1
|
Lubaba F, George M, Ahmed M, John L, Goplakrishnan AP, Shivamurthy PB, Varghese S, Pahal P, Nisar M, Ramesh P, Madar IH, Raju R. Theranostic Target NSUN2, a C(5)-Methyltransferase, Phospho-Regulatory Network Uncovered with Systematic Assembly of 805 Datasets. OMICS : A JOURNAL OF INTEGRATIVE BIOLOGY 2025. [PMID: 40126188 DOI: 10.1089/omi.2025.0025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/25/2025]
Abstract
The RNA cytosine C(5)-methyltransferase NSUN2 is involved in RNA modification and regulates gene expression and genomic stability. Beyond multiple sequence/copy number variations, NSUN2 displays altered phosphoprotein expression in various cancers and developmental disorders, thereby making it a prime molecular target of relevance to both therapeutics and diagnostics, that is, theranostics. Despite its key role in kinase-regulated pathways and broader biological processes, the phospho-regulatory network of NSUN2 remains largely unexplored. We report here a systematic assembly of 805 phosphoproteomics datasets from the literature, among which 239 datasets showed differential regulation of NSUN2 phosphopeptides and 40 ensembled phosphosites in NSUN2. Significantly, the phosphorylation sites Ser456, Ser743, and Ser751 represented NSUN2 in ∼50% of these datasets. This is notable given that the functional roles of these phosphosites have been previously underappreciated and underrepresented in the scientific literature. Therefore, we implemented a codetection/coregulation approach based on the phosphosites in other proteins that are codifferentially regulated with phosphopeptides of NSUN2. This approach led to our identification of 55 interactors, 4 potential kinases, and 7 other methylases whose phosphopeptides were codifferentially regulated with NSUN2 phosphopeptides. To the best of our knowledge, this study provides the first phosphosite-centric regulatory network model of NSUN2 to employ theranostic strategies for targeting NSUN2 in cancers and other disorders.
Collapse
Affiliation(s)
- Fathimathul Lubaba
- Centre for Integrative Omics Data Science (CIODS), Yenepoya (Deemed to be University), Mangalore, India
| | - Mejo George
- Centre for Integrative Omics Data Science (CIODS), Yenepoya (Deemed to be University), Mangalore, India
| | - Mukhtar Ahmed
- Department of Zoology, College of Science, King Saud University, Riyadh, Saudi Arabia
| | - Levin John
- Centre for Integrative Omics Data Science (CIODS), Yenepoya (Deemed to be University), Mangalore, India
| | | | | | - Susmi Varghese
- Centre for Integrative Omics Data Science (CIODS), Yenepoya (Deemed to be University), Mangalore, India
| | - Priyanka Pahal
- Centre for Integrative Omics Data Science (CIODS), Yenepoya (Deemed to be University), Mangalore, India
| | - Mahammad Nisar
- Centre for Integrative Omics Data Science (CIODS), Yenepoya (Deemed to be University), Mangalore, India
| | - Poornima Ramesh
- Centre for Integrative Omics Data Science (CIODS), Yenepoya (Deemed to be University), Mangalore, India
| | - Inamul Hasan Madar
- Centre for Integrative Omics Data Science (CIODS), Yenepoya (Deemed to be University), Mangalore, India
| | - Rajesh Raju
- Centre for Integrative Omics Data Science (CIODS), Yenepoya (Deemed to be University), Mangalore, India
| |
Collapse
|
2
|
Dey L, Chakraborty S. Supervised learning approaches for predicting Ebola-Human Protein-Protein interactions. Gene 2025; 942:149228. [PMID: 39828063 DOI: 10.1016/j.gene.2025.149228] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2024] [Revised: 12/04/2024] [Accepted: 01/07/2025] [Indexed: 01/22/2025]
Abstract
The goal of this research work is to predict protein-protein interactions (PPIs) between the Ebola virus and the host who is at risk of infection. Since there are very limited databases available on the Ebola virus; we have prepared a comprehensive database of all the PPIs between the Ebola virus and human proteins (EbolaInt). Our work focuses on the finding of some new protein-protein interactions between humans and the Ebola virus using some state- of-the-arts machine learning techniques. However, it is basically a two-class problem with a positive interacting dataset and a negative non-interacting dataset. These datasets contain various sequence-based human protein features such as structure of amino acid and conjoint triad and domain-related features. In this research, we have briefly discussed and used some well-known supervised learning approaches to predict PPIs between human proteins and Ebola virus proteins, including K-nearest neighbours (KNN), random forest (RF), support vector machine (SVM), and deep feed-forward multi-layer perceptron (DMLP) etc. We have validated our prediction results using gene ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analysis. Our goal with this prediction is to compare all other models' accuracy, precision, recall, and f1-score for predicting these PPIs. In the result section, DMLP is giving the highest accuracy along with the prediction of 2655 potential human target proteins.
Collapse
Affiliation(s)
- Lopamudra Dey
- Department of Biomedical and Clinical Sciences, Linköping University, Sweden; Department of Computer Science & Engineering, Meghnad Saha Institute of Technology, Kolkata, India
| | - Sanjay Chakraborty
- Department of Computer and Information Science (IDA), REAL, AIICS, Linköping University, Sweden; Department of Computer Science & Engineering, Techno International New Town, Kolkata, India.
| |
Collapse
|
3
|
Göktepe YE. Protein-protein interaction prediction using enhanced features with spaced conjoint triad and amino acid pairwise distance. PeerJ Comput Sci 2025; 11:e2748. [PMID: 40134873 PMCID: PMC11935777 DOI: 10.7717/peerj-cs.2748] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2024] [Accepted: 02/14/2025] [Indexed: 03/27/2025]
Abstract
Protein-protein interactions (PPIs) are pivotal in cellular processes, influencing a wide range of functions, from metabolism to immune responses. Despite the advancements in experimental techniques for PPI detection, their inherent limitations, such as high false-positive rates and significant resource demands, necessitate the development of computational approaches. This study presents a novel computational model named MFPIC (Multi-Feature Protein Interaction Classifier) for predicting PPIs, integrating enhanced sequence-based features, including a novel spaced conjoint triad (SCT) and amino acid pairwise distance (AAPD), with existing methods such as position-specific scoring matrices (PSSM) and AAindex-based features. The SCT captures complex sequence motifs by considering non-adjacent amino acid interactions, while AAPD provides critical spatial information about amino acid residues within protein sequences. The proposed model was evaluated across three benchmark datasets-Saccharomyces cerevisiae, Helicobacter pylori, and human proteins-demonstrating superior performance in comparison to state-of-the-art models. The results underscore the efficacy of integrating diverse and complementary features, achieving significant improvements in predictive accuracy, with the model achieving 95.90%, 99.33%, and 90.95% accuracy on the Saccharomyces cerevisiae, Helicobacter pylori, and human dataset, respectively. This approach not only enhances our understanding of PPI mechanisms but also offers valuable insights for the development of targeted therapeutic strategies.
Collapse
|
4
|
Hyeon DY, Nam D, Shin HJ, Jeong J, Jung E, Cho SY, Shin DH, Ku JL, Baek HJ, Yoo CW, Hong EK, Lim MC, Lee SJ, Bae YK, Kim JK, Bae J, Choi W, Kim SJ, Back S, Kang C, Madar IH, Kim H, Kim S, Kim DK, Kang J, Park GW, Park KS, Shin Y, Kim SS, Jung K, Hwang D, Lee SW, Kim JY. Proteogenomic characterization of molecular and cellular targets for treatment-resistant subtypes in locally advanced cervical cancers. Mol Cancer 2025; 24:77. [PMID: 40087745 PMCID: PMC11908047 DOI: 10.1186/s12943-025-02256-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2024] [Accepted: 02/01/2025] [Indexed: 03/17/2025] Open
Abstract
We report proteogenomic analysis of locally advanced cervical cancer (LACC). Exome-seq data revealed predominant alterations of keratinization-TP53 regulation and O-glycosylation-TP53 regulation axes in squamous and adeno-LACC, respectively, compared to in early-stage cervical cancer. Integrated clustering of mRNA, protein, and phosphorylation data identified six subtypes (Sub1-6) of LACC among which Sub3, 5, and 6 showed the treatment-resistant nature with poor local recurrence-free survival. Elevated immune and extracellular matrix (ECM) activation mediated by activated stroma (PDGFD and CXCL1high fibroblasts) characterized the immune-hot Sub3 enriched with MUC5AChigh epithelial cells (ECs). Increased epithelial-mesenchymal-transition (EMT) and ECM remodeling characterized the immune-cold squamous Sub5 enriched with PGK1 and CXCL10high ECs. We further demonstrated that CIC mutations could trigger EMT activation by upregulating ETV4, and the elevation of the immune checkpoint PVR and neutrophil-like myeloid-derived suppressive cells (FCN1 and FCGR3Bhigh macrophages) could cause suppression of T-cell activation in Sub5. Increased O-linked glycosylation of mucin characterized adeno-LACC Sub6 enriched with MUC5AChigh ECs. These results provide a battery of somatic mutations, cellular pathways, and cellular players that can be used to predict treatment-resistant LACC subtypes and can serve as potential therapeutic targets for these LACC subtypes.
Collapse
Affiliation(s)
- Do Young Hyeon
- School of Biological Sciences, Seoul National University, Seoul, 08826, Republic of Korea
| | - Dowoon Nam
- Department of Chemistry and Center for Proteogenome Research, Korea University, Seoul, 02843, Republic of Korea
| | - Hye-Jin Shin
- Research Institute and Hospital, National Cancer Center, Goyang, 10408, Republic of Korea
| | - Juhee Jeong
- Department of Anatomy and Cell Biology and Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, 03080, Republic of Korea
| | - Eunsoo Jung
- School of Biological Sciences, Seoul National University, Seoul, 08826, Republic of Korea
| | - Soo Young Cho
- Research Institute and Hospital, National Cancer Center, Goyang, 10408, Republic of Korea
| | - Dong Hoon Shin
- Research Institute and Hospital, National Cancer Center, Goyang, 10408, Republic of Korea
| | - Ja-Lok Ku
- Korean Cell Line Bank, Laboratory of Cell Biology, Cancer Research Institute, Seoul National University College of Medicine, Seoul, Korea
| | - Hye Jung Baek
- Research Institute and Hospital, National Cancer Center, Goyang, 10408, Republic of Korea
| | - Chong Woo Yoo
- Research Institute and Hospital, National Cancer Center, Goyang, 10408, Republic of Korea
| | - Eun-Kyung Hong
- Research Institute and Hospital, National Cancer Center, Goyang, 10408, Republic of Korea
| | - Myong Cheol Lim
- Research Institute and Hospital, National Cancer Center, Goyang, 10408, Republic of Korea
| | - Sang-Jin Lee
- Research Institute and Hospital, National Cancer Center, Goyang, 10408, Republic of Korea
| | - Young-Ki Bae
- Research Institute and Hospital, National Cancer Center, Goyang, 10408, Republic of Korea
| | - Jong Kwang Kim
- Research Institute and Hospital, National Cancer Center, Goyang, 10408, Republic of Korea
| | - Jingi Bae
- Department of Chemistry and Center for Proteogenome Research, Korea University, Seoul, 02843, Republic of Korea
| | - Wonyoung Choi
- Research Institute and Hospital, National Cancer Center, Goyang, 10408, Republic of Korea
| | - Su-Jin Kim
- Department of Chemistry and Center for Proteogenome Research, Korea University, Seoul, 02843, Republic of Korea
| | - Seunghoon Back
- Department of Chemistry and Center for Proteogenome Research, Korea University, Seoul, 02843, Republic of Korea
| | - Chaewon Kang
- Department of Chemistry and Center for Proteogenome Research, Korea University, Seoul, 02843, Republic of Korea
| | - Inamul Hasan Madar
- Department of Chemistry and Center for Proteogenome Research, Korea University, Seoul, 02843, Republic of Korea
| | - Hokeun Kim
- Department of Chemistry and Center for Proteogenome Research, Korea University, Seoul, 02843, Republic of Korea
| | - Suhwan Kim
- Department of Chemistry and Center for Proteogenome Research, Korea University, Seoul, 02843, Republic of Korea
| | - Duk Ki Kim
- Department of Anatomy and Cell Biology and Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, 03080, Republic of Korea
| | - Jihyung Kang
- Department of Anatomy and Cell Biology and Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, 03080, Republic of Korea
| | - Geon Woo Park
- Department of Anatomy and Cell Biology and Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, 03080, Republic of Korea
| | - Ki Seok Park
- Department of Anatomy and Cell Biology and Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, 03080, Republic of Korea
| | - Yourae Shin
- School of Biological Sciences, Seoul National University, Seoul, 08826, Republic of Korea
| | - Sang Soo Kim
- Research Institute and Hospital, National Cancer Center, Goyang, 10408, Republic of Korea.
| | - Keehoon Jung
- Department of Anatomy and Cell Biology and Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, 03080, Republic of Korea.
- Institute of Allergy and Clinical Immunology, Seoul National University Medical Research Center, Seoul, 03080, Republic of Korea.
| | - Daehee Hwang
- School of Biological Sciences, Seoul National University, Seoul, 08826, Republic of Korea.
| | - Sang-Won Lee
- Department of Chemistry and Center for Proteogenome Research, Korea University, Seoul, 02843, Republic of Korea.
| | - Joo-Young Kim
- Research Institute and Hospital, National Cancer Center, Goyang, 10408, Republic of Korea.
| |
Collapse
|
5
|
Parthaje S, Janardhanan M, Paul P, Karunakaran KB, Deb AP, Shankarappa B, Pal PK, Mahadevan A, Jain S, Viswanath B, Purushottam M. CAG Repeat Instability and Region-Specific Gene Expression Changes in the SCA12 Brain. CEREBELLUM (LONDON, ENGLAND) 2025; 24:60. [PMID: 40075006 DOI: 10.1007/s12311-025-01808-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 02/16/2025] [Indexed: 03/14/2025]
Abstract
Spinocerebellar ataxia type 12 (SCA12), an autosomal dominant cerebellar ataxia, caused by an expansion of (CAG)n in the 5' of the PPP2R2B gene on chr5q32, is common in India. The illness often manifests late in life, with diverse neurological and psychiatric symptoms, suggesting involvement of different brain regions. Prominent neuronal loss and atrophy of the cerebellum have been noted earlier. In Huntington's disease (HD), somatic instability associated with the size of the expanded CAG allele in HTT varies across regions of the brain, and influences the nature and severity of symptoms. We estimated CAG repeat size, methylation and gene expression in the PPP2R2B gene across regions in brain tissue from a person with SCA12. We also studied the regional expression of DNA repair pathway and cell cycle genes. Somatic mosaicism, manifested as CAG repeat instability, is detected across brain regions. The cerebellum showed the least somatic instability, and this was coupled with increased methylation, and lower expression, of the PPP2R2B gene. Interestingly, increased expression of DNA maintenance pathway related genes, which might partly explain the lowered DNA instability, was also observed. There was also decreased expression of cell cycle modulators, which could initiate apoptosis, and thus account for neuronal cell death seen in the brain sections. We suggest that drugs that improve DNA repeat stability, could thus be explored as a treatment option for SCA12.
Collapse
Affiliation(s)
- Shreevidya Parthaje
- Molecular Genetics Laboratory, Department of Psychiatry, National Institute of Mental Health and Neurosciences (NIMHANS), Bangalore, India
| | - Meghana Janardhanan
- Molecular Genetics Laboratory, Department of Psychiatry, National Institute of Mental Health and Neurosciences (NIMHANS), Bangalore, India
- Department of Medical Neuroscience, Dalhousie University, Halifax, Canada
- Institute of Psychiatric Phenomics and Genomics, University Hospital of Munich, Munich, Germany
| | - Pradip Paul
- Molecular Genetics Laboratory, Department of Psychiatry, National Institute of Mental Health and Neurosciences (NIMHANS), Bangalore, India
| | - Kalyani B Karunakaran
- Molecular Genetics Laboratory, Department of Psychiatry, National Institute of Mental Health and Neurosciences (NIMHANS), Bangalore, India
| | - Ashim Paul Deb
- Molecular Genetics Laboratory, Department of Psychiatry, National Institute of Mental Health and Neurosciences (NIMHANS), Bangalore, India
| | - Bhagyalakshmi Shankarappa
- Molecular Genetics Laboratory, Department of Psychiatry, National Institute of Mental Health and Neurosciences (NIMHANS), Bangalore, India
| | - Pramod Kumar Pal
- Department of Neurology, National Institute of Mental Health and Neurosciences (NIMHANS), Bangalore, India
| | - Anita Mahadevan
- Department of Neuropathology, National Institute of Mental Health and Neurosciences (NIMHANS), Bangalore, India
| | - Sanjeev Jain
- Molecular Genetics Laboratory, Department of Psychiatry, National Institute of Mental Health and Neurosciences (NIMHANS), Bangalore, India
| | - Biju Viswanath
- Molecular Genetics Laboratory, Department of Psychiatry, National Institute of Mental Health and Neurosciences (NIMHANS), Bangalore, India.
| | - Meera Purushottam
- Molecular Genetics Laboratory, Department of Psychiatry, National Institute of Mental Health and Neurosciences (NIMHANS), Bangalore, India.
| |
Collapse
|
6
|
Mahin A, Gopalakrishnan AP, Ahmed M, Nisar M, John L, Shivamurthy PB, Ummar S, Varghese S, Modi PK, Pai VR, Prasad TSK, Raju R. Orchestrating Intracellular Calcium Signaling Cascades by Phosphosite-Centric Regulatory Network: A Comprehensive Analysis on Kinases CAMKK1 and CAMKK2. OMICS : A JOURNAL OF INTEGRATIVE BIOLOGY 2025. [PMID: 40079160 DOI: 10.1089/omi.2024.0196] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/14/2025]
Abstract
Intracellular calcium signaling is a cornerstone in cell biology and a key molecular target for human health and disease. Calcium/calmodulin dependent protein kinase kinases, CAMKK1 and CAMKK2 are serine/threonine kinases that contribute to the regulation of intracellular calcium signals in response to diverse stimuli. CAMKK1 generally has stable dynamics, whereas CAMKK2 dysregulation triggers oncogenicity and neurological disorders. To differentiate the phosphosignaling hierarchy associated with predominant phosphosites of CAMKK1 and CAMKK2, we assembled and analyzed the global cellular phosphoproteome datasets. We found that predominant phosphosites in CAMKK1 and CAMKK2 are located outside the kinase domain, and their phosphomotifs are highly homologous. Further, we employed a coregulation analysis approach to these predominant phosphosites, to infer the co-occurrence patterns of phosphorylations within CAMKKs and the coregulation patterns of other protein phosphosites with CAMKK sites. We report herein that independent phosphorylations at CAMKK2 S100 and S511 increase their enzymatic activity in the presence of calcium/calmodulin. In addition, the study unveils kinase-substrate associations such as RPS6KB1 as a novel high-confidence upstream kinase of both CAMKK1 S74 and CAMKK2 S100. Further, CAMKK2 was identified as a primary orchestrator in mediating intracellular calcium signaling cascades compared to CAMKK1 based on coregulation patterns of phosphosites from proteins involved in the calcium signaling pathway. These molecular details shed promising insights into the pathophysiology of several diseases such as cancers and psychiatric disorders associated with kinase activity dysregulations of CAMKK2 and further open the avenue for novel PTM-directed therapeutic strategies to regulate CAMKK2.
Collapse
Affiliation(s)
- Althaf Mahin
- Centre for Integrative Omics Data Science (CIODS), Yenepoya (Deemed to be University), Mangalore, India
- Center for Systems Biology and Molecular Medicine (CSBMM) [an ICMR-Collaborating Centre of Excellence (ICMR-CCoE 2024)], Yenepoya Research Centre, Yenepoya (Deemed to be University), Mangalore, India
| | - Athira Perunelly Gopalakrishnan
- Centre for Integrative Omics Data Science (CIODS), Yenepoya (Deemed to be University), Mangalore, India
- Center for Systems Biology and Molecular Medicine (CSBMM) [an ICMR-Collaborating Centre of Excellence (ICMR-CCoE 2024)], Yenepoya Research Centre, Yenepoya (Deemed to be University), Mangalore, India
| | - Mukhtar Ahmed
- Department of Zoology, College of Science, King Saud University, Riyadh, Kingdom of Saudi Arabia
| | - Mahammed Nisar
- Centre for Integrative Omics Data Science (CIODS), Yenepoya (Deemed to be University), Mangalore, India
| | - Levin John
- Centre for Integrative Omics Data Science (CIODS), Yenepoya (Deemed to be University), Mangalore, India
| | | | - Samseera Ummar
- Centre for Integrative Omics Data Science (CIODS), Yenepoya (Deemed to be University), Mangalore, India
| | - Susmi Varghese
- Centre for Integrative Omics Data Science (CIODS), Yenepoya (Deemed to be University), Mangalore, India
| | - Prashant Kumar Modi
- Center for Systems Biology and Molecular Medicine (CSBMM) [an ICMR-Collaborating Centre of Excellence (ICMR-CCoE 2024)], Yenepoya Research Centre, Yenepoya (Deemed to be University), Mangalore, India
| | - Vinitha Ramanath Pai
- Department of Biochemistry, Yenepoya Medical College, Yenepoya (Deemed to be University), Mangaluru, India
| | - Thottethodi Subrahmanya Keshava Prasad
- Center for Systems Biology and Molecular Medicine (CSBMM) [an ICMR-Collaborating Centre of Excellence (ICMR-CCoE 2024)], Yenepoya Research Centre, Yenepoya (Deemed to be University), Mangalore, India
| | - Rajesh Raju
- Centre for Integrative Omics Data Science (CIODS), Yenepoya (Deemed to be University), Mangalore, India
- Center for Systems Biology and Molecular Medicine (CSBMM) [an ICMR-Collaborating Centre of Excellence (ICMR-CCoE 2024)], Yenepoya Research Centre, Yenepoya (Deemed to be University), Mangalore, India
| |
Collapse
|
7
|
Li J, Lu X, Jiang K, Tang D, Ning B, Sun F. TARSL: Triple-Attention Cross-Network Representation Learning to Predict Synthetic Lethality for Anti-Cancer Drug Discovery. IEEE J Biomed Health Inform 2025; 29:1680-1691. [PMID: 37603479 DOI: 10.1109/jbhi.2023.3306768] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/23/2023]
Abstract
Cancer is a multifaceted disease that results from co-mutations of multi biological molecules. A promising strategy for cancer therapy involves in exploiting the phenomenon of Synthetic Lethality (SL) by targeting the SL partner of cancer gene. Since traditional methods for SL prediction suffer from high-cost, time-consuming and off-targets effects, computational approaches have been efficient complementary to these methods. Most of existing approaches treat SL associations as independent of other biological interaction networks, and fail to consider other information from various biological networks. Despite some approaches have integrated different networks to capture multi-modal features of genes for SL prediction, these methods implicitly assume that all sources and levels of information contribute equally to the SL associations. As such, a comprehensive and flexible framework for learning gene cross-network representations for SL prediction is still lacking. In this work, we present a novel Triple-Attention cross-network Representation learning for SL prediction (TARSL) by capturing molecular features from heterogeneous sources. We employ three-level attention modules to consider the different contribution of multi-level information. In particular, feature-level attention can capture the correlations between molecular feature and network link, node-level attention can differentiate the importance of various neighbors, and network-level attention can concentrate on important network and reduce the effects of irrelated networks. We perform comprehensive experiments on human SL datasets and these results have proven that our model is consistently superior to baseline methods and predicted SL associations could aid in designing anti-cancer drugs.
Collapse
|
8
|
Kiouri DP, Batsis GC, Chasapis CT. Structure-Based Deep Learning Framework for Modeling Human-Gut Bacterial Protein Interactions. Proteomes 2025; 13:10. [PMID: 39982320 PMCID: PMC11843979 DOI: 10.3390/proteomes13010010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Revised: 02/09/2025] [Accepted: 02/11/2025] [Indexed: 02/22/2025] Open
Abstract
Background: The interaction network between the human host proteins and the proteins of the gut bacteria is essential for the establishment of human health, and its dysregulation directly contributes to disease development. Despite its great importance, experimental data on protein-protein interactions (PPIs) between these species are sparse due to experimental limitations. Methods: This study presents a deep learning-based framework for predicting PPIs between human and gut bacterial proteins using structural data. The framework leverages graph-based protein representations and variational autoencoders (VAEs) to extract structural embeddings from protein graphs, which are then fused through a Bi-directional Cross-Attention module to predict interactions. The model addresses common challenges in PPI datasets, such as class imbalance, using focal loss to emphasize harder-to-classify samples. Results: The results demonstrated that this framework exhibits robust performance, with high precision and recall across validation and test datasets, underscoring its generalizability. By incorporating proteoforms in the analysis, the model accounts for the structural complexity within proteomes, making predictions biologically relevant. Conclusions: These findings offer a scalable tool for investigating the interactions between the host and the gut microbiota, potentially yielding new treatment targets and diagnostics for disorders linked to the microbiome.
Collapse
Affiliation(s)
- Despoina P. Kiouri
- Institute of Chemical Biology, National Hellenic Research Foundation, 11635 Athens, Greece; (D.P.K.); (G.C.B.)
- Laboratory of Organic Chemistry, Department of Chemistry, National and Kapodistrian University of Athens, 15772 Athens, Greece
| | - Georgios C. Batsis
- Institute of Chemical Biology, National Hellenic Research Foundation, 11635 Athens, Greece; (D.P.K.); (G.C.B.)
| | - Christos T. Chasapis
- Institute of Chemical Biology, National Hellenic Research Foundation, 11635 Athens, Greece; (D.P.K.); (G.C.B.)
| |
Collapse
|
9
|
He C, Zhao Z, Wang X, Zheng H, Duan L, Zuo J. Exploring drug-target interaction prediction on cold-start scenarios via meta-learning-based graph transformer. Methods 2025; 234:10-20. [PMID: 39550022 DOI: 10.1016/j.ymeth.2024.11.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Revised: 11/07/2024] [Accepted: 11/12/2024] [Indexed: 11/18/2024] Open
Abstract
Predicting drug-target interaction (DTI) is of great importance for drug discovery and development. With the rapid development of biological and chemical technologies, computational methods for DTI prediction are becoming a promising approach. However, there are few solutions to the cold-start problem in DTI prediction scenarios, as these methods rely on existing interaction information to support their modeling. Consequently, they are unable to effectively predict DTIs for new drugs or targets with limited interaction data in the existing work. To this end, we propose a graph transformer method based on meta-learning named MGDTI (short for Meta-learning-based Graph Transformer for Drug-Target Interaction prediction) to fill this gap. Technically, we employ drug-drug similarity and target-target similarity as additional information to mitigate the scarcity of interactions. Besides, we trained MGDTI via meta-learning to be adaptive to cold-start tasks. Moreover, we employed graph transformer to prevent over-smoothing by capturing long-range dependencies. Extensive results on the benchmark dataset demonstrate that MGDTI is effective on DTI prediction under cold-start scenarios.
Collapse
Affiliation(s)
- Chengxin He
- School of Computer Science, Sichuan University, Chengdu 610065, China; College of Biomedical Engineering, Sichuan University, Chengdu 610065, China
| | - Zhenjiang Zhao
- School of Computer Science, Sichuan University, Chengdu 610065, China
| | - Xinye Wang
- School of Computer Science, Sichuan University, Chengdu 610065, China
| | - Huiru Zheng
- School of Computing, Ulster University, Belfast BT15 1ED, Northern Ireland, UK
| | - Lei Duan
- School of Computer Science, Sichuan University, Chengdu 610065, China
| | - Jie Zuo
- School of Computer Science, Sichuan University, Chengdu 610065, China.
| |
Collapse
|
10
|
Sun L, Yin Z, Lu L. ISLRWR: A network diffusion algorithm for drug-target interactions prediction. PLoS One 2025; 20:e0302281. [PMID: 39883675 PMCID: PMC11781719 DOI: 10.1371/journal.pone.0302281] [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: 12/15/2023] [Accepted: 04/01/2024] [Indexed: 02/01/2025] Open
Abstract
Machine learning techniques and computer-aided methods are now widely used in the pre-discovery tasks of drug discovery, effectively improving the efficiency of drug development and reducing the workload and cost. In this study, we used multi-source heterogeneous network information to build a network model, learn the network topology through multiple network diffusion algorithms, and obtain compressed low-dimensional feature vectors for predicting drug-target interactions (DTIs). We applied the metropolis-hasting random walk (MHRW) algorithm to improve the performance of the random walk with restart (RWR) algorithm, forming the basis by which the self-loop probability of the current node is removed. Additionally, the propagation efficiency of the MHRW was improved using the improved metropolis-hasting random walk (IMRWR) algorithm, facilitating network deep sampling. Finally, we proposed a correction of the transfer probability of the entire network after increasing the self-loop rate of isolated nodes to form the ISLRWR algorithm. Notably, the ISLRWR algorithm improved the area under the receiver operating characteristic curve (AUROC) by 7.53 and 5.72%, and the area under the precision-recall curve (AUPRC) by 5.95 and 4.19% compared to the RWR and MHRW algorithms, respectively, in predicting DTIs performance. Moreover, after excluding the interference of homologous proteins (popular drugs or targets may lead to inflated prediction results), the ISLRWR algorithm still showed a significant performance improvement.
Collapse
Affiliation(s)
- Lu Sun
- School of Mathematics, Physics and Statistics, Institute for Frontier Medical Technology, Center of Intelligent Computing and Applied Statistics, Shanghai University of Engineering Science, Shanghai, China
| | - Zhixiang Yin
- School of Mathematics, Physics and Statistics, Institute for Frontier Medical Technology, Center of Intelligent Computing and Applied Statistics, Shanghai University of Engineering Science, Shanghai, China
| | - Lin Lu
- Shanghai Xinhao Information Technology Co., Ltd., Shanghai, China
| |
Collapse
|
11
|
Zhang B, Quan L, Zhang Z, Cao L, Chen Q, Peng L, Wang J, Jiang Y, Nie L, Li G, Wu T, Lyu Q. MVCL-DTI: Predicting Drug-Target Interactions Using a Multiview Contrastive Learning Model on a Heterogeneous Graph. J Chem Inf Model 2025; 65:1009-1026. [PMID: 39812134 DOI: 10.1021/acs.jcim.4c02073] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2025]
Abstract
Accurate prediction of drug-target interactions (DTIs) is pivotal for accelerating the processes of drug discovery and drug repurposing. MVCL-DTI, a novel model leveraging heterogeneous graphs for predicting DTIs, tackles the challenge of synthesizing information from varied biological subnetworks. It integrates neighbor view, meta-path view, and diffusion view to capture semantic features and employs an attention-based contrastive learning approach, along with a multiview attention-weighted fusion module, to effectively integrate and adaptively weight the information from the different views. Tested under various conditions on benchmark data sets, including varying positive-to-negative sample ratios, conducting hard negative sampling experiments, and masking known DTIs with different ratios, as well as redundant DTIs with various similarity metrics, MVCL-DTI exhibits strong robust generalization. The model is then employed to predict novel DTIs, with a particular focus on COVID-19-related drugs, highlighting its practical applicability. Ultimately, through features visualization and computational properties analysis, we've pinpointed critical elements, including Gene Ontology and substituent nodes, along with a proper initialization strategy, underscoring their vital role in DTI prediction tasks.
Collapse
Affiliation(s)
- Bei Zhang
- School of Computer Science and Technology, Soochow University, Jiangsu 215006, China
- China Mobile (Suzhou) Software Technology Company Limited, Suzhou 215163, China
| | - Lijun Quan
- School of Computer Science and Technology, Soochow University, Jiangsu 215006, China
- Collaborative Innovation Center of Novel Software Technology and Industrialization, Jiangsu 210000, China
| | - Zhijun Zhang
- School of Computer Science and Technology, Soochow University, Jiangsu 215006, China
| | - Lexin Cao
- School of Computer Science and Technology, Soochow University, Jiangsu 215006, China
| | - Qiufeng Chen
- School of Computer Science and Technology, Soochow University, Jiangsu 215006, China
| | - Liangchen Peng
- School of Computer Science and Technology, Soochow University, Jiangsu 215006, China
| | - Junkai Wang
- School of Computer Science and Technology, Soochow University, Jiangsu 215006, China
| | - Yelu Jiang
- School of Computer Science and Technology, Soochow University, Jiangsu 215006, China
| | - Liangpeng Nie
- School of Computer Science and Technology, Soochow University, Jiangsu 215006, China
| | - Geng Li
- School of Computer Science and Technology, Soochow University, Jiangsu 215006, China
| | - Tingfang Wu
- School of Computer Science and Technology, Soochow University, Jiangsu 215006, China
- Collaborative Innovation Center of Novel Software Technology and Industrialization, Jiangsu 210000, China
| | - Qiang Lyu
- School of Computer Science and Technology, Soochow University, Jiangsu 215006, China
- Collaborative Innovation Center of Novel Software Technology and Industrialization, Jiangsu 210000, China
| |
Collapse
|
12
|
Kiouri DP, Batsis GC, Chasapis CT. Structure-Based Approaches for Protein-Protein Interaction Prediction Using Machine Learning and Deep Learning. Biomolecules 2025; 15:141. [PMID: 39858535 PMCID: PMC11763140 DOI: 10.3390/biom15010141] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2024] [Revised: 01/11/2025] [Accepted: 01/14/2025] [Indexed: 01/27/2025] Open
Abstract
Protein-Protein Interaction (PPI) prediction plays a pivotal role in understanding cellular processes and uncovering molecular mechanisms underlying health and disease. Structure-based PPI prediction has emerged as a robust alternative to sequence-based methods, offering greater biological accuracy by integrating three-dimensional spatial and biochemical features. This work summarizes the recent advances in computational approaches leveraging protein structure information for PPI prediction, focusing on machine learning (ML) and deep learning (DL) techniques. These methods not only improve predictive accuracy but also provide insights into functional sites, such as binding and catalytic residues. However, challenges such as limited high-resolution structural data and the need for effective negative sampling persist. Through the integration of experimental and computational tools, structure-based prediction paves the way for comprehensive proteomic network analysis, holding promise for advancements in drug discovery, biomarker identification, and personalized medicine. Future directions include enhancing scalability and dataset reliability to expand these approaches across diverse proteomes.
Collapse
Affiliation(s)
- Despoina P. Kiouri
- Institute of Chemical Biology, National Hellenic Research Foundation, 11635 Athens, Greece; (D.P.K.); (G.C.B.)
- Laboratory of Organic Chemistry, Department of Chemistry, National and Kapodistrian University of Athens, 15772 Athens, Greece
| | - Georgios C. Batsis
- Institute of Chemical Biology, National Hellenic Research Foundation, 11635 Athens, Greece; (D.P.K.); (G.C.B.)
| | - Christos T. Chasapis
- Institute of Chemical Biology, National Hellenic Research Foundation, 11635 Athens, Greece; (D.P.K.); (G.C.B.)
| |
Collapse
|
13
|
Yang G, Liu Y, Wen S, Chen W, Zhu X, Wang Y. DTI-MHAPR: optimized drug-target interaction prediction via PCA-enhanced features and heterogeneous graph attention networks. BMC Bioinformatics 2025; 26:11. [PMID: 39800678 PMCID: PMC11726937 DOI: 10.1186/s12859-024-06021-z] [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: 08/01/2024] [Accepted: 12/20/2024] [Indexed: 01/16/2025] Open
Abstract
Drug-target interactions (DTIs) are pivotal in drug discovery and development, and their accurate identification can significantly expedite the process. Numerous DTI prediction methods have emerged, yet many fail to fully harness the feature information of drugs and targets or address the issue of feature redundancy. We aim to refine DTI prediction accuracy by eliminating redundant features and capitalizing on the node topological structure to enhance feature extraction. To achieve this, we introduce a PCA-augmented multi-layer heterogeneous graph-based network that concentrates on key features throughout the encoding-decoding phase. Our approach initiates with the construction of a heterogeneous graph from various similarity metrics, which is then encoded via a graph neural network. We concatenate and integrate the resultant representation vectors to merge multi-level information. Subsequently, principal component analysis is applied to distill the most informative features, with the random forest algorithm employed for the final decoding of the integrated data. Our method outperforms six baseline models in terms of accuracy, as demonstrated by extensive experimentation. Comprehensive ablation studies, visualization of results, and in-depth case analyses further validate our framework's efficacy and interpretability, providing a novel tool for drug discovery that integrates multimodal features.
Collapse
Affiliation(s)
- Guang Yang
- School of Information and Artificial Intelligence, Anhui Agricultural University, Changjiang West Road, Hefei, 230036, Anhui, China
| | - Yinbo Liu
- School of Information and Artificial Intelligence, Anhui Agricultural University, Changjiang West Road, Hefei, 230036, Anhui, China
| | - Sijian Wen
- School of Information and Artificial Intelligence, Anhui Agricultural University, Changjiang West Road, Hefei, 230036, Anhui, China
| | - Wenxi Chen
- School of Information and Artificial Intelligence, Anhui Agricultural University, Changjiang West Road, Hefei, 230036, Anhui, China
| | - Xiaolei Zhu
- School of Information and Artificial Intelligence, Anhui Agricultural University, Changjiang West Road, Hefei, 230036, Anhui, China
| | - Yongmei Wang
- School of Information and Artificial Intelligence, Anhui Agricultural University, Changjiang West Road, Hefei, 230036, Anhui, China.
| |
Collapse
|
14
|
Rawal O, Turhan B, Peradejordi IF, Chandrasekar S, Kalayci S, Gnjatic S, Johnson J, Bouhaddou M, Gümüş ZH. PhosNetVis: A web-based tool for fast kinase-substrate enrichment analysis and interactive 2D/3D network visualizations of phosphoproteomics data. PATTERNS (NEW YORK, N.Y.) 2025; 6:101148. [PMID: 39896259 PMCID: PMC11783894 DOI: 10.1016/j.patter.2024.101148] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Revised: 11/12/2024] [Accepted: 12/11/2024] [Indexed: 02/04/2025]
Abstract
Protein phosphorylation involves the reversible modification of a protein (substrate) residue by another protein (kinase). Liquid chromatography-mass spectrometry studies are rapidly generating massive protein phosphorylation datasets across multiple conditions. Researchers then must infer kinases responsible for changes in phosphosites of each substrate. However, tools that infer kinase-substrate interactions (KSIs) are not optimized to interactively explore the resulting large and complex networks, significant phosphosites, and states. There is thus an unmet need for a tool that facilitates user-friendly analysis, interactive exploration, visualization, and communication of phosphoproteomics datasets. We present PhosNetVis, a web-based tool for researchers of all computational skill levels to easily infer, generate, and interactively explore KSI networks in 2D or 3D by streamlining phosphoproteomics data analysis steps within a single tool. PhostNetVis lowers barriers for researchers by rapidly generating high-quality visualizations to gain biological insights from their phosphoproteomics datasets. It is available at https://gumuslab.github.io/PhosNetVis/.
Collapse
Affiliation(s)
- Osho Rawal
- Department of Genetics and Genomics, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Berk Turhan
- Department of Genetics and Genomics, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Faculty of Engineering and Natural Sciences, Sabanci University, 34956 Istanbul, Türkiye
| | - Irene Font Peradejordi
- Department of Genetics and Genomics, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Cornell Tech, Cornell University, New York, NY 10044, USA
| | - Shreya Chandrasekar
- Department of Genetics and Genomics, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Cornell Tech, Cornell University, New York, NY 10044, USA
| | - Selim Kalayci
- Department of Genetics and Genomics, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Sacha Gnjatic
- Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Jeffrey Johnson
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Mehdi Bouhaddou
- Department of Microbiology, Immunology, and Molecular Genetics, University of California, Los Angeles, Los Angeles, CA 90095, USA
- Institute for Quantitative and Computational Biosciences, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Zeynep H. Gümüş
- Department of Genetics and Genomics, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| |
Collapse
|
15
|
Xu Y, Zhang Y, Song K, Liu J, Zhao R, Zhang X, Pei L, Li M, Chen Z, Zhang C, Wang P, Li F. ScDrugAct: a comprehensive database to dissect tumor microenvironment cell heterogeneity contributing to drug action and resistance across human cancers. Nucleic Acids Res 2025; 53:D1536-D1546. [PMID: 39526387 PMCID: PMC11701732 DOI: 10.1093/nar/gkae994] [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: 08/13/2024] [Revised: 09/27/2024] [Accepted: 10/15/2024] [Indexed: 11/16/2024] Open
Abstract
The transcriptional heterogeneity of tumor microenvironment (TME) cells is a crucial factor driving the diversity of cellular response to drug treatment and resistance. Therefore, characterizing the cells associated with drug treatment and resistance will help us understand therapeutic mechanisms, discover new therapeutic targets and facilitate precision medicine. Here, we describe a database, scDrugAct (http://bio-bigdata.hrbmu.edu.cn/scDrugAct/), which aims to establish connections among drugs, genes and cells and dissect the impact of TME cellular heterogeneity on drug action and resistance at single-cell resolution. ScDrugAct is curated with drug-cell connections between 3838 223 cells across 34 cancer types and 13 857 drugs and identifies 17 274 drug perturbation/resistance-related genes and 276 559 associations between >10 000 drugs and 53 cell types. ScDrugAct also provides multiple flexible tools to retrieve and analyze connections among drugs, genes and cells; the distribution and developmental trajectories of drug-associated cells within the TME; functional features affecting the heterogeneity of cellular responses to drug perturbation and drug resistance; the cell-specific drug-related gene network; and drug-drug similarities. ScDrugAct serves as an important resource for investigating the impact of the cellular heterogeneity of the TME on drug therapies and can help researchers understand the mechanisms of action and resistance of drugs, as well as discover therapeutic targets.
Collapse
Affiliation(s)
- Yanjun Xu
- College of Bioinformatics Science and Technology, Harbin Medical University, 157 Baojian Road, Harbin 150081, China
| | - Yifang Zhang
- College of Bioinformatics Science and Technology, Harbin Medical University, 157 Baojian Road, Harbin 150081, China
| | - Kaiyue Song
- College of Bioinformatics Science and Technology, Harbin Medical University, 157 Baojian Road, Harbin 150081, China
| | - Jiaqi Liu
- College of Bioinformatics Science and Technology, Harbin Medical University, 157 Baojian Road, Harbin 150081, China
| | - Rui Zhao
- College of Bioinformatics Science and Technology, Harbin Medical University, 157 Baojian Road, Harbin 150081, China
| | - Xiaomeng Zhang
- College of Bioinformatics Science and Technology, Harbin Medical University, 157 Baojian Road, Harbin 150081, China
| | - Liying Pei
- College of Bioinformatics Science and Technology, Harbin Medical University, 157 Baojian Road, Harbin 150081, China
| | - Mengyue Li
- College of Bioinformatics Science and Technology, Harbin Medical University, 157 Baojian Road, Harbin 150081, China
| | - Zhe Chen
- College of Bioinformatics Science and Technology, Harbin Medical University, 157 Baojian Road, Harbin 150081, China
| | - Chunlong Zhang
- College of Bioinformatics Science and Technology, Harbin Medical University, 157 Baojian Road, Harbin 150081, China
| | - Peng Wang
- College of Bioinformatics Science and Technology, Harbin Medical University, 157 Baojian Road, Harbin 150081, China
| | - Feng Li
- College of Bioinformatics Science and Technology, Harbin Medical University, 157 Baojian Road, Harbin 150081, China
| |
Collapse
|
16
|
Zou H, Li S, Guo J, Wen L, Lv C, Leng F, Chen Z, Zeng M, Xu J, Li Y, Li X. Pan-cancer analysis reveals age-associated genetic alterations in protein domains. Am J Hum Genet 2025; 112:44-58. [PMID: 39708814 PMCID: PMC11739924 DOI: 10.1016/j.ajhg.2024.11.011] [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: 07/02/2024] [Revised: 11/26/2024] [Accepted: 11/26/2024] [Indexed: 12/23/2024] Open
Abstract
Cancer incidence and mortality differ among individuals of different ages, but the functional consequences of genetic alterations remain largely unknown. We systematically characterized genetic alterations within protein domains stratified by affected individual's age and showed that the mutational effects on domains varied with age. We further identified potential age-associated driver genes with hotspots across 33 cancers. The candidate drivers involved numerous cancer-related genes that participate in various oncogenic pathways and play central roles in human protein-protein interaction (PPI) networks. We found widespread age biases in protein domains and identified the associations between hotspots and age. Age-stratified PPI networks perturbed by hotspots were constructed to illustrate the function of mutations enriched in domains. We found that hotspots in young adults were associated with premature senescence. In summary, we provided a catalog of age-associated hotspots and their perturbed networks, which may facilitate precision diagnostics and treatments for cancer.
Collapse
Affiliation(s)
- Haozhe Zou
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, Heilongjiang 150081, China
| | - Si Li
- School of Interdisciplinary Medicine and Engineering, Harbin Medical University, Harbin 150081, China
| | - Jiyu Guo
- School of Interdisciplinary Medicine and Engineering, Harbin Medical University, Harbin 150081, China
| | - Luan Wen
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, Heilongjiang 150081, China
| | - Chongwen Lv
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, Heilongjiang 150081, China
| | - Feng Leng
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, Heilongjiang 150081, China
| | - Zefeng Chen
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, Heilongjiang 150081, China
| | - Mengqian Zeng
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, Heilongjiang 150081, China
| | - Juan Xu
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, Heilongjiang 150081, China
| | - Yongsheng Li
- School of Interdisciplinary Medicine and Engineering, Harbin Medical University, Harbin 150081, China.
| | - Xia Li
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, Heilongjiang 150081, China.
| |
Collapse
|
17
|
Xie Y, Wang X, Wang P, Bi X. A pseudo-label supervised graph fusion attention network for drug–target interaction prediction. EXPERT SYSTEMS WITH APPLICATIONS 2025; 259:125264. [DOI: 10.1016/j.eswa.2024.125264] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
18
|
Wright SN, Colton S, Schaffer LV, Pillich RT, Churas C, Pratt D, Ideker T. State of the interactomes: an evaluation of molecular networks for generating biological insights. Mol Syst Biol 2025; 21:1-29. [PMID: 39653848 PMCID: PMC11697402 DOI: 10.1038/s44320-024-00077-y] [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: 09/18/2024] [Revised: 11/07/2024] [Accepted: 11/11/2024] [Indexed: 12/18/2024] Open
Abstract
Advancements in genomic and proteomic technologies have powered the creation of large gene and protein networks ("interactomes") for understanding biological systems. However, the proliferation of interactomes complicates the selection of networks for specific applications. Here, we present a comprehensive evaluation of 45 current human interactomes, encompassing protein-protein interactions as well as gene regulatory, signaling, colocalization, and genetic interaction networks. Our analysis shows that large composite networks such as HumanNet, STRING, and FunCoup are most effective for identifying disease genes, while smaller networks such as DIP, Reactome, and SIGNOR demonstrate stronger performance in interaction prediction. Our study provides a benchmark for interactomes across diverse biological applications and clarifies factors that influence network performance. Furthermore, our evaluation pipeline paves the way for continued assessment of emerging and updated interaction networks in the future.
Collapse
Affiliation(s)
- Sarah N Wright
- Department of Medicine, University of California San Diego, La Jolla, CA, 92093, USA
| | - Scott Colton
- Department of Medicine, University of California San Diego, La Jolla, CA, 92093, USA
| | - Leah V Schaffer
- Department of Medicine, University of California San Diego, La Jolla, CA, 92093, USA
| | - Rudolf T Pillich
- Department of Medicine, University of California San Diego, La Jolla, CA, 92093, USA
| | - Christopher Churas
- Department of Medicine, University of California San Diego, La Jolla, CA, 92093, USA
| | - Dexter Pratt
- Department of Medicine, University of California San Diego, La Jolla, CA, 92093, USA
| | - Trey Ideker
- Department of Medicine, University of California San Diego, La Jolla, CA, 92093, USA.
| |
Collapse
|
19
|
Huang P, Gao W, Fu C, Wang M, Li Y, Chu B, He A, Li Y, Deng X, Zhang Y, Kong Q, Yuan J, Wang H, Shi Y, Gao D, Qin R, Hunter T, Tian R. Clinical functional proteomics of intercellular signalling in pancreatic cancer. Nature 2025; 637:726-735. [PMID: 39537929 DOI: 10.1038/s41586-024-08225-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Accepted: 10/15/2024] [Indexed: 11/16/2024]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) has an atypical, highly stromal tumour microenvironment (TME) that profoundly contributes to its poor prognosis1. Here, to better understand the intercellular signalling between cancer and stromal cells directly in PDAC tumours, we developed a multidimensional proteomic strategy called TMEPro. We applied TMEPro to profile the glycosylated secreted and plasma membrane proteome of 100 human pancreatic tissue samples to a great depth, define cell type origins and identify potential paracrine cross-talk, especially that mediated through tyrosine phosphorylation. Temporal dynamics during pancreatic tumour progression were investigated in a genetically engineered PDAC mouse model. Functionally, we revealed reciprocal signalling between stromal cells and cancer cells mediated by the stromal PDGFR-PTPN11-FOS signalling axis. Furthermore, we examined the generic shedding mechanism of plasma membrane proteins in PDAC tumours and revealed that matrix-metalloprotease-mediated shedding of the AXL receptor tyrosine kinase ectodomain provides an additional dimension of intercellular signalling regulation in the PDAC TME. Importantly, the level of shed AXL has a potential correlation with lymph node metastasis, and inhibition of AXL shedding and its kinase activity showed a substantial synergistic effect in inhibiting cancer cell growth. In summary, we provide TMEPro, a generically applicable clinical functional proteomic strategy, and a comprehensive resource for better understanding the PDAC TME and facilitating the discovery of new diagnostic and therapeutic targets.
Collapse
Affiliation(s)
- Peiwu Huang
- State Key Laboratory of Medical Proteomics and Shenzhen Key Laboratory of Functional Proteomics, Department of Chemistry and Research Center for Chemical Biology and Omics Analysis, School of Science and Guangming Advanced Research Institute, Southern University of Science and Technology, Shenzhen, China
| | - Weina Gao
- State Key Laboratory of Medical Proteomics and Shenzhen Key Laboratory of Functional Proteomics, Department of Chemistry and Research Center for Chemical Biology and Omics Analysis, School of Science and Guangming Advanced Research Institute, Southern University of Science and Technology, Shenzhen, China
| | - Changying Fu
- State Key Laboratory of Medical Proteomics and Shenzhen Key Laboratory of Functional Proteomics, Department of Chemistry and Research Center for Chemical Biology and Omics Analysis, School of Science and Guangming Advanced Research Institute, Southern University of Science and Technology, Shenzhen, China
| | - Min Wang
- Department of Biliary-Pancreatic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yunguang Li
- Key Laboratory of Multi-Cell Systems, Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, Shanghai, China
| | - Bizhu Chu
- State Key Laboratory of Medical Proteomics and Shenzhen Key Laboratory of Functional Proteomics, Department of Chemistry and Research Center for Chemical Biology and Omics Analysis, School of Science and Guangming Advanced Research Institute, Southern University of Science and Technology, Shenzhen, China
| | - An He
- State Key Laboratory of Medical Proteomics and Shenzhen Key Laboratory of Functional Proteomics, Department of Chemistry and Research Center for Chemical Biology and Omics Analysis, School of Science and Guangming Advanced Research Institute, Southern University of Science and Technology, Shenzhen, China
| | - Yuan Li
- State Key Laboratory of Medical Proteomics and Shenzhen Key Laboratory of Functional Proteomics, Department of Chemistry and Research Center for Chemical Biology and Omics Analysis, School of Science and Guangming Advanced Research Institute, Southern University of Science and Technology, Shenzhen, China
| | - Xiaomei Deng
- State Key Laboratory of Medical Proteomics and Shenzhen Key Laboratory of Functional Proteomics, Department of Chemistry and Research Center for Chemical Biology and Omics Analysis, School of Science and Guangming Advanced Research Institute, Southern University of Science and Technology, Shenzhen, China
| | - Yehan Zhang
- Key Laboratory of Multi-Cell Systems, Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, Shanghai, China
| | - Qian Kong
- State Key Laboratory of Medical Proteomics and Shenzhen Key Laboratory of Functional Proteomics, Department of Chemistry and Research Center for Chemical Biology and Omics Analysis, School of Science and Guangming Advanced Research Institute, Southern University of Science and Technology, Shenzhen, China
| | - Jingxiong Yuan
- Department of Biliary-Pancreatic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Hebin Wang
- Department of Biliary-Pancreatic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yu Shi
- Molecular and Cell Biology Laboratory, Salk Institute for Biological Studies, La Jolla, CA, USA.
- Bristol Myers Squibb, San Diego, CA, USA.
| | - Dong Gao
- Key Laboratory of Multi-Cell Systems, Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, Shanghai, China.
- Institute of Cancer Research, Shenzhen Bay Laboratory, Shenzhen, China.
| | - Renyi Qin
- Department of Biliary-Pancreatic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Tony Hunter
- Molecular and Cell Biology Laboratory, Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Ruijun Tian
- State Key Laboratory of Medical Proteomics and Shenzhen Key Laboratory of Functional Proteomics, Department of Chemistry and Research Center for Chemical Biology and Omics Analysis, School of Science and Guangming Advanced Research Institute, Southern University of Science and Technology, Shenzhen, China.
| |
Collapse
|
20
|
Rao R, Gulfishan M, Kim MS, Kashyap MK. Deciphering Cancer Complexity: Integrative Proteogenomics and Proteomics Approaches for Biomarker Discovery. Methods Mol Biol 2025; 2859:211-237. [PMID: 39436604 DOI: 10.1007/978-1-0716-4152-1_12] [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: 10/23/2024]
Abstract
Proteomics has revolutionized the field of cancer biology because the use of a large number of in vivo (SILAC), in vitro (iTRAQ, ICAT, TMT, stable-isotope Dimethyl, and 18O) labeling techniques or label-free methods (spectral counting or peak intensities) coupled with mass spectrometry enables us to profile and identify dysregulated proteins in diseases such as cancer. These proteome and genome studies have led to many challenges, such as the lack of consistency or correlation between copy numbers, RNA, and protein-level data. This review covers solely mass spectrometry-based approaches used for cancer biomarker discovery. It also touches on the emerging role of oncoproteogenomics or proteogenomics in cancer biomarker discovery and how this new area is attracting the integration of genomics and proteomics areas to address some of the important questions to help impinge on the biology and pathophysiology of different malignancies to make these mass spectrometry-based studies more realistic and relevant to clinical settings.
Collapse
Affiliation(s)
- Rashmi Rao
- School of Life and Allied Health Sciences, Glocal University, Saharanpur, UP, India
| | - Mohd Gulfishan
- School of Life and Allied Health Sciences, Glocal University, Saharanpur, UP, India
| | - Min-Sik Kim
- Department of New Biology, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu-42988, Republic of Korea
| | - Manoj Kumar Kashyap
- Amity Stem Cell Institute (ASCI), Amity Medical School (AMS), Amity University Haryana, Panchgaon (Manesar), Gurugram, Haryana, India.
| |
Collapse
|
21
|
Ghasemi MR, Fateh ST, Ben-Mahmoud A, Gupta V, Stühn LG, Lesca G, Chatron N, Platzer K, Edery P, Sadeghi H, Isidor B, Cogné B, Schulz HL, Krauspe-Stübecke I, Periyasamy R, Nampoothiri S, Mirfakhraie R, Alijanpour S, Syrbe S, Pfeifer U, Spranger S, Grundmann-Hauser K, Haack TB, Papadopoulou MT, da Silva Gonçalves T, Panagiotakaki E, Arzimanoglou A, Tonekaboni SH, Rossi M, Korenke GC, Lacassie Y, Jang MH, Layman LC, Miryounesi M, Kim HG. Novel Digital Anomalies, Hippocampal Atrophy, and Mutations Expand the Genotypic and Phenotypic Spectra of CNKSR2 in the Houge Type of X-Linked Syndromic Intellectual Development Disorder (MRXSHG). Am J Med Genet A 2024:e63963. [PMID: 39707601 DOI: 10.1002/ajmg.a.63963] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2024] [Revised: 09/25/2024] [Accepted: 11/21/2024] [Indexed: 12/23/2024]
Abstract
The Houge type of X-linked syndromic intellectual developmental disorder (MRXSHG) encompasses a spectrum of neurodevelopmental disorders characterized by intellectual disability (ID), language/speech delay, attention issues, and epilepsy. These conditions arise from hemizygous or heterozygous deletions, along with point mutations, affecting CNKSR2, a gene located at Xp22.12. CNKSR2, also known as CNK2 or MAGUIN, functions as a synaptic scaffolding molecule within the neuronal postsynaptic density (PSD) of the central nervous system. It acts as a link connecting postsynaptic structural proteins, such as PSD95 and S-SCAM, by employing multiple functional domains crucial for synaptic signaling and protein-protein interactions. Predominantly expressed in dendrites, CNKSR2 is vital for dendritic spine morphogenesis in hippocampal neurons. Its loss-of-function variants result in reduced PSD size and impaired hippocampal development, affecting processes including neuronal proliferation, migration, and synaptogenesis. We present 15 patients including three from the MENA (Middle East and North Africa), a region with no documented mutations in CNKSR2. Each individual displays unique clinical presentations that encompass developmental delay, ID, language/speech delay, epilepsy, and autism. Genetic analyses revealed 14 distinct variants in CNKSR2, comprising five nonsense, three frameshift, two splice, and four missense variants, of which 13 are novel. The ACMG guidelines unanimously interpreted these 14 variants in 15 individuals as pathogenic, highlighting the detrimental impact of these CNKSR2 genetic alterations and confirming the molecular diagnosis of MRXSHG. Importantly, variants Ser767Phe and Ala827Pro may lead to proteasomal degradation or reduced PSD size, contributing to the neurodevelopmental phenotype. Furthermore, these two amino acids, along with another two affected by four missense variants, exhibit complete conservation in nine vertebrate species, illuminating their crucial role in the gene's functionality. Our study revealed unique new digital and brain phenotype, including pointed fingertips (fetal pads of fingertips), syndactyly, tapering fingers, and hippocampal atrophy. These novel clinical features in MRXSHG, combined with 13 novel variants, expand the phenotypic and genotypic spectra of MRXSHG associated with CNKSR2 mutations.
Collapse
Affiliation(s)
- Mohammad-Reza Ghasemi
- Department of Medical Genetics, Faculty of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Center for Comprehensive Genetic Services, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Sahand Tehrani Fateh
- Center for Comprehensive Genetic Services, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Faculty of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Afif Ben-Mahmoud
- Neurological Disorders Research Center, Qatar Biomedical Research Institute, Hamad Bin Khalifa University, Doha, Qatar
| | - Vijay Gupta
- Neurological Disorders Research Center, Qatar Biomedical Research Institute, Hamad Bin Khalifa University, Doha, Qatar
| | - Lara G Stühn
- Institute of Medical Genetics and Applied Genomics, University of Tuebingen, Tuebingen, Germany
| | - Gaetan Lesca
- Department of Medical Genetics, Member of the ERN EpiCARE, University Hospitals of Lyon (HCL), Lyon, France, Lyon, France
- University Claude Bernard Lyon 1, Lyon, France
| | - Nicolas Chatron
- Department of Medical Genetics, Member of the ERN EpiCARE, University Hospitals of Lyon (HCL), Lyon, France, Lyon, France
- University Claude Bernard Lyon 1, Lyon, France
| | - Konrad Platzer
- Institute of Human Genetics, University of Leipzig Medical Center, Leipzig, Germany
| | - Patrick Edery
- Department of Medical Genetics, Member of the ERN EpiCARE, University Hospitals of Lyon (HCL), Lyon, France, Lyon, France
- GENDEV Team, INSERM U1028, CNRS UMR5292, Lyon Neuroscience Research Centre, Lyon, France
| | - Hossein Sadeghi
- Genomic Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Bertrand Isidor
- Service de Génétique Médicale, CHU Nantes, Nantes Cedex 1, France
- Université de Nantes, CNRS, INSERM, l'institut du thorax, Nantes, France
| | - Benjamin Cogné
- Service de Génétique Médicale, CHU Nantes, Nantes Cedex 1, France
- Université de Nantes, CNRS, INSERM, l'institut du thorax, Nantes, France
| | | | - Ilona Krauspe-Stübecke
- Bethlehem Health Center Department of Pediatrics and Adolescent Medicine 5, Stolberg, Germany
| | - Radhakrishnan Periyasamy
- Department of Medical Genetics, Kasturba Medical College, Manipal, Manipal Academy of Higher Education, Manipal, India
| | - Sheela Nampoothiri
- Department of Pediatric Genetics, Amrita Institute of Medical Sciences & Research Centre, Cochin, India
| | - Reza Mirfakhraie
- Department of Medical Genetics, Faculty of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Sahar Alijanpour
- Department of Medical Genetics, Faculty of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Steffen Syrbe
- Division for Pediatric Epileptology, Heidelberg University Hospital, Heidelberg, Germany
| | - Ulrich Pfeifer
- Division of Child Neurology and Metabolic Medicine, Center for Child and Adolescent Medicine, Heidelberg University Hospital, Heidelberg, Germany
| | | | - Kathrin Grundmann-Hauser
- Institute of Medical Genetics and Applied Genomics, University of Tuebingen, Tuebingen, Germany
- Centre for Rare Diseases, University of Tuebingen, Tuebingen, Germany
| | - Tobias B Haack
- Institute of Medical Genetics and Applied Genomics, University of Tuebingen, Tuebingen, Germany
- Centre for Rare Diseases, University of Tuebingen, Tuebingen, Germany
| | - Maria T Papadopoulou
- Department of Paediatric Clinical Epileptology, Sleep Disorders and Functional Neurology, University Hospitals of Lyon (HCL), Member of the European Reference Network (ERN) EpiCARE, France
| | - Tayrine da Silva Gonçalves
- Department of Paediatric Clinical Epileptology, Sleep Disorders and Functional Neurology, University Hospitals of Lyon (HCL), Member of the European Reference Network (ERN) EpiCARE, France
| | - Eleni Panagiotakaki
- Department of Paediatric Clinical Epileptology, Sleep Disorders and Functional Neurology, University Hospitals of Lyon (HCL), Member of the European Reference Network (ERN) EpiCARE, France
| | - Alexis Arzimanoglou
- Department of Paediatric Clinical Epileptology, Sleep Disorders and Functional Neurology, University Hospitals of Lyon (HCL), Member of the European Reference Network (ERN) EpiCARE, France
- Sant Joan De Déu Children's Hospital, Member of the ERN EpiCARE, University of Barcelona, Institut de Recerca Sant Joan de Déu, Spain
| | - Seyed Hassan Tonekaboni
- Pediatric Neurology Excellence Center, Pediatric Neurology Department, Mofid Children Hospital, Faculty of Medicine, Shahid Beheshti University of Medical Sciences (SBMU), Tehran, Iran
| | - Massimiliano Rossi
- GENDEV Team, INSERM U1028, CNRS UMR5292, Lyon Neuroscience Research Centre, Lyon, France
- Department of Genetics, Lyon University Hospitals, Lyon, France
| | - G Christoph Korenke
- Department of Neuropediatrics, University Children's Hospital, Klinikum Oldenburg, Oldenburg, Germany
| | - Yves Lacassie
- Division of Genetics, Department of Pediatrics, Louisiana State University Health Science Center and Children's Hospital, New Orleans, Louisiana, USA
| | - Mi-Hyeon Jang
- Department of Neurosurgery, Robert Wood Johnson Medical School, Rutgers University, the State University of New Jersey, Piscataway, New Jersey, USA
| | - Lawrence C Layman
- Section of Reproductive Endocrinology, Infertility & Genetics, Department of Obstetrics & Gynecology, Augusta University, Augusta, Georgia, USA
- Department of Neuroscience and Regenerative Medicine, Augusta University, Augusta, Georgia, USA
| | - Mohammad Miryounesi
- Department of Medical Genetics, Faculty of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Center for Comprehensive Genetic Services, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Hyung-Goo Kim
- Neurological Disorders Research Center, Qatar Biomedical Research Institute, Hamad Bin Khalifa University, Doha, Qatar
- Department of Neurosurgery, Robert Wood Johnson Medical School, Rutgers University, the State University of New Jersey, Piscataway, New Jersey, USA
| |
Collapse
|
22
|
Rawal O, Turhan B, Peradejordi IF, Chandrasekar S, Kalayci S, Gnjatic S, Johnson J, Bouhaddou M, Gümüş ZH. PhosNetVis: A web-based tool for fast kinase-substrate enrichment analysis and interactive 2D/3D network visualizations of phosphoproteomics data. ARXIV 2024:arXiv:2402.05016v4. [PMID: 39010877 PMCID: PMC11247916] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 07/17/2024]
Abstract
Protein phosphorylation involves the reversible modification of a protein (substrate) residue by another protein (kinase). Liquid chromatography-mass spectrometry studies are rapidly generating massive protein phosphorylation datasets across multiple conditions. Researchers then must infer kinases responsible for changes in phosphosites of each substrate. However, tools that infer kinase-substrate interactions (KSIs) are not optimized to interactively explore the resulting large and complex networks, significant phosphosites, and states. There is thus an unmet need for a tool that facilitates user-friendly analysis, interactive exploration, visualization, and communication of phosphoproteomics datasets. We present PhosNetVis, a web-based tool for researchers of all computational skill levels to easily infer, generate and interactively explore KSI networks in 2D or 3D by streamlining phosphoproteomics data analysis steps within a single tool. PhostNetVis lowers barriers for researchers in rapidly generating high-quality visualizations to gain biological insights from their phosphoproteomics datasets. It is available at: https://gumuslab.github.io/PhosNetVis/.
Collapse
Affiliation(s)
- Osho Rawal
- Department of Genetics and Genomics, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- These authors contributed equally
| | - Berk Turhan
- Department of Genetics and Genomics, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Faculty of Engineering and Natural Sciences, Sabanci University, Istanbul 34956, Türkiye
- These authors contributed equally
| | - Irene Font Peradejordi
- Department of Genetics and Genomics, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Cornell Tech, Cornell University, New York, NY 10044, USA
| | - Shreya Chandrasekar
- Department of Genetics and Genomics, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Cornell Tech, Cornell University, New York, NY 10044, USA
| | - Selim Kalayci
- Department of Genetics and Genomics, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Sacha Gnjatic
- Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Jeffrey Johnson
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Mehdi Bouhaddou
- Department of Microbiology, Immunology, and Molecular Genetics, University of California, Los Angeles; Los Angeles, CA 90095, USA
- Institute for Quantitative and Computational Biosciences, University of California, Los Angeles; Los Angeles, CA 90095, USA
| | - Zeynep H. Gümüş
- Department of Genetics and Genomics, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Lead contact
| |
Collapse
|
23
|
Yin H, Duo H, Li S, Qin D, Xie L, Xiao Y, Sun J, Tao J, Zhang X, Li Y, Zou Y, Yang Q, Yang X, Hao Y, Li B. Unlocking biological insights from differentially expressed genes: Concepts, methods, and future perspectives. J Adv Res 2024:S2090-1232(24)00560-5. [PMID: 39647635 DOI: 10.1016/j.jare.2024.12.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2024] [Revised: 10/12/2024] [Accepted: 12/03/2024] [Indexed: 12/10/2024] Open
Abstract
BACKGROUND Identifying differentially expressed genes (DEGs) is a core task of transcriptome analysis, as DEGs can reveal the molecular mechanisms underlying biological processes. However, interpreting the biological significance of large DEG lists is challenging. Currently, gene ontology, pathway enrichment and protein-protein interaction analysis are common strategies employed by biologists. Additionally, emerging analytical strategies/approaches (such as network module analysis, knowledge graph, drug repurposing, cell marker discovery, trajectory analysis, and cell communication analysis) have been proposed. Despite these advances, comprehensive guidelines for systematically and thoroughly mining the biological information within DEGs remain lacking. AIM OF REVIEW This review aims to provide an overview of essential concepts and methodologies for the biological interpretation of DEGs, enhancing the contextual understanding. It also addresses the current limitations and future perspectives of these approaches, highlighting their broad applications in deciphering the molecular mechanism of complex diseases and phenotypes. To assist users in extracting insights from extensive datasets, especially various DEG lists, we developed DEGMiner (https://www.ciblab.net/DEGMiner/), which integrates over 300 easily accessible databases and tools. KEY SCIENTIFIC CONCEPTS OF REVIEW This review offers strong support and guidance for exploring DEGs, and also will accelerate the discovery of hidden biological insights within genomes.
Collapse
Affiliation(s)
- Huachun Yin
- College of Life Sciences, Chongqing Normal University, Chongqing 401331, PR China; Department of Neurosurgery, Xinqiao Hospital, The Army Medical University, Chongqing 400037, PR China; Department of Neurobiology, Chongqing Key Laboratory of Neurobiology, The Army Medical University, Chongqing 400038, PR China
| | - Hongrui Duo
- College of Life Sciences, Chongqing Normal University, Chongqing 401331, PR China
| | - Song Li
- Department of Neurosurgery, Xinqiao Hospital, The Army Medical University, Chongqing 400037, PR China
| | - Dan Qin
- Department of Biology, College of Science, Northeastern University, Boston, MA 02115, USA
| | - Lingling Xie
- College of Life Sciences, Chongqing Normal University, Chongqing 401331, PR China
| | - Yingxue Xiao
- College of Life Sciences, Chongqing Normal University, Chongqing 401331, PR China
| | - Jing Sun
- College of Life Sciences, Chongqing Normal University, Chongqing 401331, PR China
| | - Jingxin Tao
- College of Life Sciences, Chongqing Normal University, Chongqing 401331, PR China
| | - Xiaoxi Zhang
- College of Life Sciences, Chongqing Normal University, Chongqing 401331, PR China
| | - Yinghong Li
- Chongqing Key Laboratory of Big Data for Bio Intelligence, Chongqing University of Posts and Telecommunications, Chongqing 400065, PR China
| | - Yue Zou
- College of Life Sciences, Chongqing Normal University, Chongqing 401331, PR China
| | - Qingxia Yang
- Zhejiang Provincial Key Laboratory of Precision Diagnosis and Therapy for Major Gynecological Diseases, Women's Hospital, Zhejiang University School of Medicine, Hangzhou 310058, PR China
| | - Xian Yang
- College of Life Sciences, Chongqing Normal University, Chongqing 401331, PR China
| | - Youjin Hao
- College of Life Sciences, Chongqing Normal University, Chongqing 401331, PR China.
| | - Bo Li
- College of Life Sciences, Chongqing Normal University, Chongqing 401331, PR China.
| |
Collapse
|
24
|
Sluzala ZB, Shan Y, Elghazi L, Cárdenas EL, Hamati A, Garner AL, Fort PE. Novel mTORC2/HSPB4 Interaction: Role and Regulation of HSPB4 T148 Phosphorylation. Cells 2024; 13:2000. [PMID: 39682748 DOI: 10.3390/cells13232000] [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: 10/29/2024] [Revised: 11/23/2024] [Accepted: 11/27/2024] [Indexed: 12/18/2024] Open
Abstract
HSPB4 and HSPB5 (α-crystallins) have shown increasing promise as neuroprotective agents, demonstrating several anti-apoptotic and protective roles in disorders such as multiple sclerosis and diabetic retinopathy. HSPs are highly regulated by post-translational modification, including deamidation, glycosylation, and phosphorylation. Among them, T148 phosphorylation has been shown to regulate the structural and functional characteristics of HSPB4 and underlie, in part, its neuroprotective capacity. We recently demonstrated that this phosphorylation is reduced in retinal tissues from patients with diabetic retinopathy, raising the question of its regulation during diseases. The kinase(s) responsible for regulating this phosphorylation, however, have yet to be identified. To this end, we employed a multi-tier strategy utilizing in vitro kinome profiling, bioinformatics, and chemoproteomics to predict and discover the kinases capable of phosphorylating T148. Several kinases were identified as being capable of specifically phosphorylating T148 in vitro, and further analysis highlighted mTORC2 as a particularly strong candidate. Altogether, our data demonstrate that the HSPB4-mTORC2 interaction is multi-faceted. Our data support the role of mTORC2 as a specific kinase phosphorylating HSPB4 at T148, but also provide evidence that the HSPB4 chaperone function further strengthens the interaction. This study addresses a critical gap in our understanding of the regulatory underpinnings of T148 phosphorylation-mediated neuroprotection.
Collapse
Affiliation(s)
- Zachary B Sluzala
- Department of Ophthalmology & Visual Sciences, The University of Michigan, Ann Arbor, MI 48109, USA
| | - Yang Shan
- Department of Ophthalmology & Visual Sciences, The University of Michigan, Ann Arbor, MI 48109, USA
| | - Lynda Elghazi
- Department of Ophthalmology & Visual Sciences, The University of Michigan, Ann Arbor, MI 48109, USA
| | - Emilio L Cárdenas
- Interdepartmental Program in Medicinal Chemistry, The University of Michigan, Ann Arbor, MI 48109, USA
| | - Angelina Hamati
- Department of Ophthalmology & Visual Sciences, The University of Michigan, Ann Arbor, MI 48109, USA
| | - Amanda L Garner
- Interdepartmental Program in Medicinal Chemistry, The University of Michigan, Ann Arbor, MI 48109, USA
| | - Patrice E Fort
- Department of Ophthalmology & Visual Sciences, The University of Michigan, Ann Arbor, MI 48109, USA
- Department of Molecular & Integrative Physiology, The University of Michigan, Ann Arbor, MI 48109, USA
| |
Collapse
|
25
|
Nayar G, Altman RB. Heterogeneous network approaches to protein pathway prediction. Comput Struct Biotechnol J 2024; 23:2727-2739. [PMID: 39035835 PMCID: PMC11260399 DOI: 10.1016/j.csbj.2024.06.022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2024] [Revised: 06/17/2024] [Accepted: 06/18/2024] [Indexed: 07/23/2024] Open
Abstract
Understanding protein-protein interactions (PPIs) and the pathways they comprise is essential for comprehending cellular functions and their links to specific phenotypes. Despite the prevalence of molecular data generated by high-throughput sequencing technologies, a significant gap remains in translating this data into functional information regarding the series of interactions that underlie phenotypic differences. In this review, we present an in-depth analysis of heterogeneous network methodologies for modeling protein pathways, highlighting the critical role of integrating multifaceted biological data. It outlines the process of constructing these networks, from data representation to machine learning-driven predictions and evaluations. The work underscores the potential of heterogeneous networks in capturing the complexity of proteomic interactions, thereby offering enhanced accuracy in pathway prediction. This approach not only deepens our understanding of cellular processes but also opens up new possibilities in disease treatment and drug discovery by leveraging the predictive power of comprehensive proteomic data analysis.
Collapse
Affiliation(s)
- Gowri Nayar
- Department of Biomedical Data Science, Stanford University, United States
| | - Russ B. Altman
- Department of Biomedical Data Science, Stanford University, United States
- Department of Genetics, Stanford University, United States
- Department of Medicine, Stanford University, United States
- Department of Bioengineering, Stanford University, United States
| |
Collapse
|
26
|
Cheng X, Meng X, Chen R, Song Z, Li S, Wei S, Lv H, Zhang S, Tang H, Jiang Y, Zhang R. The molecular subtypes of autoimmune diseases. Comput Struct Biotechnol J 2024; 23:1348-1363. [PMID: 38596313 PMCID: PMC11001648 DOI: 10.1016/j.csbj.2024.03.026] [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: 11/12/2023] [Revised: 03/27/2024] [Accepted: 03/27/2024] [Indexed: 04/11/2024] Open
Abstract
Autoimmune diseases (ADs) are characterized by their complexity and a wide range of clinical differences. Despite patients presenting with similar symptoms and disease patterns, their reactions to treatments may vary. The current approach of personalized medicine, which relies on molecular data, is seen as an effective method to address the variability in these diseases. This review examined the pathologic classification of ADs, such as multiple sclerosis and lupus nephritis, over time. Acknowledging the limitations inherent in pathologic classification, the focus shifted to molecular classification to achieve a deeper insight into disease heterogeneity. The study outlined the established methods and findings from the molecular classification of ADs, categorizing systemic lupus erythematosus (SLE) into four subtypes, inflammatory bowel disease (IBD) into two, rheumatoid arthritis (RA) into three, and multiple sclerosis (MS) into a single subtype. It was observed that the high inflammation subtype of IBD, the RA inflammation subtype, and the MS "inflammation & EGF" subtype share similarities. These subtypes all display a consistent pattern of inflammation that is primarily driven by the activation of the JAK-STAT pathway, with the effective drugs being those that target this signaling pathway. Additionally, by identifying markers that are uniquely associated with the various subtypes within the same disease, the study was able to describe the differences between subtypes in detail. The findings are expected to contribute to the development of personalized treatment plans for patients and establish a strong basis for tailored approaches to treating autoimmune diseases.
Collapse
Affiliation(s)
| | | | | | - Zerun Song
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
| | - Shuai Li
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
| | - Siyu Wei
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
| | - Hongchao Lv
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
| | - Shuhao Zhang
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
| | - Hao Tang
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
| | - Yongshuai Jiang
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
| | - Ruijie Zhang
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
| |
Collapse
|
27
|
Zheng G, Wu D, Wei X, Xu D, Mao T, Yan D, Han W, Shang X, Chen Z, Qiu J, Tang K, Cao Z, Qiu T. PbsNRs: predict the potential binders and scaffolds for nuclear receptors. Brief Bioinform 2024; 26:bbae710. [PMID: 39798999 PMCID: PMC11724720 DOI: 10.1093/bib/bbae710] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Revised: 12/09/2024] [Accepted: 12/26/2024] [Indexed: 01/15/2025] Open
Abstract
Nuclear receptors (NRs) are a class of essential proteins that regulate the expression of specific genes and are associated with multiple diseases. In silico methods for prescreening potential NR binders with predictive binding ability are highly desired for NR-related drug development but are rarely reported. Here, we present the PbsNRs (Predicting binders and scaffolds for Nuclear Receptors), a user-friendly web server designed to predict the potential NR binders and scaffolds through proteochemometric modeling. The utility of PbsNRs was systemically evaluated using both chemical compounds and natural products. Results indicated that PbsNRs achieved a good prediction performance for chemical compounds on internal (ROC-AUC = 0.906, where ROC is Receiver-Operating Characteristic curve and AUC is the Area Under the Curve) and external (ROC-AUC = 0.783) datasets, outperforming both compound-ligand interaction tools and NR-specific predictors. PbsNRs also successfully identified bioactive chemical scaffolds for NRs by screening massive natural products. Moreover, the predicted bioactive and inactive natural products for NR2B1 were experimentally validated using biosensors. PbsNRs not only aids in screening potential therapeutic NR binders but also helps discover the essential molecular scaffold and guide the drug discovery for multiple NR-related diseases. The PbsNRs web server is available at http://pbsnrs.badd-cao.net.
Collapse
Affiliation(s)
- Genhui Zheng
- Institute of Clinical Science, Zhongshan Hospital, Shanghai Medical College, Shanghai Institute of Infectious Disease and Biosecurity, Intelligent Medicine Institute, School of Life Sciences, Fudan University, No. 180 Fenglin Road, Shanghai 200032, China
- Oden Institute for Computational Engineering and Sciences (ICES), University of Texas at Austin, No. 201 East 24th Street, Austin 78712, TX, United States
| | - Dingfeng Wu
- National Center, Children’s Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, No. 3333 Binsheng Road, Hangzhou 310052, China
| | - Xiuxia Wei
- School of Health Science and Engineering, University of Shanghai for Science and Technology, No. 516 Jungong Road, Yangpu District, Shanghai 200093, China
| | - Dongpo Xu
- School of Health Science and Engineering, University of Shanghai for Science and Technology, No. 516 Jungong Road, Yangpu District, Shanghai 200093, China
| | - Tiantian Mao
- School of Life Sciences and Technology, Tongji University, No. 1239 Siping Road, Shanghai 200092, China
| | - Deyu Yan
- School of Life Sciences and Technology, Tongji University, No. 1239 Siping Road, Shanghai 200092, China
| | - Wenhao Han
- School of Life Sciences and Technology, Tongji University, No. 1239 Siping Road, Shanghai 200092, China
| | - Xiaoxiao Shang
- Institute of Clinical Science, Zhongshan Hospital, Shanghai Medical College, Shanghai Institute of Infectious Disease and Biosecurity, Intelligent Medicine Institute, School of Life Sciences, Fudan University, No. 180 Fenglin Road, Shanghai 200032, China
- Department of Mathematics and Statistics, McGill University, 805 Sherbrooke Street West, Montreal H3A 0B9, Quebec, Canada
| | - Zikun Chen
- School of Life Sciences and Technology, Tongji University, No. 1239 Siping Road, Shanghai 200092, China
| | - Jingxuan Qiu
- School of Health Science and Engineering, University of Shanghai for Science and Technology, No. 516 Jungong Road, Yangpu District, Shanghai 200093, China
| | - Kailin Tang
- School of Life Sciences and Technology, Tongji University, No. 1239 Siping Road, Shanghai 200092, China
| | - Zhiwei Cao
- Institute of Clinical Science, Zhongshan Hospital, Shanghai Medical College, Shanghai Institute of Infectious Disease and Biosecurity, Intelligent Medicine Institute, School of Life Sciences, Fudan University, No. 180 Fenglin Road, Shanghai 200032, China
| | - Tianyi Qiu
- Institute of Clinical Science, Zhongshan Hospital, Shanghai Medical College, Shanghai Institute of Infectious Disease and Biosecurity, Intelligent Medicine Institute, School of Life Sciences, Fudan University, No. 180 Fenglin Road, Shanghai 200032, China
| |
Collapse
|
28
|
Wiśniewski J, Więcek K, Ali H, Pyrc K, Kula-Păcurar A, Wagner M, Chen HC. Distinguishable topology of the task-evoked functional genome networks in HIV-1 reservoirs. iScience 2024; 27:111222. [PMID: 39559761 PMCID: PMC11570469 DOI: 10.1016/j.isci.2024.111222] [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: 08/05/2024] [Revised: 10/07/2024] [Accepted: 10/18/2024] [Indexed: 11/20/2024] Open
Abstract
HIV-1 reservoirs display a heterogeneous nature, lodging both intact and defective proviruses. To deepen our understanding of such heterogeneous HIV-1 reservoirs and their functional implications, we integrated basic concepts of graph theory to characterize the composition of HIV-1 reservoirs. Our analysis revealed noticeable topological properties in networks, featuring immunologic signatures enriched by genes harboring intact and defective proviruses, when comparing antiretroviral therapy (ART)-treated HIV-1-infected individuals and elite controllers. The key variable, the rich factor, played a pivotal role in classifying distinct topological properties in networks. The host gene expression strengthened the accuracy of classification between elite controllers and ART-treated patients. Markov chain modeling for the simulation of different graph networks demonstrated the presence of an intrinsic barrier between elite controllers and non-elite controllers. Overall, our work provides a prime example of leveraging genomic approaches alongside mathematical tools to unravel the complexities of HIV-1 reservoirs.
Collapse
Affiliation(s)
- Janusz Wiśniewski
- Quantitative Virology Research Group, Population Diagnostics Center, Łukasiewicz Research Network – PORT Polish Center for Technology Development, Stabłowicka 147, 54-066 Wrocław, Poland
| | - Kamil Więcek
- Quantitative Virology Research Group, Population Diagnostics Center, Łukasiewicz Research Network – PORT Polish Center for Technology Development, Stabłowicka 147, 54-066 Wrocław, Poland
| | - Haider Ali
- Molecular Virology Group, Małopolska Centre of Biotechnology, Jagiellonian University, Gronostajowa 7A str, 30-387 Kraków, Poland
- Doctoral School of Exact and Natural Sciences, Jagiellonian University, Łojasiewicza 11, 30-348 Kraków, Poland
| | - Krzysztof Pyrc
- Virogenetics Laboratory of Virology, Małopolska Centre of Biotechnology, Jagiellonian University, Gronostajowa 7A str, 30-387 Kraków, Poland
| | - Anna Kula-Păcurar
- Molecular Virology Group, Małopolska Centre of Biotechnology, Jagiellonian University, Gronostajowa 7A str, 30-387 Kraków, Poland
| | - Marek Wagner
- Innate Immunity Research Group, Life Sciences and Biotechnology Center, Łukasiewicz Research Network – PORT Polish Center for Technology Development, Stabłowicka 147, 54-066 Wrocław, Poland
| | - Heng-Chang Chen
- Quantitative Virology Research Group, Population Diagnostics Center, Łukasiewicz Research Network – PORT Polish Center for Technology Development, Stabłowicka 147, 54-066 Wrocław, Poland
| |
Collapse
|
29
|
Lin X, Chang X, Zhang Y, Gao Z, Chi X. Automatic construction of Petri net models for computational simulations of molecular interaction network. NPJ Syst Biol Appl 2024; 10:131. [PMID: 39521772 PMCID: PMC11550427 DOI: 10.1038/s41540-024-00464-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Accepted: 10/30/2024] [Indexed: 11/16/2024] Open
Abstract
Petri nets are commonly applied in modeling biological systems. However, construction of a Petri net model for complex biological systems is often time consuming, and requires expertise in the research area, limiting their application. To address this challenge, we developed GINtoSPN, an R package that automates the conversion of multi-omics molecular interaction network extracted from the Global Integrative Network (GIN) into Petri nets in GraphML format. These GraphML files can be directly used for Signaling Petri Net (SPN) simulation. To demonstrate the utility of this tool, we built a Petri net model for neurofibromatosis type I. Simulation of NF1 gene knockout, compared to normal skin fibroblast cells, revealed persistent accumulation of Ras-GTPs as expected. Additionally, we identified several other genes substantially affected by the loss of NF1's function, exhibiting individual-specific variability. These results highlight the effectiveness of GINtoSPN in streamlining the modeling and simulation of complex biological systems.
Collapse
Affiliation(s)
- Xuefei Lin
- Department of Dermatology and Venereal Disease, Xuan Wu Hospital, Beijing, China
| | - Xiao Chang
- Department of Dermatology and Venereal Disease, Xuan Wu Hospital, Beijing, China
| | - Yizheng Zhang
- China National Center for Bioinformation, Beijing, China
- CAS Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Zhanyu Gao
- China National Center for Bioinformation, Beijing, China
- CAS Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
- HKU Li Ka Shing Faculty of Medicine, Hong Kong, China
| | - Xu Chi
- China National Center for Bioinformation, Beijing, China.
- CAS Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing, China.
| |
Collapse
|
30
|
E U, T M, A V G, D P. A comprehensive survey of drug-target interaction analysis in allopathy and siddha medicine. Artif Intell Med 2024; 157:102986. [PMID: 39326289 DOI: 10.1016/j.artmed.2024.102986] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2023] [Revised: 08/13/2024] [Accepted: 09/18/2024] [Indexed: 09/28/2024]
Abstract
Effective drug delivery is the cornerstone of modern healthcare, ensuring therapeutic compounds reach their intended targets efficiently. This paper explores the potential of personalized and holistic healthcare, driven by the synergy between traditional and allopathic medicine systems, with a specific focus on the vast reservoir of medicinal compounds found in plants rooted in the historical legacy of traditional medicine. Motivated by the desire to unlock the therapeutic potential of medicinal plants and bridge the gap between traditional and allopathic medicine, this survey delves into in-silico computational approaches for studying Drug-Target Interactions (DTI) within the contexts of allopathy and siddha medicine. The contributions of this survey are multifaceted: it offers a comprehensive overview of in-silico methods for DTI analysis in both systems, identifies common challenges in DTI studies, provides insights into future directions to advance DTI analysis, and includes a comparative analysis of DTI in allopathy and siddha medicine. The findings of this survey highlight the pivotal role of in-silico computational approaches in advancing drug research and development in both allopathy and siddha medicine, emphasizing the importance of integrating these methods to drive the future of personalized healthcare.
Collapse
Affiliation(s)
- Uma E
- Department of Information Science and Technology, College of Engineering Guindy, Chennai, India.
| | - Mala T
- Department of Information Science and Technology, College of Engineering Guindy, Chennai, India
| | - Geetha A V
- Department of Information Science and Technology, College of Engineering Guindy, Chennai, India
| | - Priyanka D
- Department of Information Science and Technology, College of Engineering Guindy, Chennai, India
| |
Collapse
|
31
|
Liu B, Tsoumakas G. Integrating Similarities via Local Interaction Consistency and Optimizing Area Under the Curve Measures via Matrix Factorization for Drug-Target Interaction Prediction. IEEE/ACM TRANSACTIONS ON COMPUTATIONAL BIOLOGY AND BIOINFORMATICS 2024; 21:2212-2225. [PMID: 39226198 DOI: 10.1109/tcbb.2024.3453499] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/05/2024]
Abstract
In drug discovery, identifying drug-target interactions (DTIs) via experimental approaches is a tedious and expensive procedure. Computational methods efficiently predict DTIs and recommend a small part of potential interacting pairs for further experimental confirmation, accelerating the drug discovery process. Although fusing heterogeneous drug and target similarities can improve the prediction ability, the existing similarity combination methods ignore the interaction consistency for neighbour entities. Furthermore, area under the precision-recall curve (AUPR) and area under the receiver operating characteristic curve (AUC) are two widely used evaluation metrics in DTI prediction. However, the two metrics are seldom considered as losses within existing DTI prediction methods. We propose a local interaction consistency (LIC) aware similarity integration method to fuse vital information from diverse views for DTI prediction models. Furthermore, we propose two matrix factorization (MF) methods that optimize AUPR and AUC using convex surrogate losses respectively, and then develop an ensemble MF approach that takes advantage of the two area under the curve metrics by combining the two single metric based MF models. Experimental results under different prediction settings show that the proposed methods outperform various competitors in terms of the metric(s) they optimize and are reliable in discovering potential new DTIs.
Collapse
|
32
|
Zhang M, Hong Y, Shen L, Xu S, Xu Y, Zhang X, Liu J, Liu X. A heterogeneous graph neural network with automatic discovery of effective metapaths for drug–target interaction prediction. FUTURE GENERATION COMPUTER SYSTEMS 2024; 160:283-294. [DOI: 10.1016/j.future.2024.05.054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
33
|
Xu B, Chen J, Wang Y, Fu Q, Lu Y. Reinforced Metapath Optimization in Heterogeneous Information Networks for Drug-Target Interaction Prediction. IEEE/ACM TRANSACTIONS ON COMPUTATIONAL BIOLOGY AND BIOINFORMATICS 2024; 21:2315-2329. [PMID: 39316496 DOI: 10.1109/tcbb.2024.3467135] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/26/2024]
Abstract
Graph neural networks offer an effective avenue for predicting drug-target interactions. In this domain, researchers have found that constructing heterogeneous information networks based on metapaths using diverse biological datasets enhances prediction performance. However, the performance of such methods is closely tied to the selection of metapaths and the compatibility between metapath subgraphs and graph neural networks. Most existing approaches still rely on fixed strategies for selecting metapaths and often fail to fully exploit node information along the metapaths, limiting the improvement in model performance. This paper introduces a novel method for predicting drug-target interactions by optimizing metapaths in heterogeneous information networks. On one hand, the method formulates the metapath optimization problem as a Markov decision process, using the enhancement of downstream network performance as a reward signal. Through iterative training of a reinforcement learning agent, a high-quality set of metapaths is learned. On the other hand, to fully leverage node information along the metapaths, the paper constructs subgraphs based on nodes along the metapaths. Different depths of subgraphs are processed using different graph convolutional neural network. The proposed method is validated using standard heterogeneous biological benchmark datasets. Experimental results on standard datasets show significant advantages over traditional methods.
Collapse
|
34
|
Ayalvari S, Kaedi M, Sehhati M. A modified multiple-criteria decision-making approach based on a protein-protein interaction network to diagnose latent tuberculosis. BMC Med Inform Decis Mak 2024; 24:319. [PMID: 39478591 PMCID: PMC11523813 DOI: 10.1186/s12911-024-02668-z] [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: 04/28/2024] [Accepted: 09/05/2024] [Indexed: 11/02/2024] Open
Abstract
BACKGROUND DNA microarrays provide informative data for transcriptional profiling and identifying gene expression signatures to help prevent progression of latent tuberculosis infection (LTBI) to active disease. However, constructing a prognostic model for distinguishing LTBI from active tuberculosis (ATB) is very challenging due to the noisy nature of data and lack of a generally stable analysis approach. METHODS In the present study, we proposed an accurate predictive model with the help of data fusion at the decision level. In this regard, results of filter feature selection and wrapper feature selection techniques were combined with multiple-criteria decision-making (MCDM) methods to select 10 genes from six microarray datasets that can be the most discriminative genes for diagnosing tuberculosis cases. As the main contribution of this study, the final ranking function was constructed by combining protein-protein interaction (PPI) network with an MCDM method (called Decision-making Trial and Evaluation Laboratory or DEMATEL) to improve the feature ranking approach. RESULTS By applying data fusion at the decision level on the 10 introduced genes in terms of fusion of classifiers of random forests (RF) and k-nearest neighbors (KNN) regarding Yager's theory, the proposed algorithm reached a sensitivity of 0.97, specificity of 0.90, and accuracy of 0.95. Finally, with the help of cumulative clustering, the genes involved in the diagnosis of latent and activated tuberculosis have been introduced. CONCLUSIONS The combination of MCDM methods and PPI networks can significantly improve the diagnosis different states of tuberculosis. CLINICAL TRIAL NUMBER Not applicable.
Collapse
Affiliation(s)
- Somayeh Ayalvari
- Faculty of Computer Engineering, University of Isfahan, Isfahan, Iran
| | - Marjan Kaedi
- Faculty of Computer Engineering, University of Isfahan, Isfahan, Iran.
| | - Mohammadreza Sehhati
- Department of Biomedical Engineering, School of Advanced Medical Technology, Isfahan University of Medical Sciences, Isfahan, Iran
| |
Collapse
|
35
|
Xiong D, Qiu Y, Zhao J, Zhou Y, Lee D, Gupta S, Torres M, Lu W, Liang S, Kang JJ, Eng C, Loscalzo J, Cheng F, Yu H. A structurally informed human protein-protein interactome reveals proteome-wide perturbations caused by disease mutations. Nat Biotechnol 2024:10.1038/s41587-024-02428-4. [PMID: 39448882 DOI: 10.1038/s41587-024-02428-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Accepted: 09/11/2024] [Indexed: 10/26/2024]
Abstract
To assist the translation of genetic findings to disease pathobiology and therapeutics discovery, we present an ensemble deep learning framework, termed PIONEER (Protein-protein InteractiOn iNtErfacE pRediction), that predicts protein-binding partner-specific interfaces for all known protein interactions in humans and seven other common model organisms to generate comprehensive structurally informed protein interactomes. We demonstrate that PIONEER outperforms existing state-of-the-art methods and experimentally validate its predictions. We show that disease-associated mutations are enriched in PIONEER-predicted protein-protein interfaces and explore their impact on disease prognosis and drug responses. We identify 586 significant protein-protein interactions (PPIs) enriched with PIONEER-predicted interface somatic mutations (termed oncoPPIs) from analysis of approximately 11,000 whole exomes across 33 cancer types and show significant associations of oncoPPIs with patient survival and drug responses. PIONEER, implemented as both a web server platform and a software package, identifies functional consequences of disease-associated alleles and offers a deep learning tool for precision medicine at multiscale interactome network levels.
Collapse
Grants
- R01GM124559 U.S. Department of Health & Human Services | NIH | National Institute of General Medical Sciences (NIGMS)
- R01GM125639 U.S. Department of Health & Human Services | NIH | National Institute of General Medical Sciences (NIGMS)
- R01GM130885 U.S. Department of Health & Human Services | NIH | National Institute of General Medical Sciences (NIGMS)
- RM1GM139738 U.S. Department of Health & Human Services | NIH | National Institute of General Medical Sciences (NIGMS)
- R01DK115398 U.S. Department of Health & Human Services | NIH | National Institute of Diabetes and Digestive and Kidney Diseases (National Institute of Diabetes & Digestive & Kidney Diseases)
- U01HG007691 U.S. Department of Health & Human Services | NIH | National Human Genome Research Institute (NHGRI)
- R01HL155107 U.S. Department of Health & Human Services | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- R01HL155096 U.S. Department of Health & Human Services | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- R01HL166137 U.S. Department of Health & Human Services | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- U54HL119145 U.S. Department of Health & Human Services | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- AHA957729 American Heart Association (American Heart Association, Inc.)
- 24MERIT1185447 American Heart Association (American Heart Association, Inc.)
- R01AG084250 U.S. Department of Health & Human Services | NIH | National Institute on Aging (U.S. National Institute on Aging)
- R56AG074001 U.S. Department of Health & Human Services | NIH | National Institute on Aging (U.S. National Institute on Aging)
- U01AG073323 U.S. Department of Health & Human Services | NIH | National Institute on Aging (U.S. National Institute on Aging)
- R01AG066707 U.S. Department of Health & Human Services | NIH | National Institute on Aging (U.S. National Institute on Aging)
- R01AG076448 U.S. Department of Health & Human Services | NIH | National Institute on Aging (U.S. National Institute on Aging)
- R01AG082118 U.S. Department of Health & Human Services | NIH | National Institute on Aging (U.S. National Institute on Aging)
- RF1AG082211 U.S. Department of Health & Human Services | NIH | National Institute on Aging (U.S. National Institute on Aging)
- R21AG083003 U.S. Department of Health & Human Services | NIH | National Institute on Aging (U.S. National Institute on Aging)
- RF1NS133812 U.S. Department of Health & Human Services | NIH | National Institute of Neurological Disorders and Stroke (NINDS)
Collapse
Affiliation(s)
- Dapeng Xiong
- Department of Computational Biology, Cornell University, Ithaca, NY, USA
- Weill Institute for Cell and Molecular Biology, Cornell University, Ithaca, NY, USA
- Center for Innovative Proteomics, Cornell University, Ithaca, NY, USA
| | - Yunguang Qiu
- Cleveland Clinic Genome Center, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
- Genomic Medicine Institute, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Junfei Zhao
- Department of Systems Biology, Herbert Irving Comprehensive Center, Columbia University, New York, NY, USA
| | - Yadi Zhou
- Cleveland Clinic Genome Center, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
- Genomic Medicine Institute, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Dongjin Lee
- Department of Computational Biology, Cornell University, Ithaca, NY, USA
- Weill Institute for Cell and Molecular Biology, Cornell University, Ithaca, NY, USA
| | - Shobhita Gupta
- Weill Institute for Cell and Molecular Biology, Cornell University, Ithaca, NY, USA
- Center for Innovative Proteomics, Cornell University, Ithaca, NY, USA
- Biophysics Program, Cornell University, Ithaca, NY, USA
| | - Mateo Torres
- Department of Computational Biology, Cornell University, Ithaca, NY, USA
- Weill Institute for Cell and Molecular Biology, Cornell University, Ithaca, NY, USA
- Center for Innovative Proteomics, Cornell University, Ithaca, NY, USA
| | - Weiqiang Lu
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, China
| | - Siqi Liang
- Department of Computational Biology, Cornell University, Ithaca, NY, USA
- Weill Institute for Cell and Molecular Biology, Cornell University, Ithaca, NY, USA
| | - Jin Joo Kang
- Department of Computational Biology, Cornell University, Ithaca, NY, USA
- Weill Institute for Cell and Molecular Biology, Cornell University, Ithaca, NY, USA
- Center for Innovative Proteomics, Cornell University, Ithaca, NY, USA
| | - Charis Eng
- Cleveland Clinic Genome Center, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
- Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine, Case Western Reserve University, Cleveland, OH, USA
- Case Comprehensive Cancer Center, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - Joseph Loscalzo
- Channing Division of Network Medicine, Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Feixiong Cheng
- Cleveland Clinic Genome Center, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA.
- Genomic Medicine Institute, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA.
- Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine, Case Western Reserve University, Cleveland, OH, USA.
- Case Comprehensive Cancer Center, Case Western Reserve University School of Medicine, Cleveland, OH, USA.
| | - Haiyuan Yu
- Department of Computational Biology, Cornell University, Ithaca, NY, USA.
- Weill Institute for Cell and Molecular Biology, Cornell University, Ithaca, NY, USA.
- Center for Innovative Proteomics, Cornell University, Ithaca, NY, USA.
| |
Collapse
|
36
|
Li L, Li H, Ishdorj TO, Zheng C, Su Y. MDNNSyn: A Multi-Modal Deep Learning Framework for Drug Synergy Prediction. IEEE J Biomed Health Inform 2024; 28:6225-6236. [PMID: 38954565 DOI: 10.1109/jbhi.2024.3421916] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/04/2024]
Abstract
Synergistic drug combination prediction tasks based on the computational models have been widely studied and applied in the cancer field. However, most of models only consider the interactions between drug pairs and specific cell lines, without taking into account the multiple biological relationships of drug-drug and cell line-cell line that also largely affect synergistic mechanisms. To this end, here we propose a multi-modal deep learning framework, termed MDNNSyn, which adequately applies multi-source information and trains multi-modal features to infer potential synergistic drug combinations. MDNNSyn extracts topology modality features by implementing the multi-layer hypergraph neural network on drug synergy hypergraph and constructs semantic modality features through similarity strategy. A multi-modal fusion network layer with gated neural network is then employed for synergy score prediction. MDNNSyn is compared to five classic and state-of-the-art prediction methods on DrugCombDB and Oncology-Screen datasets. The model achieves area under the curve (AUC) scores of 0.8682 and 0.9013 on two datasets, an improvement of 3.70 % and 2.71 % over the second-best model. Case study indicates that MDNNSyn is capable of detecting potential synergistic drug combinations.
Collapse
|
37
|
Yao W, Wei A, Xiao Z, Zhao W, Shen X, Jiang X, He T. An Improved Framework for Drug-Side Effect Associations Prediction via Counterfactual Inference-Based Data Augmentation. IEEE Trans Nanobioscience 2024; 23:540-547. [PMID: 39141449 DOI: 10.1109/tnb.2024.3443244] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/16/2024]
Abstract
Detecting side effects of drugs is a fundamental task in drug development. With the expansion of publicly available biomedical data, researchers have proposed many computational methods for predicting drug-side effect associations (DSAs), among which network-based methods attract wide attention in the biomedical field. However, the problem of data scarcity poses a great challenge for existing DSAs prediction models. Although several data augmentation methods have been proposed to address this issue, most of existing methods employ a random way to manipulate the original networks, which ignores the causality of existence of DSAs, leading to the poor performance on the task of DSAs prediction. In this paper, we propose a counterfactual inference-based data augmentation method for improving the performance of the task. First, we construct a heterogeneous information network (HIN) by integrating multiple biomedical data. Based on the community detection on the HIN, a counterfactual inference-based method is designed to derive augmented links, and an augmented HIN is obtained accordingly. Then, a meta-path-based graph neural network is applied to learn high-quality representations of drugs and side effects, on which the predicted DSAs are obtained. Finally, comprehensive experiments are conducted, and the results demonstrate the effectiveness of the proposed counterfactual inference-based data augmentation for the task of DSAs prediction.
Collapse
|
38
|
Hu X, Yi H, Cheng H, Zhao Y, Zhang D, Li J, Ruan J, Zhang J, Lu X. Multiple Heterogeneous Networks Representation With Latent Space for Synthetic Lethality Prediction. IEEE Trans Nanobioscience 2024; 23:564-571. [PMID: 39150817 DOI: 10.1109/tnb.2024.3444922] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/18/2024]
Abstract
Computational synthetic lethality (SL) method has become a promising strategy to identify SL gene pairs for targeted cancer therapy and cancer medicine development. Feature representation for integrating various biological networks is crutial to improve the identification performance. However, previous feature representation, such as matrix factorization and graph neural network, projects gene features onto latent variables by keeping a specific geometric metric. There is a lack of models of gene representational latent space with considerating multiple dimentionalities correlation and preserving latent geometric structures in both sample and feature spaces. Therefore, we propose a novel method to model gene Latent Space using matrix Tri-Factorization (LSTF) to obtain gene representation with embedding variables resulting from the potential interpretation of synthetic lethality. Meanwhile, manifold subspace regularization is applied to the tri-factorization to capture the geometrical manifold structure in the latent space with gene PPI functional and GO semantic embeddings. Then, SL gene pairs are identified by the reconstruction of the associations with gene representations in the latent space. The experimental results illustrate that LSTF is superior to other state-of-the-art methods. Case study demonstrate the effectiveness of the predicted SL associations.
Collapse
|
39
|
Gervas-Arruga J, Barba-Romero MÁ, Fernández-Martín JJ, Gómez-Cerezo JF, Segú-Vergés C, Ronzoni G, Cebolla JJ. In Silico Modeling of Fabry Disease Pathophysiology for the Identification of Early Cellular Damage Biomarker Candidates. Int J Mol Sci 2024; 25:10329. [PMID: 39408658 PMCID: PMC11477023 DOI: 10.3390/ijms251910329] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2024] [Revised: 09/19/2024] [Accepted: 09/24/2024] [Indexed: 10/20/2024] Open
Abstract
Fabry disease (FD) is an X-linked lysosomal disease whose ultimate consequences are the accumulation of sphingolipids and subsequent inflammatory events, mainly at the endothelial level. The outcomes include different nervous system manifestations as well as multiple organ damage. Despite the availability of known biomarkers, early detection of FD remains a medical need. This study aimed to develop an in silico model based on machine learning to identify candidate vascular and nervous system proteins for early FD damage detection at the cellular level. A combined systems biology and machine learning approach was carried out considering molecular characteristics of FD to create a computational model of vascular and nervous system disease. A data science strategy was applied to identify risk classifiers by using 10 K-fold cross-validation. Further biological and clinical criteria were used to prioritize the most promising candidates, resulting in the identification of 36 biomarker candidates with classifier abilities, which are easily measurable in body fluids. Among them, we propose four candidates, CAMK2A, ILK, LMNA, and KHSRP, which have high classification capabilities according to our models (cross-validated accuracy ≥ 90%) and are related to the vascular and nervous systems. These biomarkers show promise as high-risk cellular and tissue damage indicators that are potentially applicable in clinical settings, although in vivo validation is still needed.
Collapse
Affiliation(s)
| | - Miguel Ángel Barba-Romero
- Department of Internal Medicine, Albacete University Hospital, 02006 Albacete, Spain;
- Albacete Medical School, Castilla-La Mancha University, 02006 Albacete, Spain
| | | | - Jorge Francisco Gómez-Cerezo
- Department of Internal Medicine, Infanta Sofía University Hospital, 28702 Madrid, Spain;
- Faculty of Medicine, European University of Madrid, 28670 Madrid, Spain
| | | | | | | |
Collapse
|
40
|
Mazein I, Rougny A, Mazein A, Henkel R, Gütebier L, Michaelis L, Ostaszewski M, Schneider R, Satagopam V, Jensen LJ, Waltemath D, Wodke JAH, Balaur I. Graph databases in systems biology: a systematic review. Brief Bioinform 2024; 25:bbae561. [PMID: 39565895 PMCID: PMC11578065 DOI: 10.1093/bib/bbae561] [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: 04/18/2024] [Revised: 09/28/2024] [Accepted: 10/21/2024] [Indexed: 11/22/2024] Open
Abstract
Graph databases are becoming increasingly popular across scientific disciplines, being highly suitable for storing and connecting complex heterogeneous data. In systems biology, they are used as a backend solution for biological data repositories, ontologies, networks, pathways, and knowledge graph databases. In this review, we analyse all publications using or mentioning graph databases retrieved from PubMed and PubMed Central full-text search, focusing on the top 16 available graph databases, Publications are categorized according to their domain and application, focusing on pathway and network biology and relevant ontologies and tools. We detail different approaches and highlight the advantages of outstanding resources, such as UniProtKB, Disease Ontology, and Reactome, which provide graph-based solutions. We discuss ongoing efforts of the systems biology community to standardize and harmonize knowledge graph creation and the maintenance of integrated resources. Outlining prospects, including the use of graph databases as a way of communication between biological data repositories, we conclude that efficient design, querying, and maintenance of graph databases will be key for knowledge generation in systems biology and other research fields with heterogeneous data.
Collapse
Affiliation(s)
- Ilya Mazein
- Medical Informatics Laboratory, University Medicine Greifswald, Walther-Rathenau-Straße 48, Greifswald 17475, Germany
| | - Adrien Rougny
- Luxembourg Centre for Systems Biology, University of Luxembourg, 6 Avenue du Swing, Belvaux L-4367, Luxembourg
| | - Alexander Mazein
- Luxembourg Centre for Systems Biology, University of Luxembourg, 6 Avenue du Swing, Belvaux L-4367, Luxembourg
| | - Ron Henkel
- Medical Informatics Laboratory, University Medicine Greifswald, Walther-Rathenau-Straße 48, Greifswald 17475, Germany
| | - Lea Gütebier
- Medical Informatics Laboratory, University Medicine Greifswald, Walther-Rathenau-Straße 48, Greifswald 17475, Germany
| | - Lea Michaelis
- Medical Informatics Laboratory, University Medicine Greifswald, Walther-Rathenau-Straße 48, Greifswald 17475, Germany
| | - Marek Ostaszewski
- Luxembourg Centre for Systems Biology, University of Luxembourg, 6 Avenue du Swing, Belvaux L-4367, Luxembourg
| | - Reinhard Schneider
- Luxembourg Centre for Systems Biology, University of Luxembourg, 6 Avenue du Swing, Belvaux L-4367, Luxembourg
| | - Venkata Satagopam
- Luxembourg Centre for Systems Biology, University of Luxembourg, 6 Avenue du Swing, Belvaux L-4367, Luxembourg
| | - Lars Juhl Jensen
- Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Grønnegårdsvej 15, 1870 Frederiksberg C, Denmark
| | - Dagmar Waltemath
- Medical Informatics Laboratory, University Medicine Greifswald, Walther-Rathenau-Straße 48, Greifswald 17475, Germany
| | - Judith A H Wodke
- Medical Informatics Laboratory, University Medicine Greifswald, Walther-Rathenau-Straße 48, Greifswald 17475, Germany
| | - Irina Balaur
- Luxembourg Centre for Systems Biology, University of Luxembourg, 6 Avenue du Swing, Belvaux L-4367, Luxembourg
| |
Collapse
|
41
|
Zhao MX, Ding RF, Chen Q, Meng J, Li F, Fu S, Huang B, Liu Y, Ji ZL, Zhao Y. Nphos: Database and Predictor of Protein N-phosphorylation. GENOMICS, PROTEOMICS & BIOINFORMATICS 2024; 22:qzae032. [PMID: 39380205 DOI: 10.1093/gpbjnl/qzae032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Revised: 03/03/2024] [Accepted: 04/01/2024] [Indexed: 10/10/2024]
Abstract
Protein N-phosphorylation is widely present in nature and participates in various biological processes. However, current knowledge on N-phosphorylation is extremely limited compared to that on O-phosphorylation. In this study, we collected 11,710 experimentally verified N-phosphosites of 7344 proteins from 39 species and subsequently constructed the database Nphos to share up-to-date information on protein N-phosphorylation. Upon these substantial data, we characterized the sequential and structural features of protein N-phosphorylation. Moreover, after comparing hundreds of learning models, we chose and optimized gradient boosting decision tree (GBDT) models to predict three types of human N-phosphorylation, achieving mean area under the receiver operating characteristic curve (AUC) values of 90.56%, 91.24%, and 92.01% for pHis, pLys, and pArg, respectively. Meanwhile, we discovered 488,825 distinct N-phosphosites in the human proteome. The models were also deployed in Nphos for interactive N-phosphosite prediction. In summary, this work provides new insights and points for both flexible and focused investigations of N-phosphorylation. It will also facilitate a deeper and more systematic understanding of protein N-phosphorylation modification by providing a data and technical foundation. Nphos is freely available at http://www.bio-add.org/Nphos/ and http://ppodd.org.cn/Nphos/.
Collapse
Affiliation(s)
- Ming-Xiao Zhao
- Institute of Drug Discovery Technology, Ningbo University, Ningbo 315211, China
- Department of Chemical Biology, Key Laboratory for Chemical Biology of Fujian Province, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen 361005, China
| | - Ruo-Fan Ding
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen 361102, China
| | - Qiang Chen
- Zhejiang Key Laboratory of Pathophysiology, Department of Biochemistry and Molecular Biology, Health Science Center, Ningbo University, Ningbo 315211, China
| | - Junhua Meng
- BGI Genomics, BGI-Shenzhen, Shenzhen 518083, China
| | - Fulai Li
- Institute of Drug Discovery Technology, Ningbo University, Ningbo 315211, China
| | - Songsen Fu
- Institute of Drug Discovery Technology, Ningbo University, Ningbo 315211, China
| | - Biling Huang
- Institute of Drug Discovery Technology, Ningbo University, Ningbo 315211, China
| | - Yan Liu
- Department of Chemical Biology, Key Laboratory for Chemical Biology of Fujian Province, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen 361005, China
| | - Zhi-Liang Ji
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen 361102, China
| | - Yufen Zhao
- Institute of Drug Discovery Technology, Ningbo University, Ningbo 315211, China
- Department of Chemical Biology, Key Laboratory for Chemical Biology of Fujian Province, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen 361005, China
- Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology, Department of Chemistry, Tsinghua University, Beijing 100084, China
| |
Collapse
|
42
|
Cingiz MÖ. Ensemble decision of local similarity indices on the biological network for disease related gene prediction. PeerJ 2024; 12:e17975. [PMID: 39247551 PMCID: PMC11380840 DOI: 10.7717/peerj.17975] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Accepted: 08/05/2024] [Indexed: 09/10/2024] Open
Abstract
Link prediction (LP) is a task for the identification of potential, missing and spurious links in complex networks. Protein-protein interaction (PPI) networks are important for understanding the underlying biological mechanisms of diseases. Many complex networks have been constructed using LP methods; however, there are a limited number of studies that focus on disease-related gene predictions and evaluate these genes using various evaluation criteria. The main objective of the study is to investigate the effect of a simple ensemble method in disease related gene predictions. Local similarity indices (LSIs) based disease related gene predictions were integrated by a simple ensemble decision method, simple majority voting (SMV), on the PPI network to detect accurate disease related genes. Human PPI network was utilized to discover potential disease related genes using four LSIs for the gene prediction. LSIs discovered potential links between disease related genes, which were obtained from OMIM database for gastric, colorectal, breast, prostate and lung cancers. LSIs based disease related genes were ranked due to their LSI scores in descending order for retrieving the top 10, 50 and 100 disease related genes. SMV integrated four LSIs based predictions to obtain SMV based the top 10, 50 and 100 disease related genes. The performance of LSIs based and SMV based genes were evaluated separately by employing overlap analyses, which were performed with GeneCard disease-gene relation dataset and Gene Ontology (GO) terms. The GO-terms were used for biological assessment for the inferred gene lists by LSIs and SMV on all cancer types. Adamic-Adar (AA), Resource Allocation Index (RAI), and SMV based gene lists are generally achieved good performance results on all cancers in both overlap analyses. SMV also outperformed on breast cancer data. The increment in the selection of the number of the top ranked disease related genes also enhanced the performance results of SMV.
Collapse
Affiliation(s)
- Mustafa Özgür Cingiz
- Department of Computer Engineering, Faculty of Engineering and Natural Sciences, Bursa Technical University, Bursa, Turkey
| |
Collapse
|
43
|
Guo X, Song Y, Xu D, Jin X, Shang X. Genotype and Phenotype Association Analysis Based on Multi-omics Statistical Data. Curr Bioinform 2024; 19:933-942. [DOI: 10.2174/0115748936276861240109045208] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Revised: 11/29/2023] [Accepted: 12/07/2023] [Indexed: 01/03/2025]
Abstract
Background:
When using clinical data for multi-omics analysis, there are issues such as
the insufficient number of omics data types and relatively small sample size due to the protection of
patients' privacy, the requirements of data management by various institutions, and the relatively
large number of features of each omics data. This paper describes the analysis of multi-omics pathway
relationships using statistical data in the absence of clinical data.
Methods:
We proposed a novel approach to exploit easily accessible statistics in public databases.
This approach introduces phenotypic associations that are not included in the clinical data and uses
these data to build a three-layer heterogeneous network. To simplify the analysis, we decomposed
the three-layer network into double two-layer networks to predict the weights of the inter-layer associations.
By adding a hyperparameter β, the weights of the two layers of the network were
merged, and then k-fold cross-validation was used to evaluate the accuracy of this method. In calculating
the weights of the two-layer networks, the RWR with fixed restart probability was combined
with PBMDA and CIPHER to generate the PCRWR with biased weights and improved accuracy.
Results:
The area under the receiver operating characteristic curve was increased by approximately
7% in the case of the RWR with initial weights.
Conclusion:
Multi-omics statistical data were used to establish genotype and phenotype correlation
networks for analysis, which was similar to the effect of clinical multi-omics analysis.
Collapse
Affiliation(s)
- Xinpeng Guo
- School of Air and Missile Defense, Air Force Engineering University, Xi’an, 710051, People’s Republic of China
- School of Computer Science and Engineering, Northwestern Polytechnical University, Xi’an 710072, People’s Republic of China
| | - Yafei Song
- School of Air and Missile Defense, Air Force Engineering University, Xi’an, 710051, People’s Republic of China
| | - Dongyan Xu
- Department of Basic Sciences, Air Force Engineering University, Xi’an, 710051, People’s Republic
of China
| | - Xueping Jin
- School of Air and Missile Defense, Air Force Engineering University, Xi’an, 710051, People’s Republic of China
| | - Xuequn Shang
- School of Computer Science and Engineering, Northwestern Polytechnical University, Xi’an 710072, People’s
Republic of China
| |
Collapse
|
44
|
Li Z, Zhang Y, Zhou P. Temporal Protein Complex Identification Based on Dynamic Heterogeneous Protein Information Network Representation Learning. IEEE/ACM TRANSACTIONS ON COMPUTATIONAL BIOLOGY AND BIOINFORMATICS 2024; 21:1154-1164. [PMID: 38190662 DOI: 10.1109/tcbb.2024.3351078] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/10/2024]
Abstract
Protein complexes, as the fundamental units of cellular function and regulation, play a crucial role in understanding the normal physiological functions of cells. Existing methods for protein complex identification attempt to introduce other biological information on top of the protein-protein interaction (PPI) network to assist in evaluating the degree of association between proteins. However, these methods usually treat protein interaction networks as flat homogeneous static networks. They cannot distinguish the roles and importance of different types of biological information, nor can they reflect the dynamic changes of protein complexes. In recent years, heterogeneous network representation learning has achieved great success in processing complex heterogeneous information and mining deep semantics. We thus propose a temporal protein complex identification method based on Dynamic Heterogeneous Protein information network Representation Learning, DHPRL. DHPRL naturally integrates multiple types of heterogeneous biological information in the cellular temporal dimension. It simultaneously models the temporal dynamic properties of proteins and the heterogeneity of biological information to improve the understanding of protein interactions and the accuracy of complex prediction. Firstly, we construct Dynamic Heterogeneous Protein Information Network (DHPIN) by integrating temporal gene expression information and GO attribute information. Then we design a dual-view collaborative contrast mechanism. Specifically, proposing to learn protein representations from two views of DHPIN (1-hop relation view and meta-path view) to model the consistency and specificity between nearest-neighbour bio information and deeper biological semantics. The dynamic PPI network is thereafter re-weighted based on the learned protein representations. Finally, we perform protein identification on the re-weighted dynamic PPI network. Extensive experimental results demonstrate that DHPRL can effectively model complicated biological information and achieve state-of-the-art performance in most cases.
Collapse
|
45
|
Zhang B, Niu D, Zhang L, Zhang Q, Li Z. MSH-DTI: multi-graph convolution with self-supervised embedding and heterogeneous aggregation for drug-target interaction prediction. BMC Bioinformatics 2024; 25:275. [PMID: 39179993 PMCID: PMC11342675 DOI: 10.1186/s12859-024-05904-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Accepted: 08/16/2024] [Indexed: 08/26/2024] Open
Abstract
BACKGROUND The rise of network pharmacology has led to the widespread use of network-based computational methods in predicting drug target interaction (DTI). However, existing DTI prediction models typically rely on a limited amount of data to extract drug and target features, potentially affecting the comprehensiveness and robustness of features. In addition, although multiple networks are used for DTI prediction, the integration of heterogeneous information often involves simplistic aggregation and attention mechanisms, which may impose certain limitations. RESULTS MSH-DTI, a deep learning model for predicting drug-target interactions, is proposed in this paper. The model uses self-supervised learning methods to obtain drug and target structure features. A Heterogeneous Interaction-enhanced Feature Fusion Module is designed for multi-graph construction, and the graph convolutional networks are used to extract node features. With the help of an attention mechanism, the model focuses on the important parts of different features for prediction. Experimental results show that the AUROC and AUPR of MSH-DTI are 0.9620 and 0.9605 respectively, outperforming other models on the DTINet dataset. CONCLUSION The proposed MSH-DTI is a helpful tool to discover drug-target interactions, which is also validated through case studies in predicting new DTIs.
Collapse
Affiliation(s)
- Beiyi Zhang
- College of Computer Science and Technology, Qingdao University, Ningxia Road, Qingdao, 266071, Shandong, China
| | - Dongjiang Niu
- College of Computer Science and Technology, Qingdao University, Ningxia Road, Qingdao, 266071, Shandong, China
| | - Lianwei Zhang
- College of Computer Science and Technology, Qingdao University, Ningxia Road, Qingdao, 266071, Shandong, China
| | - Qiang Zhang
- College of Computer Science and Technology, Qingdao University, Ningxia Road, Qingdao, 266071, Shandong, China
| | - Zhen Li
- College of Computer Science and Technology, Qingdao University, Ningxia Road, Qingdao, 266071, Shandong, China.
| |
Collapse
|
46
|
Cebolla JJ, Giraldo P, Gómez J, Montoto C, Gervas-Arruga J. Machine Learning-Driven Biomarker Discovery for Skeletal Complications in Type 1 Gaucher Disease Patients. Int J Mol Sci 2024; 25:8586. [PMID: 39201273 PMCID: PMC11354847 DOI: 10.3390/ijms25168586] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Revised: 08/01/2024] [Accepted: 08/05/2024] [Indexed: 09/02/2024] Open
Abstract
Type 1 Gaucher disease (GD1) is a rare, autosomal recessive disorder caused by glucocerebrosidase deficiency. Skeletal manifestations represent one of the most debilitating and potentially irreversible complications of GD1. Although imaging studies are the gold standard, early diagnostic/prognostic tools, such as molecular biomarkers, are needed for the rapid management of skeletal complications. This study aimed to identify potential protein biomarkers capable of predicting the early diagnosis of bone skeletal complications in GD1 patients using artificial intelligence. An in silico study was performed using the novel Therapeutic Performance Mapping System methodology to construct mathematical models of GD1-associated complications at the protein level. Pathophysiological characterization was performed before modeling, and a data science strategy was applied to the predicted protein activity for each protein in the models to identify classifiers. Statistical criteria were used to prioritize the most promising candidates, and 18 candidates were identified. Among them, PDGFB, IL1R2, PTH and CCL3 (MIP-1α) were highlighted due to their ease of measurement in blood. This study proposes a validated novel tool to discover new protein biomarkers to support clinician decision-making in an area where medical needs have not yet been met. However, confirming the results using in vitro and/or in vivo studies is necessary.
Collapse
Affiliation(s)
| | - Pilar Giraldo
- FEETEG, 50006 Zaragoza, Spain;
- Hospital QuirónSalud Zaragoza, 50012 Zaragoza, Spain
| | | | | | | |
Collapse
|
47
|
Gabriel GC, Ganapathiraju M, Lo CW. The Role of Cilia and the Complex Genetics of Congenital Heart Disease. Annu Rev Genomics Hum Genet 2024; 25:309-327. [PMID: 38724024 DOI: 10.1146/annurev-genom-121222-105345] [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: 08/29/2024]
Abstract
Congenital heart disease (CHD) can affect up to 1% of live births, and despite abundant evidence of a genetic etiology, the genetic landscape of CHD is still not well understood. A large-scale mouse chemical mutagenesis screen for mutations causing CHD yielded a preponderance of cilia-related genes, pointing to a central role for cilia in CHD pathogenesis. The genes uncovered by the screen included genes that regulate ciliogenesis and cilia-transduced cell signaling as well as many that mediate endocytic trafficking, a cell process critical for both ciliogenesis and cell signaling. The clinical relevance of these findings is supported by whole-exome sequencing analysis of CHD patients that showed enrichment for pathogenic variants in ciliome genes. Surprisingly, among the ciliome CHD genes recovered were many that encoded direct protein-protein interactors. Assembly of the CHD genes into a protein-protein interaction network yielded a tight interactome that suggested this protein-protein interaction may have functional importance and that its disruption could contribute to the pathogenesis of CHD. In light of these and other findings, we propose that an interactome enriched for ciliome genes may provide the genomic context for the complex genetics of CHD and its often-observed incomplete penetrance and variable expressivity.
Collapse
Affiliation(s)
- George C Gabriel
- Department of Developmental Biology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA; ,
| | - Madhavi Ganapathiraju
- Carnegie Mellon University in Qatar, Doha, Qatar
- Department of Biomedical Informatics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA;
| | - Cecilia W Lo
- Department of Developmental Biology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA; ,
| |
Collapse
|
48
|
Teimouri H, Medvedeva A, Kolomeisky AB. Unraveling the role of physicochemical differences in predicting protein-protein interactions. J Chem Phys 2024; 161:045102. [PMID: 39051836 DOI: 10.1063/5.0219501] [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: 05/17/2024] [Accepted: 07/09/2024] [Indexed: 07/27/2024] Open
Abstract
The ability to accurately predict protein-protein interactions is critically important for understanding major cellular processes. However, current experimental and computational approaches for identifying them are technically very challenging and still have limited success. We propose a new computational method for predicting protein-protein interactions using only primary sequence information. It utilizes the concept of physicochemical similarity to determine which interactions will most likely occur. In our approach, the physicochemical features of proteins are extracted using bioinformatics tools for different organisms. Then they are utilized in a machine-learning method to identify successful protein-protein interactions via correlation analysis. It was found that the most important property that correlates most with the protein-protein interactions for all studied organisms is dipeptide amino acid composition (the frequency of specific amino acid pairs in a protein sequence). While current approaches often overlook the specificity of protein-protein interactions with different organisms, our method yields context-specific features that determine protein-protein interactions. The analysis is specifically applied to the bacterial two-component system that includes histidine kinase and transcriptional response regulators, as well as to the barnase-barstar complex, demonstrating the method's versatility across different biological systems. Our approach can be applied to predict protein-protein interactions in any biological system, providing an important tool for investigating complex biological processes' mechanisms.
Collapse
Affiliation(s)
- Hamid Teimouri
- Department of Chemistry, Rice University, Houston, Texas 77005, USA
- Center for Theoretical Biological Physics, Rice University, Houston, Texas 77005, USA
- Department of Chemical and Biomolecular Engineering, Rice University, Houston, Texas 77005, USA
| | - Angela Medvedeva
- Department of Chemistry, Rice University, Houston, Texas 77005, USA
- Center for Theoretical Biological Physics, Rice University, Houston, Texas 77005, USA
- Department of Chemical and Biomolecular Engineering, Rice University, Houston, Texas 77005, USA
| | - Anatoly B Kolomeisky
- Department of Chemistry, Rice University, Houston, Texas 77005, USA
- Center for Theoretical Biological Physics, Rice University, Houston, Texas 77005, USA
- Department of Chemical and Biomolecular Engineering, Rice University, Houston, Texas 77005, USA
| |
Collapse
|
49
|
Zhu Y, Ning C, Zhang N, Wang M, Zhang Y. GSRF-DTI: a framework for drug-target interaction prediction based on a drug-target pair network and representation learning on a large graph. BMC Biol 2024; 22:156. [PMID: 39020316 PMCID: PMC11256582 DOI: 10.1186/s12915-024-01949-3] [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] [Received: 09/11/2023] [Accepted: 07/01/2024] [Indexed: 07/19/2024] Open
Abstract
BACKGROUND Identification of potential drug-target interactions (DTIs) with high accuracy is a key step in drug discovery and repositioning, especially concerning specific drug targets. Traditional experimental methods for identifying the DTIs are arduous, time-intensive, and financially burdensome. In addition, robust computational methods have been developed for predicting the DTIs and are widely applied in drug discovery research. However, advancing more precise algorithms for predicting DTIs is essential to meet the stringent standards demanded by drug discovery. RESULTS We proposed a novel method called GSRF-DTI, which integrates networks with a deep learning algorithm to identify DTIs. Firstly, GSRF-DTI learned the embedding representation of drugs and targets by integrating multiple drug association information and target association information, respectively. Then, GSRF-DTI considered the influence of drug-target pair (DTP) association on DTI prediction to construct a drug-target pair network (DTP-NET). Next, we utilized GraphSAGE on DTP-NET to learn the potential features of the network and applied random forest (RF) to predict the DTIs. Furthermore, we conducted ablation experiments to validate the necessity of integrating different types of network features for identifying DTIs. It is worth noting that GSRF-DTI proposed three novel DTIs. CONCLUSIONS GSRF-DTI not only considered the influence of the interaction relationship between drug and target but also considered the impact of DTP association relationship on DTI prediction. We initially use GraphSAGE to aggregate the neighbor information of nodes for better identification. Experimental analysis on Luo's dataset and the newly constructed dataset revealed that the GSRF-DTI framework outperformed several state-of-the-art methods significantly.
Collapse
Affiliation(s)
- Yongdi Zhu
- School of Mathematics and Statistics, Shandong University, Weihai, Shandong, China
| | - Chunhui Ning
- School of Mathematics and Statistics, Shandong University, Weihai, Shandong, China
| | - Naiqian Zhang
- School of Mathematics and Statistics, Shandong University, Weihai, Shandong, China
| | - Mingyi Wang
- Department of Central Lab, Weihai Municipal Hospital, Weihai, Shandong, China.
| | - Yusen Zhang
- School of Mathematics and Statistics, Shandong University, Weihai, Shandong, China.
| |
Collapse
|
50
|
Abu-Bakar A, Ismail M, Zulkifli MZI, Zaini NAS, Shukor NIA, Harun S, Inayat-Hussain SH. Mapping the influence of hydrocarbons mixture on molecular mechanisms, involved in breast and lung neoplasms: in silico toxicogenomic data-mining. Genes Environ 2024; 46:15. [PMID: 38982523 PMCID: PMC11232146 DOI: 10.1186/s41021-024-00310-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Accepted: 06/07/2024] [Indexed: 07/11/2024] Open
Abstract
BACKGROUND Exposure to chemical mixtures inherent in air pollution, has been shown to be associated with the risk of breast and lung cancers. However, studies on the molecular mechanisms of exposure to a mixture of these pollutants, such as hydrocarbons, in the development of breast and lung cancers are scarce. We utilized in silico toxicogenomic analysis to elucidate the molecular pathways linked to both cancers that are influenced by exposure to a mixture of selected hydrocarbons. The Comparative Toxicogenomics Database and Cytoscape software were used for data mining and visualization. RESULTS Twenty-five hydrocarbons, common in air pollution with carcinogenicity classification of 1 A/B or 2 (known/presumed or suspected human carcinogen), were divided into three groups: alkanes and alkenes, halogenated hydrocarbons, and polyaromatic hydrocarbons. The in silico data-mining revealed 87 and 44 genes commonly interacted with most of the investigated hydrocarbons are linked to breast and lung cancer, respectively. The dominant interactions among the common genes are co-expression, physical interaction, genetic interaction, co-localization, and interaction in shared protein domains. Among these genes, only 16 are common in the development of both cancers. Benzo(a)pyrene and tetrachlorodibenzodioxin interacted with all 16 genes. The molecular pathways potentially affected by the investigated hydrocarbons include aryl hydrocarbon receptor, chemical carcinogenesis, ferroptosis, fluid shear stress and atherosclerosis, interleukin 17 signaling pathway, lipid and atherosclerosis, NRF2 pathway, and oxidative stress response. CONCLUSIONS Within the inherent limitations of in silico toxicogenomics tools, we elucidated the molecular pathways associated with breast and lung cancer development potentially affected by hydrocarbons mixture. Our findings indicate adaptive responses to oxidative stress and inflammatory damages are instrumental in the development of both cancers. Additionally, ferroptosis-a non-apoptotic programmed cell death driven by lipid peroxidation and iron homeostasis-was identified as a new player in these responses. Finally, AHR potential involvement in modulating IL-8, a critical gene that mediates breast cancer invasion and metastasis to the lungs, was also highlighted. A deeper understanding of the interplay between genes associated with these pathways, and other survival signaling pathways identified in this study, will provide invaluable knowledge in assessing the risk of inhalation exposure to hydrocarbons mixture. The findings offer insights into future in vivo and in vitro laboratory investigations that focus on inhalation exposure to the hydrocarbons mixture.
Collapse
Affiliation(s)
- A'edah Abu-Bakar
- Product Stewardship and Toxicology, Environment, Social Performance & Product Stewardship (ESPPS), Group Health, Safety and Environment (GHSE), Petroliam Nasional Berhad (PETRONAS), Kuala Lumpur, 50088, Malaysia.
| | - Maihani Ismail
- Product Stewardship and Toxicology, Environment, Social Performance & Product Stewardship (ESPPS), Group Health, Safety and Environment (GHSE), Petroliam Nasional Berhad (PETRONAS), Kuala Lumpur, 50088, Malaysia.
| | - M Zaqrul Ieman Zulkifli
- Product Stewardship and Toxicology, Environment, Social Performance & Product Stewardship (ESPPS), Group Health, Safety and Environment (GHSE), Petroliam Nasional Berhad (PETRONAS), Kuala Lumpur, 50088, Malaysia
| | - Nur Aini Sofiyya Zaini
- Product Stewardship and Toxicology, Environment, Social Performance & Product Stewardship (ESPPS), Group Health, Safety and Environment (GHSE), Petroliam Nasional Berhad (PETRONAS), Kuala Lumpur, 50088, Malaysia
| | - Nur Izzah Abd Shukor
- Health, Safety and Environment (HSE), KLCC Urusharta, Kuala Lumpur, 50088, Malaysia
| | - Sarahani Harun
- Institute of Systems Biology, Universiti Kebangsaan Malaysia, Bangi, Selangor, 43600 UKM, Malaysia
| | - Salmaan Hussain Inayat-Hussain
- ESPPS, GHSE, PETRONAS, Kuala Lumpur, 50088, Malaysia
- Department of Environmental Health Sciences, Yale School of Public Health, Yale University, 60 College St, New Haven, CT, 06250, USA
| |
Collapse
|