1
|
Nguyen HDT, Le TM, Jung DR, Jo Y, Choi Y, Lee D, Lee OE, Cho J, Park NJY, Seo I, Chong GO, Shin JH, Han HS. Transcriptomic analysis reveals Streptococcus agalactiae activation of oncogenic pathways in cervical adenocarcinoma. Oncol Lett 2024; 28:588. [PMID: 39411203 PMCID: PMC11474141 DOI: 10.3892/ol.2024.14720] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Accepted: 09/06/2024] [Indexed: 10/19/2024] Open
Abstract
Cervical adenocarcinoma (AC), a subtype of uterine cervical cancer (CC), poses a challenge due to its resistance to therapy and poor prognosis compared with squamous cervical carcinoma. Streptococcus agalactiae [group B Streptococcus (GBS)], a Gram-positive coccus, has been associated with cervical intraepithelial neoplasia in CC. However, the underlying mechanism interaction between GBS and CC, particularly AC, remains elusive. Leveraging The Cancer Genome Atlas public data and time-series transcriptomic data, the present study investigated the interaction between GBS and AC, revealing activation of two pivotal pathways: 'MAPK signaling pathway' and 'mTORC1 signaling'. Western blotting, reverse transcription-quantitative PCR and cell viability assays were performed to validate the activation of these pathways and their role in promoting cancer cell proliferation. Subsequently, the present study evaluated the efficacy of two anticancer drugs targeting these pathways (binimetinib and ridaforolimus) in AC cell treatment. Binimetinib demonstrated a cytostatic effect, while ridaforolimus had a modest impact on HeLa cells after 48 h of treatment, as observed in both cell viability and cytotoxicity assays. The combination of binimetinib and ridaforolimus resulted in a significantly greater cytotoxic effect compared to binimetinib or ridaforolimus monotherapy, although the synergy score indicated an additive effect. In general, the MAPK and mTORC1 signaling pathways were identified as the main pathways associated with GBS and AC cells. The combination of binimetinib and ridaforolimus could be a potential AC treatment.
Collapse
Affiliation(s)
- Hong Duc Thi Nguyen
- Department of Biomedical Science, Graduate School, Kyungpook National University, Daegu 41944, Republic of Korea
| | - Tan Minh Le
- Department of Biomedical Science, Graduate School, Kyungpook National University, Daegu 41944, Republic of Korea
| | - Da-Ryung Jung
- Department of Applied Biosciences, Kyungpook National University, Daegu 41566, Republic of Korea
| | - Youngjae Jo
- Department of Applied Biosciences, Kyungpook National University, Daegu 41566, Republic of Korea
| | - Yeseul Choi
- Department of Biomedical Science, Graduate School, Kyungpook National University, Daegu 41944, Republic of Korea
| | - Donghyeon Lee
- Department of Biomedical Science, Graduate School, Kyungpook National University, Daegu 41944, Republic of Korea
| | - Olive Em Lee
- Department of Biomedical Science, Graduate School, Kyungpook National University, Daegu 41944, Republic of Korea
| | - Junghwan Cho
- Clinical Omics Institute, Kyungpook National University, Daegu 41405, Republic of Korea
| | - Nora Jee-Young Park
- Clinical Omics Institute, Kyungpook National University, Daegu 41405, Republic of Korea
- Department of Pathology, School of Medicine, Kyungpook National University, Daegu 41944, Republic of Korea
- Department of Pathology, Kyungpook National University Chilgok Hospital, Daegu 41404, Republic of Korea
| | - Incheol Seo
- Department of Immunology, School of Medicine, Kyungpook National University, Daegu 41944, Republic of Korea
| | - Gun Oh Chong
- Clinical Omics Institute, Kyungpook National University, Daegu 41405, Republic of Korea
- Department of Obstetrics and Gynecology, School of Medicine, Kyungpook National University, Daegu 41944, Republic of Korea
- Department of Obstetrics and Gynecology, Kyungpook National University Chilgok Hospital, Daegu 41404, Republic of Korea
| | - Jae-Ho Shin
- Department of Applied Biosciences, Kyungpook National University, Daegu 41566, Republic of Korea
- Department of Integrative Biotechnology, Kyungpook National University, Daegu 41566, Republic of Korea
- Next Generation Sequencing Core Facility, Kyungpook National University, Daegu 41566, Republic of Korea
| | - Hyung Soo Han
- Department of Biomedical Science, Graduate School, Kyungpook National University, Daegu 41944, Republic of Korea
- Clinical Omics Institute, Kyungpook National University, Daegu 41405, Republic of Korea
- Department of Physiology, School of Medicine, Kyungpook National University, Daegu 41944, Republic of Korea
| |
Collapse
|
2
|
Chen F, Jing K, Zhang Z, Liu X. A review on drug repurposing applicable to obesity. Obes Rev 2024:e13848. [PMID: 39384341 DOI: 10.1111/obr.13848] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/04/2023] [Revised: 05/22/2024] [Accepted: 09/19/2024] [Indexed: 10/11/2024]
Abstract
Obesity is a major public health concern and burden on individuals and healthcare systems. Due to the challenges and limitations of lifestyle adjustments, it is advisable to consider pharmacological treatment for people affected by obesity. However, the side effects and limited efficacy of available drugs make the obesity drug market far from sufficient. Drug repurposing involves identifying new applications for existing drugs and offers some advantages over traditional drug development approaches including lower costs and shorter development timelines. This review aims to provide an overview of drug repurposing for anti-obesity medications, including the rationale for repurposing, the challenges and approaches, and the potential drugs that are being investigated for repurposing. Through advanced computational techniques, researchers can unlock the potential of repurposed drugs to tackle the global obesity epidemic. Further research, clinical trials, and collaborative efforts are essential to fully explore and leverage the potential of drug repurposing in the fight against obesity.
Collapse
Affiliation(s)
- Feng Chen
- Department of Clinical Pharmacy, School of Pharmacy, Naval Medical University, Shanghai, China
| | - Kai Jing
- Department of Clinical Pharmacy, School of Pharmacy, Naval Medical University, Shanghai, China
| | - Zhen Zhang
- Department of Clinical Pharmacy, School of Pharmacy, Naval Medical University, Shanghai, China
- Department of Nutrition and Food Hygiene, Faculty of Naval Medicine, Naval Medical University, Shanghai, China
| | - Xia Liu
- Department of Clinical Pharmacy, School of Pharmacy, Naval Medical University, Shanghai, China
| |
Collapse
|
3
|
Gonzalez G, Lin X, Herath I, Veselkov K, Bronstein M, Zitnik M. Combinatorial prediction of therapeutic perturbations using causally-inspired neural networks. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.03.573985. [PMID: 38260532 PMCID: PMC10802439 DOI: 10.1101/2024.01.03.573985] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/24/2024]
Abstract
As an alternative to target-driven drug discovery, phenotype-driven approaches identify compounds that counteract the overall disease effects by analyzing phenotypic signatures. Our study introduces a novel approach to this field, aiming to expand the search space for new therapeutic agents. We introduce PDGrapher, a causally-inspired graph neural network (GNN) designed to predict combinatorial perturbagens - sets of therapeutic targets - capable of reversing disease effects. Unlike methods that learn responses to perturbations, PDGrapher solves the inverse problem, which is to infer the perturbagens necessary to achieve a specific response - i.e., directly predicting perturbagens by learning which perturbations elicit a desired response. By encoding gene regulatory networks or protein-protein interactions, PDGrapher can predict unseen chemical or genetic perturbagens, aiding in the discovery of novel drugs or therapeutic targets. Experiments across nine cell lines with chemical perturbations show that PDGrapher successfully predicted effective perturbagens in up to 13.33% additional test samples and ranked therapeutic targets up to 35% higher than the competing methods, and the method shows competitive performance across ten genetic perturbation datasets. A key innovation of PDGrapher is its direct prediction capability, which contrasts with the indirect, computationally intensive models traditionally used in phenotype-driven drug discovery that only predict changes in phenotypes due to perturbations. The direct approach enables PDGrapher to train up to 25 times faster than methods like scGEN and CellOT, representing a considerable leap in efficiency. Our results suggest that PDGrapher can advance phenotype-driven drug discovery, offering a fast and comprehensive approach to identifying therapeutically useful perturbations.
Collapse
Affiliation(s)
- Guadalupe Gonzalez
- Imperial College London, London, UK
- Prescient Design, Genentech, South San Francisco, CA, USA
- F. Hoffmann-La Roche Ltd, Basel, Switzerland
| | - Xiang Lin
- Harvard Medical School, Boston, MA, USA
| | - Isuru Herath
- Merck & Co., South San Francisco, CA, USA
- Cornell University, Ithaca, NY, USA
| | | | | | - Marinka Zitnik
- Harvard Medical School, Boston, MA, USA
- Kempner Institute for the Study of Natural and Artificial Intelligence, Harvard University, Cambridge, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Harvard Data Science Initiative, Cambridge, MA, USA
| |
Collapse
|
4
|
Zhang J, Yuan S, Cao W, Jiang X, Yang C, Jiang C, Liu R, Yang W, Tian S. Signature Search Polestar: a comprehensive drug repurposing method evaluation assistant for customized oncogenic signature. BIOINFORMATICS (OXFORD, ENGLAND) 2024; 40:btae536. [PMID: 39213324 PMCID: PMC11398873 DOI: 10.1093/bioinformatics/btae536] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/04/2023] [Revised: 08/20/2024] [Accepted: 08/29/2024] [Indexed: 09/04/2024]
Abstract
SUMMARY The burgeoning high-throughput technologies have led to a significant surge in the scale of pharmacotranscriptomic datasets, especially for oncology. Signature search methods (SSMs), utilizing oncogenic signatures formed by differentially expressed genes through sequencing, have been instrumental in anti-cancer drug screening and identifying mechanisms of action without relying on prior knowledge. However, various studies have found that different SSMs exhibit varying performance across pharmacotranscriptomic datasets. In addition, the size of the oncogenic signature can also significantly impact the result of drug repurposing. Therefore, finding the optimal SSMs and customized oncogenic signature for a specific disease remains a challenge. To address this, we introduce Signature Search Polestar (SSP), a webserver integrating the largest pharmacotranscriptomic datasets of anti-cancer drugs from LINCS L1000 with five state-of-the-art SSMs (XSum, CMap, GSEA, ZhangScore, XCos). SSP provides three main modules: Benchmark, Robustness, and Application. Benchmark uses two indices, Area Under the Curve and Enrichment Score, based on drug annotations to evaluate SSMs at different oncogenic signature sizes. Robustness, applicable when drug annotations are insufficient, uses a performance score based on drug self-retrieval for evaluation. Application provides three screening strategies, single method, SS_all, and SS_cross, allowing users to freely utilize optimal SSMs with tailored oncogenic signature for drug repurposing. AVAILABILITY AND IMPLEMENTATION SSP is free at https://web.biotcm.net/SSP/. The current version of SSP is archived in https://doi.org/10.6084/m9.figshare.26524741.v1, allowing users to directly use or customize their own SSP webserver.
Collapse
Affiliation(s)
- Jinbo Zhang
- Department of Phytochemistry, School of Pharmacy, Second Military Medical University, Shanghai 200433, China
- Department of Pharmacy, Tianjin Rehabilitation Center of Joint Logistics Support Force, Tianjin 300110, China
| | - Shunling Yuan
- Department of Phytochemistry, School of Pharmacy, Second Military Medical University, Shanghai 200433, China
| | - Wen Cao
- Department of Phytochemistry, School of Pharmacy, Second Military Medical University, Shanghai 200433, China
| | - Xianrui Jiang
- Changhai Hospital, Second Military Medical University, Shanghai 200433, China
| | - Cheng Yang
- Department of Clinical Nutrition, Tianjin Rehabilitation Center of Joint Logistics Support Force, Tianjin 300110, China
| | - Chenchao Jiang
- Department of Pharmacy, Tianjin Rehabilitation Center of Joint Logistics Support Force, Tianjin 300110, China
| | - Runhui Liu
- Department of Phytochemistry, School of Pharmacy, Second Military Medical University, Shanghai 200433, China
| | - Wei Yang
- Department of Pharmacy, Tianjin Rehabilitation Center of Joint Logistics Support Force, Tianjin 300110, China
| | - Saisai Tian
- Department of Phytochemistry, School of Pharmacy, Second Military Medical University, Shanghai 200433, China
- Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Yantai University, Yantai 264005, China
| |
Collapse
|
5
|
Larsson P, De Rosa MC, Righino B, Olsson M, Florea BI, Forssell-Aronsson E, Kovács A, Karlsson P, Helou K, Parris TZ. Integrated transcriptomics- and structure-based drug repositioning identifies drugs with proteasome inhibitor properties. Sci Rep 2024; 14:18772. [PMID: 39138277 PMCID: PMC11322189 DOI: 10.1038/s41598-024-69465-6] [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: 12/23/2022] [Accepted: 08/05/2024] [Indexed: 08/15/2024] Open
Abstract
Computational pharmacogenomics can potentially identify new indications for already approved drugs and pinpoint compounds with similar mechanism-of-action. Here, we used an integrated drug repositioning approach based on transcriptomics data and structure-based virtual screening to identify compounds with gene signatures similar to three known proteasome inhibitors (PIs; bortezomib, MG-132, and MLN-2238). In vitro validation of candidate compounds was then performed to assess proteasomal proteolytic activity, accumulation of ubiquitinated proteins, cell viability, and drug-induced expression in A375 melanoma and MCF7 breast cancer cells. Using this approach, we identified six compounds with PI properties ((-)-kinetin-riboside, manumycin-A, puromycin dihydrochloride, resistomycin, tegaserod maleate, and thapsigargin). Although the docking scores pinpointed their ability to bind to the β5 subunit, our in vitro study revealed that these compounds inhibited the β1, β2, and β5 catalytic sites to some extent. As shown with bortezomib, only manumycin-A, puromycin dihydrochloride, and tegaserod maleate resulted in excessive accumulation of ubiquitinated proteins and elevated HMOX1 expression. Taken together, our integrated drug repositioning approach and subsequent in vitro validation studies identified six compounds demonstrating properties similar to proteasome inhibitors.
Collapse
Affiliation(s)
- Peter Larsson
- Department of Oncology, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden.
- Sahlgrenska Center for Cancer Research, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden.
| | - Maria Cristina De Rosa
- Institute of Chemical Sciences and Technologies "Giulio Natta" (SCITEC)-CNR, Rome, Italy
| | - Benedetta Righino
- Institute of Chemical Sciences and Technologies "Giulio Natta" (SCITEC)-CNR, Rome, Italy
| | - Maxim Olsson
- Department of Oncology, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Sahlgrenska Center for Cancer Research, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Bogdan Iulius Florea
- Gorlaeus Laboratories, Leiden Institute of Chemistry and Netherlands Proteomics Center, Leiden, The Netherlands
| | - Eva Forssell-Aronsson
- Sahlgrenska Center for Cancer Research, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Department of Medical Radiation Sciences, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Department of Medical Physics and Biomedical Engineering, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Anikó Kovács
- Department of Clinical Pathology, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Per Karlsson
- Department of Oncology, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Department of Oncology, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Khalil Helou
- Department of Oncology, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Sahlgrenska Center for Cancer Research, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Toshima Z Parris
- Department of Oncology, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Sahlgrenska Center for Cancer Research, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| |
Collapse
|
6
|
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
|
7
|
Kang H, Pan S, Lin S, Wang YY, Yuan N, Jia P. PharmGWAS: a GWAS-based knowledgebase for drug repurposing. Nucleic Acids Res 2024; 52:D972-D979. [PMID: 37831083 PMCID: PMC10767932 DOI: 10.1093/nar/gkad832] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Revised: 09/12/2023] [Accepted: 09/21/2023] [Indexed: 10/14/2023] Open
Abstract
Leveraging genetics insights to promote drug repurposing has become a promising and active strategy in pharmacology. Indeed, among the 50 drugs approved by FDA in 2021, two-thirds have genetically supported evidence. In this regard, the increasing amount of widely available genome-wide association studies (GWAS) datasets have provided substantial opportunities for drug repurposing based on genetics discoveries. Here, we developed PharmGWAS, a comprehensive knowledgebase designed to identify candidate drugs through the integration of GWAS data. PharmGWAS focuses on novel connections between diseases and small-molecule compounds derived using a reverse relationship between the genetically-regulated expression signature and the drug-induced signature. Specifically, we collected and processed 1929 GWAS datasets across a diverse spectrum of diseases and 724 485 perturbation signatures pertaining to a substantial 33609 molecular compounds. To obtain reliable and robust predictions for the reverse connections, we implemented six distinct connectivity methods. In the current version, PharmGWAS deposits a total of 740 227 genetically-informed disease-drug pairs derived from drug-perturbation signatures, presenting a valuable and comprehensive catalog. Further equipped with its user-friendly web design, PharmGWAS is expected to greatly aid the discovery of novel drugs, the exploration of drug combination therapies and the identification of drug resistance or side effects. PharmGWAS is available at https://ngdc.cncb.ac.cn/pharmgwas.
Collapse
Affiliation(s)
- Hongen Kang
- CAS Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences and China National Center for Bioinformation, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Siyu Pan
- CAS Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences and China National Center for Bioinformation, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Shiqi Lin
- CAS Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences and China National Center for Bioinformation, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yin-Ying Wang
- CAS Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences and China National Center for Bioinformation, Beijing 100101, China
| | - Na Yuan
- CAS Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences and China National Center for Bioinformation, Beijing 100101, China
| | - Peilin Jia
- CAS Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences and China National Center for Bioinformation, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100049, China
- National Genomics Data Center, Beijing Institute of Genomics, Chinese Academy of Sciences and China National Center for Bioinformation, Beijing 100101, China
| |
Collapse
|
8
|
Sun J, Xu M, Ru J, James-Bott A, Xiong D, Wang X, Cribbs AP. Small molecule-mediated targeting of microRNAs for drug discovery: Experiments, computational techniques, and disease implications. Eur J Med Chem 2023; 257:115500. [PMID: 37262996 PMCID: PMC11554572 DOI: 10.1016/j.ejmech.2023.115500] [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: 03/28/2023] [Revised: 05/05/2023] [Accepted: 05/15/2023] [Indexed: 06/03/2023]
Abstract
Small molecules have been providing medical breakthroughs for human diseases for more than a century. Recently, identifying small molecule inhibitors that target microRNAs (miRNAs) has gained importance, despite the challenges posed by labour-intensive screening experiments and the significant efforts required for medicinal chemistry optimization. Numerous experimentally-verified cases have demonstrated the potential of miRNA-targeted small molecule inhibitors for disease treatment. This new approach is grounded in their posttranscriptional regulation of the expression of disease-associated genes. Reversing dysregulated gene expression using this mechanism may help control dysfunctional pathways. Furthermore, the ongoing improvement of algorithms has allowed for the integration of computational strategies built on top of laboratory-based data, facilitating a more precise and rational design and discovery of lead compounds. To complement the use of extensive pharmacogenomics data in prioritising potential drugs, our previous work introduced a computational approach based on only molecular sequences. Moreover, various computational tools for predicting molecular interactions in biological networks using similarity-based inference techniques have been accumulated in established studies. However, there are a limited number of comprehensive reviews covering both computational and experimental drug discovery processes. In this review, we outline a cohesive overview of both biological and computational applications in miRNA-targeted drug discovery, along with their disease implications and clinical significance. Finally, utilizing drug-target interaction (DTIs) data from DrugBank, we showcase the effectiveness of deep learning for obtaining the physicochemical characterization of DTIs.
Collapse
Affiliation(s)
- Jianfeng Sun
- Botnar Research Centre, Nuffield Department of Orthopedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Oxford, OX3 7LD, UK.
| | - Miaoer Xu
- Department of Biology, Emory University, Atlanta, GA, 30322, USA
| | - Jinlong Ru
- Chair of Prevention of Microbial Diseases, School of Life Sciences Weihenstephan, Technical University of Munich, Freising, 85354, Germany
| | - Anna James-Bott
- Botnar Research Centre, Nuffield Department of Orthopedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Oxford, OX3 7LD, UK
| | - Dapeng Xiong
- Department of Computational Biology, Cornell University, Ithaca, NY, 14853, USA; Weill Institute for Cell and Molecular Biology, Cornell University, Ithaca, NY, 14853, USA
| | - Xia Wang
- College of Animal Science and Technology, Northwest A&F University, Yangling, 712100, China.
| | - Adam P Cribbs
- Botnar Research Centre, Nuffield Department of Orthopedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Oxford, OX3 7LD, UK.
| |
Collapse
|
9
|
Sun J, Si S, Ru J, Wang X. DeepdlncUD: Predicting regulation types of small molecule inhibitors on modulating lncRNA expression by deep learning. Comput Biol Med 2023; 163:107226. [PMID: 37450966 DOI: 10.1016/j.compbiomed.2023.107226] [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: 03/17/2023] [Revised: 05/31/2023] [Accepted: 07/01/2023] [Indexed: 07/18/2023]
Abstract
Targeting lncRNAs by small molecules (SM-lncR) to alter their expression levels has emerged as an important therapeutic modality for disease treatment. To date, no computational tools have been dedicated to predicting small molecule-mediated upregulation or downregulation of lncRNA expression. Here, we introduce DeepdlncUD, which integrates predictions of nine deep learning algorithms together, to infer the regulation types of small molecules on modulating lncRNA expression. Through systematic optimization on a training set of 771 upregulation and 739 downregulation SM-lncR pairs, each encoding 1369 sequence, representational, and physiochemical features, this method outperforms a recently released program, DeepsmirUD, by achieving 0.674 in AUC (area under the receiver operating characteristic curve), 0.722 in AUCPR (area under the precision-recall curve), 0.681 in F1-score, and 0.516 in Jaccard Index on a test set of 222 SM-lncR pairs. By extracting 125 upregulation and 46 downregulation SM-lncR pairs that involve disease-associated lncRNAs, DeepdlncUD is shown to gain an accuracy of 0.700 in the pathological context. Using connectivity scores, around half of the small molecules are correctly estimated as drugs to treat lncRNA-regulated diseases. This tool can be run at a fast speed to assist the discovery of potential small molecule drugs of lncRNA targets on a large scale. DeepdlncUD is publicly available at https://github.com/2003100127/deepdlncud.
Collapse
Affiliation(s)
- Jianfeng Sun
- Botnar Research Centre, University of Oxford, Headington, Oxford, OX3 7LD, UK.
| | - Shuyue Si
- School of Mathematics and Physics, Xi'an Jiaotong-liverpool University, Renai, Suzhou, 215028, China
| | - Jinlong Ru
- Chair of Prevention of Microbial Diseases, School of Life Sciences Weihenstephan, Technical University of Munich, 85354, Freising, Germany
| | - Xia Wang
- College of Animal Science and Technology, Northwest A&F University, Yangling, 712100, China.
| |
Collapse
|
10
|
Sapashnik D, Newman R, Pietras CM, Zhou D, Devkota K, Qu F, Kofman L, Boudreau S, Fried I, Slonim DK. Cell-specific imputation of drug connectivity mapping with incomplete data. PLoS One 2023; 18:e0278289. [PMID: 36795645 PMCID: PMC9934325 DOI: 10.1371/journal.pone.0278289] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Accepted: 11/15/2022] [Indexed: 02/17/2023] Open
Abstract
Drug repositioning allows expedited discovery of new applications for existing compounds, but re-screening vast compound libraries is often prohibitively expensive. "Connectivity mapping" is a process that links drugs to diseases by identifying compounds whose impact on expression in a collection of cells reverses the disease's impact on expression in disease-relevant tissues. The LINCS project has expanded the universe of compounds and cells for which data are available, but even with this effort, many clinically useful combinations are missing. To evaluate the possibility of repurposing drugs despite missing data, we compared collaborative filtering using either neighborhood-based or SVD imputation methods to two naive approaches via cross-validation. Methods were evaluated for their ability to predict drug connectivity despite missing data. Predictions improved when cell type was taken into account. Neighborhood collaborative filtering was the most successful method, with the best improvements in non-immortalized primary cells. We also explored which classes of compounds are most and least reliant on cell type for accurate imputation. We conclude that even for cells in which drug responses have not been fully characterized, it is possible to identify unassayed drugs that reverse in those cells the expression signatures observed in disease.
Collapse
Affiliation(s)
- Diana Sapashnik
- Department of Computer Science, Tufts University, Medford, MA, United States of America
| | - Rebecca Newman
- Department of Computer Science, Tufts University, Medford, MA, United States of America
| | | | - Di Zhou
- Department of Computer Science, Tufts University, Medford, MA, United States of America
| | - Kapil Devkota
- Department of Computer Science, Tufts University, Medford, MA, United States of America
| | - Fangfang Qu
- Department of Computer Science, Tufts University, Medford, MA, United States of America
| | - Lior Kofman
- Department of Computer Science, Tufts University, Medford, MA, United States of America
| | - Sean Boudreau
- Department of Computer Science, Tufts University, Medford, MA, United States of America
| | - Inbar Fried
- Department of Medicine, University of North Carolina School of Medicine, Chapel Hill, NC, United States of America
| | - Donna K. Slonim
- Department of Computer Science, Tufts University, Medford, MA, United States of America
- Department of Immunology, Tufts University School of Medicine, Boston, MA, United States of America
- * E-mail:
| |
Collapse
|
11
|
Sun J, Ru J, Ramos-Mucci L, Qi F, Chen Z, Chen S, Cribbs AP, Deng L, Wang X. DeepsmirUD: Prediction of Regulatory Effects on microRNA Expression Mediated by Small Molecules Using Deep Learning. Int J Mol Sci 2023; 24:1878. [PMID: 36768205 PMCID: PMC9915273 DOI: 10.3390/ijms24031878] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Revised: 12/26/2022] [Accepted: 01/12/2023] [Indexed: 01/21/2023] Open
Abstract
Aberrant miRNA expression has been associated with a large number of human diseases. Therefore, targeting miRNAs to regulate their expression levels has become an important therapy against diseases that stem from the dysfunction of pathways regulated by miRNAs. In recent years, small molecules have demonstrated enormous potential as drugs to regulate miRNA expression (i.e., SM-miR). A clear understanding of the mechanism of action of small molecules on the upregulation and downregulation of miRNA expression allows precise diagnosis and treatment of oncogenic pathways. However, outside of a slow and costly process of experimental determination, computational strategies to assist this on an ad hoc basis have yet to be formulated. In this work, we developed, to the best of our knowledge, the first cross-platform prediction tool, DeepsmirUD, to infer small-molecule-mediated regulatory effects on miRNA expression (i.e., upregulation or downregulation). This method is powered by 12 cutting-edge deep-learning frameworks and achieved AUC values of 0.843/0.984 and AUCPR values of 0.866/0.992 on two independent test datasets. With a complementarily constructed network inference approach based on similarity, we report a significantly improved accuracy of 0.813 in determining the regulatory effects of nearly 650 associated SM-miR relations, each formed with either novel small molecule or novel miRNA. By further integrating miRNA-cancer relationships, we established a database of potential pharmaceutical drugs from 1343 small molecules for 107 cancer diseases to understand the drug mechanisms of action and offer novel insight into drug repositioning. Furthermore, we have employed DeepsmirUD to predict the regulatory effects of a large number of high-confidence associated SM-miR relations. Taken together, our method shows promise to accelerate the development of potential miRNA targets and small molecule drugs.
Collapse
Affiliation(s)
- Jianfeng Sun
- College of Animal Science and Technology, Northwest A&F University, Yangling 712100, China
- Botnar Research Centre, Nuffield Department of Orthopedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Oxford OX3 7LD, UK
| | - Jinlong Ru
- Institute of Virology, Helmholtz Centre Munich—German Research Center for Environmental Health, 85764 Neuherberg, Germany
- Chair of Prevention of Microbial Diseases, School of Life Sciences Weihenstephan, Technical University of Munich, 85354 Freising, Germany
| | - Lorenzo Ramos-Mucci
- Botnar Research Centre, Nuffield Department of Orthopedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Oxford OX3 7LD, UK
| | - Fei Qi
- Institute of Genomics, School of Medicine, Huaqiao University, Xiamen 362021, China
| | - Zihao Chen
- Department of Computational Biology for Drug Discovery, Biolife Biotechnology Ltd., Zhumadian 463200, China
| | - Suyuan Chen
- Leibniz-Institut für Analytische Wissenschaften–ISAS–e.V., Otto-Hahn-Str asse 6b, 44227 Dortmund, Germany
| | - Adam P. Cribbs
- Botnar Research Centre, Nuffield Department of Orthopedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Oxford OX3 7LD, UK
| | - Li Deng
- Institute of Virology, Helmholtz Centre Munich—German Research Center for Environmental Health, 85764 Neuherberg, Germany
- Chair of Prevention of Microbial Diseases, School of Life Sciences Weihenstephan, Technical University of Munich, 85354 Freising, Germany
| | - Xia Wang
- College of Animal Science and Technology, Northwest A&F University, Yangling 712100, China
- Department of Molecular and Cellular Biology, University of Arizona, Tucson, AZ 85721, USA
| |
Collapse
|
12
|
Looi CK, Gan LL, Sim W, Hii LW, Chung FFL, Leong CO, Lim WM, Mai CW. Histone Deacetylase Inhibitors Restore Cancer Cell Sensitivity towards T Lymphocytes Mediated Cytotoxicity in Pancreatic Cancer. Cancers (Basel) 2022; 14:3709. [PMID: 35954379 PMCID: PMC9367398 DOI: 10.3390/cancers14153709] [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: 06/16/2022] [Revised: 07/27/2022] [Accepted: 07/28/2022] [Indexed: 02/04/2023] Open
Abstract
Despite medical advancements, the prognosis of pancreatic ductal adenocarcinoma (PDAC) has not improved significantly over the past 50 years. By utilising the large-scale genomic datasets available from the Australia Pancreatic Cancer Project (PACA-AU) and The Cancer Genomic Atlas Project (TCGA-PAAD), we studied the immunophenotype of PDAC in silico and identified that tumours with high cytotoxic T lymphocytes (CTL) killing activity were associated with favourable clinical outcomes. Using the STRING protein-protein interaction network analysis, the identified differentially expressed genes with low CTL killing activity were associated with TWIST/IL-6R, HDAC5, and EOMES signalling. Following Connectivity Map analysis, we identified 44 small molecules that could restore CTL sensitivity in the PDAC cells. Further high-throughput chemical library screening identified 133 inhibitors that effectively target both parental and CTL-resistant PDAC cells in vitro. Since CTL-resistant PDAC had a higher expression of histone proteins and its acetylated proteins compared to its parental cells, we further investigated the impact of histone deacetylase inhibitors (HDACi) on CTL-mediated cytotoxicity in PDAC cells in vitro, namely SW1990 and BxPC3. Further analyses revealed that givinostat and dacinostat were the two most potent HDAC inhibitors that restored CTL sensitivity in SW1990 and BxPC3 CTL-resistant cells. Through our in silico and in vitro studies, we demonstrate the novel role of HDAC inhibition in restoring CTL resistance and that combinations of HDACi with CTL may represent a promising therapeutic strategy, warranting its further detailed molecular mechanistic studies and animal studies before embarking on the clinical evaluation of these novel combined PDAC treatments.
Collapse
Affiliation(s)
- Chin-King Looi
- School of Postgraduate Studies, International Medical University, Kuala Lumpur 57000, Malaysia; (C.-K.L.); (L.-L.G.)
| | - Li-Lian Gan
- School of Postgraduate Studies, International Medical University, Kuala Lumpur 57000, Malaysia; (C.-K.L.); (L.-L.G.)
- Clinical Research Centre, Hospital Tuanku Ja’afar Seremban, Ministry of Health Malaysia, Seremban 70300, Malaysia
| | - Wynne Sim
- School of Medicine, International Medical University, Kuala Lumpur 57000, Malaysia;
| | - Ling-Wei Hii
- Center for Cancer and Stem Cell Research, Development and Innovation (IRDI), Institute for Research, International Medical University, Kuala Lumpur 57000, Malaysia; (L.-W.H.); (C.-O.L.); (W.-M.L.)
- School of Pharmacy, International Medical University, Kuala Lumpur 57000, Malaysia
| | - Felicia Fei-Lei Chung
- Department of Medical Sciences, School of Medical and Life Sciences, Sunway University, Subang Jaya 47500, Malaysia;
| | - Chee-Onn Leong
- Center for Cancer and Stem Cell Research, Development and Innovation (IRDI), Institute for Research, International Medical University, Kuala Lumpur 57000, Malaysia; (L.-W.H.); (C.-O.L.); (W.-M.L.)
- School of Pharmacy, International Medical University, Kuala Lumpur 57000, Malaysia
- AGTC Genomics, Kuala Lumpur 57000, Malaysia
| | - Wei-Meng Lim
- Center for Cancer and Stem Cell Research, Development and Innovation (IRDI), Institute for Research, International Medical University, Kuala Lumpur 57000, Malaysia; (L.-W.H.); (C.-O.L.); (W.-M.L.)
- School of Pharmacy, International Medical University, Kuala Lumpur 57000, Malaysia
- School of Pharmacy, Monash University Malaysia, Subang Jaya 47500, Malaysia
| | - Chun-Wai Mai
- Center for Cancer and Stem Cell Research, Development and Innovation (IRDI), Institute for Research, International Medical University, Kuala Lumpur 57000, Malaysia; (L.-W.H.); (C.-O.L.); (W.-M.L.)
- School of Pharmacy, International Medical University, Kuala Lumpur 57000, Malaysia
- State Key Laboratory of Oncogenes and Related Genes, Renji-Med X Clinical Stem Cell Research Center, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Pudong New District, Shanghai 200127, China
| |
Collapse
|
13
|
Domingo-Fernández D, Gadiya Y, Patel A, Mubeen S, Rivas-Barragan D, Diana CW, Misra BB, Healey D, Rokicki J, Colluru V. Causal reasoning over knowledge graphs leveraging drug-perturbed and disease-specific transcriptomic signatures for drug discovery. PLoS Comput Biol 2022; 18:e1009909. [PMID: 35213534 PMCID: PMC8906585 DOI: 10.1371/journal.pcbi.1009909] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Revised: 03/09/2022] [Accepted: 02/09/2022] [Indexed: 12/29/2022] Open
Abstract
Network-based approaches are becoming increasingly popular for drug discovery as they provide a systems-level overview of the mechanisms underlying disease pathophysiology. They have demonstrated significant early promise over other methods of biological data representation, such as in target discovery, side effect prediction and drug repurposing. In parallel, an explosion of -omics data for the deep characterization of biological systems routinely uncovers molecular signatures of disease for similar applications. Here, we present RPath, a novel algorithm that prioritizes drugs for a given disease by reasoning over causal paths in a knowledge graph (KG), guided by both drug-perturbed as well as disease-specific transcriptomic signatures. First, our approach identifies the causal paths that connect a drug to a particular disease. Next, it reasons over these paths to identify those that correlate with the transcriptional signatures observed in a drug-perturbation experiment, and anti-correlate to signatures observed in the disease of interest. The paths which match this signature profile are then proposed to represent the mechanism of action of the drug. We demonstrate how RPath consistently prioritizes clinically investigated drug-disease pairs on multiple datasets and KGs, achieving better performance over other similar methodologies. Furthermore, we present two case studies showing how one can deconvolute the predictions made by RPath as well as predict novel targets.
Collapse
Affiliation(s)
| | - Yojana Gadiya
- Enveda Biosciences, Boulder, Colorado, United States of America
| | - Abhishek Patel
- Enveda Biosciences, Boulder, Colorado, United States of America
| | - Sarah Mubeen
- Bonn-Aachen International Center for IT, Rheinische Friedrich-Wilhelms-Universität Bonn, Bonn, Germany
| | | | - Chris W. Diana
- Enveda Biosciences, Boulder, Colorado, United States of America
| | | | - David Healey
- Enveda Biosciences, Boulder, Colorado, United States of America
| | - Joe Rokicki
- Enveda Biosciences, Boulder, Colorado, United States of America
| | - Viswa Colluru
- Enveda Biosciences, Boulder, Colorado, United States of America
| |
Collapse
|
14
|
Li S, Li L, Meng X, Sun P, Liu Y, Song Y, Zhang S, Jiang C, Cai J, Zhao Z. DREAM: a database of experimentally supported protein-coding RNAs and drug associations in human cancer. Mol Cancer 2021; 20:148. [PMID: 34774049 PMCID: PMC8590212 DOI: 10.1186/s12943-021-01436-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Accepted: 09/22/2021] [Indexed: 11/10/2022] Open
Abstract
The Drug Response Gene Expression Associated Map, also referred as “DREAM” (http://bio-big-data.cn:8080/DREAM), is a manually curated database of experimentally supported protein-coding RNAs and drugs associations in human cancers. The current version of the DREAM documents 3048 entries about scientific literatures supported drug sensitivity or drug intervention related protein-coding RNAs from PubMed database and 195 high-throughput microarray data about drug sensitivity or drug intervention related protein-coding RNAs data from GEO database. Each entry in DREAM database contains detailed information on protein-coding RNA, drug, cancer, and other information including title, PubMed ID, journal, publish time. The DREAM database also provides some data visualization and online analysis services such as volcano plot, GO/KEGG enrichment function analysis, and novel drug discovery analysis. We hope the DREAM database should serve as a valuable resource for clinical practice and basic research, which could help researchers better understand the effects of protein-coding RNAs on drug response in human cancers.
Collapse
Affiliation(s)
- Shupeng Li
- Department of Neurosurgery, the Second Affiliated Hospital of Harbin Medical University, Neuroscience Institute, Heilongjiang Academy of Medical Sciences, Harbin, 150086, China
| | - Lulu Li
- Department of Neurosurgery, the Second Affiliated Hospital of Harbin Medical University, Neuroscience Institute, Heilongjiang Academy of Medical Sciences, Harbin, 150086, China
| | - Xiangqi Meng
- Department of Neurosurgery, the Second Affiliated Hospital of Harbin Medical University, Neuroscience Institute, Heilongjiang Academy of Medical Sciences, Harbin, 150086, China
| | - Penggang Sun
- Department of Neurosurgery, the Second Affiliated Hospital of Harbin Medical University, Neuroscience Institute, Heilongjiang Academy of Medical Sciences, Harbin, 150086, China
| | - Yi Liu
- Department of Neurosurgery, the Second Affiliated Hospital of Harbin Medical University, Neuroscience Institute, Heilongjiang Academy of Medical Sciences, Harbin, 150086, China
| | - Yuntang Song
- Department of Neurosurgery, the Second Affiliated Hospital of Harbin Medical University, Neuroscience Institute, Heilongjiang Academy of Medical Sciences, Harbin, 150086, China
| | - Sijia Zhang
- Department of Educational Administration, the Second Affiliated Hospital of Harbin Medical University, Harbin, 150086, China
| | - Chuanlu Jiang
- Department of Neurosurgery, the Second Affiliated Hospital of Harbin Medical University, Neuroscience Institute, Heilongjiang Academy of Medical Sciences, Harbin, 150086, China.
| | - Jinquan Cai
- Department of Neurosurgery, the Second Affiliated Hospital of Harbin Medical University, Neuroscience Institute, Heilongjiang Academy of Medical Sciences, Harbin, 150086, China. .,Department of Microbiology, Tumor and Cell Biology (MTC), Biomedicum, Karolinska Institutet, 171 65, Stockholm, Sweden.
| | - Zheng Zhao
- Beijing Neurosurgical Institute, Capital Medical University, Beijing, 100070, China.
| |
Collapse
|