1
|
Munshi S, Alarbi AM, Zheng H, Kuplicki R, Burrows K, Figueroa-Hall LK, Victor TA, Aupperle RL, Khalsa SS, Paulus MP, Teague TK, Savitz J. Increased expression of ER stress, inflammasome activation, and mitochondrial biogenesis-related genes in peripheral blood mononuclear cells in major depressive disorder. Mol Psychiatry 2025; 30:574-586. [PMID: 39174649 DOI: 10.1038/s41380-024-02695-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/05/2023] [Accepted: 08/09/2024] [Indexed: 08/24/2024]
Abstract
A subset of major depressive disorder (MDD) is characterized by immune system dysfunction, but the intracellular origin of these immune changes remains unclear. Here we tested the hypothesis that abnormalities in endoplasmic reticulum (ER) stress, inflammasome activity and mitochondrial biogenesis contribute to the development of systemic inflammation in MDD. RT-qPCR was used to measure mRNA expression of key organellar genes from peripheral blood mononuclear cells (PBMCs) isolated from 186 MDD and 67 healthy control (HC) subjects. The comparative CT (2-ΔΔCT) method was applied to quantify mRNA expression using GAPDH as the reference gene. After controlling for age, sex, BMI, and medication status using linear regression models, expression of the inflammasome (NLRC4 and NLRP3) and the ER stress (XBP1u, XBP1s, and ATF4) genes was found to be significantly increased in the MDD versus the HC group. Sensitivity analyses excluding covariates yielded similar results. After excluding outliers, expression of the inflammasome genes was no longer statistically significant but expression of the ER stress genes (XBP1u, XBP1s, and ATF4) remained significant and the mitochondrial biogenesis gene, MFN2, was significantly increased in the MDD group. NLRC4 and MFN2 were positively correlated with serum C-reactive protein concentrations, while ASC trended significant. The altered expression of inflammasome activation, ER stress, and mitochondrial biogenesis pathway components suggest that dysfunction of these organelles may play a role in the pathogenesis of MDD.
Collapse
Affiliation(s)
- Soumyabrata Munshi
- Laureate Institute for Brain Research, 6655 S. Yale Ave., Tulsa, OK, 74136, USA.
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Oklahoma Health Sciences Center, 1110 N. Stonewall Avenue, Oklahoma City, OK, 73117, USA.
| | - Ahlam M Alarbi
- Integrative Immunology Center, Department of Surgery and Department of Psychiatry, University of Oklahoma - School of Community Medicine, 4502 E. 41st St., Tulsa, OK, 74135, USA
| | - Haixia Zheng
- Laureate Institute for Brain Research, 6655 S. Yale Ave., Tulsa, OK, 74136, USA
- Oxley College of Health and Natural Sciences, The University of Tulsa, Tulsa, OK, 74199, USA
| | - Rayus Kuplicki
- Laureate Institute for Brain Research, 6655 S. Yale Ave., Tulsa, OK, 74136, USA
| | - Kaiping Burrows
- Laureate Institute for Brain Research, 6655 S. Yale Ave., Tulsa, OK, 74136, USA
| | - Leandra K Figueroa-Hall
- Laureate Institute for Brain Research, 6655 S. Yale Ave., Tulsa, OK, 74136, USA
- Oxley College of Health and Natural Sciences, The University of Tulsa, Tulsa, OK, 74199, USA
| | - Teresa A Victor
- Laureate Institute for Brain Research, 6655 S. Yale Ave., Tulsa, OK, 74136, USA
| | - Robin L Aupperle
- Laureate Institute for Brain Research, 6655 S. Yale Ave., Tulsa, OK, 74136, USA
- Oxley College of Health and Natural Sciences, The University of Tulsa, Tulsa, OK, 74199, USA
| | - Sahib S Khalsa
- Laureate Institute for Brain Research, 6655 S. Yale Ave., Tulsa, OK, 74136, USA
- Department of Psychiatry and Biobehavioral Sciences, Semel Institute for Neuroscience and Human Behavior, David Geffen School of Medicine, University of California at Los Angeles, 300 UCLA Medical Plaza, Los Angeles, CA, 90095, USA
| | - Martin P Paulus
- Laureate Institute for Brain Research, 6655 S. Yale Ave., Tulsa, OK, 74136, USA
- Oxley College of Health and Natural Sciences, The University of Tulsa, Tulsa, OK, 74199, USA
| | - T Kent Teague
- Integrative Immunology Center, Department of Surgery and Department of Psychiatry, University of Oklahoma - School of Community Medicine, 4502 E. 41st St., Tulsa, OK, 74135, USA
- Department of Biochemistry and Microbiology, Center for Health Sciences, Oklahoma State University, 1111 W. 17th St., Tulsa, OK, 74107, USA
| | - Jonathan Savitz
- Laureate Institute for Brain Research, 6655 S. Yale Ave., Tulsa, OK, 74136, USA
- Oxley College of Health and Natural Sciences, The University of Tulsa, Tulsa, OK, 74199, USA
| |
Collapse
|
2
|
Ezoe A, Shimada Y, Sawada R, Douke A, Shibata T, Kadowaki M, Yamanishi Y. Pathway-based prediction of the therapeutic effects and mode of action of custom-made multiherbal medicines. Mol Inform 2024; 43:e202400108. [PMID: 39404192 DOI: 10.1002/minf.202400108] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Revised: 06/23/2024] [Accepted: 06/24/2024] [Indexed: 11/14/2024]
Abstract
Multiherbal medicines are traditionally used as personalized medicines with custom combinations of crude drugs; however, the mechanisms of multiherbal medicines are unclear. In this study, we developed a novel pathway-based method to predict therapeutic effects and the mode of action of custom-made multiherbal medicines using machine learning. This method considers disease-related pathways as therapeutic targets and evaluates the comprehensive influence of constituent compounds on their potential target proteins in the disease-related pathways. Our proposed method enabled us to comprehensively predict new indications of 194 Kampo medicines for 87 diseases. Using Kampo-induced transcriptomic data, we demonstrated that Kampo constituent compounds stimulated the disease-related proteins and a customized Kampo formula enhanced the efficacy compared with an existing Kampo formula. The proposed method will be useful for discovering effective Kampo medicines and optimizing custom-made multiherbal medicines in practice.
Collapse
Affiliation(s)
- Akihiro Ezoe
- Department of Bioscience and Bioinformatics, Kyushu Institute of Technology, Iizuka, Fukuoka, 820-8502, Japan
- Plant Genomic Network Research Team, RIKEN Center for Sustainable Resource Science (CSRS), Yokohama, Kanagawa, 230-0045, Japan
| | - Yuki Shimada
- Department of Bioscience and Bioinformatics, Kyushu Institute of Technology, Iizuka, Fukuoka, 820-8502, Japan
| | - Ryusuke Sawada
- Department of Bioscience and Bioinformatics, Kyushu Institute of Technology, Iizuka, Fukuoka, 820-8502, Japan
- Department of Pharmacology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Kita-ku, Okayama, 700-8558, Japan
| | - Akihiro Douke
- Department of Bioscience and Bioinformatics, Kyushu Institute of Technology, Iizuka, Fukuoka, 820-8502, Japan
| | - Tomokazu Shibata
- Department of Bioscience and Bioinformatics, Kyushu Institute of Technology, Iizuka, Fukuoka, 820-8502, Japan
| | - Makoto Kadowaki
- Research Center for Pre-Disease Science, University of Toyama, Sugitani, Toyama, 930-0194, Japan
| | - Yoshihiro Yamanishi
- Department of Bioscience and Bioinformatics, Kyushu Institute of Technology, Iizuka, Fukuoka, 820-8502, Japan
- Graduate School of Informatics, Nagoya University, Chikusa, Nagoya, 464-8601, Japan
| |
Collapse
|
3
|
Zhang L, Li Y, Hu W, Gao S, Tang Y, Sun L, Jiang N, Xiao Z, Han L, Zhou W. Computational identification of mitochondrial dysfunction biomarkers in severe SARS-CoV-2 infection: Facilitating therapeutic applications of phytomedicine. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 131:155784. [PMID: 38878325 DOI: 10.1016/j.phymed.2024.155784] [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: 12/11/2023] [Revised: 03/18/2024] [Accepted: 04/13/2024] [Indexed: 06/25/2024]
Abstract
BACKGROUND Currently, SARS-CoV-2 has not disappeared and continues to prevail worldwide, with the ongoing risk of mutations and the potential for severe COVID-19. The impairment of monocyte mitochondrial function caused by SARS-CoV-2, leading to a metabolic and immune dysregulation, is a crucial factor in the development of severe COVID-19. PURPOSE Discover effective phytomedicines based on mitochondrial-related biomarkers in severe SARS-CoV-2 infection. METHODS Firstly, differential gene analysis and gene set enrichment analysis (GSEA) were conducted on monocytes datasets to identify genes and pathways distinguishing severe patients from uninfected individuals. Then, GO and KEGG enrichment analysis on the differentially expressed genes (DEGs) obtained. Take the DEGs and intersect them with the MitoCarta 3.0 gene set to obtain the differentially expressed mitochondrial-related genes (DE-MRGs). Subsequently, machine learning algorithms were employed to screen potential mitochondrial dysfunction biomarkers for severe COVID-19 based on score values. ROC curves were then plotted to assess the distinguish capability of the biomarkers, followed by validation using two additional independent datasets. Next, the effects of the identified biomarkers on metabolic pathways and immune cells were explored through Gene Set Variation Analysis (GSVA) and CIBERSORT. Finally, potential nature products for severe COVID-19 were screened from the expression profile dataset based on dysregulated mitochondrial-related genes, followed by in vitro experimental validation. RESULTS There are 1812 DEGs and 17 dysregulated mitochondrial processes between severe COVID-19 patients and uninfected individuals. A total of 77 DE-MRGs were identified, and the potential biomarkers were identified as RECQL4, PYCR1, PIF1, POLQ, and GLDC. In both the training and validation sets, the area under the ROC curve (AUC) for these five biomarkers was greater than 0.9. And they did not show significant changes in mild to moderate patients (p > 0.05), indicating their ability to effectively distinguish severe COVID-19. These biomarkers exhibit a highly significant correlation with the dysregulated metabolic processes (p < 0.05) and immune cell imbalance (p < 0.05) in severe patients, as demonstrated by GSVA and CIBERSORT algorithms. Curcumin has the highest score in the predictive model based on transcriptomic data from 496 natural compounds (p = 0.02; ES = 0.90). Pre-treatment with curcumin for 8 h has been shown to alleviate mitochondrial membrane potential damage caused by the SARS-CoV-2 S1 protein (p < 0.05) and reduce elevated levels of reactive oxygen species (ROS) (p < 0.01). CONCLUSION The results of this study indicate a significant correlation between severe SARS-CoV-2 infection and mitochondrial dysfunction. The proposed mitochondrial dysfunction biomarkers identified in this study are associated with the disease progression, metabolic and immune changes in severe SARS-CoV-2 infected patients. Curcumin has a potential role in preventing severe COVID-19 by protecting mitochondrial function. Our findings provide new strategies for predicting the prognosis and enabling early intervention in SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Lihui Zhang
- Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; Beijing Institute of Pharmacology & Toxicology, Beijing 100850, China; State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology & Toxicology, Beijing 100850, China
| | - Yuehan Li
- Beijing Institute of Pharmacology & Toxicology, Beijing 100850, China; State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology & Toxicology, Beijing 100850, China
| | - Wanting Hu
- Beijing Institute of Pharmacology & Toxicology, Beijing 100850, China; State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology & Toxicology, Beijing 100850, China
| | - Shengqiao Gao
- Beijing Institute of Pharmacology & Toxicology, Beijing 100850, China; State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology & Toxicology, Beijing 100850, China
| | - Yiran Tang
- Beijing Institute of Pharmacology & Toxicology, Beijing 100850, China; State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology & Toxicology, Beijing 100850, China
| | - Lei Sun
- Beijing Institute of Pharmacology & Toxicology, Beijing 100850, China; State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology & Toxicology, Beijing 100850, China
| | - Ning Jiang
- Beijing Institute of Pharmacology & Toxicology, Beijing 100850, China; State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology & Toxicology, Beijing 100850, China
| | - Zhiyong Xiao
- Beijing Institute of Pharmacology & Toxicology, Beijing 100850, China; State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology & Toxicology, Beijing 100850, China
| | - Lu Han
- Beijing Institute of Pharmacology & Toxicology, Beijing 100850, China; State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology & Toxicology, Beijing 100850, China.
| | - Wenxia Zhou
- Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; Beijing Institute of Pharmacology & Toxicology, Beijing 100850, China; State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology & Toxicology, Beijing 100850, China.
| |
Collapse
|
4
|
Li SQ, Xu WT, Yin YX, Wei HT, Li KZ, Xie MZ, Lv F, Xie LY, Hu BL. SNHG4-mediated PTEN destabilization confers oxaliplatin resistance in colorectal cancer cells by inhibiting ferroptosis. Apoptosis 2024; 29:835-848. [PMID: 38573492 DOI: 10.1007/s10495-024-01948-3] [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] [Accepted: 02/18/2024] [Indexed: 04/05/2024]
Abstract
Oxaliplatin resistance poses a significant challenge in colorectal cancer (CRC) therapy, necessitating further investigation into the underlying molecular mechanisms. This study aimed to elucidate the regulatory role of SNHG4 in oxaliplatin resistance and ferroptosis in CRC. Our findings revealed that treatment with oxaliplatin led to downregulation of SNHG4 expression in CRC cells, while resistant CRC cells exhibited higher levels of SNHG4 compared to parental cells. Silencing SNHG4 attenuated oxaliplatin resistance and reduced the expression of resistance-related proteins MRD1 and MPR1. Furthermore, induction of ferroptosis effectively diminished oxaliplatin resistance in both parental and resistant CRC cells. Notably, ferroptosis induction resulted in decreased SNHG4 expression, whereas SNHG4 overexpression suppressed ferroptosis. Through FISH, RIP, and RNA pull-down assays, we identified the cytoplasmic localization of both SNHG4 and PTEN, establishing that SNHG4 directly targets PTEN, thereby reducing mRNA stability in CRC cells. Silencing PTEN abrogated the impact of SNHG4 on oxaliplatin resistance and ferroptosis in CRC cells. In vivo experiments further validated the influence of SNHG4 on oxaliplatin resistance and ferroptosis in CRC cells through PTEN regulation. In conclusion, SNHG4 promotes resistance to oxaliplatin in CRC cells by suppressing ferroptosis through instability of PTEN, thus serves as a target for patients with oxaliplatin-base chemoresistance.
Collapse
Affiliation(s)
- Si-Qi Li
- Department of Research, Guangxi Medical University Cancer Hospital, No. 71 Hedi Road, Nanning, 530021, Guangxi, China
| | - Wen-Ting Xu
- Department of Research, Guangxi Medical University Cancer Hospital, No. 71 Hedi Road, Nanning, 530021, Guangxi, China
| | - Yi-Xin Yin
- Department of Research, Guangxi Medical University Cancer Hospital, No. 71 Hedi Road, Nanning, 530021, Guangxi, China
| | - Hao-Tang Wei
- Department of Gastrointestinal Surgery, Third Affiliated Hospital of Guangxi Medical University, Guangxi, 530031, China
| | - Ke-Zhi Li
- Department of Research, Guangxi Medical University Cancer Hospital, No. 71 Hedi Road, Nanning, 530021, Guangxi, China
| | - Ming-Zhi Xie
- Department of Chemotherapy, Guangxi Medical University Cancer Hospital, Nanning, 530021, Guangxi, China
| | - Feng Lv
- Department of Research, Guangxi Medical University Cancer Hospital, No. 71 Hedi Road, Nanning, 530021, Guangxi, China
| | - Li-Ye Xie
- Department of Research, Guangxi Medical University Cancer Hospital, No. 71 Hedi Road, Nanning, 530021, Guangxi, China
| | - Bang-Li Hu
- Department of Research, Guangxi Medical University Cancer Hospital, No. 71 Hedi Road, Nanning, 530021, Guangxi, China.
| |
Collapse
|
5
|
Gonzalez Gomez C, Rosa-Calatrava M, Fouret J. Optimizing in silico drug discovery: simulation of connected differential expression signatures and applications to benchmarking. Brief Bioinform 2024; 25:bbae299. [PMID: 38935068 PMCID: PMC11210109 DOI: 10.1093/bib/bbae299] [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: 03/28/2024] [Revised: 05/19/2024] [Accepted: 06/11/2024] [Indexed: 06/28/2024] Open
Abstract
BACKGROUND We present a novel simulation method for generating connected differential expression signatures. Traditional methods have struggled with the lack of reliable benchmarking data and biases in drug-disease pair labeling, limiting the rigorous benchmarking of connectivity-based approaches. OBJECTIVE Our aim is to develop a simulation method based on a statistical framework that allows for adjustable levels of parametrization, especially the connectivity, to generate a pair of interconnected differential signatures. This could help to address the issue of benchmarking data availability for connectivity-based drug repurposing approaches. METHODS We first detailed the simulation process and how it reflected real biological variability and the interconnectedness of gene expression signatures. Then, we generated several datasets to enable the evaluation of different existing algorithms that compare differential expression signatures, providing insights into their performance and limitations. RESULTS Our findings demonstrate the ability of our simulation to produce realistic data, as evidenced by correlation analyses and the log2 fold-change distribution of deregulated genes. Benchmarking reveals that methods like extreme cosine similarity and Pearson correlation outperform others in identifying connected signatures. CONCLUSION Overall, our method provides a reliable tool for simulating differential expression signatures. The data simulated by our tool encompass a wide spectrum of possibilities to challenge and evaluate existing methods to estimate connectivity scores. This may represent a critical gap in connectivity-based drug repurposing research because reliable benchmarking data are essential for assessing and advancing in the development of new algorithms. The simulation tool is available as a R package (General Public License (GPL) license) at https://github.com/cgonzalez-gomez/cosimu.
Collapse
Affiliation(s)
- Catalina Gonzalez Gomez
- CIRI, Centre International de Recherche en Infectiologie, Team VirPath, Univ Lyon, 14 Inserm, U1111, Université Claude Bernard Lyon 1, CNRS, UMR5308, ENS de Lyon, F-15 69007 Lyon, Rhône-Alpes, France
- International Associated Laboratory RespiVir France—Canada, Centre de Recherche en Infectiologie, Faculté de Médecine RTH Laennec 69008 Lyon, Université Claude Bernard Lyon 1, Université de Lyon, INSERM, CNRS, ENS de Lyon, France, Centre Hospitalier Universitaire de Québec - Université Laval, QC G1V 4G2 Québec, Canada
- Nexomis, Faculté de Médecine RTH Laennec, Université Claude Bernard Lyon 1, Université de Lyon, 7 Rue Guillaume Paradin, 69008 Lyon, Rhône-Alpes, France
- Signia Therapeutics, 60 Avenue Rockefeller, 69008 Lyon, Rhône-Alpes, France
| | - Manuel Rosa-Calatrava
- CIRI, Centre International de Recherche en Infectiologie, Team VirPath, Univ Lyon, 14 Inserm, U1111, Université Claude Bernard Lyon 1, CNRS, UMR5308, ENS de Lyon, F-15 69007 Lyon, Rhône-Alpes, France
- International Associated Laboratory RespiVir France—Canada, Centre de Recherche en Infectiologie, Faculté de Médecine RTH Laennec 69008 Lyon, Université Claude Bernard Lyon 1, Université de Lyon, INSERM, CNRS, ENS de Lyon, France, Centre Hospitalier Universitaire de Québec - Université Laval, QC G1V 4G2 Québec, Canada
- Nexomis, Faculté de Médecine RTH Laennec, Université Claude Bernard Lyon 1, Université de Lyon, 7 Rue Guillaume Paradin, 69008 Lyon, Rhône-Alpes, France
- VirNext, Faculté de Médecine RTH Laennec, Université Claude Bernard Lyon 1, Université de Lyon, 7 Rue Guillaume Paradin, 69008 Lyon, Rhône-Alpes, France
| | - Julien Fouret
- CIRI, Centre International de Recherche en Infectiologie, Team VirPath, Univ Lyon, 14 Inserm, U1111, Université Claude Bernard Lyon 1, CNRS, UMR5308, ENS de Lyon, F-15 69007 Lyon, Rhône-Alpes, France
- International Associated Laboratory RespiVir France—Canada, Centre de Recherche en Infectiologie, Faculté de Médecine RTH Laennec 69008 Lyon, Université Claude Bernard Lyon 1, Université de Lyon, INSERM, CNRS, ENS de Lyon, France, Centre Hospitalier Universitaire de Québec - Université Laval, QC G1V 4G2 Québec, Canada
- Nexomis, Faculté de Médecine RTH Laennec, Université Claude Bernard Lyon 1, Université de Lyon, 7 Rue Guillaume Paradin, 69008 Lyon, Rhône-Alpes, France
- Signia Therapeutics, 60 Avenue Rockefeller, 69008 Lyon, Rhône-Alpes, France
| |
Collapse
|
6
|
Hossein Nowroozzadeh M, Yousefi M, Abuali M, Sanie-Jahromi F. Effect of adalimumab as an anti-inflammatory agent on gene expression of retinal pigment epithelial cells. Biomed Pharmacother 2024; 174:116568. [PMID: 38599062 DOI: 10.1016/j.biopha.2024.116568] [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: 01/28/2024] [Revised: 03/31/2024] [Accepted: 04/04/2024] [Indexed: 04/12/2024] Open
Abstract
Adalimumab (ADA) is an anti-inflammatory antibody that has FDA approval as a systemic medication for treating noninfectious uveitis. It is also provisionally being investigated as an intravitreal injection for various retinal conditions. This study aimed to assess the effect of ADA on apoptotic, inflammatory, and fibrogenesis gene expression at mRNA and protein levels in retinal pigment epithelial (RPE) cells. RPEs were treated with serial concentrations of ADA (0.5x, x, 2x, and 4x; [x = 250 µg/mL]) for 24 hours. MTT assay was done and the mRNA and protein expressions were quantified using real-time PCR and ELISA assay, respectively. The mRNA levels of IL-1b and IL-6 were significantly increased in ADA-treated RPEs at 0.5x and x concentrations. However, the increase in cytokine secretion was observed only in IL-1b at x concentration. TGF-β was significantly upregulated in the 0.5x and 4x doses of ADA both at mRNA and protein levels. MTT assay, along with an unchanged BCL-2/BAX ratio confirmed the safety of ADA on RPEs at all studied concentrations. In conclusion, despite its safety, the 2x concentration of ADA was the only dose that did not ignite the expression of any of the studied inflammatory and fibrogenesis genes. This dosage, which is roughly equal to 2 mg intravitreal dose in a clinical setting, might be referred to as a reference starting point for future in-vivo studies in ocular conditions.
Collapse
Affiliation(s)
- M Hossein Nowroozzadeh
- Poostchi Ophthalmology Research Center, Department of Ophthalmology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Islamic Republic of Iran.
| | - Mojtaba Yousefi
- Poostchi Ophthalmology Research Center, Department of Ophthalmology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Islamic Republic of Iran.
| | - Mostafa Abuali
- Poostchi Ophthalmology Research Center, Department of Ophthalmology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Islamic Republic of Iran.
| | - Fatemeh Sanie-Jahromi
- Poostchi Ophthalmology Research Center, Department of Ophthalmology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Islamic Republic of Iran.
| |
Collapse
|
7
|
Jiang L, Qu S, Yu Z, Wang J, Liu X. MOASL: Predicting drug mechanism of actions through similarity learning with transcriptomic signature. Comput Biol Med 2024; 169:107853. [PMID: 38104518 DOI: 10.1016/j.compbiomed.2023.107853] [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: 07/19/2023] [Revised: 11/02/2023] [Accepted: 12/11/2023] [Indexed: 12/19/2023]
Abstract
Understanding the mechanisms of actions (MOAs) of compounds is crucial in drug discovery. A common step in drug MOAs annotation is to query the dysregulated gene signatures induced by drugs in a reference library of pre-defined signatures. However, traditional similarity-based computational strategies face challenges when dealing with high-dimensional and noisy transcriptional signature data. To address this issue, we introduce MOASL (MOAs prediction via Similarity Learning), a novel approach that contrastive to learn similarity embeddings among signatures with shared MOAs automatically. We evaluated the accuracy of signature matching on various transcriptional activity score (TAS) datasets and individual cell lines by using MOASL. The results show MOASL achieved higher performance over several statistical and machine learning methods. Furthermore, we provided the rationale of our model by visualizing the signature annotation procedure. Using MOASL, the MOAs label of query signature could be conveniently defined by calculating the similarity between the query embedding and the reference embeddings. Finally, we applied MOASL to repurpose thousands of compounds as glucocorticoid receptor (GR) agonists, accurately identifying 8 out of the top 10 compounds. MOASL is conveniently accessible on GitHub at https://github.com/jianglikun/MOASL, empowering researchers and practitioners in the field of drug discovery to predict the MOAs of drug.
Collapse
Affiliation(s)
- Likun Jiang
- Department of Computer Science, Xiamen University, Xiamen 361005, PR China; National Institute for Data Science in Health and Medicine, Xiamen University, Xiamen 361005, PR China
| | - Susu Qu
- Academy for Advanced Interdisciplinary Studies, Peking University, Beijing 100871, PR China; Chinese Institute for Brain Research, Beijing 102206, PR China
| | - Zhengqiu Yu
- National Institute for Data Science in Health and Medicine, Xiamen University, Xiamen 361005, PR China; School of Medicine, Xiamen University, Xiamen 361005, PR China
| | - Jianmin Wang
- The Interdisciplinary Graduate Program in Integrative Biotechnology and Translational Medicine, Yonsei University, Incheon 21983, South Korea
| | - Xiangrong Liu
- Department of Computer Science, Xiamen University, Xiamen 361005, PR China; National Institute for Data Science in Health and Medicine, Xiamen University, Xiamen 361005, PR China.
| |
Collapse
|
8
|
Qu X, Du G, Hu J, Cai Y. Graph-DTI: A New Model for Drug-target Interaction Prediction Based on Heterogenous Network Graph Embedding. Curr Comput Aided Drug Des 2024; 20:1013-1024. [PMID: 37448360 DOI: 10.2174/1573409919666230713142255] [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: 12/21/2022] [Revised: 05/04/2023] [Accepted: 05/26/2023] [Indexed: 07/15/2023]
Abstract
BACKGROUND In this study, we aimed to develop a new end-to-end learning model called Graph-Drug-Target Interaction (DTI), which integrates various types of information in the heterogeneous network data, and to explore automatic learning of the topology-maintaining representations of drugs and targets, thereby effectively contributing to the prediction of DTI. Precise predictions of DTI can guide drug discovery and development. Most machine learning algorithms integrate multiple data sources and combine them with common embedding methods. However, the relationship between the drugs and target proteins is not well reported. Although some existing studies have used heterogeneous network graphs for DTI prediction, there are many limitations in the neighborhood information between the nodes in the heterogeneous network graphs. We studied the drug-drug interaction (DDI) and DTI from DrugBank Version 3.0, protein-protein interaction (PPI) from the human protein reference database Release 9, drug structure similarity from Morgan fingerprints of radius 2 and calculated by RDKit, and protein sequence similarity from Smith-Waterman score. METHODS Our study consists of three major components. First, various drugs and target proteins were integrated, and a heterogeneous network was established based on a series of data sets. Second, the graph neural networks-inspired graph auto-encoding method was used to extract high-order structural information from the heterogeneous networks, thereby revealing the description of nodes (drugs and proteins) and their topological neighbors. Finally, potential DTI prediction was made, and the obtained samples were sent to the classifier for secondary classification. RESULTS The performance of Graph-DTI and all baseline methods was evaluated using the sums of the area under the precision-recall curve (AUPR) and the area under the receiver operating characteristic curve (AUC). The results indicated that Graph-DTI outperformed the baseline methods in both performance results. CONCLUSION Compared with other baseline DTI prediction methods, the results showed that Graph-DTI had better prediction performance. Additionally, in this study, we effectively classified drugs corresponding to different targets and vice versa. The above findings showed that Graph-DTI provided a powerful tool for drug research, development, and repositioning. Graph- DTI can serve as a drug development and repositioning tool more effectively than previous studies that did not use heterogeneous network graph embedding.
Collapse
Affiliation(s)
- Xiaohan Qu
- School of Medical Information Engineering, Guangdong Pharmaceutical University, Guangzhou, China
| | - Guoxia Du
- School of Medical Information Engineering, Guangdong Pharmaceutical University, Guangzhou, China
| | - Jing Hu
- School of Medical Information Engineering, Guangdong Pharmaceutical University, Guangzhou, China
| | - Yongming Cai
- School of Medical Information Engineering, Guangdong Pharmaceutical University, Guangzhou, China
- Guangdong Provincial Traditional Chinese Medicine Precision Medicine Big Data Engineering Technology Research Center, Guangzhou, China
| |
Collapse
|
9
|
Mizuno T, Kusuhara H. Investigation of normalization procedures for transcriptome profiles of compounds oriented toward practical study design. J Toxicol Sci 2024; 49:249-259. [PMID: 38825484 DOI: 10.2131/jts.49.249] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/04/2024]
Abstract
The transcriptome profile is a representative phenotype-based descriptor of compounds, widely acknowledged for its ability to effectively capture compound effects. However, the presence of batch differences is inevitable. Despite the existence of sophisticated statistical methods, many of them presume a substantial sample size. How should we design a transcriptome analysis to obtain robust compound profiles, particularly in the context of small datasets frequently encountered in practical scenarios? This study addresses this question by investigating the normalization procedures for transcriptome profiles, focusing on the baseline distribution employed in deriving biological responses as profiles. Firstly, we investigated two large GeneChip datasets, comparing the impact of different normalization procedures. Through an evaluation of the similarity between response profiles of biological replicates within each dataset and the similarity between response profiles of the same compound across datasets, we revealed that the baseline distribution defined by all samples within each batch under batch-corrected condition is a good choice for large datasets. Subsequently, we conducted a simulation to explore the influence of the number of control samples on the robustness of response profiles across datasets. The results offer insights into determining the suitable quantity of control samples for diminutive datasets. It is crucial to acknowledge that these conclusions stem from constrained datasets. Nevertheless, we believe that this study enhances our understanding of how to effectively leverage transcriptome profiles of compounds and promotes the accumulation of essential knowledge for the practical application of such profiles.
Collapse
Affiliation(s)
- Tadahaya Mizuno
- Laboratory of Molecular Pharmacokinetics, Graduate School of Pharmaceutical Sciences, The University of Tokyo
| | - Hiroyuki Kusuhara
- Laboratory of Molecular Pharmacokinetics, Graduate School of Pharmaceutical Sciences, The University of Tokyo
| |
Collapse
|
10
|
Lin YW, Kao HJ, Chen WT, Kao CF, Wu JY, Chen YT, Lee YC. Cell-based screen identifies porphyrins as FGFR3 activity inhibitors with therapeutic potential for achondroplasia and cancer. JCI Insight 2023; 8:e171257. [PMID: 37824212 PMCID: PMC10721322 DOI: 10.1172/jci.insight.171257] [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: 04/06/2023] [Accepted: 10/10/2023] [Indexed: 10/14/2023] Open
Abstract
Overactive fibroblast growth factor receptor 3 (FGFR3) signaling drives pathogenesis in a variety of cancers and a spectrum of short-limbed bone dysplasias, including the most common form of human dwarfism, achondroplasia (ACH). Targeting FGFR3 activity holds great promise as a therapeutic approach for treatment of these diseases. Here, we established a receptor/adaptor translocation assay system that can specifically monitor FGFR3 activation, and we applied it to identify FGFR3 modulators from complex natural mixtures. An FGFR3-suppressing plant extract of Amaranthus viridis was identified from the screen, and 2 bioactive porphyrins, pheophorbide a (Pa) and pyropheophorbide a, were sequentially isolated from the extract and functionally characterized. Further analysis showed that Pa reduced excessive FGFR3 signaling by decreasing its half-life in FGFR3-overactivated multiple myeloma cells and chondrocytes. In an ex vivo culture system, Pa alleviated defective long bone growth in humanized ACH mice (FGFR3ACH mice). Overall, our study presents an approach to discovery and validation of plant extracts or drug candidates that target FGFR3 activation. The compounds identified by this approach may have applications as therapeutics for FGFR3-associated cancers and skeletal dysplasias.
Collapse
Affiliation(s)
- Yun-Wen Lin
- Institute of Cellular and Organismic Biology, Academia Sinica, Taipei, Taiwan
- Graduate Institute of Life Sciences, National Defense Medical Center, Taipei, Taiwan
| | - Hsiao-Jung Kao
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Wei-Ting Chen
- Institute of Cellular and Organismic Biology, Academia Sinica, Taipei, Taiwan
| | - Cheng-Fu Kao
- Institute of Cellular and Organismic Biology, Academia Sinica, Taipei, Taiwan
| | - Jer-Yuarn Wu
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Yuan-Tsong Chen
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
- Department of Pediatrics, Duke University Medical Center, Durham, North Carolina, USA
| | - Yi-Ching Lee
- Institute of Cellular and Organismic Biology, Academia Sinica, Taipei, Taiwan
- Graduate Institute of Life Sciences, National Defense Medical Center, Taipei, Taiwan
| |
Collapse
|
11
|
Wang J, Novick S. DOSE-L1000: unveiling the intricate landscape of compound-induced transcriptional changes. Bioinformatics 2023; 39:btad683. [PMID: 37952162 PMCID: PMC10663987 DOI: 10.1093/bioinformatics/btad683] [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: 08/26/2023] [Revised: 10/31/2023] [Accepted: 11/10/2023] [Indexed: 11/14/2023] Open
Abstract
MOTIVATION The LINCS L1000 project has collected gene expression profiles for thousands of compounds across a wide array of concentrations, cell lines, and time points. However, conventional analysis methods often fall short in capturing the rich information encapsulated within the L1000 transcriptional dose-response data. RESULTS We present DOSE-L1000, a database that unravels the potency and efficacy of compound-gene pairs and the intricate landscape of compound-induced transcriptional changes. Our study uses the fitting of over 140 million generalized additive models and robust linear models, spanning the complete spectrum of compounds and landmark genes within the LINCS L1000 database. This systematic approach provides quantitative insights into differential gene expression and the potency and efficacy of compound-gene pairs across diverse cellular contexts. Through examples, we showcase the application of DOSE-L1000 in tasks such as cell line and compound comparisons, along with clustering analyses and predictions of drug-target interactions. DOSE-L1000 fosters applications in drug discovery, accelerating the transition to omics-driven drug development. AVAILABILITY AND IMPLEMENTATION DOSE-L1000 is publicly available at https://doi.org/10.5281/zenodo.8286375.
Collapse
Affiliation(s)
- Junmin Wang
- Data Sciences and Quantitative Biology, Discovery Sciences, Biopharmaceuticals R&D, AstraZeneca, Gaithersburg, MD 20878, United States
| | - Steven Novick
- Global Statistical Sciences, Eli Lilly, Indianapolis, IN 46285, United States
| |
Collapse
|
12
|
Mitchell DC, Kuljanin M, Li J, Van Vranken JG, Bulloch N, Schweppe DK, Huttlin EL, Gygi SP. A proteome-wide atlas of drug mechanism of action. Nat Biotechnol 2023; 41:845-857. [PMID: 36593396 PMCID: PMC11069389 DOI: 10.1038/s41587-022-01539-0] [Citation(s) in RCA: 62] [Impact Index Per Article: 31.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2022] [Accepted: 09/30/2022] [Indexed: 01/03/2023]
Abstract
Defining the cellular response to pharmacological agents is critical for understanding the mechanism of action of small molecule perturbagens. Here, we developed a 96-well-plate-based high-throughput screening infrastructure for quantitative proteomics and profiled 875 compounds in a human cancer cell line with near-comprehensive proteome coverage. Examining the 24-h proteome changes revealed ligand-induced changes in protein expression and uncovered rules by which compounds regulate their protein targets while identifying putative dihydrofolate reductase and tankyrase inhibitors. We used protein-protein and compound-compound correlation networks to uncover mechanisms of action for several compounds, including the adrenergic receptor antagonist JP1302, which we show disrupts the FACT complex and degrades histone H1. By profiling many compounds with overlapping targets covering a broad chemical space, we linked compound structure to mechanisms of action and highlighted off-target polypharmacology for molecules within the library.
Collapse
Affiliation(s)
- Dylan C Mitchell
- Department of Cell Biology, Harvard Medical School, Boston, MA, USA
| | - Miljan Kuljanin
- Department of Cell Biology, Harvard Medical School, Boston, MA, USA
| | - Jiaming Li
- Department of Cell Biology, Harvard Medical School, Boston, MA, USA
| | | | - Nathan Bulloch
- Department of Cell Biology, Harvard Medical School, Boston, MA, USA
| | - Devin K Schweppe
- Department of Genome Sciences, University of Washington, Seattle, WA, USA
| | - Edward L Huttlin
- Department of Cell Biology, Harvard Medical School, Boston, MA, USA
| | - Steven P Gygi
- Department of Cell Biology, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
13
|
Bracchetti L, Cocci P, Palermo FA. Multiple Aspects of the Fight against the Red Palm Weevil in an Urban Area: Study Case, San Benedetto del Tronto (Central Italy). INSECTS 2023; 14:502. [PMID: 37367318 DOI: 10.3390/insects14060502] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Revised: 05/26/2023] [Accepted: 05/26/2023] [Indexed: 06/28/2023]
Abstract
The fight against alien invasive insect pests of plants in the urban environment often affects varied sectors of the economy, landscape gardening, public health, and ecology. This paper focuses on the evolution of the red palm weevil in San Benedetto del Tronto, a coastal urban area in central Italy. We investigated the evolution of this insect pest of palm trees in the 2013-2020 period, considering both the effectiveness of the chemicals used and their potentially harmful effects. With a multidisciplinary approach, we carried out a spatio-temporal analysis of the extent and mode of pest spread over time using historical aerial photos, freely available remote sensing images, and field surveys integrated in a GIS environment. We also assessed the toxicity risk associated with the chemicals used to protect the palms from the red weevil. The fight against this weevil is now concentrated in specific areas such as parks, roads, villas, hotels, farmhouses, and nurseries. The preventive chemical treatments applied are very effective in preserving the palms, but they show a toxic potential for all organisms. We discuss current local management of this pest, focusing on several aspects involved in the fight against this beetle in an urban area.
Collapse
Affiliation(s)
- Luca Bracchetti
- Unità di Ricerca e Didattica San Benedetto del Tronto (URDiS), University of Camerino, Via A. Scipioni, 6, 63034 San Benedetto del Tronto, Italy
| | - Paolo Cocci
- School of Biosciences and Veterinary Medicine, University of Camerino, Via Gentile III da Varano, 62032 Camerino, Italy
| | - Francesco Alessandro Palermo
- School of Biosciences and Veterinary Medicine, University of Camerino, Via Gentile III da Varano, 62032 Camerino, Italy
| |
Collapse
|
14
|
Graves OK, Kim W, Özcan M, Ashraf S, Turkez H, Yuan M, Zhang C, Mardinoglu A, Li X. Discovery of drug targets and therapeutic agents based on drug repositioning to treat lung adenocarcinoma. Biomed Pharmacother 2023; 161:114486. [PMID: 36906970 DOI: 10.1016/j.biopha.2023.114486] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Revised: 02/20/2023] [Accepted: 03/07/2023] [Indexed: 03/12/2023] Open
Abstract
BACKGROUND Lung adenocarcinoma (LUAD) is the one of the most common subtypes in lung cancer. Although various targeted therapies have been used in the clinical practice, the 5-year overall survival rate of patients is still low. Thus, it is urgent to identify new therapeutic targets and develop new drugs for the treatment of the LUAD patients. METHODS Survival analysis was used to identify the prognostic genes. Gene co-expression network analysis was used to identify the hub genes driving the tumor development. A profile-based drug repositioning approach was used to repurpose the potentially useful drugs for targeting the hub genes. MTT and LDH assay were used to measure the cell viability and drug cytotoxicity, respectively. Western blot was used to detect the expression of the proteins. FINDINGS We identified 341 consistent prognostic genes from two independent LUAD cohorts, whose high expression was associated with poor survival outcomes of patients. Among them, eight genes were identified as hub genes due to their high centrality in the key functional modules in the gene-co-expression network analysis and these genes were associated with the various hallmarks of cancer (e.g., DNA replication and cell cycle). We performed drug repositioning analysis for three of the eight genes (CDCA8, MCM6, and TTK) based on our drug repositioning approach. Finally, we repurposed five drugs for inhibiting the protein expression level of each target gene and validated the drug efficacy by performing in vitro experiments. INTERPRETATION We found the consensus targetable genes for the treatment of LUAD patients with different races and geographic characteristics. We also proved the feasibility of our drug repositioning approach for the development of new drugs for disease treatment.
Collapse
Affiliation(s)
| | - Woonghee Kim
- Science for Life Laboratory, KTH-Royal Institute of Technology, Stockholm SE-17165, Sweden.
| | - Mehmet Özcan
- Science for Life Laboratory, KTH-Royal Institute of Technology, Stockholm SE-17165, Sweden.
| | - Sajda Ashraf
- Trustlife Labs, Drug Research & Development Center, 34774 Istanbul, Turkey.
| | - Hasan Turkez
- Trustlife Labs, Drug Research & Development Center, 34774 Istanbul, Turkey.
| | - Meng Yuan
- Science for Life Laboratory, KTH-Royal Institute of Technology, Stockholm SE-17165, Sweden.
| | - Cheng Zhang
- Science for Life Laboratory, KTH-Royal Institute of Technology, Stockholm SE-17165, Sweden.
| | - Adil Mardinoglu
- Science for Life Laboratory, KTH-Royal Institute of Technology, Stockholm SE-17165, Sweden; Centre for Host-Microbiome Interactions, Faculty of Dentistry, Oral & Craniofacial Sciences, King's College London, London SE1 9RT, UK.
| | - Xiangyu Li
- Science for Life Laboratory, KTH-Royal Institute of Technology, Stockholm SE-17165, Sweden; Bash Biotech Inc, 600 West Broadway, Suite 700, San Diego, CA 92101, USA; Guangzhou Laboratory, Guangzhou 510005, China.
| |
Collapse
|
15
|
Shah I, Bundy J, Chambers B, Everett LJ, Haggard D, Harrill J, Judson RS, Nyffeler J, Patlewicz G. Navigating Transcriptomic Connectivity Mapping Workflows to Link Chemicals with Bioactivities. Chem Res Toxicol 2022; 35:1929-1949. [PMID: 36301716 PMCID: PMC10483698 DOI: 10.1021/acs.chemrestox.2c00245] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Screening new compounds for potential bioactivities against cellular targets is vital for drug discovery and chemical safety. Transcriptomics offers an efficient approach for assessing global gene expression changes, but interpreting chemical mechanisms from these data is often challenging. Connectivity mapping is a potential data-driven avenue for linking chemicals to mechanisms based on the observation that many biological processes are associated with unique gene expression signatures (gene signatures). However, mining the effects of a chemical on gene signatures for biological mechanisms is challenging because transcriptomic data contain thousands of noisy genes. New connectivity mapping approaches seeking to distinguish signal from noise continue to be developed, spurred by the promise of discovering chemical mechanisms, new drugs, and disease targets from burgeoning transcriptomic data. Here, we analyze these approaches in terms of diverse transcriptomic technologies, public databases, gene signatures, pattern-matching algorithms, and statistical evaluation criteria. To navigate the complexity of connectivity mapping, we propose a harmonized scheme to coherently organize and compare published workflows. We first standardize concepts underlying transcriptomic profiles and gene signatures based on various transcriptomic technologies such as microarrays, RNA-Seq, and L1000 and discuss the widely used data sources such as Gene Expression Omnibus, ArrayExpress, and MSigDB. Next, we generalize connectivity mapping as a pattern-matching task for finding similarity between a query (e.g., transcriptomic profile for new chemical) and a reference (e.g., gene signature of known target). Published pattern-matching approaches fall into two main categories: vector-based use metrics like correlation, Jaccard index, etc., and aggregation-based use parametric and nonparametric statistics (e.g., gene set enrichment analysis). The statistical methods for evaluating the performance of different approaches are described, along with comparisons reported in the literature on benchmark transcriptomic data sets. Lastly, we review connectivity mapping applications in toxicology and offer guidance on evaluating chemical-induced toxicity with concentration-response transcriptomic data. In addition to serving as a high-level guide and tutorial for understanding and implementing connectivity mapping workflows, we hope this review will stimulate new algorithms for evaluating chemical safety and drug discovery using transcriptomic data.
Collapse
Affiliation(s)
- Imran Shah
- Center for Computational Toxicology and Exposure, Office of Research and Development, US. Environmental Protection Agency, Research Triangle Park, North Carolina 27711, USA
| | - Joseph Bundy
- Center for Computational Toxicology and Exposure, Office of Research and Development, US. Environmental Protection Agency, Research Triangle Park, North Carolina 27711, USA
| | - Bryant Chambers
- Center for Computational Toxicology and Exposure, Office of Research and Development, US. Environmental Protection Agency, Research Triangle Park, North Carolina 27711, USA
| | - Logan J. Everett
- Center for Computational Toxicology and Exposure, Office of Research and Development, US. Environmental Protection Agency, Research Triangle Park, North Carolina 27711, USA
| | - Derik Haggard
- Center for Computational Toxicology and Exposure, Office of Research and Development, US. Environmental Protection Agency, Research Triangle Park, North Carolina 27711, USA
| | - Joshua Harrill
- Center for Computational Toxicology and Exposure, Office of Research and Development, US. Environmental Protection Agency, Research Triangle Park, North Carolina 27711, USA
| | - Richard S. Judson
- Center for Computational Toxicology and Exposure, Office of Research and Development, US. Environmental Protection Agency, Research Triangle Park, North Carolina 27711, USA
| | - Johanna Nyffeler
- Center for Computational Toxicology and Exposure, Office of Research and Development, US. Environmental Protection Agency, Research Triangle Park, North Carolina 27711, USA
- Oak Ridge Institute for Science and Education (ORISE) Postdoctoral Fellow, Oak Ridge, Tennessee, 37831, US
| | - Grace Patlewicz
- Center for Computational Toxicology and Exposure, Office of Research and Development, US. Environmental Protection Agency, Research Triangle Park, North Carolina 27711, USA
| |
Collapse
|
16
|
Iwata M, Kosai K, Ono Y, Oki S, Mimori K, Yamanishi Y. Regulome-based characterization of drug activity across the human diseasome. NPJ Syst Biol Appl 2022; 8:44. [DOI: 10.1038/s41540-022-00255-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Accepted: 10/21/2022] [Indexed: 11/09/2022] Open
Abstract
AbstractDrugs are expected to recover the cell system away from the impaired state to normalcy through disease treatment. However, the understanding of gene regulatory machinery underlying drug activity or disease pathogenesis is far from complete. Here, we perform large-scale regulome analysis for various diseases in terms of gene regulatory machinery. Transcriptome signatures were converted into regulome signatures of transcription factors by integrating publicly available ChIP-seq data. Regulome-based correlations between diseases and their approved drugs were much clearer than the transcriptome-based correlations. For example, an inverse correlation was observed for cancers, whereas a positive correlation was observed for immune system diseases. After demonstrating the usefulness of the regulome-based drug discovery method in terms of accuracy and applicability, we predicted new drugs for nonsmall cell lung cancer and validated the anticancer activity in vitro. The proposed method is useful for understanding disease–disease relationships and drug discovery.
Collapse
|
17
|
Fernández-Torras A, Duran-Frigola M, Bertoni M, Locatelli M, Aloy P. Integrating and formatting biomedical data as pre-calculated knowledge graph embeddings in the Bioteque. Nat Commun 2022; 13:5304. [PMID: 36085310 PMCID: PMC9463154 DOI: 10.1038/s41467-022-33026-0] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Accepted: 08/30/2022] [Indexed: 12/25/2022] Open
Abstract
Biomedical data is accumulating at a fast pace and integrating it into a unified framework is a major challenge, so that multiple views of a given biological event can be considered simultaneously. Here we present the Bioteque, a resource of unprecedented size and scope that contains pre-calculated biomedical descriptors derived from a gigantic knowledge graph, displaying more than 450 thousand biological entities and 30 million relationships between them. The Bioteque integrates, harmonizes, and formats data collected from over 150 data sources, including 12 biological entities (e.g., genes, diseases, drugs) linked by 67 types of associations (e.g., 'drug treats disease', 'gene interacts with gene'). We show how Bioteque descriptors facilitate the assessment of high-throughput protein-protein interactome data, the prediction of drug response and new repurposing opportunities, and demonstrate that they can be used off-the-shelf in downstream machine learning tasks without loss of performance with respect to using original data. The Bioteque thus offers a thoroughly processed, tractable, and highly optimized assembly of the biomedical knowledge available in the public domain.
Collapse
Affiliation(s)
- Adrià Fernández-Torras
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, Barcelona, Catalonia, Spain
| | - Miquel Duran-Frigola
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, Barcelona, Catalonia, Spain
- Ersilia Open Source Initiative, Cambridge, UK
| | - Martino Bertoni
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, Barcelona, Catalonia, Spain
| | - Martina Locatelli
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, Barcelona, Catalonia, Spain
| | - Patrick Aloy
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, Barcelona, Catalonia, Spain.
- Institució Catalana de Recerca i Estudis Avançats (ICREA), Barcelona, Catalonia, Spain.
| |
Collapse
|
18
|
Kamath A, Srinivasamurthy SK, Chowta MN, Ullal SD, Daali Y, Chakradhara Rao US. Role of Drug Transporters in Elucidating Inter-Individual Variability in Pediatric Chemotherapy-Related Toxicities and Response. Pharmaceuticals (Basel) 2022; 15:990. [PMID: 36015138 PMCID: PMC9415926 DOI: 10.3390/ph15080990] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Revised: 08/01/2022] [Accepted: 08/04/2022] [Indexed: 11/16/2022] Open
Abstract
Pediatric cancer treatment has evolved significantly in recent decades. The implementation of risk stratification strategies and the selection of evidence-based chemotherapy combinations have improved survival outcomes. However, there is large interindividual variability in terms of chemotherapy-related toxicities and, sometimes, the response among this population. This variability is partly attributed to the functional variability of drug-metabolizing enzymes (DME) and drug transporters (DTS) involved in the process of absorption, distribution, metabolism and excretion (ADME). The DTS, being ubiquitous, affects drug disposition across membranes and has relevance in determining chemotherapy response in pediatric cancer patients. Among the factors affecting DTS function, ontogeny or maturation is important in the pediatric population. In this narrative review, we describe the role of drug uptake/efflux transporters in defining pediatric chemotherapy-treatment-related toxicities and responses. Developmental differences in DTS and the consequent implications are also briefly discussed for the most commonly used chemotherapeutic drugs in the pediatric population.
Collapse
Affiliation(s)
- Ashwin Kamath
- Department of Pharmacology, Kasturba Medical College, Mangalore, Manipal Academy of Higher Education, Manipal 575001, India
| | - Suresh Kumar Srinivasamurthy
- Department of Pharmacology, Ras Al Khaimah College of Medical Sciences, Ras Al Khaimah Medical and Health Sciences University, Ras Al Khaimah P.O. Box 11172, United Arab Emirates
| | - Mukta N. Chowta
- Department of Pharmacology, Kasturba Medical College, Mangalore, Manipal Academy of Higher Education, Manipal 575001, India
| | - Sheetal D. Ullal
- Department of Pharmacology, Kasturba Medical College, Mangalore, Manipal Academy of Higher Education, Manipal 575001, India
| | - Youssef Daali
- Department of Anaesthesiology, Pharmacology, Intensive Care and Emergency Medicine, Division of Clinical Pharmacology and Toxicology, Geneva University Hospitals, 1205 Geneva, Switzerland
| | - Uppugunduri S. Chakradhara Rao
- Department of Pharmacology, Kasturba Medical College, Mangalore, Manipal Academy of Higher Education, Manipal 575001, India
- CANSEARCH Research Platform in Pediatric Oncology and Hematology, Department of Pediatrics, Gynecology and Obstetrics, University of Geneva, 1205 Geneva, Switzerland
| |
Collapse
|
19
|
Zhang M, Liu Y, Liu Y, Hou S, Li H, Ma Y, Wang C, Chen X. A Potential Indicator ARRDC2 Has Feasibility to Evaluate Prognosis and Immune Microenvironment in Ovarian Cancer. Front Genet 2022; 13:815082. [PMID: 35664304 PMCID: PMC9157644 DOI: 10.3389/fgene.2022.815082] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Accepted: 05/02/2022] [Indexed: 12/04/2022] Open
Abstract
Background: The abnormal expression of α-arrestin protein family plays a key regulatory role in the occurrence and development of many cancers, including colorectal cancer and cervical cancer, and is inseparable from changes in the tumor immune microenvironment. However, the role of ARRDC2, an important member of this family, in the malignant biological process of ovarian cancer (OC) has not been reported, and its role in the change of the immune microenvironment is also unknown. Methods: In this study, HPA, TCGA, GEO and other databases were used to explore the role of ARRDC2 in the prognosis assessment of ovarian cancer. Then, GO, KEGG analysis and GSEA analysis of the biological processes and cell signaling pathways that ARRDC2 may be involved in activated or inhibited. In addition, the TIMER and TISIDB database were used to conduct in-depth research on the role of ARRDC2 in the change of the immune microenvironment of ovarian cancer. The CMap database explored and screened drugs that may be used for treatment. Through cell transfection, CCK-8, Ki-67 immunofluorescence, wound healing, transwell and clone formation assay, the effect of ARRDC2 knockdown on the malignant biological behavior of OC cells were explored. Results: There were significant differences between OC and ARRDC2 mRNA and protein levels. High ARRDC2 expression level is associated with poor overall survival and can be used as an independent prognostic factor. Interestingly, ARRDC2 expression is positively correlated with B cells, Neutrophils, Dendritic cells and CD8+ T cells, signifying that ARRDC2 may be related to infiltration of immune cells. ARRDC2 and its co-expressed genes are enriched in cell signaling pathways related to the immune system. We explored two possible drugs for the treatment of ovarian cancer. Finally, the results of in vitro experiments indicated that knockdown of ARRDC2 may inhibit malignant phenotypes such as proliferation and migration of OC cells. Conclusion: The differentially expressed ARRDC2 may be a potential prognostic indicator and can be used as a novel biomarker for exploring the immune microenvironment of ovarian cancer.
Collapse
Affiliation(s)
- Mengjun Zhang
- Department of Gynecology, Harbin Medical University Cancer Hospital, Harbin, China
| | - Yunduo Liu
- Department of Gynecology, Harbin Medical University Cancer Hospital, Harbin, China
| | - Yuan Liu
- Department of Gynecology, Harbin Medical University Cancer Hospital, Harbin, China
| | - Siyu Hou
- Department of Gynecology, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
| | - Hao Li
- Department of Gynecology, Harbin Medical University Cancer Hospital, Harbin, China
| | - Ying Ma
- Department of Gynecology, Harbin Medical University Cancer Hospital, Harbin, China
| | - Can Wang
- Department of Gynecology, Harbin Medical University Cancer Hospital, Harbin, China
| | - Xiuwei Chen
- Department of Gynecology, Harbin Medical University Cancer Hospital, Harbin, China
| |
Collapse
|
20
|
Gao QH, Wen B, Kang Y, Zhang WM. Pump-free microfluidic magnetic levitation approach for density-based cell characterization. Biosens Bioelectron 2022; 204:114052. [PMID: 35149454 DOI: 10.1016/j.bios.2022.114052] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Revised: 01/22/2022] [Accepted: 01/27/2022] [Indexed: 11/16/2022]
Abstract
Magnetic levitation (MagLev) provides a simple but promising method for density-based analysis and detection down to the individual cell level. However, each existing MagLev configuration for the single-cell density measurement, mainly consisting of a capillary (∼50 mm) placed between two magnets, yields a fairly low sample utilization because of no knowledge about the sample cells in the regions other than the limited microscope vision. Moreover, the quantitative analysis may be affected due to the unclearly defined measurement area, which is specifically associated with the uneven magnetization of magnets, cell size, degree of aggregation. In this work, we explore a pump-free microfluidic magnetic levitation approach for density-based cell characterization, enabling sensitive and effective cellular density measurement on small sample volumes. The microfluidic MagLev comprises a pump-free microfluidic chip placed between two ring magnets with like poles facing. With no external pumps, connectors or control facility, much smaller amounts of fluids (∼4 μL) could be driven automatically in the entire microchannel in 16 s. Based on the pump-free mechanism, unique density signatures of cells from different lineages (ARPE-19, HCT116, HeLa, HT1080, Huh7) are characterized by monitoring the levitation profiles. Furthermore, variation in density of A549 lung cancer cells subjected to a drug treatment are observed in our platform, allowing evaluation of the efficacy of the drug treatment at the individual cell level. Thereby, the proposed pump-free microfluidic MagLev platform, a low-cost, fully automatic and portable design for label-free density-based cell characterization, provides a universal detection tool that operates efficiently within small-volume environments.
Collapse
Affiliation(s)
- Qiu-Hua Gao
- State Key Laboratory of Mechanical System and Vibration, School of Mechanical Engineering, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Baiqing Wen
- School of Biomedical Engineering, Bio-ID Center, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Yani Kang
- School of Biomedical Engineering, Bio-ID Center, Shanghai Jiao Tong University, Shanghai, 200240, China.
| | - Wen-Ming Zhang
- State Key Laboratory of Mechanical System and Vibration, School of Mechanical Engineering, Shanghai Jiao Tong University, Shanghai, 200240, China.
| |
Collapse
|
21
|
Liu Z, Ren Z, Zhang C, Qian R, Wang H, Wang J, Zhang W, Liu B, Lian X, Wang Y, Guo Y, Gao Y. ELK3: A New Molecular Marker for the Diagnosis and Prognosis of Glioma. Front Oncol 2022; 11:608748. [PMID: 34976781 PMCID: PMC8716454 DOI: 10.3389/fonc.2021.608748] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Accepted: 11/25/2021] [Indexed: 12/21/2022] Open
Abstract
ETS transcription factor ELK3 (ELK3), a novel oncogene, affects pathological processes and progression of many cancers in human tissues. However, it remains unclear whether ELK3, as a key gene, affects the pathological process of gliomas and the prognosis of patients with gliomas. This study aimed to comprehensively and systematically reveal the correlation between ELK3 and the malignant progression of gliomas by analyzing clinical sample information stored in multiple databases. We revealed the putative mechanism of ELK3 involvement in malignant gliomas progression and identified a new and efficient biomarker for glioma diagnosis and targeted therapy. Based on the sample data from multiple databases and real-time quantitative polymerase chain reaction (RT-qPCR), the abnormally high expression of ELK3 in gliomas was confirmed. Kaplan-Meier and Cox regression analyses demonstrated that a high ELK3 expression was markedly associated with low patient survival and served as an independent biomarker of gliomas. Wilcox and Kruskal-Wallis tests revealed that expression of ELK3 was positively correlated with several clinical characteristics of patients with gliomas, such as age, WHO classification, and recurrence. Moreover, Cell Counting Kit‐8 (CCK-8), immunofluorescence, and wound healing assays confirmed that ELK3 overexpression markedly promoted the proliferation and migration of glioma cells. Finally, gene set enrichment analysis (GSEA) and western blotting confirmed that overexpression of ELK3 regulated the JAK–STAT signaling pathway and upregulate the expression of signal transducer and activator of transcription 3 (STAT3) and phosphorylated STAT3 (P-STAT3) to promote the malignant transition of gliomas. Therefore, ELK3 may serve as an efficient biomarker for the diagnosis and prognosis of gliomas and it can also be used as a therapeutic target to improve the poor prognosis of patients with gliomas.
Collapse
Affiliation(s)
- Zhendong Liu
- Department of Surgery of Spine and Spinal Cord, Henan Provincial People's Hospital; People's Hospital of Zhengzhou University, People's Hospital of Henan University, Zhengzhou, China
| | - Zhishuai Ren
- People's Hospital of Zhengzhou University, Henan Provincial People's Hospital, Zhengzhou, Henan, China
| | - Cheng Zhang
- North Broward Preparatory School, Nord Anglia Education, Coconut Creek, FL, United States
| | - Rongjun Qian
- Department of Neurosurgery, Henan Provincial People's Hospital, Zhengzhou, China
| | - Hongbo Wang
- People's Hospital of Henan University, Henan Provincial People's Hospital, Zhengzhou, China
| | - Jialin Wang
- People's Hospital of Zhengzhou University, Henan Provincial People's Hospital, Zhengzhou, Henan, China
| | - Wang Zhang
- Department of Neurosurgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Binfeng Liu
- People's Hospital of Zhengzhou University, Henan Provincial People's Hospital, Zhengzhou, Henan, China
| | - Xiaoyu Lian
- People's Hospital of Zhengzhou University, Henan Provincial People's Hospital, Zhengzhou, Henan, China
| | - Yanbiao Wang
- People's Hospital of Zhengzhou University, Henan Provincial People's Hospital, Zhengzhou, Henan, China
| | - Yuqi Guo
- Department of Obstetrics and Gynecology, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, People's Hospital of Henan University, Zhengzhou, China.,Henan International Joint Laboratory for Gynecological Oncology and Nanomedicine, Zhengzhou, China
| | - Yanzheng Gao
- Department of Surgery of Spine and Spinal Cord, Henan Provincial People's Hospital; People's Hospital of Zhengzhou University, People's Hospital of Henan University, Zhengzhou, China
| |
Collapse
|
22
|
Mirza N, Stevelink R, Taweel B, Koeleman BPC, Marson AG. Using common genetic variants to find drugs for common epilepsies. Brain Commun 2021; 3:fcab287. [PMID: 34988442 PMCID: PMC8710935 DOI: 10.1093/braincomms/fcab287] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Revised: 09/17/2021] [Accepted: 10/20/2021] [Indexed: 12/18/2022] Open
Abstract
Better drugs are needed for common epilepsies. Drug repurposing offers the potential of significant savings in the time and cost of developing new treatments. In order to select the best candidate drug(s) to repurpose for a disease, it is desirable to predict the relative clinical efficacy that drugs will have against the disease. Common epilepsy can be divided into different types and syndromes. Different antiseizure medications are most effective for different types and syndromes of common epilepsy. For predictions of antiepileptic efficacy to be clinically translatable, it is essential that the predictions are specific to each form of common epilepsy, and reflect the patterns of drug efficacy observed in clinical studies and practice. These requirements are not fulfilled by previously published drug predictions for epilepsy. We developed a novel method for predicting the relative efficacy of drugs against any common epilepsy, by using its Genome-Wide Association Study summary statistics and drugs' activity data. The methodological advancement in our technique is that the drug predictions for a disease are based upon drugs' effects on the function and abundance of proteins, and the magnitude and direction of those effects, relative to the importance, degree and direction of the proteins' dysregulation in the disease. We used this method to predict the relative efficacy of all drugs, licensed for any condition, against each of the major types and syndromes of common epilepsy. Our predictions are concordant with findings from real-world experience and randomized clinical trials. Our method predicts the efficacy of existing antiseizure medications against common epilepsies; in this prediction, our method outperforms the best alternative existing method: area under receiver operating characteristic curve (mean ± standard deviation) 0.83 ± 0.03 and 0.63 ± 0.04, respectively. Importantly, our method predicts which antiseizure medications are amongst the more efficacious in clinical practice, and which antiseizure medications are amongst the less efficacious in clinical practice, for each of the main syndromes of common epilepsy, and it predicts the distinct order of efficacy of individual antiseizure medications in clinical trials of different common epilepsies. We identify promising candidate drugs for each of the major syndromes of common epilepsy. We screen five promising predicted drugs in an animal model: each exerts a significant dose-dependent effect upon seizures. Our predictions are a novel resource for selecting suitable candidate drugs that could potentially be repurposed for each of the major syndromes of common epilepsy. Our method is potentially generalizable to other complex diseases.
Collapse
Affiliation(s)
- Nasir Mirza
- Department of Pharmacology & Therapeutics, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool L69 3GE, UK
| | - Remi Stevelink
- Department of Genetics, Center for Molecular Medicine, University Medical Center Utrecht, Utrecht 3584 CX, the Netherlands; member of the ERN EpiCARE
- Department of Child Neurology, University Medical Center Utrecht Brain Center, Utrecht 3584 CX, the Netherlands
| | - Basel Taweel
- School of Medicine, University of Liverpool, Liverpool L69 3GE, UK
| | - Bobby P C Koeleman
- Department of Genetics, Center for Molecular Medicine, University Medical Center Utrecht, Utrecht 3584 CX, the Netherlands; member of the ERN EpiCARE
| | - Anthony G Marson
- Department of Pharmacology & Therapeutics, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool L69 3GE, UK
| |
Collapse
|
23
|
Dafniet B, Cerisier N, Boezio B, Clary A, Ducrot P, Dorval T, Gohier A, Brown D, Audouze K, Taboureau O. Development of a chemogenomics library for phenotypic screening. J Cheminform 2021; 13:91. [PMID: 34819133 PMCID: PMC8611952 DOI: 10.1186/s13321-021-00569-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2021] [Accepted: 11/06/2021] [Indexed: 12/03/2022] Open
Abstract
With the development of advanced technologies in cell-based phenotypic screening, phenotypic drug discovery (PDD) strategies have re-emerged as promising approaches in the identification and development of novel and safe drugs. However, phenotypic screening does not rely on knowledge of specific drug targets and needs to be combined with chemical biology approaches to identify therapeutic targets and mechanisms of actions induced by drugs and associated with an observable phenotype. In this study, we developed a system pharmacology network integrating drug-target-pathway-disease relationships as well as morphological profile from an existing high content imaging-based high-throughput phenotypic profiling assay known as “Cell Painting”. Furthermore, from this network, a chemogenomic library of 5000 small molecules that represent a large and diverse panel of drug targets involved in diverse biological effects and diseases has been developed. Such a platform and a chemogenomic library could assist in the target identification and mechanism deconvolution of some phenotypic assays. The usefulness of the platform is illustrated through examples.
Collapse
Affiliation(s)
- Bryan Dafniet
- Université de Paris, INSERM U1133, CNRS UMR8251, 75006, Paris, France
| | - Natacha Cerisier
- Université de Paris, INSERM U1133, CNRS UMR8251, 75006, Paris, France
| | - Batiste Boezio
- Université de Paris, INSERM U1133, CNRS UMR8251, 75006, Paris, France
| | - Anaelle Clary
- Institut de Recherche Servier, 125 Chemin de Ronde, 78290, Croissy-sur-Seine, France
| | - Pierre Ducrot
- Institut de Recherche Servier, 125 Chemin de Ronde, 78290, Croissy-sur-Seine, France
| | - Thierry Dorval
- Institut de Recherche Servier, 125 Chemin de Ronde, 78290, Croissy-sur-Seine, France
| | - Arnaud Gohier
- Institut de Recherche Servier, 125 Chemin de Ronde, 78290, Croissy-sur-Seine, France
| | - David Brown
- Institut de Recherche Servier, 125 Chemin de Ronde, 78290, Croissy-sur-Seine, France
| | - Karine Audouze
- Université de Paris, INSERM UMR S-1124, 75006, Paris, France
| | - Olivier Taboureau
- Université de Paris, INSERM U1133, CNRS UMR8251, 75006, Paris, France.
| |
Collapse
|
24
|
Hunter KS, Miller A, Mentink-Kane M, Davies SJ. Schistosome AMPK Is Required for Larval Viability and Regulates Glycogen Metabolism in Adult Parasites. Front Microbiol 2021; 12:726465. [PMID: 34539616 PMCID: PMC8440919 DOI: 10.3389/fmicb.2021.726465] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2021] [Accepted: 08/12/2021] [Indexed: 11/25/2022] Open
Abstract
On entering the mammalian host, schistosomes transition from a freshwater environment where resources are scarce, to an environment where there is an unlimited supply of glucose, their preferred energy substrate. Adult schistosome glycolytic activity consumes almost five times the parasite's dry weight in glucose per day to meet the parasite's energy demands, and the schistosome glycolytic enzymes and mechanisms for glucose uptake that sustain this metabolic activity have previously been identified. However, little is known of the parasite processes that regulate schistosome glucose metabolism. We previously described the Schistosoma mansoni ortholog of 5' AMP-Activated Protein Kinase (AMPK), which is a central regulator of energy metabolism in eukaryotes, and characterized the developmental regulation of its expression and activity in S. mansoni. Here we sought to explore the function of AMPK in schistosomes and test whether it regulates parasite glycolysis. Adult schistosomes mounted a compensatory response to chemical inhibition of AMPK α, resulting in increased AMPK α protein abundance and activity. RNAi inhibition of AMPK α expression, however, suggests that AMPK α is not required for adult schistosome viability in vitro. Larval schistosomula, on the other hand, are sensitive to chemical AMPK α inhibition, and this correlates with inactivity of the AMPK α gene in this life cycle stage that precludes a compensatory response to AMPK inhibition. While our data indicate that AMPK is not essential in adult schistosomes, our results suggest that AMPK regulates adult worm glycogen stores, influencing both glycogen utilization and synthesis. AMPK may therefore play a role in the ability of adult schistosomes to survive in vivo stressors such as transient glucose deprivation and oxidative stress. These findings suggest that AMPK warrants further investigation as a potential drug target, especially for interventions aimed at preventing establishment of a schistosome infection.
Collapse
Affiliation(s)
- Kasandra S Hunter
- Department of Microbiology and Immunology, F. Edward Hébert School of Medicine, Uniformed Services University, Bethesda, MD, United States
| | - André Miller
- Schistosomiasis Resource Center, Biomedical Research Institute, Rockville, MD, United States
| | - Margaret Mentink-Kane
- Schistosomiasis Resource Center, Biomedical Research Institute, Rockville, MD, United States
| | - Stephen J Davies
- Department of Microbiology and Immunology, F. Edward Hébert School of Medicine, Uniformed Services University, Bethesda, MD, United States
| |
Collapse
|
25
|
Frost J, Rocha S, Ciulli A. Von Hippel-Lindau (VHL) small-molecule inhibitor binding increases stability and intracellular levels of VHL protein. J Biol Chem 2021; 297:100910. [PMID: 34174286 PMCID: PMC8313594 DOI: 10.1016/j.jbc.2021.100910] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Revised: 06/15/2021] [Accepted: 06/22/2021] [Indexed: 11/24/2022] Open
Abstract
Von Hippel-Lindau (VHL) disease is characterized by frequent mutation of VHL protein, a tumor suppressor that functions as the substrate recognition subunit of a Cullin2 RING E3 ligase complex (CRL2VHL). CRL2VHL plays important roles in oxygen sensing by targeting hypoxia-inducible factor-alpha (HIF-α) subunits for ubiquitination and degradation. VHL is also commonly hijacked by bifunctional molecules such as proteolysis-targeting chimeras to induce degradation of target molecules. We previously reported the design and characterization of VHL inhibitors VH032 and VH298 that block the VHL:HIF-α interaction, activate the HIF transcription factor, and induce a hypoxic response, which can be beneficial to treat anemia and mitochondrial diseases. How these compounds affect the global cellular proteome remains unknown. Here, we use unbiased quantitative MS to identify the proteomic changes elicited by the VHL inhibitor compared with hypoxia or the broad-spectrum prolyl-hydroxylase domain enzyme inhibitor IOX2. Our results demonstrate that VHL inhibitors selectively activate the HIF response similar to the changes induced in hypoxia and IOX2 treatment. Interestingly, VHL inhibitors were found to specifically upregulate VHL itself. Our analysis revealed that this occurs via protein stabilization of VHL isoforms and not via changes in transcript levels. Increased VHL levels upon VH298 treatment resulted in turn in reduced levels of HIF-1α protein. This work demonstrates the specificity of VHL inhibitors and reveals different antagonistic effects upon their acute versus prolonged treatment in cells. These findings suggest that therapeutic use of VHL inhibitors may not produce overt side effects from HIF stabilization as previously thought.
Collapse
Affiliation(s)
- Julianty Frost
- Division of Biological Chemistry and Drug Discovery, School of Life Sciences, University of Dundee, Dundee, Scotland, United Kingdom; Center for Gene Regulation and Expression, School of Life Sciences, University of Dundee, Dundee, Scotland, United Kingdom; Department of Molecular Physiology and Cell Signalling, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool, United Kingdom
| | - Sonia Rocha
- Center for Gene Regulation and Expression, School of Life Sciences, University of Dundee, Dundee, Scotland, United Kingdom; Department of Molecular Physiology and Cell Signalling, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool, United Kingdom.
| | - Alessio Ciulli
- Division of Biological Chemistry and Drug Discovery, School of Life Sciences, University of Dundee, Dundee, Scotland, United Kingdom.
| |
Collapse
|
26
|
He B, Hou F, Ren C, Bing P, Xiao X. A Review of Current In Silico Methods for Repositioning Drugs and Chemical Compounds. Front Oncol 2021; 11:711225. [PMID: 34367996 PMCID: PMC8340770 DOI: 10.3389/fonc.2021.711225] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Accepted: 07/07/2021] [Indexed: 12/23/2022] Open
Abstract
Drug repositioning is a new way of applying the existing therapeutics to new disease indications. Due to the exorbitant cost and high failure rate in developing new drugs, the continued use of existing drugs for treatment, especially anti-tumor drugs, has become a widespread practice. With the assistance of high-throughput sequencing techniques, many efficient methods have been proposed and applied in drug repositioning and individualized tumor treatment. Current computational methods for repositioning drugs and chemical compounds can be divided into four categories: (i) feature-based methods, (ii) matrix decomposition-based methods, (iii) network-based methods, and (iv) reverse transcriptome-based methods. In this article, we comprehensively review the widely used methods in the above four categories. Finally, we summarize the advantages and disadvantages of these methods and indicate future directions for more sensitive computational drug repositioning methods and individualized tumor treatment, which are critical for further experimental validation.
Collapse
Affiliation(s)
- Binsheng He
- Academician Workstation, Changsha Medical University, Changsha, China
| | - Fangxing Hou
- Queen Mary School, Nanchang University, Jiangxi, China
| | - Changjing Ren
- School of Science, Dalian Maritime University, Dalian, China.,Genies Beijing Co., Ltd., Beijing, China
| | - Pingping Bing
- Academician Workstation, Changsha Medical University, Changsha, China
| | - Xiangzuo Xiao
- Department of Radiology, The First Affiliated Hospital of Nanchang University, Jiangxi, China
| |
Collapse
|
27
|
Samart K, Tuyishime P, Krishnan A, Ravi J. Reconciling multiple connectivity scores for drug repurposing. Brief Bioinform 2021; 22:6278144. [PMID: 34013329 PMCID: PMC8597919 DOI: 10.1093/bib/bbab161] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2020] [Revised: 04/02/2021] [Accepted: 04/07/2021] [Indexed: 12/16/2022] Open
Abstract
The basis of several recent methods for drug repurposing is the key principle that an
efficacious drug will reverse the disease molecular ‘signature’ with minimal side effects.
This principle was defined and popularized by the influential ‘connectivity map’ study in
2006 regarding reversal relationships between disease- and drug-induced gene expression
profiles, quantified by a disease-drug ‘connectivity score.’ Over the past 15 years,
several studies have proposed variations in calculating connectivity scores toward
improving accuracy and robustness in light of massive growth in reference drug profiles.
However, these variations have been formulated inconsistently using various notations and
terminologies even though they are based on a common set of conceptual and statistical
ideas. Therefore, we present a systematic reconciliation of multiple disease-drug
similarity metrics (\documentclass[12pt]{minimal}
\usepackage{amsmath}
\usepackage{wasysym}
\usepackage{amsfonts}
\usepackage{amssymb}
\usepackage{amsbsy}
\usepackage{upgreek}
\usepackage{mathrsfs}
\setlength{\oddsidemargin}{-69pt}
\begin{document}
}{}$ES$\end{document}, \documentclass[12pt]{minimal}
\usepackage{amsmath}
\usepackage{wasysym}
\usepackage{amsfonts}
\usepackage{amssymb}
\usepackage{amsbsy}
\usepackage{upgreek}
\usepackage{mathrsfs}
\setlength{\oddsidemargin}{-69pt}
\begin{document}
}{}$css$\end{document},
\documentclass[12pt]{minimal}
\usepackage{amsmath}
\usepackage{wasysym}
\usepackage{amsfonts}
\usepackage{amssymb}
\usepackage{amsbsy}
\usepackage{upgreek}
\usepackage{mathrsfs}
\setlength{\oddsidemargin}{-69pt}
\begin{document}
}{}$Sum$\end{document}, \documentclass[12pt]{minimal}
\usepackage{amsmath}
\usepackage{wasysym}
\usepackage{amsfonts}
\usepackage{amssymb}
\usepackage{amsbsy}
\usepackage{upgreek}
\usepackage{mathrsfs}
\setlength{\oddsidemargin}{-69pt}
\begin{document}
}{}$Cosine$\end{document},
\documentclass[12pt]{minimal}
\usepackage{amsmath}
\usepackage{wasysym}
\usepackage{amsfonts}
\usepackage{amssymb}
\usepackage{amsbsy}
\usepackage{upgreek}
\usepackage{mathrsfs}
\setlength{\oddsidemargin}{-69pt}
\begin{document}
}{}$XSum$\end{document}, \documentclass[12pt]{minimal}
\usepackage{amsmath}
\usepackage{wasysym}
\usepackage{amsfonts}
\usepackage{amssymb}
\usepackage{amsbsy}
\usepackage{upgreek}
\usepackage{mathrsfs}
\setlength{\oddsidemargin}{-69pt}
\begin{document}
}{}$XCor$\end{document},
\documentclass[12pt]{minimal}
\usepackage{amsmath}
\usepackage{wasysym}
\usepackage{amsfonts}
\usepackage{amssymb}
\usepackage{amsbsy}
\usepackage{upgreek}
\usepackage{mathrsfs}
\setlength{\oddsidemargin}{-69pt}
\begin{document}
}{}$XSpe$\end{document}, \documentclass[12pt]{minimal}
\usepackage{amsmath}
\usepackage{wasysym}
\usepackage{amsfonts}
\usepackage{amssymb}
\usepackage{amsbsy}
\usepackage{upgreek}
\usepackage{mathrsfs}
\setlength{\oddsidemargin}{-69pt}
\begin{document}
}{}$XCos$\end{document},
\documentclass[12pt]{minimal}
\usepackage{amsmath}
\usepackage{wasysym}
\usepackage{amsfonts}
\usepackage{amssymb}
\usepackage{amsbsy}
\usepackage{upgreek}
\usepackage{mathrsfs}
\setlength{\oddsidemargin}{-69pt}
\begin{document}
}{}$EWCos$\end{document}) and connectivity scores
(\documentclass[12pt]{minimal}
\usepackage{amsmath}
\usepackage{wasysym}
\usepackage{amsfonts}
\usepackage{amssymb}
\usepackage{amsbsy}
\usepackage{upgreek}
\usepackage{mathrsfs}
\setlength{\oddsidemargin}{-69pt}
\begin{document}
}{}$CS$\end{document}, \documentclass[12pt]{minimal}
\usepackage{amsmath}
\usepackage{wasysym}
\usepackage{amsfonts}
\usepackage{amssymb}
\usepackage{amsbsy}
\usepackage{upgreek}
\usepackage{mathrsfs}
\setlength{\oddsidemargin}{-69pt}
\begin{document}
}{}$RGES$\end{document},
\documentclass[12pt]{minimal}
\usepackage{amsmath}
\usepackage{wasysym}
\usepackage{amsfonts}
\usepackage{amssymb}
\usepackage{amsbsy}
\usepackage{upgreek}
\usepackage{mathrsfs}
\setlength{\oddsidemargin}{-69pt}
\begin{document}
}{}$NCS$\end{document}, \documentclass[12pt]{minimal}
\usepackage{amsmath}
\usepackage{wasysym}
\usepackage{amsfonts}
\usepackage{amssymb}
\usepackage{amsbsy}
\usepackage{upgreek}
\usepackage{mathrsfs}
\setlength{\oddsidemargin}{-69pt}
\begin{document}
}{}$WCS$\end{document},
\documentclass[12pt]{minimal}
\usepackage{amsmath}
\usepackage{wasysym}
\usepackage{amsfonts}
\usepackage{amssymb}
\usepackage{amsbsy}
\usepackage{upgreek}
\usepackage{mathrsfs}
\setlength{\oddsidemargin}{-69pt}
\begin{document}
}{}$Tau$\end{document}, \documentclass[12pt]{minimal}
\usepackage{amsmath}
\usepackage{wasysym}
\usepackage{amsfonts}
\usepackage{amssymb}
\usepackage{amsbsy}
\usepackage{upgreek}
\usepackage{mathrsfs}
\setlength{\oddsidemargin}{-69pt}
\begin{document}
}{}$CSS$\end{document},
\documentclass[12pt]{minimal}
\usepackage{amsmath}
\usepackage{wasysym}
\usepackage{amsfonts}
\usepackage{amssymb}
\usepackage{amsbsy}
\usepackage{upgreek}
\usepackage{mathrsfs}
\setlength{\oddsidemargin}{-69pt}
\begin{document}
}{}$EMUDRA$\end{document}) by defining them using consistent
notation and terminology. In addition to providing clarity and deeper insights, this
coherent definition of connectivity scores and their relationships provides a unified
scheme that newer methods can adopt, enabling the computational drug-development community
to compare and investigate different approaches easily. To facilitate the continuous and
transparent integration of newer methods, this article will be available as a live
document (https://jravilab.github.io/connectivity_scores) coupled with a GitHub
repository (https://github.com/jravilab/connectivity_scores) that any researcher can
build on and push changes to.
Collapse
Affiliation(s)
- Kewalin Samart
- Computational Mathematics, and Computational Math, Science & Engineering at Michigan State University, East Lansing, MI, USA
| | - Phoebe Tuyishime
- College of Agriculture and Natural Resources at Michigan State University, East Lansing, MI, USA
| | - Arjun Krishnan
- Departments of Computational Math, Science & Engineering, and Biochemistry & Molecular Biology at Michigan State University, East Lansing, MI, USA
| | - Janani Ravi
- Pathobiology and Diagnostic Investigation at Michigan State University, East Lansing, MI, USA
| |
Collapse
|
28
|
Gao S, Han L, Luo D, Liu G, Xiao Z, Shan G, Zhang Y, Zhou W. Modeling drug mechanism of action with large scale gene-expression profiles using GPAR, an artificial intelligence platform. BMC Bioinformatics 2021; 22:17. [PMID: 33413089 PMCID: PMC7788535 DOI: 10.1186/s12859-020-03915-6] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Accepted: 11/30/2020] [Indexed: 01/03/2023] Open
Abstract
Background Querying drug-induced gene expression profiles with machine learning method is an effective way for revealing drug mechanism of actions (MOAs), which is strongly supported by the growth of large scale and high-throughput gene expression databases. However, due to the lack of code-free and user friendly applications, it is not easy for biologists and pharmacologists to model MOAs with state-of-art deep learning approach. Results In this work, a newly developed online collaborative tool, Genetic profile-activity relationship (GPAR) was built to help modeling and predicting MOAs easily via deep learning. The users can use GPAR to customize their training sets to train self-defined MOA prediction models, to evaluate the model performances and to make further predictions automatically. Cross-validation tests show GPAR outperforms Gene set enrichment analysis in predicting MOAs. Conclusion GPAR can serve as a better approach in MOAs prediction, which may facilitate researchers to generate more reliable MOA hypothesis.
Collapse
Affiliation(s)
- Shengqiao Gao
- Beijing Institute of Pharmacology and Toxicology, State Key Laboratory of Toxicology and Medical Countermeasures, Beijing, 100850, China
| | - Lu Han
- Beijing Institute of Pharmacology and Toxicology, State Key Laboratory of Toxicology and Medical Countermeasures, Beijing, 100850, China
| | - Dan Luo
- Beijing Institute of Pharmacology and Toxicology, State Key Laboratory of Toxicology and Medical Countermeasures, Beijing, 100850, China
| | - Gang Liu
- Beijing Institute of Pharmacology and Toxicology, State Key Laboratory of Toxicology and Medical Countermeasures, Beijing, 100850, China
| | - Zhiyong Xiao
- Beijing Institute of Pharmacology and Toxicology, State Key Laboratory of Toxicology and Medical Countermeasures, Beijing, 100850, China
| | - Guangcun Shan
- School of Instrumentation Science and Opto-Electronics Engineering and Beijing Advanced Innovation Center for Big Data-Based Precision Medicine, Beihang University, Beijing, 100083, China
| | - Yongxiang Zhang
- Beijing Institute of Pharmacology and Toxicology, State Key Laboratory of Toxicology and Medical Countermeasures, Beijing, 100850, China.
| | - Wenxia Zhou
- Beijing Institute of Pharmacology and Toxicology, State Key Laboratory of Toxicology and Medical Countermeasures, Beijing, 100850, China.
| |
Collapse
|
29
|
Shi W, Chen X, Deng L. A Review of Recent Developments and Progress in Computational Drug Repositioning. Curr Pharm Des 2021; 26:3059-3068. [PMID: 31951162 DOI: 10.2174/1381612826666200116145559] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2019] [Accepted: 01/09/2020] [Indexed: 12/27/2022]
Abstract
Computational drug repositioning is an efficient approach towards discovering new indications for existing drugs. In recent years, with the accumulation of online health-related information and the extensive use of biomedical databases, computational drug repositioning approaches have achieved significant progress in drug discovery. In this review, we summarize recent advancements in drug repositioning. Firstly, we explicitly demonstrated the available data source information which is conducive to identifying novel indications. Furthermore, we provide a summary of the commonly used computing approaches. For each method, we briefly described techniques, case studies, and evaluation criteria. Finally, we discuss the limitations of the existing computing approaches.
Collapse
Affiliation(s)
- Wanwan Shi
- School of Computer Science and Engineering, Central South University, Changsha, China
| | - Xuegong Chen
- School of Computer Science and Engineering, Central South University, Changsha, China
| | - Lei Deng
- School of Computer Science and Engineering, Central South University, Changsha, China
| |
Collapse
|
30
|
Iyer KR, Camara K, Daniel-Ivad M, Trilles R, Pimentel-Elardo SM, Fossen JL, Marchillo K, Liu Z, Singh S, Muñoz JF, Kim SH, Porco JA, Cuomo CA, Williams NS, Ibrahim AS, Edwards JE, Andes DR, Nodwell JR, Brown LE, Whitesell L, Robbins N, Cowen LE. An oxindole efflux inhibitor potentiates azoles and impairs virulence in the fungal pathogen Candida auris. Nat Commun 2020; 11:6429. [PMID: 33353950 PMCID: PMC7755909 DOI: 10.1038/s41467-020-20183-3] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2020] [Accepted: 11/12/2020] [Indexed: 12/11/2022] Open
Abstract
Candida auris is an emerging fungal pathogen that exhibits resistance to multiple drugs, including the most commonly prescribed antifungal, fluconazole. Here, we use a combinatorial screening approach to identify a bis-benzodioxolylindolinone (azoffluxin) that synergizes with fluconazole against C. auris. Azoffluxin enhances fluconazole activity through the inhibition of efflux pump Cdr1, thus increasing intracellular fluconazole levels. This activity is conserved across most C. auris clades, with the exception of clade III. Azoffluxin also inhibits efflux in highly azole-resistant strains of Candida albicans, another human fungal pathogen, increasing their susceptibility to fluconazole. Furthermore, azoffluxin enhances fluconazole activity in mice infected with C. auris, reducing fungal burden. Our findings suggest that pharmacologically targeting Cdr1 in combination with azoles may be an effective strategy to control infection caused by azole-resistant isolates of C. auris.
Collapse
Affiliation(s)
- Kali R Iyer
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
| | - Kaddy Camara
- Department of Chemistry and Center for Molecular Discovery (BU-CMD), Boston University, Boston, MA, USA
- Clark+Elbing LLP, Boston, MA, USA
| | | | - Richard Trilles
- Department of Chemistry and Center for Molecular Discovery (BU-CMD), Boston University, Boston, MA, USA
| | | | - Jen L Fossen
- Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
- Department of Medical Microbiology and Immunology, University of Wisconsin, Madison, WI, USA
| | - Karen Marchillo
- Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
- Department of Medical Microbiology and Immunology, University of Wisconsin, Madison, WI, USA
| | - Zhongle Liu
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
| | - Shakti Singh
- Division of Infectious Disease, The Lundquist Institute for Biomedical Innovation Los Angeles Biomedical Research Institute at Harbor-University of California, Los Angeles (UCLA) Medical Center, Torrance, CA, USA
| | - José F Muñoz
- Infectious Disease and Microbiome Program, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Sang Hu Kim
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
| | - John A Porco
- Department of Chemistry and Center for Molecular Discovery (BU-CMD), Boston University, Boston, MA, USA
| | - Christina A Cuomo
- Infectious Disease and Microbiome Program, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Noelle S Williams
- Department of Biochemistry, University of Texas Southwestern Medical School, Dallas, TX, USA
| | - Ashraf S Ibrahim
- Division of Infectious Disease, The Lundquist Institute for Biomedical Innovation Los Angeles Biomedical Research Institute at Harbor-University of California, Los Angeles (UCLA) Medical Center, Torrance, CA, USA
- David Geffen School of Medicine, UCLA, Los Angeles, CA, USA
| | - John E Edwards
- Division of Infectious Disease, The Lundquist Institute for Biomedical Innovation Los Angeles Biomedical Research Institute at Harbor-University of California, Los Angeles (UCLA) Medical Center, Torrance, CA, USA
- David Geffen School of Medicine, UCLA, Los Angeles, CA, USA
| | - David R Andes
- Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
- Department of Medical Microbiology and Immunology, University of Wisconsin, Madison, WI, USA
| | - Justin R Nodwell
- Department of Biochemistry, University of Toronto, Toronto, ON, Canada
| | - Lauren E Brown
- Department of Chemistry and Center for Molecular Discovery (BU-CMD), Boston University, Boston, MA, USA
| | - Luke Whitesell
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
| | - Nicole Robbins
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
| | - Leah E Cowen
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
31
|
Ortiz-Meoz RF, Wang L, Matico R, Rutkowska-Klute A, De la Rosa M, Bedard S, Midgett R, Strohmer K, Thomson D, Zhang C, Mebrahtu M, Guss J, Totoritis R, Consler T, Campobasso N, Taylor D, Lewis T, Weaver K, Muelbaier M, Seal J, Dunham R, Kazmierski W, Favre D, Bergamini G, Shewchuk L, Rendina A, Zhang G. Characterization of Apo-Form Selective Inhibition of Indoleamine 2,3-Dioxygenase*. Chembiochem 2020; 22:516-522. [PMID: 32974990 DOI: 10.1002/cbic.202000298] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Revised: 09/23/2020] [Indexed: 01/01/2023]
Abstract
Indoleamine-2,3-dioxygenase 1 (IDO1) is a heme-containing enzyme that catalyzes the rate-limiting step in the kynurenine pathway of tryptophan (TRP) metabolism. As it is an inflammation-induced immunoregulatory enzyme, pharmacological inhibition of IDO1 activity is currently being pursued as a potential therapeutic tool for the treatment of cancer and other disease states. As such, a detailed understanding of the mechanism of action of IDO1 inhibitors with various mechanisms of inhibition is of great interest. Comparison of an apo-form-binding IDO1 inhibitor (GSK5628) to the heme-coordinating compound, epacadostat (Incyte), allows us to explore the details of the apo-binding inhibition of IDO1. Herein, we demonstrate that GSK5628 inhibits IDO1 by competing with heme for binding to a heme-free conformation of the enzyme (apo-IDO1), whereas epacadostat coordinates its binding with the iron atom of the IDO1 heme cofactor. Comparison of these two compounds in cellular systems reveals a long-lasting inhibitory effect of GSK5628, previously undescribed for other known IDO1 inhibitors. Detailed characterization of this apo-binding mechanism for IDO1 inhibition might help design superior inhibitors or could confer a unique competitive advantage over other IDO1 inhibitors vis-à-vis specificity and pharmacokinetic parameters.
Collapse
Affiliation(s)
- Rodrigo F Ortiz-Meoz
- Drug Design and Selection, GlaxoSmithKline, 1250 S. Collegeville Road, Collegeville, PA, 19426, USA
| | - Liping Wang
- Drug Design and Selection, GlaxoSmithKline, 1250 S. Collegeville Road, Collegeville, PA, 19426, USA
| | - Rosalie Matico
- Drug Design and Selection, GlaxoSmithKline, 1250 S. Collegeville Road, Collegeville, PA, 19426, USA
| | | | - Martha De la Rosa
- Infectious Diseases TAU, GlaxoSmithKline Five Moore Drive, Research Triangle Park, NC 27709, USA
| | - Sabrina Bedard
- Drug Design and Selection, GlaxoSmithKline, 1250 S. Collegeville Road, Collegeville, PA, 19426, USA
| | - Robert Midgett
- Drug Design and Selection, GlaxoSmithKline, 1250 S. Collegeville Road, Collegeville, PA, 19426, USA
| | - Katrin Strohmer
- Cellzome GmbH, GlaxoSmithKline, Meyerhofstrasse 1, 69117, Heidelberg, Germany
| | - Douglas Thomson
- Cellzome GmbH, GlaxoSmithKline, Meyerhofstrasse 1, 69117, Heidelberg, Germany
| | - Cunyu Zhang
- Drug Design and Selection, GlaxoSmithKline, 1250 S. Collegeville Road, Collegeville, PA, 19426, USA
| | - Makda Mebrahtu
- Drug Design and Selection, GlaxoSmithKline, 1250 S. Collegeville Road, Collegeville, PA, 19426, USA
| | - Jeffrey Guss
- Drug Design and Selection, GlaxoSmithKline, 1250 S. Collegeville Road, Collegeville, PA, 19426, USA
| | - Rachel Totoritis
- Drug Design and Selection, GlaxoSmithKline, 1250 S. Collegeville Road, Collegeville, PA, 19426, USA
| | - Thomas Consler
- Drug Design and Selection, GlaxoSmithKline, 1250 S. Collegeville Road, Collegeville, PA, 19426, USA
| | - Nino Campobasso
- Drug Design and Selection, GlaxoSmithKline, 1250 S. Collegeville Road, Collegeville, PA, 19426, USA
| | - David Taylor
- Drug Design and Selection, GlaxoSmithKline, 1250 S. Collegeville Road, Collegeville, PA, 19426, USA
| | - Tia Lewis
- Drug Design and Selection, GlaxoSmithKline, 1250 S. Collegeville Road, Collegeville, PA, 19426, USA
| | - Kurt Weaver
- Drug Design and Selection, GlaxoSmithKline, 1250 S. Collegeville Road, Collegeville, PA, 19426, USA
| | - Marcel Muelbaier
- Cellzome GmbH, GlaxoSmithKline, Meyerhofstrasse 1, 69117, Heidelberg, Germany
| | - John Seal
- Drug Design and Selection, GlaxoSmithKline, 1250 S. Collegeville Road, Collegeville, PA, 19426, USA
| | - Richard Dunham
- Infectious Diseases TAU, GlaxoSmithKline Five Moore Drive, Research Triangle Park, NC 27709, USA
| | - Wieslaw Kazmierski
- Infectious Diseases TAU, GlaxoSmithKline Five Moore Drive, Research Triangle Park, NC 27709, USA
| | - David Favre
- Infectious Diseases TAU, GlaxoSmithKline Five Moore Drive, Research Triangle Park, NC 27709, USA
| | - Giovanna Bergamini
- Cellzome GmbH, GlaxoSmithKline, Meyerhofstrasse 1, 69117, Heidelberg, Germany
| | - Lisa Shewchuk
- Drug Design and Selection, GlaxoSmithKline, 1250 S. Collegeville Road, Collegeville, PA, 19426, USA
| | - Alan Rendina
- Drug Design and Selection, GlaxoSmithKline, 1250 S. Collegeville Road, Collegeville, PA, 19426, USA
| | - Guofeng Zhang
- Drug Design and Selection, GlaxoSmithKline, 1250 S. Collegeville Road, Collegeville, PA, 19426, USA
| |
Collapse
|
32
|
Chen J, Wong KC. RNCE: network integration with reciprocal neighbors contextual encoding for multi-modal drug community study on cancer targets. Brief Bioinform 2020; 22:5861765. [PMID: 32577712 DOI: 10.1093/bib/bbaa118] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2020] [Revised: 04/29/2020] [Indexed: 11/14/2022] Open
Abstract
Mining drug targets and mechanisms of action (MoA) for novel anticancer drugs from pharmacogenomic data is a path to enhance the drug discovery efficiency. Recent approaches have successfully attempted to discover targets/MoA by characterizing drug similarities and communities with integrative methods on multi-modal or multi-omics drug information. However, the sparse and imbalanced community size structure of the drug network is seldom considered in recent approaches. Consequently, we developed a novel network integration approach accounting for network structure by a reciprocal nearest neighbor and contextual information encoding (RNCE) approach. In addition, we proposed a tailor-made clustering algorithm to perform drug community detection on drug networks. RNCE and spectral clustering are proved to outperform state-of-the-art approaches in a series of tests, including network similarity tests and community detection tests on two drug databases. The observed improvement of RNCE can contribute to the field of drug discovery and the related multi-modal/multi-omics integrative studies. Availabilityhttps://github.com/WINGHARE/RNCE.
Collapse
Affiliation(s)
- Junyi Chen
- Department of Computer Science, City University of Hong Kong
| | - Ka-Chun Wong
- Department of Computer Science, City University of Hong Kong
| |
Collapse
|
33
|
Lin K, Li L, Dai Y, Wang H, Teng S, Bao X, Lu ZJ, Wang D. A comprehensive evaluation of connectivity methods for L1000 data. Brief Bioinform 2019; 21:2194-2205. [DOI: 10.1093/bib/bbz129] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2019] [Revised: 08/26/2019] [Accepted: 09/14/2019] [Indexed: 01/08/2023] Open
Abstract
Abstract
The methodologies for evaluating similarities between gene expression profiles of different perturbagens are the key to understanding mechanisms of actions (MoAs) of unknown compounds and finding new indications for existing drugs. L1000-based next-generation Connectivity Map (CMap) data is more than a thousand-fold scale-up of the CMap pilot dataset. Although several systematic evaluations have been performed individually to assess the accuracy of the methodologies for the CMap pilot study, the performance of these methodologies needs to be re-evaluated for the L1000 data. Here, using the drug–drug similarities from the Drug Repurposing Hub database as a benchmark standard, we evaluated six popular published methods for the prediction performance of drug–drug relationships based on the partial area under the receiver operating characteristic (ROC) curve at false positive rates of 0.001, 0.005 and 0.01 (AUC0.001, AUC0.005 and AUC0.01). The similarity evaluating algorithm called ZhangScore was generally superior to other methods and exhibited the highest accuracy at the gene signature sizes ranging from 10 to 200. Further, we tested these methods with an experimentally derived gene signature related to estrogen in breast cancer cells, and the results confirmed that ZhangScore was more accurate than other methods. Moreover, based on scoring results of ZhangScore for the gene signature of TOP2A knockdown, in addition to well-known TOP2A inhibitors, we identified a number of potential inhibitors and at least two of them were the subject of previous investigation. Our studies provide potential guidelines for researchers to choose the suitable connectivity method. The six connectivity methods used in this report have been implemented in R package (https://github.com/Jasonlinchina/RCSM).
Collapse
Affiliation(s)
- Kequan Lin
- School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Lu Li
- School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Yifei Dai
- School of Medicine, Tsinghua University, Beijing 100084, China
| | - Huili Wang
- School of Medicine, Tsinghua University, Beijing 100084, China
| | - Shuaishuai Teng
- School of Medicine, Tsinghua University, Beijing 100084, China
| | - Xilinqiqige Bao
- International Mongolian Hospital of Inner Mongolia, Hohhot 010065, China
| | - Zhi John Lu
- School of Life Sciences, Tsinghua University, Beijing 100084, China
- Center of Synthetic & Systems Biology, Tsinghua University, Beijing 100084, China
- MOE Key Laboratory of Bioinformatics, Tsinghua University, Beijing 100084, China
| | - Dong Wang
- School of Medicine, Tsinghua University, Beijing 100084, China
- Center of Synthetic & Systems Biology, Tsinghua University, Beijing 100084, China
- National Collaborative Innovation Center for Biotherapy, Tsinghua University, Beijing 100084, China
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| |
Collapse
|
34
|
Cury NM, Capitão RM, Almeida RDCBD, Artico LL, Corrêa JR, Simão dos Santos EF, Yunes JA, Correia CRD. Synthesis and evaluation of 2-carboxy indole derivatives as potent and selective anti-leukemic agents. Eur J Med Chem 2019; 181:111570. [DOI: 10.1016/j.ejmech.2019.111570] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2019] [Revised: 07/26/2019] [Accepted: 07/28/2019] [Indexed: 12/29/2022]
|
35
|
Li YH, Yu CY, Li XX, Zhang P, Tang J, Yang Q, Fu T, Zhang X, Cui X, Tu G, Zhang Y, Li S, Yang F, Sun Q, Qin C, Zeng X, Chen Z, Chen YZ, Zhu F. Therapeutic target database update 2018: enriched resource for facilitating bench-to-clinic research of targeted therapeutics. Nucleic Acids Res 2019; 46:D1121-D1127. [PMID: 29140520 PMCID: PMC5753365 DOI: 10.1093/nar/gkx1076] [Citation(s) in RCA: 386] [Impact Index Per Article: 64.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2017] [Accepted: 11/10/2017] [Indexed: 12/25/2022] Open
Abstract
Extensive efforts have been directed at the discovery, investigation and clinical monitoring of targeted therapeutics. These efforts may be facilitated by the convenient access of the genetic, proteomic, interactive and other aspects of the therapeutic targets. Here, we describe an update of the Therapeutic target database (TTD) previously featured in NAR. This update includes: (i) 2000 drug resistance mutations in 83 targets and 104 target/drug regulatory genes, which are resistant to 228 drugs targeting 63 diseases (49 targets of 61 drugs with patient prevalence data); (ii) differential expression profiles of 758 targets in the disease-relevant drug-targeted tissue of 12 615 patients of 70 diseases; (iii) expression profiles of 629 targets in the non-targeted tissues of 2565 healthy individuals; (iv) 1008 target combinations of 1764 drugs and the 1604 target combination of 664 multi-target drugs; (v) additional 48 successful, 398 clinical trial and 21 research targets, 473 approved, 812 clinical trial and 1120 experimental drugs, and (vi) ICD-10-CM and ICD-9-CM codes for additional 482 targets and 262 drugs against 98 disease conditions. This update makes TTD more useful for facilitating the patient focused research, discovery and clinical investigations of the targeted therapeutics. TTD is accessible at http://bidd.nus.edu.sg/group/ttd/ttd.asp.
Collapse
Affiliation(s)
- Ying Hong Li
- Bioinformatics and Drug Design Group, Department of Pharmacy and Center for Computational Science and Engineering, National University of Singapore, Singapore 117543, Singapore.,Innovative Drug Research and Bioinformatics Group, School of Pharmaceutical Sciences, Chongqing University, Chongqing 401331, China
| | - Chun Yan Yu
- Bioinformatics and Drug Design Group, Department of Pharmacy and Center for Computational Science and Engineering, National University of Singapore, Singapore 117543, Singapore.,Innovative Drug Research and Bioinformatics Group, School of Pharmaceutical Sciences, Chongqing University, Chongqing 401331, China
| | - Xiao Xu Li
- Bioinformatics and Drug Design Group, Department of Pharmacy and Center for Computational Science and Engineering, National University of Singapore, Singapore 117543, Singapore.,Innovative Drug Research and Bioinformatics Group, School of Pharmaceutical Sciences, Chongqing University, Chongqing 401331, China
| | - Peng Zhang
- Bioinformatics and Drug Design Group, Department of Pharmacy and Center for Computational Science and Engineering, National University of Singapore, Singapore 117543, Singapore
| | - Jing Tang
- Innovative Drug Research and Bioinformatics Group, School of Pharmaceutical Sciences, Chongqing University, Chongqing 401331, China
| | - Qingxia Yang
- Innovative Drug Research and Bioinformatics Group, School of Pharmaceutical Sciences, Chongqing University, Chongqing 401331, China
| | - Tingting Fu
- Innovative Drug Research and Bioinformatics Group, School of Pharmaceutical Sciences, Chongqing University, Chongqing 401331, China
| | - Xiaoyu Zhang
- Innovative Drug Research and Bioinformatics Group, School of Pharmaceutical Sciences, Chongqing University, Chongqing 401331, China
| | - Xuejiao Cui
- Innovative Drug Research and Bioinformatics Group, School of Pharmaceutical Sciences, Chongqing University, Chongqing 401331, China
| | - Gao Tu
- Innovative Drug Research and Bioinformatics Group, School of Pharmaceutical Sciences, Chongqing University, Chongqing 401331, China
| | - Yang Zhang
- Innovative Drug Research and Bioinformatics Group, School of Pharmaceutical Sciences, Chongqing University, Chongqing 401331, China
| | - Shuang Li
- Innovative Drug Research and Bioinformatics Group, School of Pharmaceutical Sciences, Chongqing University, Chongqing 401331, China
| | - Fengyuan Yang
- Innovative Drug Research and Bioinformatics Group, School of Pharmaceutical Sciences, Chongqing University, Chongqing 401331, China
| | - Qiu Sun
- Innovative Drug Research and Bioinformatics Group, School of Pharmaceutical Sciences, Chongqing University, Chongqing 401331, China
| | - Chu Qin
- Bioinformatics and Drug Design Group, Department of Pharmacy and Center for Computational Science and Engineering, National University of Singapore, Singapore 117543, Singapore
| | - Xian Zeng
- Bioinformatics and Drug Design Group, Department of Pharmacy and Center for Computational Science and Engineering, National University of Singapore, Singapore 117543, Singapore
| | - Zhe Chen
- Zhejiang Key Laboratory of Gastro-intestinal Pathophysiology, Zhejiang Hospital of Traditional Chinese Medicine, Zhejiang Chinese Medical University, Hangzhou 310006, China
| | - Yu Zong Chen
- Bioinformatics and Drug Design Group, Department of Pharmacy and Center for Computational Science and Engineering, National University of Singapore, Singapore 117543, Singapore
| | - Feng Zhu
- Bioinformatics and Drug Design Group, Department of Pharmacy and Center for Computational Science and Engineering, National University of Singapore, Singapore 117543, Singapore.,Innovative Drug Research and Bioinformatics Group, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China.,Innovative Drug Research and Bioinformatics Group, School of Pharmaceutical Sciences, Chongqing University, Chongqing 401331, China
| |
Collapse
|
36
|
Wang Z, Lachmann A, Keenan AB, Ma'ayan A. L1000FWD: fireworks visualization of drug-induced transcriptomic signatures. Bioinformatics 2019; 34:2150-2152. [PMID: 29420694 DOI: 10.1093/bioinformatics/bty060] [Citation(s) in RCA: 110] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2017] [Accepted: 02/05/2018] [Indexed: 11/12/2022] Open
Abstract
Motivation As part of the NIH Library of Integrated Network-based Cellular Signatures program, hundreds of thousands of transcriptomic signatures were generated with the L1000 technology, profiling the response of human cell lines to over 20 000 small molecule compounds. This effort is a promising approach toward revealing the mechanisms-of-action (MOA) for marketed drugs and other less studied potential therapeutic compounds. Results L1000 fireworks display (L1000FWD) is a web application that provides interactive visualization of over 16 000 drug and small-molecule induced gene expression signatures. L1000FWD enables coloring of signatures by different attributes such as cell type, time point, concentration, as well as drug attributes such as MOA and clinical phase. Signature similarity search is implemented to enable the search for mimicking or opposing signatures given as input of up and down gene sets. Each point on the L1000FWD interactive map is linked to a signature landing page, which provides multifaceted knowledge from various sources about the signature and the drug. Notably such information includes most frequent diagnoses, co-prescribed drugs and age distribution of prescriptions as extracted from the Mount Sinai Health System electronic medical records. Overall, L1000FWD serves as a platform for identifying functions for novel small molecules using unsupervised clustering, as well as for exploring drug MOA. Availability and implementation L1000FWD is freely accessible at: http://amp.pharm.mssm.edu/L1000FWD. Supplementary information Supplementary data are available at Bioinformatics online.
Collapse
Affiliation(s)
- Zichen Wang
- Department of Pharmacological Sciences, BD2K-LINCS Data Coordination and Integration Center, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Alexander Lachmann
- Department of Pharmacological Sciences, BD2K-LINCS Data Coordination and Integration Center, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Alexandra B Keenan
- Department of Pharmacological Sciences, BD2K-LINCS Data Coordination and Integration Center, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Avi Ma'ayan
- Department of Pharmacological Sciences, BD2K-LINCS Data Coordination and Integration Center, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| |
Collapse
|
37
|
Keenan AB, Wojciechowicz ML, Wang Z, Jagodnik KM, Jenkins SL, Lachmann A, Ma'ayan A. Connectivity Mapping: Methods and Applications. Annu Rev Biomed Data Sci 2019. [DOI: 10.1146/annurev-biodatasci-072018-021211] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Connectivity mapping resources consist of signatures representing changes in cellular state following systematic small-molecule, disease, gene, or other form of perturbations. Such resources enable the characterization of signatures from novel perturbations based on similarity; provide a global view of the space of many themed perturbations; and allow the ability to predict cellular, tissue, and organismal phenotypes for perturbagens. A signature search engine enables hypothesis generation by finding connections between query signatures and the database of signatures. This framework has been used to identify connections between small molecules and their targets, to discover cell-specific responses to perturbations and ways to reverse disease expression states with small molecules, and to predict small-molecule mimickers for existing drugs. This review provides a historical perspective and the current state of connectivity mapping resources with a focus on both methodology and community implementations.
Collapse
Affiliation(s)
- Alexandra B. Keenan
- Department of Pharmacological Sciences and Mount Sinai Center for Bioinformatics, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Megan L. Wojciechowicz
- Department of Pharmacological Sciences and Mount Sinai Center for Bioinformatics, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Zichen Wang
- Department of Pharmacological Sciences and Mount Sinai Center for Bioinformatics, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Kathleen M. Jagodnik
- Department of Pharmacological Sciences and Mount Sinai Center for Bioinformatics, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Sherry L. Jenkins
- Department of Pharmacological Sciences and Mount Sinai Center for Bioinformatics, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Alexander Lachmann
- Department of Pharmacological Sciences and Mount Sinai Center for Bioinformatics, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Avi Ma'ayan
- Department of Pharmacological Sciences and Mount Sinai Center for Bioinformatics, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| |
Collapse
|
38
|
Silverbush D, Sharan R. A systematic approach to orient the human protein-protein interaction network. Nat Commun 2019; 10:3015. [PMID: 31289271 PMCID: PMC6617457 DOI: 10.1038/s41467-019-10887-6] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2018] [Accepted: 06/06/2019] [Indexed: 11/16/2022] Open
Abstract
The protein-protein interaction (PPI) network of an organism serves as a skeleton for its signaling circuitry, which mediates cellular response to environmental and genetic cues. Understanding this circuitry could improve the prediction of gene function and cellular behavior in response to diverse signals. To realize this potential, one has to comprehensively map PPIs and their directions of signal flow. While the quality and the volume of identified human PPIs improved dramatically over the last decade, the directions of these interactions are still mostly unknown, thus precluding subsequent prediction and modeling efforts. Here we present a systematic approach to orient the human PPI network using drug response and cancer genomic data. We provide a diffusion-based method for the orientation task that significantly outperforms existing methods. The oriented network leads to improved prioritization of cancer driver genes and drug targets compared to the state-of-the-art unoriented network.
Collapse
Affiliation(s)
- Dana Silverbush
- The Blavatnik School of Computer Science, Tel Aviv University, Tel Aviv, 69978, Israel
| | - Roded Sharan
- The Blavatnik School of Computer Science, Tel Aviv University, Tel Aviv, 69978, Israel.
| |
Collapse
|
39
|
Turanli B, Zhang C, Kim W, Benfeitas R, Uhlen M, Arga KY, Mardinoglu A. Discovery of therapeutic agents for prostate cancer using genome-scale metabolic modeling and drug repositioning. EBioMedicine 2019; 42:386-396. [PMID: 30905848 PMCID: PMC6491384 DOI: 10.1016/j.ebiom.2019.03.009] [Citation(s) in RCA: 54] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2018] [Revised: 02/28/2019] [Accepted: 03/04/2019] [Indexed: 12/27/2022] Open
Abstract
BACKGROUND Genome-scale metabolic models (GEMs) offer insights into cancer metabolism and have been used to identify potential biomarkers and drug targets. Drug repositioning is a time- and cost-effective method of drug discovery that can be applied together with GEMs for effective cancer treatment. METHODS In this study, we reconstruct a prostate cancer (PRAD)-specific GEM for exploring prostate cancer metabolism and also repurposing new therapeutic agents that can be used in development of effective cancer treatment. We integrate global gene expression profiling of cell lines with >1000 different drugs through the use of prostate cancer GEM and predict possible drug-gene interactions. FINDINGS We identify the key reactions with altered fluxes based on the gene expression changes and predict the potential drug effect in prostate cancer treatment. We find that sulfamethoxypyridazine, azlocillin, hydroflumethiazide, and ifenprodil can be repurposed for the treatment of prostate cancer based on an in silico cell viability assay. Finally, we validate the effect of ifenprodil using an in vitro cell assay and show its inhibitory effect on a prostate cancer cell line. INTERPRETATION Our approach demonstate how GEMs can be used to predict therapeutic agents for cancer treatment based on drug repositioning. Besides, it paved a way and shed a light on the applicability of computational models to real-world biomedical or pharmaceutical problems.
Collapse
Affiliation(s)
- Beste Turanli
- Science for Life Laboratory, KTH - Royal Institute of Technology, Stockholm SE-17121, Sweden; Department of Bioengineering, Marmara University, Istanbul, Turkey; Department of Bioengineering, Istanbul Medeniyet University, Istanbul, Turkey
| | - Cheng Zhang
- Science for Life Laboratory, KTH - Royal Institute of Technology, Stockholm SE-17121, Sweden
| | - Woonghee Kim
- Science for Life Laboratory, KTH - Royal Institute of Technology, Stockholm SE-17121, Sweden
| | - Rui Benfeitas
- Science for Life Laboratory, KTH - Royal Institute of Technology, Stockholm SE-17121, Sweden
| | - Mathias Uhlen
- Science for Life Laboratory, KTH - Royal Institute of Technology, Stockholm SE-17121, Sweden
| | | | - Adil Mardinoglu
- Science for Life Laboratory, KTH - Royal Institute of Technology, Stockholm SE-17121, Sweden; Department of Biology and Biological Engineering, Chalmers University of Technology, Gothenburg SE-41296, Sweden; Centre for Host-Microbiome Interactions, Faculty of Dentistry, Oral & Craniofacial Sciences, King's College London, London, United Kingdom.
| |
Collapse
|
40
|
Wang YY, Cui C, Qi L, Yan H, Zhao XM. DrPOCS: Drug Repositioning Based on Projection Onto Convex Sets. IEEE/ACM TRANSACTIONS ON COMPUTATIONAL BIOLOGY AND BIOINFORMATICS 2019; 16:154-162. [PMID: 29993698 DOI: 10.1109/tcbb.2018.2830384] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
Drug repositioning, i.e., identifying new indications for known drugs, has attracted a lot of attentions recently and is becoming an effective strategy in drug development. In literature, several computational approaches have been proposed to identify potential indications of old drugs based on various types of data sources. In this paper, by formulating the drug-disease associations as a low-rank matrix, we propose a novel method, namely DrPOCS, to identify candidate indications of old drugs based on projection onto convex sets (POCS). With the integration of drug structure and disease phenotype information, DrPOCS predicts potential associations between drugs and diseases with matrix completion. Benchmarking results demonstrate that our proposed approach outperforms popular existing approaches with high accuracy. In addition, a number of novel predicted indications are validated with various types of evidences, indicating the predictive power of our proposed approach.
Collapse
|
41
|
Musa A, Ghoraie LS, Zhang SD, Glazko G, Yli-Harja O, Dehmer M, Haibe-Kains B, Emmert-Streib F. A review of connectivity map and computational approaches in pharmacogenomics. Brief Bioinform 2018; 19:506-523. [PMID: 28069634 PMCID: PMC5952941 DOI: 10.1093/bib/bbw112] [Citation(s) in RCA: 102] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Large-scale perturbation databases, such as Connectivity Map (CMap) or Library of Integrated Network-based Cellular Signatures (LINCS), provide enormous opportunities for computational pharmacogenomics and drug design. A reason for this is that in contrast to classical pharmacology focusing at one target at a time, the transcriptomics profiles provided by CMap and LINCS open the door for systems biology approaches on the pathway and network level. In this article, we provide a review of recent developments in computational pharmacogenomics with respect to CMap and LINCS and related applications.
Collapse
Affiliation(s)
- Aliyu Musa
- Predictive Medicine and Analytics Lab, Department of Signal Processing, Tampere University of Technology, Tampere, Finland
| | - Laleh Soltan Ghoraie
- Bioinformatics and Computational Genomics Laboratory, Princess Margaret Cancer Center, University Health Network, Toronto, ON, Canada
| | - Shu-Dong Zhang
- Northern Ireland Centre for Stratified Medicine, Biomedical Sciences Research Institute, University of Ulster, C-TRIC Building, Altnagelvin Area Hospital, Glenshane Road, Derry/Londonderry, Northern Ireland, UK
| | - Galina Glazko
- University of Rochester Department of Biostatistics and Computational Biology, Rochester, New York, USA
| | - Olli Yli-Harja
- Computational Systems Biology, Department of Signal Processing, Tampere University of Technology, Tampere, Finland
| | - Matthias Dehmer
- Institute for Bioinformatics and Translational Research, UMIT- The Health and Life Sciences University, Eduard Wallnoefer Zentrum 1, Hall in Tyrol, Austria
| | - Benjamin Haibe-Kains
- Bioinformatics and Computational Genomics Laboratory, Princess Margaret Cancer Center, University Health Network, Toronto, ON, Canada
- Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada
- Department of Computer Science, University of Toronto, Toronto, ON, Canada
- Ontario Institute of Cancer Research, Toronto, ON, Canada
| | - Frank Emmert-Streib
- Predictive Medicine and Analytics Lab, Department of Signal Processing, Tampere University of Technology, Tampere, Finland
| |
Collapse
|
42
|
Musa A, Ghoraie LS, Zhang SD, Glazko G, Yli-Harja O, Dehmer M, Haibe-Kains B, Emmert-Streib F. A review of connectivity map and computational approaches in pharmacogenomics. Brief Bioinform 2018. [PMID: 28069634 DOI: 10.1093/bib] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/14/2023] Open
Abstract
Large-scale perturbation databases, such as Connectivity Map (CMap) or Library of Integrated Network-based Cellular Signatures (LINCS), provide enormous opportunities for computational pharmacogenomics and drug design. A reason for this is that in contrast to classical pharmacology focusing at one target at a time, the transcriptomics profiles provided by CMap and LINCS open the door for systems biology approaches on the pathway and network level. In this article, we provide a review of recent developments in computational pharmacogenomics with respect to CMap and LINCS and related applications.
Collapse
Affiliation(s)
- Aliyu Musa
- Predictive Medicine and Analytics Lab, Department of Signal Processing, Tampere University of Technology, Tampere, Finland
| | - Laleh Soltan Ghoraie
- Bioinformatics and Computational Genomics Laboratory, Princess Margaret Cancer Center, University Health Network, Toronto, ON, Canada
| | - Shu-Dong Zhang
- Northern Ireland Centre for Stratified Medicine, Biomedical Sciences Research Institute, University of Ulster, C-TRIC Building, Altnagelvin Area Hospital, Glenshane Road, Derry/Londonderry BT47 6SB, Northern Ireland, UK
| | - Galina Glazko
- University of Rochester Department of Biostatistics and Computational Biology, Rochester, New York 14642, USA
| | - Olli Yli-Harja
- Computational Systems Biology, Department of Signal Processing, Tampere University of Technology, Tampere, Finland
| | - Matthias Dehmer
- Institute for Bioinformatics and Translational Research, UMIT- The Health and Life Sciences University, Eduard Wallnoefer Zentrum 1, 6060 Hall in Tyrol, Austria
| | - Benjamin Haibe-Kains
- Bioinformatics and Computational Genomics Laboratory, Princess Margaret Cancer Center, University Health Network, Toronto, ON, Canada
- Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada
- Department of Computer Science, University of Toronto, Toronto, ON, Canada
- Ontario Institute of Cancer Research, Toronto, ON, Canada
| | - Frank Emmert-Streib
- Predictive Medicine and Analytics Lab, Department of Signal Processing, Tampere University of Technology, Tampere, Finland
| |
Collapse
|
43
|
Zhou X, Wang M, Katsyv I, Irie H, Zhang B. EMUDRA: Ensemble of Multiple Drug Repositioning Approaches to improve prediction accuracy. Bioinformatics 2018; 34:3151-3159. [PMID: 29688306 PMCID: PMC6138000 DOI: 10.1093/bioinformatics/bty325] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2017] [Accepted: 04/20/2018] [Indexed: 01/31/2023] Open
Abstract
Motivation Availability of large-scale genomic, epigenetic and proteomic data in complex diseases makes it possible to objectively and comprehensively identify the therapeutic targets that can lead to new therapies. The Connectivity Map has been widely used to explore novel indications of existing drugs. However, the prediction accuracy of the existing methods, such as Kolmogorov-Smirnov statistic remains low. Here we present a novel high-performance drug repositioning approach that improves over the state-of-the-art methods. Results We first designed an expression weighted cosine (EWCos) method to minimize the influence of the uninformative expression changes and then developed an ensemble approach termed ensemble of multiple drug repositioning approaches (EMUDRA) to integrate EWCos and three existing state-of-the-art methods. EMUDRA significantly outperformed individual drug repositioning methods when applied to simulated and independent evaluation datasets. We predicted using EMUDRA and experimentally validated an antibiotic rifabutin as an inhibitor of cell growth in triple negative breast cancer. EMUDRA can identify drugs that more effectively target disease gene signatures and will thus be a useful tool for identifying novel therapies for complex diseases and predicting new indications for existing drugs. Availability and implementation The EMUDRA R package is available at doi: 10.7303/syn11510888. Supplementary information Supplementary data are available at Bioinformatics online.
Collapse
Affiliation(s)
- Xianxiao Zhou
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA,Icahn Institute for Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Minghui Wang
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA,Icahn Institute for Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Igor Katsyv
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA,Icahn Institute for Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, NY, USA,Medical Scientist Training Program, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Hanna Irie
- Division of Hematology and Medical Oncology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA,Department of Oncological Sciences, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Bin Zhang
- To whom correspondence should be addressed. ,
| |
Collapse
|
44
|
Sirci F, Napolitano F, Pisonero-Vaquero S, Carrella D, Medina DL, di Bernardo D. Comparing structural and transcriptional drug networks reveals signatures of drug activity and toxicity in transcriptional responses. NPJ Syst Biol Appl 2017; 3:23. [PMID: 28861278 PMCID: PMC5572457 DOI: 10.1038/s41540-017-0022-3] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2017] [Revised: 06/27/2017] [Accepted: 07/07/2017] [Indexed: 02/07/2023] Open
Abstract
We performed an integrated analysis of drug chemical structures and drug-induced transcriptional responses. We demonstrated that a network representing three-dimensional structural similarities among 5452 compounds can be used to automatically group together drugs with similar scaffolds, physicochemical parameters and mode-of-action. We compared the structural network to a network representing transcriptional similarities among a subset of 1309 drugs for which transcriptional response were available in the Connectivity Map data set. Analysis of structurally similar, but transcriptionally different drugs sharing the same MOA enabled us to detect and remove weak and noisy transcriptional responses, greatly enhancing the reliability of transcription-based approaches to drug discovery and drug repositioning. Cardiac glycosides exhibited the strongest transcriptional responses with a significant induction of pathways related to epigenetic regulation, which suggests an epigenetic mechanism of action for these drugs. Drug classes with the weakest transcriptional responses tended to induce expression of cytochrome P450 enzymes, hinting at drug-induced drug resistance. Analysis of transcriptionally similar, but structurally different drugs with unrelated MOA, led us to the identification of a 'toxic' transcriptional signature indicative of lysosomal stress (lysosomotropism) and lipid accumulation (phospholipidosis) partially masking the target-specific transcriptional effects of these drugs. We found that this transcriptional signature is shared by 258 compounds and it is associated to the activation of the transcription factor TFEB, a master regulator of lysosomal biogenesis and autophagy. Finally, we built a predictive Random Forest model of these 258 compounds based on 128 physicochemical parameters, which should help in the early identification of potentially toxic drug candidates. Transcriptional responses to drug treatment can reveal mechanism of action and off-target effects thus enabling drug repositioning, but only if measured in the appropriate cells at clinically relevant concentrations. A team led by Diego di Bernardo and Diego Medina generated a network representing structural similarities among compounds to automatically group together drugs with similar scaffolds and MOA. By comparing the structural drug network with a transcriptional drug network based on similarities in transcriptional response, the team observed broad differences between the two. This observation led to the identification of a transcriptional signature related lysosomal stress and phospholipidosis, and a physicochemical model to identify such compounds. These results provide general guidelines to prevent erroneous conclusion when using transcriptional responses of small molecules for drug discovery and drug repositioning
Collapse
Affiliation(s)
- Francesco Sirci
- Telethon Institute of Genetics and Medicine (TIGEM), System Biology and Bioinformatics lab. and High Content Screening facility, Via Campi Flegrei 34, 80078 Pozzuoli (NA), Italy
| | - Francesco Napolitano
- Telethon Institute of Genetics and Medicine (TIGEM), System Biology and Bioinformatics lab. and High Content Screening facility, Via Campi Flegrei 34, 80078 Pozzuoli (NA), Italy
| | - Sandra Pisonero-Vaquero
- Telethon Institute of Genetics and Medicine (TIGEM), System Biology and Bioinformatics lab. and High Content Screening facility, Via Campi Flegrei 34, 80078 Pozzuoli (NA), Italy
| | - Diego Carrella
- Telethon Institute of Genetics and Medicine (TIGEM), System Biology and Bioinformatics lab. and High Content Screening facility, Via Campi Flegrei 34, 80078 Pozzuoli (NA), Italy
| | - Diego L Medina
- Telethon Institute of Genetics and Medicine (TIGEM), System Biology and Bioinformatics lab. and High Content Screening facility, Via Campi Flegrei 34, 80078 Pozzuoli (NA), Italy
| | - Diego di Bernardo
- Telethon Institute of Genetics and Medicine (TIGEM), System Biology and Bioinformatics lab. and High Content Screening facility, Via Campi Flegrei 34, 80078 Pozzuoli (NA), Italy.,Department of Chemical, Materials and Industrial Production Engineering, University of Naples Federico II, Piazzale Tecchio 80, 80125 Naples, Italy
| |
Collapse
|
45
|
Vilar S, Hripcsak G. The role of drug profiles as similarity metrics: applications to repurposing, adverse effects detection and drug-drug interactions. Brief Bioinform 2017; 18:670-681. [PMID: 27273288 PMCID: PMC6078166 DOI: 10.1093/bib/bbw048] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2016] [Revised: 04/18/2016] [Indexed: 12/30/2022] Open
Abstract
Explosion of the availability of big data sources along with the development in computational methods provides a useful framework to study drugs' actions, such as interactions with pharmacological targets and off-targets. Databases related to protein interactions, adverse effects and genomic profiles are available to be used for the construction of computational models. In this article, we focus on the description of biological profiles for drugs that can be used as a system to compare similarity and create methods to predict and analyze drugs' actions. We highlight profiles constructed with different biological data, such as target-protein interactions, gene expression measurements, adverse effects and disease profiles. We focus on the discovery of new targets or pathways for drugs already in the pharmaceutical market, also called drug repurposing, in the interaction with off-targets responsible for adverse reactions and in drug-drug interaction analysis. The current and future applications, strengths and challenges facing all these methods are also discussed. Biological profiles or signatures are an important source of data generation to deeply analyze biological actions with important implications in drug-related studies.
Collapse
Affiliation(s)
- Santiago Vilar
- Corresponding author: Santiago Vilar, Department of Biomedical Informatics, Columbia University Medical Center, New York, NY 10032, USA. E-mail: ; George Hripcsak, Department of Biomedical Informatics, Columbia University Medical Center, New York, NY 10032, USA. E-mail:
| | - George Hripcsak
- Corresponding author: Santiago Vilar, Department of Biomedical Informatics, Columbia University Medical Center, New York, NY 10032, USA. E-mail: ; George Hripcsak, Department of Biomedical Informatics, Columbia University Medical Center, New York, NY 10032, USA. E-mail:
| |
Collapse
|
46
|
Licciardello MP, Ringler A, Markt P, Klepsch F, Lardeau CH, Sdelci S, Schirghuber E, Müller AC, Caldera M, Wagner A, Herzog R, Penz T, Schuster M, Boidol B, Dürnberger G, Folkvaljon Y, Stattin P, Ivanov V, Colinge J, Bock C, Kratochwill K, Menche J, Bennett KL, Kubicek S. A combinatorial screen of the CLOUD uncovers a synergy targeting the androgen receptor. Nat Chem Biol 2017; 13:771-778. [DOI: 10.1038/nchembio.2382] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2015] [Accepted: 03/02/2017] [Indexed: 12/24/2022]
|
47
|
El-Hachem N, Gendoo DMA, Ghoraie LS, Safikhani Z, Smirnov P, Chung C, Deng K, Fang A, Birkwood E, Ho C, Isserlin R, Bader GD, Goldenberg A, Haibe-Kains B. Integrative Cancer Pharmacogenomics to Infer Large-Scale Drug Taxonomy. Cancer Res 2017; 77:3057-3069. [PMID: 28314784 DOI: 10.1158/0008-5472.can-17-0096] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2017] [Revised: 02/27/2017] [Accepted: 03/13/2017] [Indexed: 11/16/2022]
Abstract
Identification of drug targets and mechanism of action (MoA) for new and uncharacterized anticancer drugs is important for optimization of treatment efficacy. Current MoA prediction largely relies on prior information including side effects, therapeutic indication, and chemoinformatics. Such information is not transferable or applicable for newly identified, previously uncharacterized small molecules. Therefore, a shift in the paradigm of MoA predictions is necessary toward development of unbiased approaches that can elucidate drug relationships and efficiently classify new compounds with basic input data. We propose here a new integrative computational pharmacogenomic approach, referred to as Drug Network Fusion (DNF), to infer scalable drug taxonomies that rely only on basic drug characteristics toward elucidating drug-drug relationships. DNF is the first framework to integrate drug structural information, high-throughput drug perturbation, and drug sensitivity profiles, enabling drug classification of new experimental compounds with minimal prior information. DNF taxonomy succeeded in identifying pertinent and novel drug-drug relationships, making it suitable for investigating experimental drugs with potential new targets or MoA. The scalability of DNF facilitated identification of key drug relationships across different drug categories, providing a flexible tool for potential clinical applications in precision medicine. Our results support DNF as a valuable resource to the cancer research community by providing new hypotheses on compound MoA and potential insights for drug repurposing. Cancer Res; 77(11); 3057-69. ©2017 AACR.
Collapse
Affiliation(s)
- Nehme El-Hachem
- Integrative Computational Systems Biology, Institut de Recherches Cliniques de Montréal, Montreal, Quebec, Canada.,Department of Biomedical Sciences. Université de Montréal, Montreal, Quebec, Canada
| | - Deena M A Gendoo
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada.,Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada
| | - Laleh Soltan Ghoraie
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada.,Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada
| | - Zhaleh Safikhani
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada.,Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada
| | - Petr Smirnov
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - Christina Chung
- Department of Computer Science, University of Toronto, Toronto, Ontario, Canada
| | - Kenan Deng
- Department of Computer Science, University of Toronto, Toronto, Ontario, Canada
| | - Ailsa Fang
- Department of Computer Science, University of Toronto, Toronto, Ontario, Canada
| | - Erin Birkwood
- School of Computer Science, McGill University, Montreal, Quebec, Canada
| | - Chantal Ho
- Department of Computer Science, University of Toronto, Toronto, Ontario, Canada
| | - Ruth Isserlin
- Department of Computer Science, University of Toronto, Toronto, Ontario, Canada
| | - Gary D Bader
- Department of Computer Science, University of Toronto, Toronto, Ontario, Canada.,The Donnelly Centre, Toronto, Ontario, Canada.,The Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Ontario, Canada
| | - Anna Goldenberg
- Department of Computer Science, University of Toronto, Toronto, Ontario, Canada.,Hospital for Sick Children, Toronto, Ontario, Canada
| | - Benjamin Haibe-Kains
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada. .,Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada.,Department of Computer Science, University of Toronto, Toronto, Ontario, Canada.,Ontario Institute of Cancer Research, Toronto, Ontario, Canada
| |
Collapse
|
48
|
Elucidating the modes of action for bioactive compounds in a cell-specific manner by large-scale chemically-induced transcriptomics. Sci Rep 2017; 7:40164. [PMID: 28071740 PMCID: PMC5223214 DOI: 10.1038/srep40164] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2016] [Accepted: 12/01/2016] [Indexed: 12/17/2022] Open
Abstract
The identification of the modes of action of bioactive compounds is a major challenge in chemical systems biology of diseases. Genome-wide expression profiling of transcriptional responses to compound treatment for human cell lines is a promising unbiased approach for the mode-of-action analysis. Here we developed a novel approach to elucidate the modes of action of bioactive compounds in a cell-specific manner using large-scale chemically-induced transcriptome data acquired from the Library of Integrated Network-based Cellular Signatures (LINCS), and analyzed 16,268 compounds and 68 human cell lines. First, we performed pathway enrichment analyses of regulated genes to reveal active pathways among 163 biological pathways. Next, we explored potential target proteins (including primary targets and off-targets) with cell-specific transcriptional similarity using chemical-protein interactome. Finally, we predicted new therapeutic indications for 461 diseases based on the target proteins. We showed the usefulness of the proposed approach in terms of prediction coverage, interpretation, and large-scale applicability, and validated the new prediction results experimentally by an in vitro cellular assay. The approach has a high potential for advancing drug discovery and repositioning.
Collapse
|
49
|
Cammalleri M, Dal Monte M, Locri F, Marsili S, Lista L, De Rosa M, Pavone V, Rusciano D, Bagnoli P. Diabetic Retinopathy in the Spontaneously Diabetic Torii Rat: Pathogenetic Mechanisms and Preventive Efficacy of Inhibiting the Urokinase-Type Plasminogen Activator Receptor System. J Diabetes Res 2017; 2017:2904150. [PMID: 29464181 PMCID: PMC5804371 DOI: 10.1155/2017/2904150] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/24/2017] [Revised: 10/20/2017] [Accepted: 11/13/2017] [Indexed: 12/26/2022] Open
Abstract
The spontaneously diabetic Torii (SDT) rat is of increasing preclinical interest because of its similarities to human type 2 diabetic retinopathy (DR). The system formed by urokinase-type plasminogen activator (uPA) and its receptor (uPAR) is a player in blood-retinal barrier (BRB) breakdown in DR. Here, we investigated whether in SDT rats, preventive administration of UPARANT, an inhibitor of the uPAR pathway, counteracts the retinal impairment in response to chronic hyperglycemia. Electroretinogram (ERG) monitoring was followed over time. Fluorescein-dextran microscopy, CD31 immunohistochemistry, quantitative PCR, ELISA, Evans blue perfusion, and Western blot were also used. UPARANT prevented ERG dysfunction, upregulation of vascular endothelial growth factor and fibroblast growth factor-2, BRB leakage, gliosis, and retinal cell death. The mechanisms underlying UPARANT benefits were studied comparing them with the acute streptozotocin (STZ) model in which UPARANT is known to inhibit DR signs. In SDT rats, but not in the STZ model, UPARANT downregulated the expression of uPAR and its membrane partners. In both models, UPARANT reduced the levels of transcription factors coupled to inflammation or inflammatory factors themselves. These findings may help to establish the uPAR system as putative target for the development of novel drugs that may prevent type 2 DR.
Collapse
Affiliation(s)
- Maurizio Cammalleri
- Department of Biology, University of Pisa, Via San Zeno 31, 56127 Pisa, Italy
| | - Massimo Dal Monte
- Department of Biology, University of Pisa, Via San Zeno 31, 56127 Pisa, Italy
| | - Filippo Locri
- Department of Biology, University of Pisa, Via San Zeno 31, 56127 Pisa, Italy
| | - Stefania Marsili
- Department of Biology, University of Pisa, Via San Zeno 31, 56127 Pisa, Italy
| | - Liliana Lista
- Department of Biology, University of Napoli Federico II, Complesso Universitario Monte Sant'Angelo, Via Cinthia, Edificio 7, 80126 Napoli, Italy
| | - Mario De Rosa
- Department of Experimental Medicine, Second University of Napoli, Via Santa Maria di Costantinopoli 16, 80138 Napoli, Italy
| | - Vincenzo Pavone
- Department of Biology, University of Napoli Federico II, Complesso Universitario Monte Sant'Angelo, Via Cinthia, Edificio 7, 80126 Napoli, Italy
| | - Dario Rusciano
- Sooft Fidia Pharma, Contrada Molino 17, 63833 Montegiorgio, Italy
| | - Paola Bagnoli
- Department of Biology, University of Pisa, Via San Zeno 31, 56127 Pisa, Italy
| |
Collapse
|
50
|
A Systematic Framework for Drug Repositioning from Integrated Omics and Drug Phenotype Profiles Using Pathway-Drug Network. BIOMED RESEARCH INTERNATIONAL 2016; 2016:7147039. [PMID: 28127549 PMCID: PMC5233404 DOI: 10.1155/2016/7147039] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/17/2016] [Revised: 10/12/2016] [Accepted: 10/20/2016] [Indexed: 12/23/2022]
Abstract
Drug repositioning offers new clinical indications for old drugs. Recently, many computational approaches have been developed to repurpose marketed drugs in human diseases by mining various of biological data including disease expression profiles, pathways, drug phenotype expression profiles, and chemical structure data. However, despite encouraging results, a comprehensive and efficient computational drug repositioning approach is needed that includes the high-level integration of available resources. In this study, we propose a systematic framework employing experimental genomic knowledge and pharmaceutical knowledge to reposition drugs for a specific disease. Specifically, we first obtain experimental genomic knowledge from disease gene expression profiles and pharmaceutical knowledge from drug phenotype expression profiles and construct a pathway-drug network representing a priori known associations between drugs and pathways. To discover promising candidates for drug repositioning, we initialize node labels for the pathway-drug network using identified disease pathways and known drugs associated with the phenotype of interest and perform network propagation in a semisupervised manner. To evaluate our method, we conducted some experiments to reposition 1309 drugs based on four different breast cancer datasets and verified the results of promising candidate drugs for breast cancer by a two-step validation procedure. Consequently, our experimental results showed that the proposed framework is quite useful approach to discover promising candidates for breast cancer treatment.
Collapse
|