Sweden
| | - Eduard Calpe Bosch
- Department of Molecular Biology, Umeå University, Umeå, Sweden
- The Laboratory for Molecular Infection Medicine Sweden (MIMS), Umeå University, Umeå, Sweden
- Umeå Centre for Microbial Research (UCMR), Umeå University, Umeå, Sweden
| | - María Rayón Díaz
- Department of Molecular Biology, Umeå University, Umeå, Sweden
- The Laboratory for Molecular Infection Medicine Sweden (MIMS), Umeå University, Umeå, Sweden
- Umeå Centre for Microbial Research (UCMR), Umeå University, Umeå, Sweden
| | - Moa Lundgren
- Department of Molecular Biology, Umeå University, Umeå, Sweden
- The Laboratory for Molecular Infection Medicine Sweden (MIMS), Umeå University, Umeå, Sweden
- Umeå Centre for Microbial Research (UCMR), Umeå University, Umeå, Sweden
| | - Karin van der Wal
- Department of Molecular Biology, Umeå University, Umeå, Sweden
- The Laboratory for Molecular Infection Medicine Sweden (MIMS), Umeå University, Umeå, Sweden
- Umeå Centre for Microbial Research (UCMR), Umeå University, Umeå, Sweden
| | - Shaochun Zhu
- The Laboratory for Molecular Infection Medicine Sweden (MIMS), Umeå University, Umeå, Sweden
- Umeå Centre for Microbial Research (UCMR), Umeå University, Umeå, Sweden
- Department of Chemistry, Umeå University, Umeå, Sweden
| | - André Mateus
- The Laboratory for Molecular Infection Medicine Sweden (MIMS), Umeå University, Umeå, Sweden
- Umeå Centre for Microbial Research (UCMR), Umeå University, Umeå, Sweden
- Department of Chemistry, Umeå University, Umeå, Sweden
| | - Bjoern O. Schroeder
- Department of Molecular Biology, Umeå University, Umeå, Sweden
- The Laboratory for Molecular Infection Medicine Sweden (MIMS), Umeå University, Umeå, Sweden
- Umeå Centre for Microbial Research (UCMR), Umeå University, Umeå, Sweden
| | - Jeremy R. Lohman
- Department of Biochemistry and Molecular Biology, Michigan State University, East Lansing, Michigan, United States of America
| | - Barbara S. Sixt
- Department of Molecular Biology, Umeå University, Umeå, Sweden
- The Laboratory for Molecular Infection Medicine Sweden (MIMS), Umeå University, Umeå, Sweden
- Umeå Centre for Microbial Research (UCMR), Umeå University, Umeå, Sweden
| |
Collapse
|
13
|
Mohammed I, Sagurthi SR. Current Approaches and Strategies Applied in First-in-class Drug Discovery. ChemMedChem 2025; 20:e202400639. [PMID: 39648151 DOI: 10.1002/cmdc.202400639] [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: 08/16/2024] [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
|
14
|
Samowitz P, Radnai L, Vaissiere T, Michaelson SD, Rojas C, Mitchell R, Kilinc M, Edwards A, Shumate J, Hawkins R, Fernandez-Vega V, Spicer TP, Scampavia L, Kamenecka T, Miller CA, Rumbaugh G. The Endo-GeneScreen Platform Identifies Drug-Like Probes that Regulate Endogenous Protein Levels within Physiological Contexts. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.13.643156. [PMID: 40161629 PMCID: PMC11952490 DOI: 10.1101/2025.03.13.643156] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
Traditional phenotypic drug discovery platforms have suffered from poor scalability and a lack of mechanistic understanding of newly discovered phenotypic probes. To address this, we created Endo- GeneScreen (EGS), a high-throughput enabled screening platform that identifies bioactive small molecules capable of regulating endogenous protein expression encoded by any preselected target gene within a biologically appropriate context. As a proof-of-concept, EGS successfully identified drug candidates that up-regulate endogenous expression of neuronal Syngap1, a gene that causes a neurodevelopmental disorder when haploinsufficient. For example, SR-1815, a previously unknown and undescribed kinase inhibitor, alleviated major cellular consequences of Syngap1 loss-of-function by restoring normal SynGAP protein levels and dampening neuronal hyperactivity within haploinsufficient neurons. Moreover, we demonstrate that EGS assays accelerate preclinical development of identified drug candidates and facilitate mode-of-action deconvolution studies. Thus, EGS identifies first-in-class bioactive small molecule probes that promote biological discovery and precision therapeutic development.
Collapse
|
15
|
Wen H, Lu H, Zhou Z, Sun K, Huang Y, Zeng J, Wang Y, Luo L, Xu C, Xu J, Zhang X, Wang X, Eeltink S, Zhang B. Large Scale Printing of Robust HPLC Medium via Layer-by-Layer Stereolithography. Anal Chem 2025; 97:5014-5021. [PMID: 39947930 DOI: 10.1021/acs.analchem.4c05587] [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: 03/12/2025]
Abstract
The manufacture of high-performance liquid chromatography (HPLC) medium has long been viewed as an art rather than science; this raised a great challenge in securing separation consistency, method transferability, and scaling-up in purification of biomolecules. Herein, we report a large scale layer-by-layer manufacturing strategy for a high performance chromatography medium utilizing 3D-printing technology. Combining stereolithography 3D printing and porogenic chemistry, the strategy enables parallel production of high-performance separation medium in diverse scales, shapes, and throughput. Between 1,000 printed devices, high performance consistency was demonstrated by column-to-column and batch-to-batch reproducibility (coefficient of variation of retention time, 2.04%). Fast separations of intact proteins were realized in reversed-phase chromatography: within 1 min, resolution > 1.5 was achieved, and nondenatured antibody separation was realized in hydrophobic interaction chromatography. Purification of native proteins was directly amplified by 3 orders of magnitude: 12 mg of hemeproteins was isolated in 8 min at negligible scaling-up cost, supporting liter-scale processing of fermentation within 7 h on one 20 mm i.d. printed column. With advantages in automatic and parallel production capacity, high-fidelity microstructure across dimensions, and highly efficient method transfer and scaling-up, the stereolithographically printed high performance chromatography medium may open a new path to speeding up separation and purification processes from primary analysis to mass-purification of biomolecular entities, as demanded in the biosynthesis and pharmaceutical industries.
Collapse
Affiliation(s)
- Hanrong Wen
- Department of Chemistry and the MOE Key Laboratory of Spectrochemical Analysis & Instrumentation, College of Chemistry and Chemical Engineering, State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, Xiamen University, Xiamen 361005, China
- Department of Chemical Engineering, Vrije Universiteit Brussel, Brussels 1050, Belgium
| | - Haonan Lu
- Department of Chemistry and the MOE Key Laboratory of Spectrochemical Analysis & Instrumentation, College of Chemistry and Chemical Engineering, State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, Xiamen University, Xiamen 361005, China
| | - Zhuoheng Zhou
- Department of Chemistry and the MOE Key Laboratory of Spectrochemical Analysis & Instrumentation, College of Chemistry and Chemical Engineering, State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, Xiamen University, Xiamen 361005, China
- Department of Chemical Engineering, Vrije Universiteit Brussel, Brussels 1050, Belgium
| | - Kaiyue Sun
- Department of Chemistry and the MOE Key Laboratory of Spectrochemical Analysis & Instrumentation, College of Chemistry and Chemical Engineering, State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, Xiamen University, Xiamen 361005, China
| | - Yinjia Huang
- Department of Chemistry and the MOE Key Laboratory of Spectrochemical Analysis & Instrumentation, College of Chemistry and Chemical Engineering, State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, Xiamen University, Xiamen 361005, China
| | - Juxing Zeng
- Department of Chemistry and the MOE Key Laboratory of Spectrochemical Analysis & Instrumentation, College of Chemistry and Chemical Engineering, State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, Xiamen University, Xiamen 361005, China
| | - Yuchen Wang
- Department of Chemistry and the MOE Key Laboratory of Spectrochemical Analysis & Instrumentation, College of Chemistry and Chemical Engineering, State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, Xiamen University, Xiamen 361005, China
| | - Lianzhong Luo
- Fujian Province Universities and Colleges Engineering Research Center for Marine Biopharmaceutical Resource Utilization, Xiamen Medical College, Xiamen 361023, China
| | - Chen Xu
- HaoQi Separation & Purification Technologies, Xiamen 361102, China
| | - Jianzhong Xu
- HaoQi Separation & Purification Technologies, Xiamen 361102, China
| | - Xin Zhang
- Anhui Wanyi Science and Technology Co. Ltd, Hefei 230088, China
| | | | - Sebastiaan Eeltink
- Department of Chemical Engineering, Vrije Universiteit Brussel, Brussels 1050, Belgium
| | - Bo Zhang
- Department of Chemistry and the MOE Key Laboratory of Spectrochemical Analysis & Instrumentation, College of Chemistry and Chemical Engineering, State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, Xiamen University, Xiamen 361005, China
| |
Collapse
|
16
|
Qiao Y, Liang J, Jiang D. State of the ART: Drug Screening Reveals Artesunate as a Promising Anti-Fibrosis Therapy. JOURNAL OF RESPIRATORY BIOLOGY AND TRANSLATIONAL MEDICINE 2025; 2:10023. [PMID: 39925974 PMCID: PMC11800322 DOI: 10.70322/jrbtm.2024.10023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/11/2025]
Abstract
Fibrosis is a progressive pathological process that severely impairs normal organ function. Current treatments for fibrosis are extremely limited, with no curative approaches available. In a recent article published in Cell, Zhang and colleagues employed drug screening using ACTA2 reporter iPSC-derived cardiac fibroblasts and identified artesunate as a potent antifibrotic drug by targeting MD2/TLR4 signaling. This study provides new insights into strategies for exploiting existing drugs to treat fibrosis.
Collapse
Affiliation(s)
- Yujie Qiao
- Division of Pulmonary, Women’s Guild Lung Institute,
Department of Medicine, Los Angeles, CA 90048, USA
- Department of Respiratory and Critical Care Medicine,
Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Jiurong Liang
- Division of Pulmonary, Women’s Guild Lung Institute,
Department of Medicine, Los Angeles, CA 90048, USA
| | - Dianhua Jiang
- Division of Pulmonary, Women’s Guild Lung Institute,
Department of Medicine, Los Angeles, CA 90048, USA
- Department of Biomedical Sciences, Cedars-Sinai Medical
Center, Los Angeles, CA 90048, USA
| |
Collapse
|
17
|
Zhao Y, Yuan C, Shi Y, Liu X, Luo L, Zhang L, Pešić M, Yao H, Li L. Drug screening approaches for small-molecule compounds in cancer-targeted therapy. J Drug Target 2025; 33:368-383. [PMID: 39575843 DOI: 10.1080/1061186x.2024.2427185] [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: 05/30/2024] [Revised: 09/30/2024] [Accepted: 10/27/2024] [Indexed: 02/08/2025]
Abstract
Small-molecule compounds exhibit distinct pharmacological properties and clinical effectiveness. Over the past decade, advances in covalent drug discovery have led to successful small-molecule drugs, such as EGFR, BTK, and KRAS (G12C) inhibitors, for cancer therapy. Researchers are paying more attention to refining drug screening methods aiming for high throughput, fast speed, high specificity, and accuracy. Therefore, the discovery and development of small-molecule drugs has been facilitated by significantly reducing screening time and financial resources, and increasing promising lead compounds compared with traditional methods. This review aims to introduce classical and emerging methods for screening small-molecule compounds in targeted cancer therapy. It includes classification, principles, advantages, disadvantages, and successful applications, serving as valuable references for subsequent researchers.
Collapse
Affiliation(s)
- Yelin Zhao
- State Key Laboratory of Respiratory Health and Multimorbidity, NHC Key Laboratory of Biotechnology for Microbial Drugs, Department of Oncology, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Chenyu Yuan
- State Key Laboratory of Respiratory Health and Multimorbidity, NHC Key Laboratory of Biotechnology for Microbial Drugs, Department of Oncology, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yuchen Shi
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Xiaohong Liu
- Guang'anmen Hospital, Chinese Academy of Chinese Medical Sciences, Xicheng District, Beijing, China
| | - Liaoxin Luo
- School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, China
| | - Li Zhang
- State Key Laboratory of Respiratory Health and Multimorbidity, NHC Key Laboratory of Biotechnology for Microbial Drugs, Department of Oncology, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Milica Pešić
- Department of Neurobiology, Institute for Biological Research, 'Siniša Stanković'- National Institute of the Republic of Serbia, University of Belgrade, Belgrade, Serbia
| | - Hongjuan Yao
- State Key Laboratory of Respiratory Health and Multimorbidity, NHC Key Laboratory of Biotechnology for Microbial Drugs, Department of Oncology, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Liang Li
- State Key Laboratory of Respiratory Health and Multimorbidity, NHC Key Laboratory of Biotechnology for Microbial Drugs, Department of Oncology, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
18
|
Sahu MR, Ingale SR, Kontham R. Total synthesis of linear lipodepsipeptide kavaratamide A and its C25-epimer. Org Biomol Chem 2025; 23:1819-1822. [PMID: 39807835 DOI: 10.1039/d4ob01970a] [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: 01/16/2025]
Abstract
We report the stereoselective total synthesis of kavaratamide A, a linear lipodepsipeptide from the cyanobacterium Moorena bouillonii (collected in Kavaratti, India), and its unnatural C25-epimer. The convergent approach employs Keck asymmetric allylation to construct the chiral β-hydroxy carboxylic acid fragment [(3S)-HDA; 3-hydroxydecanoic acid], while the peptide unit was assembled from L-Val, N-Me-L-Ala, (S)-Hiva, and (S)-iPr-O-Me-pyr using well-orchestrated coupling methods to prevent racemization. Modifications to the Keck allylation conditions enabled the synthesis of the C25-epimer with good yield. Cytotoxicity of kavaratamide A and C25-epi-kavaratamide A, assessed using the MTT assay, demonstrated moderate activity against HepG2 and PANC-1 cell lines.
Collapse
Affiliation(s)
- Manas Ranjan Sahu
- Organic Chemistry Division, CSIR-National Chemical Laboratory, Dr Homi Bhabha Road, Pune-411008, India.
| | - Sudhir R Ingale
- Organic Chemistry Division, CSIR-National Chemical Laboratory, Dr Homi Bhabha Road, Pune-411008, India.
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad-201002, India
| | - Ravindar Kontham
- Organic Chemistry Division, CSIR-National Chemical Laboratory, Dr Homi Bhabha Road, Pune-411008, India.
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad-201002, India
| |
Collapse
|
19
|
Zhang S, Liu K, Liu Y, Hu X, Gu X. The role and application of bioinformatics techniques and tools in drug discovery. Front Pharmacol 2025; 16:1547131. [PMID: 40017606 PMCID: PMC11865229 DOI: 10.3389/fphar.2025.1547131] [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: 12/17/2024] [Accepted: 01/27/2025] [Indexed: 03/01/2025] Open
Abstract
The process of drug discovery and development is both lengthy and intricate, demanding a substantial investment of time and financial resources. Bioinformatics techniques and tools can not only accelerate the identification of drug targets and the screening and refinement of drug candidates, but also facilitate the characterization of side effects and the prediction of drug resistance. High-throughput data from genomics, transcriptomics, proteomics, and metabolomics make significant contributions to mechanics-based drug discovery and drug reuse. This paper summarizes bioinformatics technologies and tools in drug research and development and their roles and applications in drug research and development, aiming to provide references for the development of new drugs and the realization of precision medicine.
Collapse
Affiliation(s)
- Shujun Zhang
- Department of Infectious Diseases, The First Affiliated Hospital, College of Clinical Medicine, Henan University of Science and Technology, Luoyang, Henan, China
| | - Kaijie Liu
- Department of Infectious Diseases, The First Affiliated Hospital, College of Clinical Medicine, Henan University of Science and Technology, Luoyang, Henan, China
| | - Yafeng Liu
- Department of Infectious Diseases, The First Affiliated Hospital, College of Clinical Medicine, Henan University of Science and Technology, Luoyang, Henan, China
| | - Xinjun Hu
- Department of Infectious Diseases, The First Affiliated Hospital, College of Clinical Medicine, Henan University of Science and Technology, Luoyang, Henan, China
- Henan Medical Key Laboratory of Gastrointestinal Microecology and Hepatology, Luoyang, China
| | - Xinyu Gu
- Department of Oncology, The First Affiliated Hospital, College of Clinical Medicine, Henan University of Science and Technology, Luoyang, Henan, China
| |
Collapse
|
20
|
Ilg MM, Lapthorn AR, Harding SL, Minhas T, Koduri G, Bustin SA, Cellek S. Development of a phenotypic screening assay to measure activation of cancer-associated fibroblasts. Front Pharmacol 2025; 16:1526495. [PMID: 40017592 PMCID: PMC11865240 DOI: 10.3389/fphar.2025.1526495] [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: 11/11/2024] [Accepted: 01/27/2025] [Indexed: 03/01/2025] Open
Abstract
Background In cancer metastasis, tumor cells condition distant tissues to create a supportive environment, or metastatic niche, by driving the activation of cancer-associated fibroblasts (CAFs). These CAFs remodel the extracellular matrix, creating a microenvironment that supports tumor growth and compromises immune cell function, enabling cancer cells to evade immune detection. Consequently, targeting the activation of CAFs has been proposed as a therapeutic strategy to hinder metastatic spread. Our objective was to develop the first in vitro phenotypic screening assay capable of assessing this activation process. Methods Human primary lung fibroblasts were co-cultured with highly invasive breast cancer cells (MDA-MB-231) to identify changes in the expression of selected genes using RT-qPCR. An In-Cell ELISA (ICE)-based assay using human lung fibroblasts, MDA-MB-231 cells and human monocytes (THP-1 cells) was developed to measure the activation of CAFs. Another ELISA assay was used to measure released osteopontin. Results When lung fibroblast were co-cultured with MDA-MB-231 cells, among the 10 selected genes, the genes for osteopontin (SPP1), insulin like growth factor 1 (IGF1), periostin (POSTN) and α-smooth muscle actin (α-SMA, ACTA2) elicited the greatest fold change (55-, 37-, 8- and 5-fold respectively). Since osteopontin, IGF-1 and periostin are secreted proteins and α-SMA is an intracellular cytoskeleton protein, α-SMA was chosen to be the readout biomarker for the ICE assay. When fibroblasts were co-cultured with MDA-MB-231 cells and monocytes in the 96 well ICE assay, α-SMA expression was increased 2.3-fold yielding a robust Z' of 0.56. A secondary, low throughput assay was developed by measuring the release of osteopontin which showed a 6-fold increase when fibroblasts were co-cultured with MDA-MB-231 cells and monocytes. Discussion This phenotypic assay is the first to measure the activation of CAFs in a 96-well format, making it suitable for medium-to high-throughput screening of potential therapeutic compounds. By focusing on observable cellular phenotypic changes rather than targeting specific molecular pathways, this assay allows for a broader and unbiased identification of compounds capable of modulating CAF activation.
Collapse
Affiliation(s)
- Marcus M. Ilg
- Fibrosis Research Group, Medical Technology Research Centre, Anglia Ruskin University, Chelmsford, United Kingdom
| | - Alice R. Lapthorn
- Fibrosis Research Group, Medical Technology Research Centre, Anglia Ruskin University, Chelmsford, United Kingdom
| | - Sophie L. Harding
- Fibrosis Research Group, Medical Technology Research Centre, Anglia Ruskin University, Chelmsford, United Kingdom
| | - Tariq Minhas
- The Essex Cardiothoracic Centre, Basildon University Hospital, Basildon, United Kingdom
| | - Gouri Koduri
- Southend University Hospital NHS Foundation Trust, Westcliff-on-Sea, United Kingdom
| | - Stephen A. Bustin
- Fibrosis Research Group, Medical Technology Research Centre, Anglia Ruskin University, Chelmsford, United Kingdom
| | - Selim Cellek
- Fibrosis Research Group, Medical Technology Research Centre, Anglia Ruskin University, Chelmsford, United Kingdom
| |
Collapse
|
21
|
Swinney DC. Lessons learned from phenotypic drug discovery efforts. Expert Opin Drug Discov 2025; 20:141-144. [PMID: 39673450 DOI: 10.1080/17460441.2024.2442741] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2024] [Revised: 12/01/2024] [Accepted: 12/11/2024] [Indexed: 12/16/2024]
|
22
|
Farha MA, Tu MM, Brown ED. Important challenges to finding new leads for new antibiotics. Curr Opin Microbiol 2025; 83:102562. [PMID: 39603107 DOI: 10.1016/j.mib.2024.102562] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Revised: 10/15/2024] [Accepted: 10/28/2024] [Indexed: 11/29/2024]
Abstract
Identification of new antibiotics remains a huge challenge. The last antibiotic of new chemical class and mechanism was discovered more than 30 years ago. Advances since have been largely incremental modifications to a limited number of chemical scaffolds. Discovering and developing truly new antibiotics is challenging: the science is complex, and the development process is time consuming and expensive. Herein, we focus on the discovery phase of modern antibacterial research and development. We argue that antibacterial discovery has been challenged by a poor understanding of bacterial permeability, by generic in vitro conventions that ignore the host, and by the inherent complexity of bacterial systems. Together, these factors have colluded to challenge modern, industrial, and reductionist approaches to antibiotic discovery. Nevertheless, advances in our understanding of many of these obstacles, including a new appreciation for the complexity of both host and pathogen biology, bode well for future efforts.
Collapse
Affiliation(s)
- Maya A Farha
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario L8N 3Z5, Canada; Michael G. DeGroote Institute of Infectious Disease Research, McMaster University, Hamilton, Ontario L8N 3Z5, Canada
| | - Megan M Tu
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario L8N 3Z5, Canada; Michael G. DeGroote Institute of Infectious Disease Research, McMaster University, Hamilton, Ontario L8N 3Z5, Canada
| | - Eric D Brown
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario L8N 3Z5, Canada; Michael G. DeGroote Institute of Infectious Disease Research, McMaster University, Hamilton, Ontario L8N 3Z5, Canada.
| |
Collapse
|
23
|
Seal S, Trapotsi MA, Spjuth O, Singh S, Carreras-Puigvert J, Greene N, Bender A, Carpenter AE. Cell Painting: a decade of discovery and innovation in cellular imaging. Nat Methods 2025; 22:254-268. [PMID: 39639168 PMCID: PMC11810604 DOI: 10.1038/s41592-024-02528-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Accepted: 09/24/2024] [Indexed: 12/07/2024]
Abstract
Modern quantitative image analysis techniques have enabled high-throughput, high-content imaging experiments. Image-based profiling leverages the rich information in images to identify similarities or differences among biological samples, rather than measuring a few features, as in high-content screening. Here, we review a decade of advancements and applications of Cell Painting, a microscopy-based cell-labeling assay aiming to capture a cell's state, introduced in 2013 to optimize and standardize image-based profiling. Cell Painting's ability to capture cellular responses to various perturbations has expanded owing to improvements in the protocol, adaptations for different perturbations, and enhanced methodologies for feature extraction, quality control, and batch-effect correction. Cell Painting is a versatile tool that has been used in various applications, alone or with other -omics data, to decipher the mechanism of action of a compound, its toxicity profile, and other biological effects. Future advances will likely involve computational and experimental techniques, new publicly available datasets, and integration with other high-content data types.
Collapse
Affiliation(s)
- Srijit Seal
- Broad Institute of MIT and Harvard, Cambridge, MA, USA.
- Yusuf Hamied Department of Chemistry, University of Cambridge, Cambridge, UK.
| | - Maria-Anna Trapotsi
- Imaging and Data Analytics, Clinical Pharmacology & Safety Sciences, R&D, AstraZeneca, Cambridge, UK.
| | - Ola Spjuth
- Department of Pharmaceutical Biosciences and Science for Life Laboratory, Uppsala University, Uppsala, Sweden
- Phenaros Pharmaceuticals AB, Uppsala, Sweden
| | | | - Jordi Carreras-Puigvert
- Department of Pharmaceutical Biosciences and Science for Life Laboratory, Uppsala University, Uppsala, Sweden
- Phenaros Pharmaceuticals AB, Uppsala, Sweden
| | - Nigel Greene
- Imaging and Data Analytics, Clinical Pharmacology & Safety Sciences, R&D, AstraZeneca, Waltham, MA, USA
| | - Andreas Bender
- Yusuf Hamied Department of Chemistry, University of Cambridge, Cambridge, UK
- STAR-UBB Institute, Babeş-Bolyai University, Cluj-Napoca, Romania
| | | |
Collapse
|
24
|
Liu BHM, Lin Y, Long X, Hung SW, Gaponova A, Ren F, Zhavoronkov A, Pun FW, Wang CC. Utilizing AI for the Identification and Validation of Novel Therapeutic Targets and Repurposed Drugs for Endometriosis. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2406565. [PMID: 39666559 PMCID: PMC11792045 DOI: 10.1002/advs.202406565] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Revised: 10/08/2024] [Indexed: 12/14/2024]
Abstract
Endometriosis affects over 190 million women globally, and effective therapies are urgently needed to address the burden of endometriosis on women's health. Using an artificial intelligence (AI)-driven target discovery platform, two unreported therapeutic targets, guanylate-binding protein 2 (GBP2) and hematopoietic cell kinase (HCK) are identified, along with a drug repurposing target, integrin beta 2 (ITGB2) for the treatment of endometriosis. GBP2, HCK, and ITGB2 are upregulated in human endometriotic specimens. siRNA-mediated knockdown of GBP2 and HCK significantly reduced cell viability and proliferation while stimulating apoptosis in endometrial stromal cells. In subcutaneous and intraperitoneal endometriosis mouse models, siRNAs targeting GBP2 and HCK notably reduced lesion volume and weight, with decreased proliferation and increased apoptosis within lesions. Both subcutaneous and intraperitoneal administration of Lifitegrast, an approved ITGB2 antagonist, effectively suppresses lesion growth. Collectively, these data present Lifitegrast as a previously unappreciated intervention for endometriosis treatment and identify GBP2 and HCK as novel druggable targets in endometriosis treatment. This study underscores AI's potential to accelerate the discovery of novel drug targets and facilitate the repurposing of treatment modalities for endometriosis.
Collapse
Affiliation(s)
- Bonnie Hei Man Liu
- Insilico Medicine Hong Kong Ltd.Unit 310, 3/F, Building 8W, Hong Kong Science and Technology ParkHong KongChina
| | - Yuezhen Lin
- Department of Obstetrics and GynaecologyThe Chinese University of Hong KongHong KongChina
| | - Xi Long
- Insilico Medicine Hong Kong Ltd.Unit 310, 3/F, Building 8W, Hong Kong Science and Technology ParkHong KongChina
| | - Sze Wan Hung
- Department of Obstetrics and GynaecologyThe Chinese University of Hong KongHong KongChina
| | - Anna Gaponova
- Insilico Medicine Hong Kong Ltd.Unit 310, 3/F, Building 8W, Hong Kong Science and Technology ParkHong KongChina
| | - Feng Ren
- Insilico Medicine Shanghai Ltd.9F, Chamtime Plaza Block C, Lane 2889, Jinke Road, Pudong New AreaShanghai201203China
| | - Alex Zhavoronkov
- Insilico Medicine Hong Kong Ltd.Unit 310, 3/F, Building 8W, Hong Kong Science and Technology ParkHong KongChina
- Buck Institute for Research on Aging8001 Redwood Blvd.NovatoCA94945USA
| | - Frank W. Pun
- Insilico Medicine Hong Kong Ltd.Unit 310, 3/F, Building 8W, Hong Kong Science and Technology ParkHong KongChina
| | - Chi Chiu Wang
- Department of Obstetrics and GynaecologyThe Chinese University of Hong KongHong KongChina
- Reproduction and DevelopmentLi Ka Shing Institute of Health SciencesThe Chinese University of Hong KongHong KongChina
- School of Biomedical SciencesThe Chinese University of Hong KongHong KongChina
- Chinese University of Hong Kong‐Sichuan University Joint Laboratory in Reproductive MedicineThe Chinese University of Hong KongHong KongChina
- State Key Laboratory of Chinese Medicine ModernizationInnovation Center of Yangtze River Delta Zhejiang UniversityJiaxing314102China
| |
Collapse
|
25
|
Seymour S, Cadena I, Johnson M, Thakkar R, Jenne M, Adem I, Almer A, Frankovic R, Spence D, Haddadin A, Fogg KC. Empowering High Throughput Screening of 3D Models: Automated Dispensing of Cervical and Endometrial Cancer Cells. Cell Mol Bioeng 2025; 18:71-82. [PMID: 39949489 PMCID: PMC11813830 DOI: 10.1007/s12195-024-00841-y] [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: 04/01/2024] [Accepted: 12/17/2024] [Indexed: 02/16/2025] Open
Abstract
Purpose Cervical and endometrial cancers pose significant challenges in women's healthcare due to their high mortality rates and limited treatment options. High throughput screening (HTS) of cervical and endometrial cancer in vitro models offers a promising avenue for drug repurposing and broadening patient treatment options. Traditional two-dimensional (2D) cell-based screenings have limited capabilities to capture crucial multicellular interactions, that are improved upon in three dimensional (3D) multicellular tissue engineered models. However, manual fabrication of the 3D platforms is both time consuming and subject to variability. Thus, the goal of this study was to utilize automated cell dispensing to fabricate 3D cell-based HTS platforms using the HP D100 Single Cell Dispenser to dispense cervical and endometrial cancer cells. Methods We evaluated the effects of automated dispensing of the cancer cell lines by tuning the dispensing protocol to align with cell size measured in solution and the minimum cell number for acceptable cell viability and proliferation. We modified our previously reported coculture models of cervical and endometrial cancer to be in a 384 well plate format and measured microvessel length and cancer cell invasion. Results Automatically and manually dispensed cells were directly compared revealing minimal differences between the dispensing methods. These findings suggest that automated dispensing of cancer cells minimally affects cell behavior and can be deployed to decrease in vitro model fabrication time. Conclusions By streamlining the manufacturing process, automated dispensing holds promise for enhancing efficiency and scalability of 3D in vitro HTS platforms, ultimately contributing to advancement in cancer research and treatment. Supplementary Information The online version contains supplementary material available at 10.1007/s12195-024-00841-y.
Collapse
Affiliation(s)
- Samantha Seymour
- School of Chemical, Biological, and Environmental Engineering, Oregon State University, Corvallis, OR 97330 USA
| | - Ines Cadena
- School of Chemical, Biological, and Environmental Engineering, Oregon State University, Corvallis, OR 97330 USA
| | | | - Riya Thakkar
- School of Chemical, Biological, and Environmental Engineering, Oregon State University, Corvallis, OR 97330 USA
| | - Molly Jenne
- School of Chemical, Biological, and Environmental Engineering, Oregon State University, Corvallis, OR 97330 USA
| | - Iman Adem
- School of Chemical, Biological, and Environmental Engineering, Oregon State University, Corvallis, OR 97330 USA
| | - Alyssa Almer
- School of Chemical, Biological, and Environmental Engineering, Oregon State University, Corvallis, OR 97330 USA
| | - Rachael Frankovic
- School of Chemical, Biological, and Environmental Engineering, Oregon State University, Corvallis, OR 97330 USA
| | - Danielle Spence
- School of Chemical, Biological, and Environmental Engineering, Oregon State University, Corvallis, OR 97330 USA
| | - Andrea Haddadin
- School of Chemical, Biological, and Environmental Engineering, Oregon State University, Corvallis, OR 97330 USA
| | - Kaitlin C. Fogg
- School of Chemical, Biological, and Environmental Engineering, Oregon State University, Corvallis, OR 97330 USA
| |
Collapse
|
26
|
Gonzalez G, Lin X, Herath I, Veselkov K, Bronstein M, Zitnik M. Combinatorial prediction of therapeutic perturbations using causally-inspired neural networks. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.01.03.573985. [PMID: 38260532 PMCID: PMC10802439 DOI: 10.1101/2024.01.03.573985] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/24/2024]
Abstract
Phenotype-driven approaches identify disease-counteracting compounds by analyzing the phenotypic signatures that distinguish diseased from healthy states. These approaches can guide the discovery of targeted perturbations, including small-molecule drugs and genetic interventions, that modulate disease phenotypes toward healthier states. Here, we introduce PDGrapher, a causally inspired graph neural network (GNN) designed to predict combinatorial perturbagens (sets of therapeutic targets) capable of reversing disease phenotypes. Unlike methods that learn how perturbations alter phenotypes, PDGrapher solves the inverse problem of directly predicting the perturbagens needed to achieve a desired response. PDGrapher is a GNN that embeds disease cell states into gene regulatory or protein-protein interaction networks, learns a latent representation of these states, and identifies the optimal combinatorial perturbations that most effectively shift the diseased state toward the desired treated state within that latent space. In experiments in nine cell lines with chemical perturbations, PDGrapher identified effective per-turbagens in up to 13.33% more test samples than competing methods and achieved a normalized discounted cumulative gain of up to 0.12 higher to classify therapeutic targets. It also demonstrated competitive performance on ten genetic perturbation datasets. A key advantage of PDGrapher is its direct prediction paradigm, in contrast to the indirect and computationally intensive models traditionally employed in phenotype-driven research. This approach accelerates training by up to 25 times compared to existing methods. PDGrapher provides a fast approach for identifying therapeutic perturbations and advancing phenotype-driven drug discovery.
Collapse
Affiliation(s)
- Guadalupe Gonzalez
- Imperial College London, London, UK
- Prescient Design, Genentech, South San Francisco, CA, USA
- F. Hoffmann-La Roche Ltd, Basel, Switzerland
| | - Xiang Lin
- Harvard Medical School, Boston, MA, USA
| | - Isuru Herath
- Merck & Co., South San Francisco, CA, USA
- Cornell University, Ithaca, NY, USA
| | | | | | - Marinka Zitnik
- Harvard Medical School, Boston, MA, USA
- Kempner Institute for the Study of Natural and Artificial Intelligence, Harvard University, Cambridge, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Harvard Data Science Initiative, Cambridge, MA, USA
| |
Collapse
|
27
|
OSADA H. Chemical biology research in RIKEN NPDepo aimed at agricultural applications. PROCEEDINGS OF THE JAPAN ACADEMY. SERIES B, PHYSICAL AND BIOLOGICAL SCIENCES 2025; 101:8-31. [PMID: 39805590 PMCID: PMC11808203 DOI: 10.2183/pjab.101.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/31/2024] [Accepted: 10/23/2024] [Indexed: 01/16/2025]
Abstract
This review outlines research on chemical biology using mainly microbial metabolites for agricultural applications. We established the RIKEN Natural Products Depository (NPDepo), housing many microbial metabolites, to support academic researchers who focus on drug discovery. We studied methods to stimulate secondary metabolism in microorganisms to collect various microbial products. The switch of secondary metabolism in microorganisms changes depending on the culture conditions. We discovered compounds that activate biosynthetic gene clusters in actinomycetes and filamentous fungi. Using these compounds, we succeeded in inducing the production of active compounds. Two approaches for screening bioactive compounds are described. One is phenotypic screening to explore antifungal compounds assisted by artificial intelligence (AI). AI can distinguish the morphological changes induced by antifungal compounds in filamentous fungi. The other is the chemical array method for detecting interactions between compounds and target proteins. Our chemical biology approach yielded many new compounds as fungicide candidates.
Collapse
Affiliation(s)
- Hiroyuki OSADA
- Institute of Microbial Chemistry (BIKAKEN), Tokyo, Japan
- RIKEN Center for Sustainable Resource Science, Wako, Saitama, Japan
| |
Collapse
|
28
|
Manoharan S, Perumal E. Chemotherapeutic potential of radotinib against blood and solid tumors: A beacon of hope in drug repurposing. Bioorg Chem 2025; 154:108017. [PMID: 39647393 DOI: 10.1016/j.bioorg.2024.108017] [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: 08/15/2024] [Revised: 11/08/2024] [Accepted: 11/28/2024] [Indexed: 12/10/2024]
Abstract
Tyrosine kinase inhibitors (TKIs) represent a pivotal class of targeted therapies in oncology, with multiple generations developed to address diverse molecular targets. Imatinib is the first TKI developed to target the BCR-ABL1 chimeric protein, which is the key driver oncogene implicated in Philadelphia chromosome-positive chronic myeloid leukemia (CML). Several second-generation tyrosine kinase inhibitors (2GTKIs), such as nilotinib, dasatinib, bosutinib, and radotinib (RTB), followed the groundbreaking introduction of imatinib. RTB occupies the unique position of being the least explored member of this class. While nilotinib, dasatinib, and bosutinib have garnered significant attention and extensive research focus, RTB remains relatively uncharted in comparison to its counterparts. Fundamental drug characteristics, such as the pharmacokinetic and pharmacodynamic properties of RTB, remain unavailable in existing sources. Compared to other 2GTKIs, RTB has been less utilized in combinatorial drug studies, and no investigations have been reported on its effects on solid tumors to date. However, the effects of RTB have been studied in acute myeloid leukemia (AML), multiple myeloma (MM), Parkinson's disease, and idiopathic pulmonary fibrosis (IPF). Although RTB has been investigated in some conditions, these studies are still in their preliminary stages and are comparatively lesser than studies on other 2GTKIs. This review is the first attempt that extensively presents a compilation of data on RTB and describes its therapeutic potential against blood and solid tumors. Further investigations on RTB could expand its chemotherapeutic usage in various solid tumors and enhance the possibility of drug repurposing in cancer therapy.
Collapse
Affiliation(s)
- Suryaa Manoharan
- Molecular Toxicology Laboratory, Department of Biotechnology, Bharathiar University, Coimbatore 641046, India
| | - Ekambaram Perumal
- Molecular Toxicology Laboratory, Department of Biotechnology, Bharathiar University, Coimbatore 641046, India.
| |
Collapse
|
29
|
Attri M, Raghav A, Sinha J. Revolutionising Neurological Therapeutics: Investigating Drug Repurposing Strategies. CNS & NEUROLOGICAL DISORDERS DRUG TARGETS 2025; 24:115-131. [PMID: 39323347 DOI: 10.2174/0118715273329531240911075309] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Revised: 07/08/2024] [Accepted: 07/15/2024] [Indexed: 09/27/2024]
Abstract
Repurposing drugs (DR) has become a viable approach to hasten the search for cures for neurodegenerative diseases (NDs). This review examines different off-target and on-target drug discovery techniques and how they might be used to find possible treatments for non-diagnostic depressions. Off-target strategies look at the known or unknown side effects of currently approved drugs for repositioning, whereas on-target strategies connect disease pathways to targets that can be treated with drugs. The review highlights the potential of experimental and computational methodologies, such as machine learning, proteomic techniques, network and genomics-based approaches, and in silico screening, in uncovering new drug-disease correlations. It also looks at difficulties and failed attempts at drug repurposing for NDs, highlighting the necessity of exact and standardised procedures to increase success rates. This review's objectives are to address the purpose of drug repurposing in human disorders, particularly neurological diseases, and to provide an overview of repurposing candidates that are presently undergoing clinical trials for neurological conditions, along with any possible causes and early findings. We then include a list of drug repurposing strategies, restrictions, and difficulties for upcoming research.
Collapse
Affiliation(s)
- Meenakshi Attri
- School of Medical & Allied Sciences, K.R. Mangalam University, Gurugram, Haryana 122103, India
| | - Asha Raghav
- Department of Pharmaceutics, School of Health Sciences, Sushant University, Gurugram, Haryana 122003, India
| | - Jyoti Sinha
- Department of Pharmaceutics, School of Health Sciences, Sushant University, Gurugram, Haryana 122003, India
| |
Collapse
|
30
|
Farkas E, McKay GA, Hu LT, Nekouei M, Ho P, Moreira W, Chan CC, Dam LC, Auclair K, Gruenheid S, Whyte L, Dedon P, Nguyen D. Bioluminescent Pseudomonas aeruginosa and Escherichia coli for whole-cell screening of antibacterial and adjuvant compounds. Sci Rep 2024; 14:31039. [PMID: 39730767 DOI: 10.1038/s41598-024-81926-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2024] [Accepted: 11/29/2024] [Indexed: 12/29/2024] Open
Abstract
Continued efforts to discover new antibacterial molecules are critical to achieve a robust pre-clinical pipeline for new antibiotics. Screening of compound or natural product extract libraries remains a widespread approach and can benefit from the development of whole cell assays that are robust, simple and versatile, and allow for high throughput testing of antibacterial activity. In this study, we created and validated two bioluminescent reporter strains for high-throughput screening, one in Pseudomonas aeruginosa, and another in a hyperporinated and efflux-deficient Escherichia coli. We show that the bioluminescent strains have a large dynamic range that closely correlates with cell viability and is superior to conventional optical density (OD600) measurements, can detect dose-dependent antibacterial activity and be used for different drug discovery applications. We evaluated the assays' performance characteristics (signal to background ratio, signal window, Z' robust) and demonstrated their potential utility for antibiotic drug discovery in two examples. The P. aeruginosa bioluminescent reporter was used in a pilot screen of 960 repurposed compound libraries to identify adjuvants that potentiate the fluoroquinolone antibiotic ofloxacin. The E. coli bioluminescent reporter was used to test the antibacterial activity of bioactive bacterial supernatants and assist with bioassay-guided fractionation of the crude extracts.
Collapse
Affiliation(s)
- Eszter Farkas
- Department of Microbiology and Immunology, McGill University, Montreal, QC, Canada
- Meakins-Christie Laboratories, Research Institute of the McGill University Health Centre (RI-MUHC), Montreal, QC, Canada
| | - Geoffrey A McKay
- Meakins-Christie Laboratories, Research Institute of the McGill University Health Centre (RI-MUHC), Montreal, QC, Canada
| | - Lin Tao Hu
- Department of Microbiology and Immunology, McGill University, Montreal, QC, Canada
| | - Mina Nekouei
- Department of Chemistry, McGill University, Montreal, QC, Canada
| | - Peying Ho
- Antimicrobial Resistance Interdisciplinary Research Group (AMR IRG), Singapore- Massachusetts Institute of Technology Alliance for Research and Technology (SMART) Centre, Singapore, Singapore
| | - Wilfried Moreira
- Antimicrobial Resistance Interdisciplinary Research Group (AMR IRG), Singapore- Massachusetts Institute of Technology Alliance for Research and Technology (SMART) Centre, Singapore, Singapore
- Singapore Centre for Environmental Life Science Engineering (SCELSE), Singapore, Singapore
| | - Chia Ching Chan
- Antimicrobial Resistance Interdisciplinary Research Group (AMR IRG), Singapore- Massachusetts Institute of Technology Alliance for Research and Technology (SMART) Centre, Singapore, Singapore
| | - Linh Chi Dam
- Antimicrobial Resistance Interdisciplinary Research Group (AMR IRG), Singapore- Massachusetts Institute of Technology Alliance for Research and Technology (SMART) Centre, Singapore, Singapore
- Cardiovascular and Metabolic Disorders Programme, Duke-NUS Medical School, Singapore, Singapore
- National Heart Research Institute Singapore, National Heart Centre Singapore, Singapore, Singapore
| | - Karine Auclair
- Department of Chemistry, McGill University, Montreal, QC, Canada
| | - Samantha Gruenheid
- Department of Microbiology and Immunology, McGill University, Montreal, QC, Canada
| | - Lyle Whyte
- Department of Natural Resource Sciences, McGill University, Montreal, QC, Canada
| | - Peter Dedon
- Antimicrobial Resistance Interdisciplinary Research Group (AMR IRG), Singapore- Massachusetts Institute of Technology Alliance for Research and Technology (SMART) Centre, Singapore, Singapore
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Dao Nguyen
- Department of Microbiology and Immunology, McGill University, Montreal, QC, Canada.
- Meakins-Christie Laboratories, Research Institute of the McGill University Health Centre (RI-MUHC), Montreal, QC, Canada.
- Department of Medicine, McGill University, Montreal, QC, Canada.
| |
Collapse
|
31
|
Ogasawara D, Konrad DB, Tan ZY, Carey KL, Luo J, Won SJ, Li H, Carter TR, DeMeester KE, Njomen E, Schreiber SL, Xavier RJ, Melillo B, Cravatt BF. Chemical tools to expand the ligandable proteome: Diversity-oriented synthesis-based photoreactive stereoprobes. Cell Chem Biol 2024; 31:2138-2155.e32. [PMID: 39547236 PMCID: PMC11837778 DOI: 10.1016/j.chembiol.2024.10.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Revised: 09/09/2024] [Accepted: 10/18/2024] [Indexed: 11/17/2024]
Abstract
Chemical proteomics enables the global analysis of small molecule-protein interactions in native biological systems and has emerged as a versatile approach for ligand discovery. The range of small molecules explored by chemical proteomics has, however, remained limited. Here, we describe a diversity-oriented synthesis (DOS)-inspired library of stereochemically defined compounds bearing diazirine and alkyne units for UV light-induced covalent modification and click chemistry enrichment of interacting proteins, respectively. We find that these "photo-stereoprobes" interact in a stereoselective manner with hundreds of proteins from various structural and functional classes in human cells and demonstrate that these interactions can form the basis for high-throughput screening-compatible NanoBRET assays. Integrated phenotypic screening and chemical proteomics identified photo-stereoprobes that modulate autophagy by engaging the mitochondrial serine protease CLPP. Our findings show the utility of DOS-inspired photo-stereoprobes for expanding the ligandable proteome, furnishing target engagement assays, and facilitating the discovery and characterization of bioactive compounds in phenotypic screens.
Collapse
Affiliation(s)
- Daisuke Ogasawara
- Department of Chemistry, The Scripps Research Institute, La Jolla, CA 92037, USA.
| | - David B Konrad
- Department of Chemistry, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Zher Yin Tan
- Immunology Program, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Chemical Biology and Therapeutics Science Program, Broad Institute, Cambridge, MA 02142, USA; Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA 02138, USA
| | - Kimberly L Carey
- Immunology Program, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Jessica Luo
- Immunology Program, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Sang Joon Won
- Department of Chemistry, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Haoxin Li
- Department of Chemistry, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Trever R Carter
- Department of Chemistry, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Kristen E DeMeester
- Department of Chemistry, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Evert Njomen
- Department of Chemistry, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Stuart L Schreiber
- Chemical Biology and Therapeutics Science Program, Broad Institute, Cambridge, MA 02142, USA; Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA 02138, USA
| | - Ramnik J Xavier
- Immunology Program, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Department of Molecular Biology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA; Center for Computational and Integrative Biology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Bruno Melillo
- Department of Chemistry, The Scripps Research Institute, La Jolla, CA 92037, USA.
| | - Benjamin F Cravatt
- Department of Chemistry, The Scripps Research Institute, La Jolla, CA 92037, USA.
| |
Collapse
|
32
|
Chen S, Liu J, Tang N, Zeng Y. Cancer phenomics research hotspots and development trends: a bibliometric analysis from 2000 to 2023. Discov Oncol 2024; 15:811. [PMID: 39695032 DOI: 10.1007/s12672-024-01710-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Accepted: 12/13/2024] [Indexed: 12/20/2024] Open
Abstract
The emerging field of cancer phenomics provides comprehensive insights into tumor heterogeneity, promoting advances in personalized oncology. This study explores current research hotspots and future development trends in cancer phenomics through a bibliometric analysis of research from 2000 to 2023. Using data from the Web of Science Core Collection, we analyzed 1260 publications to identify global contributions and collaborative networks. Employing CiteSpace and VOSviewer tools, we examined research trends, highlighting disease progression, multi-omics integration, and phenotypic drug discovery as major focus areas. Key findings reveal that the United States, China, and the United Kingdom are leading contributors, with top institutions such as Harvard Medical School advancing research and fostering international collaboration. Additionally, the analysis underscores the prominence of double-positive (DP) T cells and natural killer (NK) cells in cancer immunology, showcasing their potential roles in phenotypic screening and cancer therapeutics. Despite advancements, the study notes ongoing challenges in translating phenomics research to clinical applications, suggesting that enhanced global partnerships and technological integration are essential. This analysis offers valuable perspectives for future research and highlights phenomics' transformative potential in precision oncology, advocating for its role in advancing cancer diagnosis, treatment, and prevention.
Collapse
Affiliation(s)
- Shupeng Chen
- School of Clinical Medicine, Jiangxi University of Chinese Medicine, Nanchang, 330004, China
| | - Jie Liu
- School of Clinical Medicine, Jiangxi University of Chinese Medicine, Nanchang, 330004, China
| | - Nana Tang
- Hematology Department, Affiliated Hospital of Jiangxi University of Traditional Chinese Medicine, Nanchang, 330006, Jiangxi, China
| | - Yingjian Zeng
- Hematology Department, Affiliated Hospital of Jiangxi University of Traditional Chinese Medicine, Nanchang, 330006, Jiangxi, China.
| |
Collapse
|
33
|
Sangeetha B, Leroy KI, Udaya Kumar B. Harnessing Bioluminescence: A Comprehensive Review of In Vivo Imaging for Disease Monitoring and Therapeutic Intervention. Cell Biochem Funct 2024; 42:e70020. [PMID: 39673353 DOI: 10.1002/cbf.70020] [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: 07/05/2024] [Revised: 11/13/2024] [Accepted: 11/15/2024] [Indexed: 12/16/2024]
Abstract
The technique of using naturally occurring light-emitting reactants (photoproteins and luciferases] that have been extracted from a wide range of animals is known as bioluminescence imaging, or BLI. This imaging offers important details on the location and functional state of regenerative cells inserted into various disease-modeling animals. Reports on gene expression patterns, cell motions, and even the actions of individual biomolecules in whole tissues and live animals have all been made possible by bioluminescence. Generally speaking, bioluminescent light in animals may be found down to a few centimetres, while the precise limit depends on the signal's brightness and the detector's sensitivity. We can now spatiotemporally visualize cell behaviors in any body region of a living animal in a time frame process, including proliferation, apoptosis, migration, and immunological responses, thanks to BLI. The biological applications of in vivo BLI in nondestructively monitoring biological processes in intact small animal models are reviewed in this work, along with some of the advancements that will make BLI a more versatile molecular imaging tool.
Collapse
Affiliation(s)
- B Sangeetha
- Department of Biotechnology, St Joseph's College of Engineering, Chennai, Tamilnadu, India
| | - K I Leroy
- Department of Biotechnology, St Joseph's College of Engineering, Chennai, Tamilnadu, India
| | - B Udaya Kumar
- Department of Biotechnology, St Joseph's College of Engineering, Chennai, Tamilnadu, India
| |
Collapse
|
34
|
Wang R, Chen K, Liu S, Ren R, Hou H, Zeng Q, Zhang Y, Liu Y. Design, synthesis and biological evaluation of novel oxazole derivatives as potential hypoglycemic agents. Bioorg Med Chem 2024; 114:117961. [PMID: 39437535 DOI: 10.1016/j.bmc.2024.117961] [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: 08/23/2024] [Revised: 10/10/2024] [Accepted: 10/14/2024] [Indexed: 10/25/2024]
Abstract
A series of 2,4-disubstituted-oxazole derivatives have been designed and synthesized based on compound 3a, a promising lead compound developed in our lab. Among these derivatives, the optimized compound 5k exhibited potent hypoglycemic activity, increasing glucose consumption by 60 % in HepG2 cells compared to the solvent control, and its activity was higher than that of metformin. Further investigation indicated that compound 5k exhibited negligible cytotoxic effects at a concentration of 25 μM in HepG2 and 3T3-L1 cells and showed moderate inhibitory activity against various subtypes of human cytochrome P450 subtypes. An oral glucose tolerance test confirmed that 5k is an effective hypoglycemic agent. Additionally, mechanistic studies suggested that 5k may exert its hypoglycemic activity through the activation of the AMPK pathway.
Collapse
Affiliation(s)
- Ruifeng Wang
- Department of Endocrinology, First Hospital of Shanxi Medical University, Shanxi Medical University, Taiyuan 030001, China; School of Pharmacy, Shanxi Medical University, Taiyuan 030001, China; Medicinal Basic Research Innovation Center of Chronic Kidney Disease, Ministry of Education, Shanxi Medical University, Taiyuan 030001, China
| | - Ke Chen
- School of Pharmacy, Shanxi Medical University, Taiyuan 030001, China; Medicinal Basic Research Innovation Center of Chronic Kidney Disease, Ministry of Education, Shanxi Medical University, Taiyuan 030001, China
| | - Shuihua Liu
- School of Pharmacy, Shanxi Medical University, Taiyuan 030001, China; Medicinal Basic Research Innovation Center of Chronic Kidney Disease, Ministry of Education, Shanxi Medical University, Taiyuan 030001, China
| | - Ruyue Ren
- School of Pharmacy, Shanxi Medical University, Taiyuan 030001, China; Medicinal Basic Research Innovation Center of Chronic Kidney Disease, Ministry of Education, Shanxi Medical University, Taiyuan 030001, China
| | - Hongbao Hou
- Department of Pharmacology, Shanxi Medical University, Taiyuan 030001, China; Medicinal Basic Research Innovation Center of Chronic Kidney Disease, Ministry of Education, Shanxi Medical University, Taiyuan 030001, China
| | - Qingxuan Zeng
- Department of Pharmacology, Shanxi Medical University, Taiyuan 030001, China; Medicinal Basic Research Innovation Center of Chronic Kidney Disease, Ministry of Education, Shanxi Medical University, Taiyuan 030001, China
| | - Yi Zhang
- Department of Pharmacology, Shanxi Medical University, Taiyuan 030001, China; Medicinal Basic Research Innovation Center of Chronic Kidney Disease, Ministry of Education, Shanxi Medical University, Taiyuan 030001, China.
| | - Yunfeng Liu
- Department of Endocrinology, First Hospital of Shanxi Medical University, Shanxi Medical University, Taiyuan 030001, China.
| |
Collapse
|
35
|
Shahwan M, Khan MS, Zuberi A, Altwaijry N, Shamsi A. Structure-guided drug repurposing identifies aristospan as a potential inhibitor of β-lactamase: insights from virtual screening and molecular dynamics simulations. Front Pharmacol 2024; 15:1459822. [PMID: 39568577 PMCID: PMC11576302 DOI: 10.3389/fphar.2024.1459822] [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: 07/04/2024] [Accepted: 10/24/2024] [Indexed: 11/22/2024] Open
Abstract
The rise of β-Lactamase mediated antibiotic resistance is a major concern for public health; hence, there is an urgent need to find new treatment approaches. Structure-guided drug repurposing offers a promising approach to swiftly deliver essential therapeutics in the fight against escalating antibiotic resistance. Here, a structure-guided virtual screening approach was used involving drug profiling, molecular docking, and molecular dynamics (MD) simulation to identify existing drugs against β-Lactamase-associated drug resistance. We exploited a large panel of FDA-approved drugs to an extensive in silico analysis to ascertain their ability to inhibit β-Lactamase. First, molecular docking investigations were performed to assess the binding affinities and interactions of screened molecules with the active site of β-Lactamase enzymes. Out of all the screened candidates, Aristospan was identified to possess promising characteristics, which include appropriate drug profiles, high binding specificity, and efficiency towards the binding pocket of β-Lactamase. Further analysis showed that Aristospan possesses several desirable biological characteristics and tends to bind to the β-Lactamase binding site. To explore the interactions further, the best docking pose of Aristospan was selected for MD simulations to assess the thermodynamic stability of the drug-enzyme complex and its conformational changes over 500 ns. The MD simulations in independent replica runs demonstrated that the β-Lactamase-Aristospan complex was stable in the 500 ns trajectory. These enlightening results suggest that Aristospan may harbor the potential for further evolution into a possible β-Lactamase inhibitor, with potential applications in overcoming antibiotic resistance in both Gram-positive and Gram-negative bacteria.
Collapse
Affiliation(s)
- Moyad Shahwan
- Center for Medical and Bio-Allied Health Sciences Research, Ajman University, Ajman, United Arab Emirates
- College of Pharmacy and Health Sciences, Ajman University, Ajman, United Arab Emirates
| | - Mohd Shahnawaz Khan
- Department of Biochemistry, College of Science, King Saud University, Riyadh, Saudi Arabia
| | - Azna Zuberi
- Division of Reproductive Science in Medicine, Department of Obstetrics and Gynecology, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| | - Nojood Altwaijry
- Department of Biochemistry, College of Science, King Saud University, Riyadh, Saudi Arabia
| | - Anas Shamsi
- Center for Medical and Bio-Allied Health Sciences Research, Ajman University, Ajman, United Arab Emirates
| |
Collapse
|
36
|
Phanchana M, Pipatthana M, Phetruen T, Konpetch P, Prangthip P, Harnvoravongchai P, Sripong C, Singhakaew S, Wongphayak S, Chankhamhaengdecha S, Janvilisri T. Identification and preclinical evaluation of MMV676558 as a promising therapeutic candidate against Clostridioides difficile. Biomed Pharmacother 2024; 180:117469. [PMID: 39321508 DOI: 10.1016/j.biopha.2024.117469] [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: 07/12/2024] [Revised: 09/03/2024] [Accepted: 09/19/2024] [Indexed: 09/27/2024] Open
Abstract
Clostridioides difficile, a gram-positive, toxin-producing, spore-forming anaerobe, is a major cause of antibiotic-associated diarrhoea. The bacterium's intrinsic drug resistance limits current treatment options to fidaxomicin and vancomycin for initial episodes, with anti-toxin B monoclonal antibody or faecal microbiota transplantation recommended for complicated or recurrent cases. This underscores the urgent need for novel therapeutics. In this study, we screened the MMV Pathogen Box at a 10 µM concentration against C. difficile R20291. Primary hits were evaluated for minimum inhibitory concentrations (MIC), killing kinetics, and biofilm inhibition. Bacterial cytological profiling (BCP) and transmission electron microscopy (TEM) were employed to study the mode of action. MMV676558 was further tested in a mouse model to assess survival, histopathology, and gut microbiota effects. We identified nineteen hits that inhibited over 50 % of C. difficile growth. MIC assays revealed three hits with MICs below 16 µg/mL: MMV676558, MMV688755, and MMV690027. These hits were effective against various C. difficile ribotypes. Killing kinetics were comparable or superior to vancomycin and fidaxomicin, and biofilm assays showed inhibitory effects. BCP and TEM analyses suggested membrane function disruption as the mode of action. Furthermore, MMV676558 demonstrated a protective effect in mice, with favourable histopathology and gut microbiota profiles. Given the urgent threat posed by C. difficile antibiotic resistance, discovering new treatments is a top priority. Our study identified three promising hits from the MMV Pathogen Box, with MMV676558 showing significant in vivo potential for further evaluation.
Collapse
Affiliation(s)
- Matthew Phanchana
- Department of Molecular Tropical Medicine and Genetics, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Methinee Pipatthana
- Department of Microbiology, Faculty of Public Health, Mahidol University, Bangkok, Thailand
| | - Tanaporn Phetruen
- Department of Biochemistry, Faculty of Science, Mahidol University, Bangkok, Thailand
| | - Pattanai Konpetch
- Department of Biochemistry, Faculty of Science, Mahidol University, Bangkok, Thailand
| | - Pattaneeya Prangthip
- Department of Tropical Nutrition and Food Science, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | | | | | - Sombat Singhakaew
- Department of Biology, Faculty of Science, Mahidol University, Bangkok, Thailand
| | | | | | - Tavan Janvilisri
- Department of Biochemistry, Faculty of Science, Mahidol University, Bangkok, Thailand.
| |
Collapse
|
37
|
Cai D, Xu X, Zeng W, Wang Z, Chen C, Mo Y, Meekrathok P, Wang D, Peng P, Peng Z, Qiu J. Deoxyarbutin targets mitochondria to trigger p53-dependent senescence of glioblastoma cells. Free Radic Biol Med 2024; 224:382-392. [PMID: 39209136 DOI: 10.1016/j.freeradbiomed.2024.08.027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Revised: 08/21/2024] [Accepted: 08/22/2024] [Indexed: 09/04/2024]
Abstract
Cellular senescence is a natural barrier of the transition from premalignant cells to invasive cancer. Pharmacological induction of senescence has been proposed as a possible anticancer strategy. In this study, we found that deoxyarbutin inhibited the growth of glioblastoma (GBM) cells by inducing cellular senescence, independent of tyrosinase expression. Instead, deoxyarbutin induced mitochondrial oxidative stress and damage. These aberrant mitochondria were key to the p53-dependent senescence of GBM cells. Facilitating autophagy or mitigating mitochondrial oxidative stress both suppressed p53 expression and alleviated cellular senescence induced by deoxyarbutin. Thus, our study reveals that deoxyarbutin induces mitochondrial oxidative stress to trigger the p53-dependent senescence of GBM cells. Importantly, deoxyarbutin treatment resulted in accumulation of p53, induction of cellular senescence, and inhibition of tumor growth in a subcutaneous tumor model of mouse. In conclusion, our study reveals that deoxyarbutin has therapeutic potential for GBM by inducing mitochondrial oxidative stress for p53-dependent senescence of GBM cells.
Collapse
Affiliation(s)
- Dongjing Cai
- Hunan Key Laboratory of Molecular Precision Medicine, Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, 410008, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Xia Xu
- Department of General Practice, Xiangya Hospital, Central South University, Changsha, 410008, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Weiqian Zeng
- Hunan Key Laboratory of Molecular Precision Medicine, Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, 410008, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Zheng Wang
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, 410008, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Cheng Chen
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, 410008, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Yunan Mo
- Hunan Key Laboratory of Molecular Precision Medicine, Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, 410008, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Piyanat Meekrathok
- Hunan Key Laboratory of Molecular Precision Medicine, Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, 410008, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Dandan Wang
- Hunan Key Laboratory of Molecular Precision Medicine, Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, 410008, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Pengwei Peng
- Hunan Key Laboratory of Molecular Precision Medicine, Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, 410008, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Zhigang Peng
- Hunan Key Laboratory of Molecular Precision Medicine, Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, 410008, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Jian Qiu
- Hunan Key Laboratory of Molecular Precision Medicine, Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, 410008, China; MOE Key Lab of Rare Pediatric Diseases & Hunan Key Laboratory of Medical Genetics & Hunan Key Laboratory of Animal Models for Human Diseases, School of Life Sciences, Central South University, Changsha, 410008, China; NHC Key Laboratory of Cancer Proteomics & State Local Joint Engineering Laboratory for Anticancer Drugs, Xiangya Hospital, Central South University, Changsha, 410008, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, China.
| |
Collapse
|
38
|
Cui X, Li X, Zheng H, Su Y, Zhang S, Li M, Hao X, Zhang S, Hu Z, Xia Z, Shi C, Xu Y, Mao C. Human midbrain organoids: a powerful tool for advanced Parkinson's disease modeling and therapy exploration. NPJ Parkinsons Dis 2024; 10:189. [PMID: 39428415 PMCID: PMC11491477 DOI: 10.1038/s41531-024-00799-8] [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: 01/06/2023] [Accepted: 10/02/2024] [Indexed: 10/22/2024] Open
Abstract
Parkinson's disease (PD) is a neurodegenerative disorder marked by the loss of dopaminergic neurons in the substantia nigra. Despite progress, the pathogenesis remains unclear. Human midbrain organoids (hMLOs) have emerged as a promising model for studying PD, drug screening, and potential treatments. This review discusses the development of hMLOs, their application in PD research, and current challenges in organoid construction, highlighting possible optimization strategies.
Collapse
Affiliation(s)
- Xin Cui
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
- Academy of Medical Sciences of Zhengzhou University, Zhengzhou, China
- Henan Key Laboratory of Cerebrovascular Diseases, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
| | - Xinwei Li
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
- Academy of Medical Sciences of Zhengzhou University, Zhengzhou, China
- Henan Key Laboratory of Cerebrovascular Diseases, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
| | - Huimin Zheng
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
- Henan Key Laboratory of Cerebrovascular Diseases, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
| | - Yun Su
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
- Henan Key Laboratory of Cerebrovascular Diseases, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
| | - Shuyu Zhang
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
- Neuro-Intensive Care Unit, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Mengjie Li
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
- Henan Key Laboratory of Cerebrovascular Diseases, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
| | - Xiaoyan Hao
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
- Henan Key Laboratory of Cerebrovascular Diseases, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
| | - Shuo Zhang
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
- Academy of Medical Sciences of Zhengzhou University, Zhengzhou, China
- Henan Key Laboratory of Cerebrovascular Diseases, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
| | - Zhengwei Hu
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
- Academy of Medical Sciences of Zhengzhou University, Zhengzhou, China
- Henan Key Laboratory of Cerebrovascular Diseases, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
| | - Zongping Xia
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
- Henan Key Laboratory of Cerebrovascular Diseases, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
- Clinical Systems Biology Laboratories, Zhengzhou University, Zhengzhou, China
| | - Changhe Shi
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
- Henan Key Laboratory of Cerebrovascular Diseases, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
- Institute of Neuroscience, Zhengzhou University, Zhengzhou, China
| | - Yuming Xu
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China.
- Henan Key Laboratory of Cerebrovascular Diseases, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China.
- Institute of Neuroscience, Zhengzhou University, Zhengzhou, China.
| | - Chengyuan Mao
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China.
- Henan Key Laboratory of Cerebrovascular Diseases, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China.
| |
Collapse
|
39
|
Yun C, Li N, Zhang Y, Fang T, Ma J, Zheng Z, Zhou S, Cai X. Glucose Transporter-Targeting Chimeras Enabling Tumor-Selective Degradation of Secreted and Membrane Proteins. ACS Chem Biol 2024; 19:2254-2263. [PMID: 39374326 DOI: 10.1021/acschembio.4c00584] [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: 10/09/2024]
Abstract
Tumor-selective degradation of target proteins has the potential to offer superior therapeutic benefits with maximized therapeutic windows and minimized off-target effects. However, the development of effective lysosome-targeted degradation platforms for achieving selective protein degradation in tumors remains a substantial challenge. Cancer cells depend on certain solute carrier (SLC) transporters to acquire extracellular nutrients to sustain their metabolism and growth. This current study exploits facilitative glucose transporters (GLUTs), a group of SLC transporters widely overexpressed in numerous types of cancer, to drive the endocytosis and lysosomal degradation of target proteins in tumor cells. GLUT-targeting chimeras (GTACs) were generated by conjugating multiple glucose ligands to an antibody specific for the target protein. We demonstrate that the constructed GTACs can induce the internalization and lysosomal degradation of the extracellular and membrane proteins streptavidin, tumor necrosis factor-alpha (TNF-α), and human epidermal growth factor receptor 2 (HER2). Compared with the parent antibody, the GTAC exhibited higher potency in inhibiting the growth of tumor cells in vitro and enhanced tumor-targeting capacity in a tumor-bearing mouse model. Thus, the GTAC platform represents a novel degradation strategy that harnesses an SLC transporter for tumor-selective depletion of secreted and membrane proteins of interest.
Collapse
Affiliation(s)
- Chengyu Yun
- School of Pharmaceutical Sciences, Sun Yat-sen University, 132 East Outer Ring Road, Guangzhou 510006, China
| | - Na Li
- School of Pharmaceutical Sciences, Sun Yat-sen University, 132 East Outer Ring Road, Guangzhou 510006, China
| | - Yishu Zhang
- School of Pharmaceutical Sciences, Sun Yat-sen University, 132 East Outer Ring Road, Guangzhou 510006, China
| | - Tong Fang
- School of Pharmaceutical Sciences, Sun Yat-sen University, 132 East Outer Ring Road, Guangzhou 510006, China
| | - Jing Ma
- School of Pharmaceutical Sciences, Sun Yat-sen University, 132 East Outer Ring Road, Guangzhou 510006, China
| | - Zhenting Zheng
- School of Pharmaceutical Sciences, Sun Yat-sen University, 132 East Outer Ring Road, Guangzhou 510006, China
| | - Subing Zhou
- School of Pharmaceutical Sciences, Sun Yat-sen University, 132 East Outer Ring Road, Guangzhou 510006, China
| | - Xiaoqing Cai
- School of Pharmaceutical Sciences, Sun Yat-sen University, 132 East Outer Ring Road, Guangzhou 510006, China
| |
Collapse
|
40
|
Liu N, Kattan WE, Mead BE, Kummerlowe C, Cheng T, Ingabire S, Cheah JH, Soule CK, Vrcic A, McIninch JK, Triana S, Guzman M, Dao TT, Peters JM, Lowder KE, Crawford L, Amini AP, Blainey PC, Hahn WC, Cleary B, Bryson B, Winter PS, Raghavan S, Shalek AK. Scalable, compressed phenotypic screening using pooled perturbations. Nat Biotechnol 2024:10.1038/s41587-024-02403-z. [PMID: 39375446 DOI: 10.1038/s41587-024-02403-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2023] [Accepted: 08/26/2024] [Indexed: 10/09/2024]
Abstract
High-throughput phenotypic screens using biochemical perturbations and high-content readouts are constrained by limitations of scale. To address this, we establish a method of pooling exogenous perturbations followed by computational deconvolution to reduce required sample size, labor and cost. We demonstrate the increased efficiency of compressed experimental designs compared to conventional approaches through benchmarking with a bioactive small-molecule library and a high-content imaging readout. We then apply compressed screening in two biological discovery campaigns. In the first, we use early-passage pancreatic cancer organoids to map transcriptional responses to a library of recombinant tumor microenvironment protein ligands, uncovering reproducible phenotypic shifts induced by specific ligands distinct from canonical reference signatures and correlated with clinical outcome. In the second, we identify the pleotropic modulatory effects of a chemical compound library with known mechanisms of action on primary human peripheral blood mononuclear cell immune responses. In sum, our approach empowers phenotypic screens with information-rich readouts to advance drug discovery efforts and basic biological inquiry.
Collapse
Affiliation(s)
- Nuo Liu
- Institute for Medical Engineering and Science (IMES) and Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA, USA
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, USA
- Program in Computational and Systems Biology, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Walaa E Kattan
- Institute for Medical Engineering and Science (IMES) and Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA, USA
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, USA
| | - Benjamin E Mead
- Institute for Medical Engineering and Science (IMES) and Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA, USA
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, USA
| | - Conner Kummerlowe
- Institute for Medical Engineering and Science (IMES) and Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA, USA
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, USA
- Program in Computational and Systems Biology, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Thomas Cheng
- Institute for Medical Engineering and Science (IMES) and Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA, USA
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, USA
| | - Sarah Ingabire
- Institute for Medical Engineering and Science (IMES) and Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA, USA
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, USA
| | - Jaime H Cheah
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Center for the Development of Therapeutics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Christian K Soule
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Center for the Development of Therapeutics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Anita Vrcic
- Center for the Development of Therapeutics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Jane K McIninch
- Center for the Development of Therapeutics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Sergio Triana
- Institute for Medical Engineering and Science (IMES) and Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA, USA
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, USA
| | - Manuel Guzman
- Institute for Medical Engineering and Science (IMES) and Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA, USA
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, USA
| | - Tyler T Dao
- Institute for Medical Engineering and Science (IMES) and Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA, USA
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, USA
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Joshua M Peters
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, USA
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Kristen E Lowder
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Dana Farber Cancer Institute, Boston, MA, USA
| | - Lorin Crawford
- Microsoft Research, Cambridge, MA, USA
- Center for Computational Biology, Brown University, Providence, RI, USA
- Department of Biostatistics, Brown University, Providence, RI, USA
| | | | - Paul C Blainey
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - William C Hahn
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Dana Farber Cancer Institute, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Brian Cleary
- Faculty of Computing and Data Sciences, Boston University, Boston, MA, USA
- Department of Biomedical Engineering, Boston University, Boston, MA, USA
- Department of Biology, Boston University, Boston, MA, USA
| | - Bryan Bryson
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, USA
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | | | - Srivatsan Raghavan
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Dana Farber Cancer Institute, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Alex K Shalek
- Institute for Medical Engineering and Science (IMES) and Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA, USA.
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA.
- Broad Institute of MIT and Harvard, Cambridge, MA, USA.
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, USA.
- Program in Computational and Systems Biology, Massachusetts Institute of Technology, Cambridge, MA, USA.
- Program in Immunology, Harvard Medical School, Boston, MA, USA.
- Harvard Stem Cell Institute, Cambridge, MA, USA.
| |
Collapse
|
41
|
Wang T, Yang C, Tang Y, Wen K, Ma Y, Chen Y, Li Z, Zhao Y, Zhu S, Meng X, Du S, Miao Z, Wei W, Deng H. Development of a new paradigm model for deciphering action mechanism of Danhong injection using a combination of isothermal shift assay and database interrogation. Chin Med 2024; 19:136. [PMID: 39369254 PMCID: PMC11452974 DOI: 10.1186/s13020-024-01017-6] [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: 06/28/2024] [Accepted: 09/28/2024] [Indexed: 10/07/2024] Open
Abstract
BACKGROUND Identification of active components of traditional Chinese Medicine (TCM) and their respective targets is important for understanding the mechanisms underlying TCM efficacy. However, there are still no effective technical methods to achieve this. METHODS Herein, we have established a method for rapidly identifying targets of a specific TCM and interrogating the targets with their corresponding active components based on Isothermal Shift Assay (iTSA) and database interrogation. RESULTS We optimized iTSA workflow and identified 110 targets for Danhong injection (DHI) which is used as an effective remedy for cardiovascular and cerebrovascular diseases. Moreover, we identified the targets of the nine major ingredients found in DHI. Database interrogation found that the potential targets for DHI, in which we verified that ADK as the target for salvianolic acid A and ALDH1B1 as the target for protocatechualdehyde in DHI, respectively. CONCLUSION Overall, we established a novel paradigm model for the identification of targets and their respective ingredients in DHI, which facilitates the discovery of drug candidates and targets for improving disease management and contributes to revealing the underlying mechanisms of TCM and fostering TCM development and modernization.
Collapse
Affiliation(s)
- Tianxiang Wang
- MOE Key Laboratory of Bioinformatics, Center for Synthetic and Systematic Biology, School of Life Sciences, Tsinghua University, Beijing, 100084, People's Republic of China
| | - Changmei Yang
- MOE Key Laboratory of Bioinformatics, Center for Synthetic and Systematic Biology, School of Life Sciences, Tsinghua University, Beijing, 100084, People's Republic of China
| | - Yuxiang Tang
- MOE Key Laboratory of Bioinformatics, Center for Synthetic and Systematic Biology, School of Life Sciences, Tsinghua University, Beijing, 100084, People's Republic of China
| | - Ke Wen
- MOE Key Laboratory of Bioinformatics, Center for Synthetic and Systematic Biology, School of Life Sciences, Tsinghua University, Beijing, 100084, People's Republic of China
| | - Yuxin Ma
- MOE Key Laboratory of Bioinformatics, Center for Synthetic and Systematic Biology, School of Life Sciences, Tsinghua University, Beijing, 100084, People's Republic of China
| | - Yuling Chen
- MOE Key Laboratory of Bioinformatics, Center for Synthetic and Systematic Biology, School of Life Sciences, Tsinghua University, Beijing, 100084, People's Republic of China
| | - Zhiqiang Li
- MOE Key Laboratory of Bioinformatics, Center for Synthetic and Systematic Biology, School of Life Sciences, Tsinghua University, Beijing, 100084, People's Republic of China
| | - Yujiao Zhao
- MOE Key Laboratory of Bioinformatics, Center for Synthetic and Systematic Biology, School of Life Sciences, Tsinghua University, Beijing, 100084, People's Republic of China
| | - Songbiao Zhu
- Chinese Institutes for Medical Research, Beijing, China
| | - Xianbin Meng
- MOE Key Laboratory of Bioinformatics, Center for Synthetic and Systematic Biology, School of Life Sciences, Tsinghua University, Beijing, 100084, People's Republic of China
| | - Sijing Du
- MOE Key Laboratory of Bioinformatics, Center for Synthetic and Systematic Biology, School of Life Sciences, Tsinghua University, Beijing, 100084, People's Republic of China
| | - Zelong Miao
- MOE Key Laboratory of Bioinformatics, Center for Synthetic and Systematic Biology, School of Life Sciences, Tsinghua University, Beijing, 100084, People's Republic of China
| | - Wei Wei
- Wangjing hospital of China Academy of Chinese Medical Sciences, Beijing, China.
| | - Haiteng Deng
- MOE Key Laboratory of Bioinformatics, Center for Synthetic and Systematic Biology, School of Life Sciences, Tsinghua University, Beijing, 100084, People's Republic of China.
| |
Collapse
|
42
|
Flickinger KM, Wilson KM, Rossiter NJ, Hunger AL, Vishwasrao PV, Lee TD, Mellado Fritz CA, Richards RM, Hall MD, Cantor JR. Conditional lethality profiling reveals anticancer mechanisms of action and drug-nutrient interactions. SCIENCE ADVANCES 2024; 10:eadq3591. [PMID: 39365851 PMCID: PMC11451515 DOI: 10.1126/sciadv.adq3591] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Accepted: 08/29/2024] [Indexed: 10/06/2024]
Abstract
Chemical screens across hundreds of cell lines have shown that the drug sensitivities of human cancers can vary by genotype or lineage. However, most drug discovery studies have relied on culture media that poorly reflect metabolite levels in human blood. Here, we perform drug screens in traditional and Human Plasma-Like Medium (HPLM). Sets of compounds that show conditional anticancer activity span different phases of global development and include non-oncology drugs. Comparisons of the synthetic and serum-derived components that comprise typical media trace sets of conditional phenotypes to nucleotide synthesis substrates. We also characterize a unique dual mechanism for brivudine, a compound approved for antiviral use. Brivudine selectively impairs cell growth in low folate conditions by targeting two enzymes involved in one-carbon metabolism. Cataloged gene essentiality data further suggest that conditional phenotypes for other compounds are linked to off-target effects. Our findings establish general strategies for identifying drug-nutrient interactions and mechanisms of action by exploiting conditional lethality in cancer cells.
Collapse
Affiliation(s)
- Kyle M. Flickinger
- Morgridge Institute for Research, Madison, WI 53715, USA
- Department of Biochemistry, University of Wisconsin–Madison, Madison, WI 53706, USA
| | - Kelli M. Wilson
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD, 20850, USA
| | - Nicholas J. Rossiter
- Cellular and Molecular Biology Program, University of Michigan, Ann Arbor, MI 48109, USA
| | - Andrea L. Hunger
- Department of Biochemistry, University of Wisconsin–Madison, Madison, WI 53706, USA
| | - Paresh V. Vishwasrao
- Division of Hematology, Oncology, and Bone Marrow Transplant, University of Wisconsin–Madison, Madison, WI 53706, USA
| | - Tobie D. Lee
- Early Translation Branch, National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD, 20850, USA
| | - Carlos A. Mellado Fritz
- Morgridge Institute for Research, Madison, WI 53715, USA
- Department of Biochemistry, University of Wisconsin–Madison, Madison, WI 53706, USA
| | - Rebecca M. Richards
- Division of Hematology, Oncology, and Bone Marrow Transplant, University of Wisconsin–Madison, Madison, WI 53706, USA
| | - Matthew D. Hall
- Early Translation Branch, National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD, 20850, USA
| | - Jason R. Cantor
- Morgridge Institute for Research, Madison, WI 53715, USA
- Department of Biochemistry, University of Wisconsin–Madison, Madison, WI 53706, USA
- Department of Biomedical Engineering, University of Wisconsin–Madison, Madison, WI 53706, USA
- Carbone Cancer Center, University of Wisconsin–Madison, Madison, WI 53792, USA
| |
Collapse
|
43
|
Min Y, Wei Y, Wang P, Wang X, Li H, Wu N, Bauer S, Zheng S, Shi Y, Wang Y, Wu J, Zhao D, Zeng J. From Static to Dynamic Structures: Improving Binding Affinity Prediction with Graph-Based Deep Learning. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2405404. [PMID: 39206846 PMCID: PMC11516055 DOI: 10.1002/advs.202405404] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Revised: 07/29/2024] [Indexed: 09/04/2024]
Abstract
Accurate prediction of protein-ligand binding affinities is an essential challenge in structure-based drug design. Despite recent advances in data-driven methods for affinity prediction, their accuracy is still limited, partially because they only take advantage of static crystal structures while the actual binding affinities are generally determined by the thermodynamic ensembles between proteins and ligands. One effective way to approximate such a thermodynamic ensemble is to use molecular dynamics (MD) simulation. Here, an MD dataset containing 3,218 different protein-ligand complexes is curated, and Dynaformer, a graph-based deep learning model is further developed to predict the binding affinities by learning the geometric characteristics of the protein-ligand interactions from the MD trajectories. In silico experiments demonstrated that the model exhibits state-of-the-art scoring and ranking power on the CASF-2016 benchmark dataset, outperforming the methods hitherto reported. Moreover, in a virtual screening on heat shock protein 90 (HSP90) using Dynaformer, 20 candidates are identified and their binding affinities are further experimentally validated. Dynaformer displayed promising results in virtual drug screening, revealing 12 hit compounds (two are in the submicromolar range), including several novel scaffolds. Overall, these results demonstrated that the approach offer a promising avenue for accelerating the early drug discovery process.
Collapse
Affiliation(s)
- Yaosen Min
- Institute for Interdisciplinary Information SciencesTsinghua UniversityBeijing100084China
| | - Ye Wei
- Institute for Interdisciplinary Information SciencesTsinghua UniversityBeijing100084China
| | - Peizhuo Wang
- Institute for Interdisciplinary Information SciencesTsinghua UniversityBeijing100084China
- School of Life Science and TechnologyXidian UniversityXi'an710071ShaanxiChina
| | - Xiaoting Wang
- School of MedicineTsinghua UniversityBeijing100084China
| | - Han Li
- Institute for Interdisciplinary Information SciencesTsinghua UniversityBeijing100084China
| | - Nian Wu
- Institute for Interdisciplinary Information SciencesTsinghua UniversityBeijing100084China
| | - Stefan Bauer
- Department of Intelligent SystemsKTHStockholm10044Sweden
| | | | - Yu Shi
- Microsoft Research AsiaBeijing100080China
| | - Yingheng Wang
- Department of Electrical EngineeringTsinghua UniversityBeijing100084China
| | - Ji Wu
- Department of Electrical EngineeringTsinghua UniversityBeijing100084China
| | - Dan Zhao
- Institute for Interdisciplinary Information SciencesTsinghua UniversityBeijing100084China
| | - Jianyang Zeng
- School of EngineeringWestlake UniversityHangzhou310030China
- Research Center for Industries of the FutureWestlake UniversityHangzhou310030China
- Present address:
Westlake Laboratory of Life Sciences and BiomedicineWestlake UniversityHangzhou310024China
| |
Collapse
|
44
|
Singh SK, Bhattacharjee M, Unni B, Kashyap RS, Malik A, Akhtar S, Fatima S. In silico testing to identify compounds that inhibit ClfA and ClfB binding to the host for the formulation of future drugs against Staphylococcus aureus colonization and infection. Front Cell Infect Microbiol 2024; 14:1422500. [PMID: 39411322 PMCID: PMC11475578 DOI: 10.3389/fcimb.2024.1422500] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Accepted: 08/30/2024] [Indexed: 10/19/2024] Open
Abstract
Introduction Staphylococcus aureus is a highly resistant pathogen. It has multiple virulence factors, which makes it one of the most pathogenic bacteria for humankind. The vast increase in antibiotic resistance in these bacteria is a warning of existing healthcare policies. Most of the available antibiotics are ineffective due to resistance; this situation requires the development of drugs that target specific proteins and are not susceptible to resistance. Methods In this study, we identified a compound that acts as an antagonist of ClfA and ClfB by inhibiting their binding to host cells. Results The shortlisted compound's binding activity was tested by docking and molecular dynamics during its interaction with proteins. The identified compound has excellent binding energy with both ClfA (-10.11 kcal/mol) and ClfB (-11.11 kcal/mol). Discussion The molecular dynamics of the protein and compound were stable and promising for further in vitro and in vivo tests. The performance of our compound was tested and compared with that of the control molecule allantodapsone, which was reported in a previous study as a pan inhibitor of the clumping factor. An ADMET study of our selected compound revealed its reliable drug likeliness. This compound is an ideal candidate for in vitro studies.
Collapse
Affiliation(s)
| | | | - Balagopalan Unni
- Faculty of Sciences, Assam Downtown University, Guwahati, Assam, India
| | - Rajpal Singh Kashyap
- Department of Research, Central India Institute of Medical Science, Nagpur, Maharasthra, India
| | - Abdul Malik
- College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Suhail Akhtar
- Department of Biochemistry, Andrew Taylor Still University of Health Science, Kirksville, MO, United States
| | - Sabiha Fatima
- College of Applied Medical Sciences, King Saud University, Riyadh, Saudi Arabia
| |
Collapse
|
45
|
Nguyen HT, Rissanen SL, Peltokangas M, Laakkonen T, Kettunen J, Barthod L, Sivakumar R, Palojärvi A, Junttila P, Talvitie J, Bassis M, Nickels SL, Kalvala S, Ilina P, Tammela P, Lehtonen S, Schwamborn JC, Mosser S, Singh P. Highly scalable and standardized organ-on-chip platform with TEER for biological barrier modeling. Tissue Barriers 2024; 12:2315702. [PMID: 38346163 PMCID: PMC11583584 DOI: 10.1080/21688370.2024.2315702] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2023] [Revised: 01/02/2024] [Accepted: 01/15/2024] [Indexed: 11/22/2024] Open
Abstract
The development of new therapies is hampered by the lack of predictive, and patient-relevant in vitro models. Organ-on-chip (OOC) technologies can potentially recreate physiological features and hold great promise for tissue and disease modeling. However, the non-standardized design of these chips and perfusion control systems has been a barrier to quantitative high-throughput screening (HTS). Here we present a scalable OOC microfluidic platform for applied kinetic in vitro assays (AKITA) that is applicable for high, medium, and low throughput. Its standard 96-well plate and 384-well plate layouts ensure compatibility with existing laboratory workflows and high-throughput data collection and analysis tools. The AKITA plate is optimized for the modeling of vascularized biological barriers, primarily the blood-brain barrier, skin, and lung, with precise flow control on a custom rocker. The integration of trans-epithelial electrical resistance (TEER) sensors allows rapid and repeated monitoring of barrier integrity over long time periods. Together with automated liquid handling and compound permeability testing analyses, we demonstrate the flexibility of the AKITA platform for establishing human-relevant models for preclinical drug and precision medicine's efficacy, toxicity, and permeability under near-physiological conditions.
Collapse
Affiliation(s)
- Hoang-Tuan Nguyen
- Finnadvance Ltd, Oulu, Finland
- Faculty of Biochemistry and Molecular Medicine, and Biocenter Oulu, University of Oulu, Oulu, Finland
| | | | | | | | | | | | | | | | | | | | - Michele Bassis
- Developmental and Cellular Biology, Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, Esch-sur-Alzette, Luxembourg
| | - Sarah L. Nickels
- Developmental and Cellular Biology, Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, Esch-sur-Alzette, Luxembourg
| | - Sara Kalvala
- A. I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Polina Ilina
- Drug Research Program, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland
| | - Päivi Tammela
- Drug Research Program, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland
| | - Sarka Lehtonen
- A. I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
- Neuroscience Center, University of Helsinki, Helsinki, Finland
| | - Jens C. Schwamborn
- Developmental and Cellular Biology, Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, Esch-sur-Alzette, Luxembourg
| | | | | |
Collapse
|
46
|
Bai Q, Shao E, Ma D, Jiao B, Scheetz SD, Hartnett-Scott KA, Ilin VA, Aizenman E, Kofler J, Burton EA. A human Tau expressing zebrafish model of progressive supranuclear palsy identifies Brd4 as a regulator of microglial synaptic elimination. Nat Commun 2024; 15:8195. [PMID: 39294122 PMCID: PMC11410960 DOI: 10.1038/s41467-024-52173-0] [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: 07/11/2024] [Accepted: 08/28/2024] [Indexed: 09/20/2024] Open
Abstract
Progressive supranuclear palsy (PSP) is an incurable neurodegenerative disease characterized by 4-repeat (0N/4R)-Tau protein accumulation in CNS neurons. We generated transgenic zebrafish expressing human 0N/4R-Tau to investigate PSP pathophysiology. Tau zebrafish replicated multiple features of PSP, including: decreased survival; hypokinesia; impaired optokinetic responses; neurodegeneration; neuroinflammation; synapse loss; and Tau hyperphosphorylation, misfolding, mislocalization, insolubility, truncation, and oligomerization. Using automated assays, we screened 147 small molecules for activity in rescuing neurological deficits in Tau zebrafish. (+)JQ1, a bromodomain inhibitor, improved hypokinesia, survival, microgliosis, and brain synapse elimination. A heterozygous brd4+/- mutant reducing expression of the bromodomain protein Brd4 similarly rescued these phenotypes. Microglial phagocytosis of synaptic material was decreased by (+)JQ1 in both Tau zebrafish and rat primary cortical cultures. Microglia in human PSP brains expressed Brd4. Our findings implicate Brd4 as a regulator of microglial synaptic elimination in tauopathy and provide an unbiased approach for identifying mechanisms and therapeutic targets in PSP.
Collapse
Affiliation(s)
- Qing Bai
- Department of Neurology, University of Pittsburgh, Pittsburgh, PA, 15213, USA
- Pittsburgh Institute for Neurodegenerative Diseases, University of Pittsburgh, Pittsburgh, PA, 15213, USA
| | - Enhua Shao
- Department of Neurology, University of Pittsburgh, Pittsburgh, PA, 15213, USA
- Pittsburgh Institute for Neurodegenerative Diseases, University of Pittsburgh, Pittsburgh, PA, 15213, USA
- Tsinghua University School of Medicine, Beijing, China
| | - Denglei Ma
- Department of Neurology, University of Pittsburgh, Pittsburgh, PA, 15213, USA
- Pittsburgh Institute for Neurodegenerative Diseases, University of Pittsburgh, Pittsburgh, PA, 15213, USA
| | - Binxuan Jiao
- Department of Neurology, University of Pittsburgh, Pittsburgh, PA, 15213, USA
- Pittsburgh Institute for Neurodegenerative Diseases, University of Pittsburgh, Pittsburgh, PA, 15213, USA
- Tsinghua University School of Medicine, Beijing, China
| | - Seth D Scheetz
- Department of Neurology, University of Pittsburgh, Pittsburgh, PA, 15213, USA
- Pittsburgh Institute for Neurodegenerative Diseases, University of Pittsburgh, Pittsburgh, PA, 15213, USA
| | - Karen A Hartnett-Scott
- Pittsburgh Institute for Neurodegenerative Diseases, University of Pittsburgh, Pittsburgh, PA, 15213, USA
- Department of Neurobiology, University of Pittsburgh, Pittsburgh, PA, 15213, USA
| | - Vladimir A Ilin
- Department of Neurology, University of Pittsburgh, Pittsburgh, PA, 15213, USA
- Pittsburgh Institute for Neurodegenerative Diseases, University of Pittsburgh, Pittsburgh, PA, 15213, USA
| | - Elias Aizenman
- Pittsburgh Institute for Neurodegenerative Diseases, University of Pittsburgh, Pittsburgh, PA, 15213, USA
- Department of Neurobiology, University of Pittsburgh, Pittsburgh, PA, 15213, USA
| | - Julia Kofler
- Department of Pathology, University of Pittsburgh, Pittsburgh, PA, 15213, USA
- Alzheimer's Disease Research Center, University of Pittsburgh, Pittsburgh, PA, 15213, USA
| | - Edward A Burton
- Department of Neurology, University of Pittsburgh, Pittsburgh, PA, 15213, USA.
- Pittsburgh Institute for Neurodegenerative Diseases, University of Pittsburgh, Pittsburgh, PA, 15213, USA.
- Geriatrics Research, Education and Clinical Center, Pittsburgh VA Healthcare System, Pittsburgh, PA, 15240, USA.
| |
Collapse
|
47
|
Ma J, Zhang W, Rahimialiabadi S, Ganesh NU, Sun Z, Parvez S, Peterson RT, Yeh JRJ. Instantaneous visual genotyping and facile site-specific transgenesis via CRISPR-Cas9 and phiC31 integrase. Biol Open 2024; 13:bio061666. [PMID: 39225039 PMCID: PMC11391820 DOI: 10.1242/bio.061666] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2024] [Accepted: 08/01/2024] [Indexed: 09/04/2024] Open
Abstract
Here, we introduce 'TICIT', targeted integration by CRISPR-Cas9 and integrase technologies, which utilizes the site-specific DNA recombinase - phiC31 integrase - to insert large DNA fragments into CRISPR-Cas9 target loci. This technique, which relies on first knocking in a 39-basepair phiC31 landing site via CRISPR-Cas9, enables researchers to repeatedly perform site-specific transgenesis at the exact genomic location with high precision and efficiency. We applied this approach to devise a method for the instantaneous determination of a zebrafish's genotype simply by examining its color. When a zebrafish mutant line must be propagated as heterozygotes due to homozygous lethality, employing this method allows facile identification of a population of homozygous mutant embryos even before the mutant phenotypes manifest. Thus, it should facilitate various downstream applications, such as large-scale chemical screens. We demonstrated that TICIT could also create reporter fish driven by an endogenous promoter. Further, we identified a landing site in the tyrosinase gene that could support transgene expression in a broad spectrum of tissue and cell types. In sum, TICIT enables site-specific DNA integration without requiring complex donor DNA construction. It can yield consistent transgene expression, facilitate diverse applications in zebrafish, and may be applicable to cells in culture and other model organisms.
Collapse
Affiliation(s)
- Junyan Ma
- Department of Basic Medical Science, Quanzhou Medical College, Quanzhou, Fujian 362011, China
- Cardiovascular Research Center, Massachusetts General Hospital, Charlestown, MA 02129, USA and Department of Medicine, Harvard Medical School, Boston, MA 02115, USA
| | - Weiting Zhang
- Cardiovascular Research Center, Massachusetts General Hospital, Charlestown, MA 02129, USA and Department of Medicine, Harvard Medical School, Boston, MA 02115, USA
| | - Simin Rahimialiabadi
- Department of Pharmacology and Toxicology, University of Utah, Salt Lake City, UT 84112, USA
| | - Nikkitha Umesh Ganesh
- Cardiovascular Research Center, Massachusetts General Hospital, Charlestown, MA 02129, USA and Department of Medicine, Harvard Medical School, Boston, MA 02115, USA
| | - Zhengwang Sun
- Center for Immunology and Inflammatory Disease, Massachusetts General Hospital, Charlestown, MA 02129, USA
| | - Saba Parvez
- Department of Pharmacology and Toxicology, University of Utah, Salt Lake City, UT 84112, USA
| | - Randall T. Peterson
- Department of Pharmacology and Toxicology, University of Utah, Salt Lake City, UT 84112, USA
| | - Jing-Ruey Joanna Yeh
- Cardiovascular Research Center, Massachusetts General Hospital, Charlestown, MA 02129, USA and Department of Medicine, Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
48
|
Vishwakarma S, Hernandez-Hernandez S, Ballester PJ. Graph neural networks are promising for phenotypic virtual screening on cancer cell lines. Biol Methods Protoc 2024; 9:bpae065. [PMID: 39502795 PMCID: PMC11537795 DOI: 10.1093/biomethods/bpae065] [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: 07/03/2024] [Revised: 08/20/2024] [Accepted: 09/02/2024] [Indexed: 11/08/2024] Open
Abstract
Artificial intelligence is increasingly driving early drug design, offering novel approaches to virtual screening. Phenotypic virtual screening (PVS) aims to predict how cancer cell lines respond to different compounds by focusing on observable characteristics rather than specific molecular targets. Some studies have suggested that deep learning may not be the best approach for PVS. However, these studies are limited by the small number of tested molecules as well as not employing suitable performance metrics and dissimilar-molecules splits better mimicking the challenging chemical diversity of real-world screening libraries. Here we prepared 60 datasets, each containing approximately 30 000-50 000 molecules tested for their growth inhibitory activities on one of the NCI-60 cancer cell lines. We conducted multiple performance evaluations of each of the five machine learning algorithms for PVS on these 60 problem instances. To provide even a more comprehensive evaluation, we used two model validation types: the random split and the dissimilar-molecules split. Overall, about 14 440 training runs aczross datasets were carried out per algorithm. The models were primarily evaluated using hit rate, a more suitable metric in VS contexts. The results show that all models are more challenged by test molecules that are substantially different from those in the training data. In both validation types, the D-MPNN algorithm, a graph-based deep neural network, was found to be the most suitable for building predictive models for this PVS problem.
Collapse
Affiliation(s)
- Sachin Vishwakarma
- Evotec SAS (France), Toulouse, France
- Centre de Recherche en Cancérologie de Marseille, Marseille 13009, France
| | | | - Pedro J Ballester
- Department of Bioengineering, Imperial College London, London SW7 2AZ, United Kingdom
| |
Collapse
|
49
|
Schuhmacher A, Gassmann O, Hinder M, Hartl D. Comparative analysis of FDA approvals by top 20 pharma companies (2014-2023). Drug Discov Today 2024; 29:104128. [PMID: 39097219 DOI: 10.1016/j.drudis.2024.104128] [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: 06/17/2024] [Revised: 07/22/2024] [Accepted: 07/30/2024] [Indexed: 08/05/2024]
Abstract
This article addresses the research and development (R&D) productivity challenge of the pharmaceutical industry, focusing on United States Food and Drug Administration (FDA)-related new drug approvals of the top 20 pharmaceutical companies (2014-2023). We evaluated the degree of innovation in new drugs to determine the innovativeness of these leading companies. A key finding of our analysis is the decline in the number of new drugs approved by the FDA for these leading companies over the investigated time period. This trend suggests that some of the leading companies are losing ground in R&D innovation, raising concerns about their ability to sustain competitive advantage, ensure long-term market success, and maintain viable business models.
Collapse
Affiliation(s)
- Alexander Schuhmacher
- Technische Hochschule Ingolstadt, THI Business School, Esplanade 10, D-85049 Ingolstadt, Germany; University of St. Gallen, Institute of Technology Management, Dufourstrasse 40a, CH-9000 St. Gallen, Switzerland.
| | - Oliver Gassmann
- University of St. Gallen, Institute of Technology Management, Dufourstrasse 40a, CH-9000 St. Gallen, Switzerland
| | - Markus Hinder
- Novartis, Development, Patient Safety, Forum 1, CH-4002 Basel, Switzerland; Fresenius University of Applied Sciences, Moritzstr. 17a, D-65185 Wiesbaden, Germany
| | - Dominik Hartl
- University of Tübingen, Hoppe-Seyler-Strasse 1, D-72076 Tübingen, Germany; Granite Bio, Aeschenvorstadt 36, CH-4051 Basel, Switzerland
| |
Collapse
|
50
|
Dee W, Sequeira I, Lobley A, Slabaugh G. Cell-vision fusion: A Swin transformer-based approach for predicting kinase inhibitor mechanism of action from Cell Painting data. iScience 2024; 27:110511. [PMID: 39175778 PMCID: PMC11340608 DOI: 10.1016/j.isci.2024.110511] [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: 12/22/2023] [Revised: 04/08/2024] [Accepted: 07/11/2024] [Indexed: 08/24/2024] Open
Abstract
Image-based profiling of the cellular response to drug compounds has proven effective at characterizing the morphological changes resulting from perturbation experiments. As data availability increases, however, there are growing demands for novel deep-learning methods. We applied the SwinV2 computer vision architecture to predict the mechanism of action of 10 kinase inhibitor compounds directly from Cell Painting images. This method outperforms the standard approach of using image-based profiles (IBP)-multidimensional feature set representations generated by bioimaging software. Furthermore, our fusion approach-cell-vision fusion, combining three different data modalities, images, IBPs, and chemical structures-achieved 69.79% accuracy and 70.56% F1 score, 4.20% and 5.49% higher, respectively, than the best-performing IBP method. We provide three techniques, specific to Cell Painting images, which enable deep-learning architectures to train effectively and demonstrate approaches to combat the significant batch effects present in large Cell Painting datasets.
Collapse
Affiliation(s)
- William Dee
- Digital Environment Research Institute (DERI), Queen Mary University of London, London E1 1HH, UK
- Centre for Oral Immunobiology and Regenerative Medicine, Barts Centre for Squamous Cancer, Institute of Dentistry, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London E1 2AD, UK
- Exscientia Plc, The Schrödinger Building Oxford Science Park, Oxford OX4 4GE, UK
| | - Ines Sequeira
- Centre for Oral Immunobiology and Regenerative Medicine, Barts Centre for Squamous Cancer, Institute of Dentistry, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London E1 2AD, UK
| | - Anna Lobley
- Exscientia Plc, The Schrödinger Building Oxford Science Park, Oxford OX4 4GE, UK
| | - Gregory Slabaugh
- Digital Environment Research Institute (DERI), Queen Mary University of London, London E1 1HH, UK
| |
Collapse
|