1
|
Venturini P, Faria PL, Cordeiro JV. AI and omics technologies in biobanking: Applications and challenges for public health. Public Health 2025; 243:105726. [PMID: 40315692 DOI: 10.1016/j.puhe.2025.105726] [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: 10/19/2024] [Revised: 03/10/2025] [Accepted: 04/08/2025] [Indexed: 05/04/2025]
Abstract
OBJECTIVES Considering the growing intersection of biobanks, artificial intelligence (AI) and omics research, and their critical impact on public health, this study aimed to explore the current and future public health implications and challenges of AI and omics-driven innovations in biobanking. STUDY DESIGN Narrative literature review. METHODS A structured literature search was conducted in Scopus, PubMed, Web of Science and IEEExplore databases using relevant search terms. Additional references were identified through backward and forward citation chaining. Key themes were aggregated and analysed through thematic analysis. RESULTS Thirty-seven studies were selected for analysis, leading to the identification and categorisation of key developments. Several key technical, ethical and implementation challenges were also identified, including AI model selection, data accessibility, variability and quality issues, lack of robust and standardised validation methods, explainability, accountability, lack of transparency, algorithmic bias, privacy, security and fairness issues, and governance model selection. Based on these results, potential future scenarios of AI and omics integration in biobanking and their related public health implications were considered. CONCLUSIONS While AI and omics-driven innovations in biobanking offer specific transformative public health benefits, addressing their technical, ethical and implementation challenges is crucial. Robust regulatory frameworks, feasible governance models, access to quality data, interdisciplinary collaboration, and transparent and validated AI systems are essential to maximise benefits and mitigate risks. Further research and policy development are needed to support the responsible integration of these technologies in biobanking and public health.
Collapse
Affiliation(s)
- Pedro Venturini
- NOVA National School of Public Health, NOVA University Lisbon, Lisbon, Portugal.
| | - Paula Lobato Faria
- NOVA National School of Public Health, NOVA University Lisbon, Lisbon, Portugal; NOVA National School of Public Health, Public Health Research Centre, Comprehensive Health Research Center, CHRC, REAL, CCAL, NOVA University Lisbon, Avenida Padre Cruz, Lisbon, 1600-560, Portugal; Interdisciplinary Center of Social Sciences, NOVA University of Lisbon, Portugal
| | - João V Cordeiro
- NOVA National School of Public Health, Public Health Research Centre, Comprehensive Health Research Center, CHRC, REAL, CCAL, NOVA University Lisbon, Avenida Padre Cruz, Lisbon, 1600-560, Portugal; Interdisciplinary Center of Social Sciences, NOVA University of Lisbon, Portugal
| |
Collapse
|
2
|
Yao L, Shah SR, Ozer A, Zhang J, Pan X, Xia T, Fangal VD, Leung AKY, Wei M, Lis JT, Yu H. High-resolution reconstruction of cell-type specific transcriptional regulatory processes from bulk sequencing samples. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.04.02.646189. [PMID: 40291712 PMCID: PMC12026507 DOI: 10.1101/2025.04.02.646189] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/30/2025]
Abstract
Biological systems exhibit remarkable heterogeneity, characterized by intricate interplay among diverse cell types. Resolving the regulatory processes of specific cell types is crucial for delineating developmental mechanisms and disease etiologies. While single-cell sequencing methods such as scRNA-seq and scATAC-seq have revolutionized our understanding of individual cellular functions, adapting bulk genome-wide assays to achieve single-cell resolution of other genomic features remains a significant technical challenge. Here, we introduce Deep-learning-based DEconvolution of Tissue profiles with Accurate Interpretation of Locus-specific Signals (DeepDETAILS), a novel quasi-supervised framework to reconstruct cell-type-specific genomic signals with base-pair precision. DeepDETAILS' core innovation lies in its ability to perform cross-modality deconvolution using scATAC-seq reference libraries for other bulk datasets, benefiting from the affordability and availability of scATAC-seq data. DeepDETAILS enables high-resolution mapping of genomic signals across diverse cell types, with great versatility for various omics datasets, including nascent transcript sequencing (such as PRO-cap and PRO-seq) and ChIP-seq for chromatin modifications. Our results demonstrate that DeepDETAILS significantly outperformed traditional statistical deconvolution methods. Using DeepDETAILS, we developed a comprehensive compendium of high-resolution nascent transcription and histone modification signals across 39 diverse human tissues and 86 distinct cell types. Furthermore, we applied our compendium to fine-map risk variants associated with Primary Sclerosing Cholangitis (PSC), a progressive cholestatic liver disorder, and revealed a potential etiology of the disease. Our tool and compendium provide invaluable insights into cellular complexity, opening new avenues for studying biological processes in various contexts.
Collapse
|
3
|
Stojchevski R, Sutanto EA, Sutanto R, Hadzi-Petrushev N, Mladenov M, Singh SR, Sinha JK, Ghosh S, Yarlagadda B, Singh KK, Verma P, Sengupta S, Bhaskar R, Avtanski D. Translational Advances in Oncogene and Tumor-Suppressor Gene Research. Cancers (Basel) 2025; 17:1008. [PMID: 40149342 PMCID: PMC11940485 DOI: 10.3390/cancers17061008] [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: 02/10/2025] [Revised: 03/10/2025] [Accepted: 03/15/2025] [Indexed: 03/29/2025] Open
Abstract
Cancer, characterized by the uncontrolled proliferation of cells, is one of the leading causes of death globally, with approximately one in five people developing the disease in their lifetime. While many driver genes were identified decades ago, and most cancers can be classified based on morphology and progression, there is still a significant gap in knowledge about genetic aberrations and nuclear DNA damage. The study of two critical groups of genes-tumor suppressors, which inhibit proliferation and promote apoptosis, and oncogenes, which regulate proliferation and survival-can help to understand the genomic causes behind tumorigenesis, leading to more personalized approaches to diagnosis and treatment. Aberration of tumor suppressors, which undergo two-hit and loss-of-function mutations, and oncogenes, activated forms of proto-oncogenes that experience one-hit and gain-of-function mutations, are responsible for the dysregulation of key signaling pathways that regulate cell division, such as p53, Rb, Ras/Raf/ERK/MAPK, PI3K/AKT, and Wnt/β-catenin. Modern breakthroughs in genomics research, like next-generation sequencing, have provided efficient strategies for mapping unique genomic changes that contribute to tumor heterogeneity. Novel therapeutic approaches have enabled personalized medicine, helping address genetic variability in tumor suppressors and oncogenes. This comprehensive review examines the molecular mechanisms behind tumor-suppressor genes and oncogenes, the key signaling pathways they regulate, epigenetic modifications, tumor heterogeneity, and the drug resistance mechanisms that drive carcinogenesis. Moreover, the review explores the clinical application of sequencing techniques, multiomics, diagnostic procedures, pharmacogenomics, and personalized treatment and prevention options, discussing future directions for emerging technologies.
Collapse
Affiliation(s)
- Radoslav Stojchevski
- Friedman Diabetes Institute, Lenox Hill Hospital, Northwell Health, New York, NY 10022, USA;
- Feinstein Institutes for Medical Research, Manhasset, NY 11030, USA
- Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY 11549, USA
| | - Edward Agus Sutanto
- CUNY School of Medicine, The City College of New York, 160 Convent Avenue, New York, NY 10031, USA;
| | - Rinni Sutanto
- New York Institute of Technology College of Osteopathic Medicine, Glen Head, NY 11545, USA;
| | - Nikola Hadzi-Petrushev
- Faculty of Natural Sciences and Mathematics, Institute of Biology, Ss. Cyril and Methodius University, 1000 Skopje, North Macedonia; (N.H.-P.)
| | - Mitko Mladenov
- Faculty of Natural Sciences and Mathematics, Institute of Biology, Ss. Cyril and Methodius University, 1000 Skopje, North Macedonia; (N.H.-P.)
| | - Sajal Raj Singh
- GloNeuro, Sector 107, Vishwakarma Road, Noida 201301, Uttar Pradesh, India (J.K.S.)
| | - Jitendra Kumar Sinha
- GloNeuro, Sector 107, Vishwakarma Road, Noida 201301, Uttar Pradesh, India (J.K.S.)
| | - Shampa Ghosh
- GloNeuro, Sector 107, Vishwakarma Road, Noida 201301, Uttar Pradesh, India (J.K.S.)
| | | | - Krishna Kumar Singh
- Symbiosis Centre for Information Technology (SCIT), Rajiv Gandhi InfoTech Park, Hinjawadi, Pune 411057, Maharashtra, India;
| | - Prashant Verma
- School of Management, BML Munjal University, NH8, Sidhrawali, Gurugram 122413, Haryana, India
| | - Sonali Sengupta
- Department of Gastroenterology, All India Institute of Medical Sciences (AIIMS), New Delhi 110029, India
| | - Rakesh Bhaskar
- School of Chemical Engineering, Yeungnam University, Gyeongsan 38541, Republic of Korea
- Research Institute of Cell Culture, Yeungnam University, Gyeongsan 38541, Republic of Korea
| | - Dimiter Avtanski
- Friedman Diabetes Institute, Lenox Hill Hospital, Northwell Health, New York, NY 10022, USA;
- Feinstein Institutes for Medical Research, Manhasset, NY 11030, USA
- Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY 11549, USA
| |
Collapse
|
4
|
Melograna F, Sudhakar P, Yousefi B, Caenepeel C, Falony G, Vieira-Silva S, Krishnamoorthy S, Fardo D, Verstockt B, Raes J, Vermeire S, Van Steen K. Individual-network based predictions of microbial interaction signatures for response to biological therapies in IBD patients. Front Mol Biosci 2025; 11:1490533. [PMID: 39944755 PMCID: PMC11813754 DOI: 10.3389/fmolb.2024.1490533] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2024] [Accepted: 12/31/2024] [Indexed: 03/06/2025] Open
Abstract
Inflammatory Bowel Disease (IBD), which includes Ulcerative Colitis (UC) and Crohn's Disease (CD), is marked by dysbiosis of the gut microbiome. Despite therapeutic interventions with biological agents like Vedolizumab, Ustekinumab, and anti-TNF agents, the variability in clinical, histological, and molecular responses remains significant due to inter-individual and inter-population differences. This study introduces a novel approach using Individual Specific Networks (ISNs) derived from faecal microbial measurements of IBD patients across multiple cohorts. These ISNs, constructed from baseline and follow-up data post-treatment, successfully predict therapeutic outcomes based on endoscopic remission criteria. Our analysis revealed that ISNs characterised by core gut microbial families, including Lachnospiraceae and Ruminococcaceae, are predictive of treatment responses. We identified significant changes in abundance levels of specific bacterial genera in response to treatment, confirming the robustness of ISNs in capturing both linear and non-linear microbiota signals. Utilising network topological metrics, we further validated these findings, demonstrating that critical microbial features identified through ISNs can differentiate responders from non-responders with respect to various therapeutic outcomes. The study highlights the potential of ISNs to provide individualised insights into microbiota-driven therapeutic responses, emphasising the need for larger cohort studies to enhance the accuracy of molecular biomarkers. This innovative methodology paves the way for more personalised and effective treatment strategies in managing IBD.
Collapse
Affiliation(s)
- Federico Melograna
- BIO3 Laboratory for Systems Medicine, Department of Human Genetics, KU Leuven, Leuven, Belgium
| | - Padhmanand Sudhakar
- Department of Biotechnology, Kumaraguru College Technology, Coimbatore, Tamil Nadu, India
| | - Behnam Yousefi
- Institute of Medical Systems Biology, Center for Biomedical AI (bAIome), Center for Molecular Neurobiology (ZMNH), University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- German Center for Child and Adolescent Health (DZKJ), Partner Site Hamburg, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Clara Caenepeel
- Department of Gastroenterology and Hepatology, University Hospitals Leuven, KU Leuven, Leuven, Belgium
| | - Gwen Falony
- Laboratory of Molecular Bacteriology, Department of Microbiology and Immunology, Rega Institute, Katholieke Universiteit Leuven, Leuven, Belgium
- Center for Microbiology, Vlaams Instituut voor Biotechnologie (VIB), Leuven, Belgium
- Institute of Medical Microbiology and Hygiene and Research Center for Immunotherapy (FZI), University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Sara Vieira-Silva
- Laboratory of Molecular Bacteriology, Department of Microbiology and Immunology, Rega Institute, Katholieke Universiteit Leuven, Leuven, Belgium
- Institute of Medical Microbiology and Hygiene and Research Center for Immunotherapy (FZI), University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
- Institute of Molecular Biology (IMB), Mainz, Germany
| | | | - David Fardo
- College of Public Health, University of Kentucky, Lexington, KY, United States
| | - Bram Verstockt
- Department of Gastroenterology and Hepatology, University Hospitals Leuven, KU Leuven, Leuven, Belgium
- KU Leuven Department of Chronic Diseases and Metabolism, Translational Research Center for Gastrointestinal Disorders (TARGID), Leuven, Belgium
| | - Jeroen Raes
- Laboratory of Molecular Bacteriology, Department of Microbiology and Immunology, Rega Institute, Katholieke Universiteit Leuven, Leuven, Belgium
- Center for Microbiology, Vlaams Instituut voor Biotechnologie (VIB), Leuven, Belgium
| | - Severine Vermeire
- Department of Gastroenterology and Hepatology, University Hospitals Leuven, KU Leuven, Leuven, Belgium
- KU Leuven Department of Chronic Diseases and Metabolism, Translational Research Center for Gastrointestinal Disorders (TARGID), Leuven, Belgium
| | - Kristel Van Steen
- BIO3 Laboratory for Systems Medicine, Department of Human Genetics, KU Leuven, Leuven, Belgium
| |
Collapse
|
5
|
Dongare DB, Nishad SS, Mastoli SY, Saraf SA, Srivastava N, Dey A. High-throughput sequencing: a breakthrough in molecular diagnosis for precision medicine. Funct Integr Genomics 2025; 25:22. [PMID: 39838192 DOI: 10.1007/s10142-025-01529-w] [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: 10/09/2024] [Revised: 01/03/2025] [Accepted: 01/07/2025] [Indexed: 01/23/2025]
Abstract
High-resolution insights into the nucleotide arrangement within an organism's genome are pivotal for deciphering its genetic composition, function, and evolutionary trajectory. Over the years, nucleic acid sequencing has been instrumental in driving significant advancements in genomics and molecular biology. The advent of high-throughput or next-generation sequencing (NGS) technologies has revolutionized whole genome sequencing, revealing novel and intriguing features of genomes, such as single nucleotide polymorphisms and lethal mutations in both coding and non-coding regions. These platforms provide a practical approach to comprehensively identifying and analyzing whole genomes with remarkable throughput, accuracy, and scalability within a short time frame. The resulting data holds immense potential for enhancing healthcare systems, developing novel and personalized therapies, and preparing for future pandemics and outbreaks. Given the wide array of available high-throughput sequencing platforms, selecting the appropriate technology based on specific needs is crucial. However, there is limited information regarding sample preparation, sequencing principles, and output data to facilitate a comparative evaluation of these platforms. This review details various NGS technologies and approaches, examining their advantages, limitations, and future potential. Despite being in their early stages and facing challenges, ongoing advancements in NGS are expected to yield significant future benefits.
Collapse
Affiliation(s)
- Dipali Barku Dongare
- Department of Biotechnology, National Institute of Pharmaceutical Education and Research (NIPER)-Raebareli, Lucknow, 226002, India
| | - Shaik Shireen Nishad
- Department of Biotechnology, National Institute of Pharmaceutical Education and Research (NIPER)-Raebareli, Lucknow, 226002, India
| | - Sakshi Y Mastoli
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER)-Raebareli, Lucknow, 226002, India
| | - Shubhini A Saraf
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER)-Raebareli, Lucknow, 226002, India
| | - Nidhi Srivastava
- Department of Biotechnology, National Institute of Pharmaceutical Education and Research (NIPER)-Raebareli, Lucknow, 226002, India
| | - Abhishek Dey
- Department of Biotechnology, National Institute of Pharmaceutical Education and Research (NIPER)-Raebareli, Lucknow, 226002, India.
| |
Collapse
|
6
|
Guo M, Ye X, Huang D, Sakurai T. Robust feature learning using contractive autoencoders for multi-omics clustering in cancer subtyping. Methods 2025; 233:52-60. [PMID: 39577512 DOI: 10.1016/j.ymeth.2024.11.013] [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: 07/20/2024] [Revised: 10/04/2024] [Accepted: 11/18/2024] [Indexed: 11/24/2024] Open
Abstract
Cancer can manifest in virtually any tissue or organ, necessitating precise subtyping of cancer patients to enhance diagnosis, treatment, and prognosis. With the accumulation of vast amounts of omics data, numerous studies have focused on integrating multi-omics data for cancer subtyping using clustering techniques. However, due to the heterogeneity of different omics data, extracting important features to effectively integrate these data for accurate clustering analysis remains a significant challenge. This study proposes a new multi-omics clustering framework for cancer subtyping, which utilizes contractive autoencoder to extract robust features. By encouraging the learned representation to be less sensitive to small changes, the contractive autoencoder learns robust feature representations from different omics. To incorporate survival information into the clustering analysis, Cox proportional hazards regression is used to further select the key features significantly associated with survival for integration. Finally, we utilize K-means clustering on the integrated feature to obtain the clustering result. The proposed framework is evaluated on ten different cancer datasets across four levels of omics data and compared to other existing methods. The experimental results indicate that the proposed framework effectively integrates the four omics datasets and outperforms other methods, achieving higher C-index scores and showing more significant differences between survival curves. Additionally, differential gene analysis and pathway enrichment analysis are performed to further demonstrate the effectiveness of the proposed method framework.
Collapse
Affiliation(s)
- Mengke Guo
- Department of Computer Science, University of Tsukuba, Tsukuba 3058577, Japan
| | - Xiucai Ye
- Department of Computer Science, University of Tsukuba, Tsukuba 3058577, Japan.
| | - Dong Huang
- Department of Computer Science, University of Tsukuba, Tsukuba 3058577, Japan.
| | - Tetsuya Sakurai
- Department of Computer Science, University of Tsukuba, Tsukuba 3058577, Japan
| |
Collapse
|
7
|
Popova L, Carabetta VJ. The Use of Next-Generation Sequencing in Personalized Medicine. Methods Mol Biol 2025; 2866:287-315. [PMID: 39546209 DOI: 10.1007/978-1-0716-4192-7_16] [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: 11/17/2024]
Abstract
The revolutionary progress in development of next-generation sequencing (NGS) technologies has made it possible to deliver accurate genomic information in a timely manner. Over the past several years, NGS has transformed biomedical and clinical research and found its application in the field of personalized medicine. Here we discuss the rise of personalized medicine and the history of NGS. We discuss current applications and uses of NGS in medicine, including infectious diseases, oncology, genomic medicine, and dermatology. We provide a brief discussion of selected studies where NGS was used to respond to wide variety of questions in biomedical research and clinical medicine. Finally, we discuss the challenges of implementing NGS into routine clinical use.
Collapse
Affiliation(s)
- Liya Popova
- Department of Biomedical Sciences, Cooper Medical School of Rowan University, Camden, NJ, USA
| | - Valerie J Carabetta
- Department of Biomedical Sciences, Cooper Medical School of Rowan University, Camden, NJ, USA.
| |
Collapse
|
8
|
Elphinstone B, Walshe J, Nicol D, Taylor M. Commercialisation fears and preferred forms of governance: a mixed methods investigation to identify a trusted Australian genomics repository. Front Public Health 2024; 12:1508261. [PMID: 39735747 PMCID: PMC11671527 DOI: 10.3389/fpubh.2024.1508261] [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: 10/09/2024] [Accepted: 11/25/2024] [Indexed: 12/31/2024] Open
Abstract
This study aimed to identify operating conditions and governance mechanisms that would help to facilitate trust in, and willingness to donate to, a hypothetical Australian national genomic repository for health research where commercial use of data is permitted. Semi-structured telephone interviews with members of the Australian public (N = 39) clarified perceived risks and preferred repository conditions. These insights were subsequently tested experimentally in a national sample (N = 1,117). Contrary to what was expected based on the interviews, when certain baseline operating conditions were included (e.g., public management, data access committee to ensure data is restricted to human health research), none of the additional tested governance mechanisms (e.g., financial penalties for misuse) increased trust or donation willingness. Thus, providing suitable baseline conditions are in place, a feasible Australian genomic repository may not require external oversight or new legislation to optimize recruitment, even if commercial users are anticipated.
Collapse
Affiliation(s)
- Brad Elphinstone
- Department of Psychological Sciences, Swinburne University of Technology, Hawthorn, VIC, Australia
| | - Jarrod Walshe
- Department of Psychological Sciences, Swinburne University of Technology, Hawthorn, VIC, Australia
| | - Dianne Nicol
- Faculty of Law, University of Tasmania, Hobart, TAS, Australia
| | - Mark Taylor
- Melbourne Law School, The University of Melbourne, Parkville, VIC, Australia
| |
Collapse
|
9
|
Li T, Li M, Wu Y, Li Y. Visualization Methods for DNA Sequences: A Review and Prospects. Biomolecules 2024; 14:1447. [PMID: 39595624 PMCID: PMC11592258 DOI: 10.3390/biom14111447] [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: 10/17/2024] [Revised: 11/08/2024] [Accepted: 11/12/2024] [Indexed: 11/28/2024] Open
Abstract
The efficient analysis and interpretation of biological sequence data remain major challenges in bioinformatics. Graphical representation, as an emerging and effective visualization technique, offers a more intuitive method for analyzing DNA sequences. However, many visualization approaches are dispersed across research databases, requiring urgent organization, integration, and analysis. Additionally, no single visualization method excels in all aspects. To advance these methods, knowledge graphs and advanced machine learning techniques have become key areas of exploration. This paper reviews the current 2D and 3D DNA sequence visualization methods and proposes a new research direction focused on constructing knowledge graphs for biological sequence visualization, explaining the relevant theories, techniques, and models involved. Additionally, we summarize machine learning techniques applicable to sequence visualization, such as graph embedding methods and the use of convolutional neural networks (CNNs) for processing graphical representations. These machine learning techniques and knowledge graphs aim to provide valuable insights into computational biology, bioinformatics, genomic computing, and evolutionary analysis. The study serves as an important reference for improving intelligent search systems, enriching knowledge bases, and enhancing query systems related to biological sequence visualization, offering a comprehensive framework for future research.
Collapse
Affiliation(s)
- Tan Li
- School of Physics and Electronic Information, Gannan Normal University, Ganzhou 341000, China; (T.L.); (Y.L.)
| | - Mengshan Li
- School of Physics and Electronic Information, Gannan Normal University, Ganzhou 341000, China; (T.L.); (Y.L.)
| | - Yan Wu
- School of Mathematics and Computer Science, Gannan Normal University, Ganzhou 341000, China;
| | - Yelin Li
- School of Physics and Electronic Information, Gannan Normal University, Ganzhou 341000, China; (T.L.); (Y.L.)
| |
Collapse
|
10
|
Nazari E, Khalili-Tanha G, Pourali G, Khojasteh-Leylakoohi F, Azari H, Dashtiahangar M, Fiuji H, Yousefli Z, Asadnia A, Maftooh M, Akbarzade H, Nassiri M, Hassanian SM, Ferns GA, Peters GJ, Giovannetti E, Batra J, Khazaei M, Avan A. The diagnostic and prognostic value of C1orf174 in colorectal cancer. BIOIMPACTS : BI 2024; 15:30566. [PMID: 40256241 PMCID: PMC12008501 DOI: 10.34172/bi.30566] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Revised: 09/12/2024] [Accepted: 09/23/2024] [Indexed: 04/22/2025]
Abstract
Introduction Colorectal cancer (CRC) is among the lethal cancers, indicating the need for the identification of novel biomarkers for the detection of patients in earlier stages. RNA and microRNA sequencing were analyzed using bioinformatics and machine learning algorithms to identify differentially expressed genes (DEGs), followed by validation in CRC patients. Methods The genome-wide RNA sequencing of 631 samples, comprising 398 patients and 233 normal cases was extracted from the Cancer Genome Atlas (TCGA). The DEGs were identified using DESeq package in R. Survival analysis was evaluated using Kaplan-Meier analysis to identify prognostic biomarkers. Predictive biomarkers were determined by machine learning algorithms such as Deep learning, Decision Tree, and Support Vector Machine. The biological pathways, protein-protein interaction (PPI), the co-expression of DEGs, and the correlation between DEGs and clinical data were evaluated. Additionally, the diagnostic markers were assessed with a combioROC package. Finally, the candidate tope score gene was validated by Real-time PCR in CRC patients. Results The survival analysis revealed five novel prognostic genes, including KCNK13, C1orf174, CLEC18A, SRRM5, and GPR89A. Thirty-nine upregulated, 40 downregulated genes, and 20 miRNAs were detected by SVM with high accuracy and AUC. The upregulation of KRT20 and FAM118A genes and the downregulation of LRAT and PROZ genes had the highest coefficient in the advanced stage. Furthermore, our findings showed that three miRNAs (mir-19b-1, mir-326, and mir-330) upregulated in the advanced stage. C1orf174, as a novel gene, was validated using RT-PCR in CRC patients. The combineROC curve analysis indicated that the combination of C1orf174-AKAP4-DIRC1-SKIL-Scan29A4 can be considered as diagnostic markers with sensitivity, specificity, and AUC values of 0.90, 0.94, and 0.92, respectively. Conclusion Machine learning algorithms can be used to Identify key dysregulated genes/miRNAs involved in the pathogenesis of diseases, leading to the detection of patients in earlier stages. Our data also demonstrated the prognostic value of C1orf174 in colorectal cancer.
Collapse
Affiliation(s)
- Elham Nazari
- Proteomics Research Center, Faculty of Paramedical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Ghazaleh Khalili-Tanha
- Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- Medical Genetics Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Ghazaleh Pourali
- Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | | | - Hanieh Azari
- Medical Genetics Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | | | - Hamid Fiuji
- Department of Medical Oncology, Cancer Center Amsterdam, Amsterdam U.M.C., VU. University Medical Center (VUMC), Amsterdam, The Netherlands
| | - Zahra Yousefli
- Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- Medical Genetics Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Alireza Asadnia
- Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- Medical Genetics Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mina Maftooh
- Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- College of Medicine, University of Warith Al-Anbiyaa, Karbala, Iraq
| | - Hamed Akbarzade
- Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mohammadreza Nassiri
- Recombinant Proteins Research Group, The Research Institute of Biotechnology, Ferdowsi University of Mashhad, Mashhad, Iran
| | - Seyed Mahdi Hassanian
- Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Gordon A Ferns
- Brighton & Sussex Medical School, Division of Medical Education, Falmer, Brighton, Sussex BN1 9PH, UK
| | - Godefridus J Peters
- Department of Medical Oncology, Cancer Center Amsterdam, Amsterdam U.M.C., VU. University Medical Center (VUMC), Amsterdam, The Netherlands
- Professor In Biochemistry, Medical University of Gdansk,Gdansk, Poland
| | - Elisa Giovannetti
- Department of Medical Oncology, Cancer Center Amsterdam, Amsterdam U.M.C., VU. University Medical Center (VUMC), Amsterdam, The Netherlands
- Cancer Pharmacology Lab, AIRC Start up Unit, Fondazione Pisana per La Scienza, Pisa, Italy
| | - Jyotsna Batra
- Centre for Genomics and Personalised Health, Queensland University of Technology, Brisbane 4059, Australia
- Faculty of Health, School of Biomedical Sciences, Queensland University of Technology, Brisbane 4059, Australia
| | - Majid Khazaei
- Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Amir Avan
- Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- Faculty of Health, School of Biomedical Sciences, Queensland University of Technology, Brisbane 4059, Australia
| |
Collapse
|
11
|
Huizer K, Banga IK, Kumar RM, Muthukumar S, Prasad S. Dynamic Real-Time Biosensing Enabled Biorhythm Tracking for Psychiatric Disorders. WILEY INTERDISCIPLINARY REVIEWS. NANOMEDICINE AND NANOBIOTECHNOLOGY 2024; 16:e2021. [PMID: 39654328 DOI: 10.1002/wnan.2021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Revised: 10/09/2024] [Accepted: 11/02/2024] [Indexed: 01/12/2025]
Abstract
This review article explores the transformative potential of dynamic, real-time biosensing in biorhythm tracking for psychiatric disorders. Psychiatric diseases, characterized by a complex, heterogeneous, and multifactorial pathophysiology, pose challenges in both diagnosis and treatment. Common denominators in the pathophysiology of psychiatric diseases include disruptions in the stress response, sleep-wake cycle, energy metabolism, and immune response: all of these are characterized by a strong biorhythmic regulation (e.g., circadian), leading to dynamic changes in the levels of biomarkers involved. Technological and practical limitations have hindered the analysis of such dynamic processes to date. The integration of biosensors marks a paradigm shift in psychiatric research. These advanced technologies enable multiplex, non-invasive, and near-continuous analysis of biorhythmic biomarkers in real time, overcoming the constraints of conventional approaches. Focusing on the regulation of the stress response, sleep/wake cycle, energy metabolism, and immune response, biosensing allows for a deeper understanding of the heterogeneous and multifactorial pathophysiology of psychiatric diseases. The potential applications of nanobiosensing in biorhythm tracking, however, extend beyond observation. Continuous monitoring of biomarkers can provide a foundation for personalized medicine in Psychiatry, and allow for the transition from syndromal diagnostic entities to pathophysiology-based psychiatric diagnoses. This evolution promises enhanced disease tracking, early relapse prediction, and tailored disease management and treatment strategies. As non-invasive biosensing continues to advance, its integration into biorhythm tracking holds promise not only to unravel the intricate etiology of psychiatric disorders but also for ushering in a new era of precision medicine, ultimately improving the outcomes and quality of life for individuals grappling with these challenging conditions.
Collapse
Affiliation(s)
- Karin Huizer
- Parnassia Academy, Parnassia Psychiatric Institute, Hague, The Netherlands
- Department of Pathology, Erasmus Medical Center, Rotterdam, The Netherlands
| | | | | | | | | |
Collapse
|
12
|
de Brito MWD, de Carvalho SS, Mota MBDS, Mesquita RD. RNA-seq validation: software for selection of reference and variable candidate genes for RT-qPCR. BMC Genomics 2024; 25:697. [PMID: 39014352 PMCID: PMC11251314 DOI: 10.1186/s12864-024-10511-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: 03/12/2024] [Accepted: 06/06/2024] [Indexed: 07/18/2024] Open
Abstract
BACKGROUND Real-time quantitative PCR (RT-qPCR) is one of the most widely used gene expression analyses for validating RNA-seq data. This technique requires reference genes that are stable and highly expressed, at least across the different biological conditions present in the transcriptome. Reference and variable candidate gene selection is often neglected, leading to misinterpretation of the results. RESULTS We developed a software named "Gene Selector for Validation" (GSV), which identifies the best reference and variable candidate genes for validation within a quantitative transcriptome. This tool also filters the candidate genes concerning the RT-qPCR assay detection limit. GSV was compared with other software using synthetic datasets and performed better, removing stable low-expression genes from the reference candidate list and creating the variable-expression validation list. GSV software was used on a real case, an Aedes aegypti transcriptome. The top GSV reference candidate genes were selected for RT-qPCR analysis, confirming that eiF1A and eiF3j were the most stable genes tested. The tool also confirmed that traditional mosquito reference genes were less stable in the analyzed samples, highlighting the possibility of inappropriate choices. A meta-transcriptome dataset with more than ninety thousand genes was also processed successfully. CONCLUSION The GSV tool is a time and cost-effective tool that can be used to select reference and validation candidate genes from the biological conditions present in transcriptomic data.
Collapse
Affiliation(s)
- Márcio Wilson Dias de Brito
- Programa de Pós-graduação em Biologia Computacional e Sistemas, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro, Brazil
- RioGen Tecnologia, Rio de Janeiro, Brazil
| | | | - Maria Beatriz Dos Santos Mota
- RioGen Tecnologia, Rio de Janeiro, Brazil
- Departamento de Bioquímica, Instituto de Química, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Rafael Dias Mesquita
- Programa de Pós-graduação em Biologia Computacional e Sistemas, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro, Brazil.
- Departamento de Bioquímica, Instituto de Química, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil.
- Instituto Nacional de Ciência e Tecnologia em Entomologia Molecular, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil.
| |
Collapse
|
13
|
Xie M, Kuang Y, Song M, Bao E. Subtype-MGTP: a cancer subtype identification framework based on multi-omics translation. Bioinformatics 2024; 40:btae360. [PMID: 38857453 PMCID: PMC11194476 DOI: 10.1093/bioinformatics/btae360] [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: 12/20/2023] [Revised: 04/30/2024] [Accepted: 06/07/2024] [Indexed: 06/12/2024] Open
Abstract
MOTIVATION The identification of cancer subtypes plays a crucial role in cancer research and treatment. With the rapid development of high-throughput sequencing technologies, there has been an exponential accumulation of cancer multi-omics data. Integrating multi-omics data has emerged as a cost-effective and efficient strategy for cancer subtyping. While current methods primarily rely on genomics data, protein expression data offers a closer representation of phenotype. Therefore, integrating protein expression data holds promise for enhancing subtyping accuracy. However, the scarcity of protein expression data compared to genomics data presents a challenge in its direct incorporation into existing methods. Moreover, striking a balance between omics-specific learning and cross-omics learning remains a prevalent challenge in current multi-omics integration methods. RESULTS We introduce Subtype-MGTP, a novel cancer subtyping framework based on the translation of Multiple Genomics To Proteomics. Subtype-MGTP comprises two modules: a translation module, which leverages available protein data to translate multi-type genomics data into predicted protein expression data, and an improved deep subspace clustering module, which integrates contrastive learning to cluster the predicted protein data, yielding refined subtyping results. Extensive experiments conducted on benchmark datasets demonstrate that Subtype-MGTP outperforms nine state-of-the-art cancer subtyping methods. The interpretability of clustering results is further supported by the clinical and survival analysis. Subtype-MGTP also exhibits strong robustness against varying rates of missing protein data and demonstrates distinct advantages in integrating multi-omics data with imbalanced multi-omics data. AVAILABILITY AND IMPLEMENTATION The code and results are available at https://github.com/kybinn/Subtype-MGTP.
Collapse
Affiliation(s)
- Minzhu Xie
- College of Information Science and Engineering, Hunan Normal University, Changsha 410081, China
- Key Laboratory of Computing and Stochastic Mathematics (Ministry of Education), Changsha 410081, China
- College of Mathematics and Statistics, Hunan Normal University, Changsha 410081, China
| | - Yabin Kuang
- College of Information Science and Engineering, Hunan Normal University, Changsha 410081, China
| | - Mengyun Song
- College of Information Science and Engineering, Hunan Normal University, Changsha 410081, China
| | - Ergude Bao
- School of Software Engineering, Beijing Jiaotong University, Beijing 100044, China
| |
Collapse
|
14
|
Cammayo-Fletcher PLT, Flores RA, Nguyen BT, Altanzul B, Fernandez-Colorado CP, Kim WH, Devi RM, Kim S, Min W. Identification of Critical Immune Regulators and Potential Interactions of IL-26 in Riemerella anatipestifer-Infected Ducks by Transcriptome Analysis and Profiling. Microorganisms 2024; 12:973. [PMID: 38792803 PMCID: PMC11123779 DOI: 10.3390/microorganisms12050973] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Revised: 05/02/2024] [Accepted: 05/09/2024] [Indexed: 05/26/2024] Open
Abstract
Riemerella anatipestifer (RA) is an economically important pathogen in the duck industry worldwide that causes high mortality and morbidity in infected birds. We previously found that upregulated IL-17A expression in ducks infected with RA participates in the pathogenesis of the disease, but this mechanism is not linked to IL-23, which primarily promotes Th17 cell differentiation and proliferation. RNA sequencing analysis was used in this study to investigate other mechanisms of IL-17A upregulation in RA infection. A possible interaction of IL-26 and IL-17 was discovered, highlighting the potential of IL-26 as a novel upstream cytokine that can regulate IL-17A during RA infection. Additionally, this process identified several important pathways and genes related to the complex networks and potential regulation of the host immune response in RA-infected ducks. Collectively, these findings not only serve as a roadmap for our understanding of RA infection and the development of new immunotherapeutic approaches for this disease, but they also provide an opportunity to understand the immune system of ducks.
Collapse
Affiliation(s)
- Paula Leona T. Cammayo-Fletcher
- College of Veterinary Medicine & Institute of Animal Medicine, Gyeongsang National University, Jinju 52828, Republic of Korea; (P.L.T.C.-F.); (R.A.F.); (B.T.N.); (B.A.); (W.H.K.); (S.K.)
| | - Rochelle A. Flores
- College of Veterinary Medicine & Institute of Animal Medicine, Gyeongsang National University, Jinju 52828, Republic of Korea; (P.L.T.C.-F.); (R.A.F.); (B.T.N.); (B.A.); (W.H.K.); (S.K.)
| | - Binh T. Nguyen
- College of Veterinary Medicine & Institute of Animal Medicine, Gyeongsang National University, Jinju 52828, Republic of Korea; (P.L.T.C.-F.); (R.A.F.); (B.T.N.); (B.A.); (W.H.K.); (S.K.)
| | - Bujinlkham Altanzul
- College of Veterinary Medicine & Institute of Animal Medicine, Gyeongsang National University, Jinju 52828, Republic of Korea; (P.L.T.C.-F.); (R.A.F.); (B.T.N.); (B.A.); (W.H.K.); (S.K.)
| | - Cherry P. Fernandez-Colorado
- Department of Veterinary Paraclinical Sciences, College of Veterinary Medicine, University of the Philippines Los Baños, Los Baños 4031, Philippines;
| | - Woo H. Kim
- College of Veterinary Medicine & Institute of Animal Medicine, Gyeongsang National University, Jinju 52828, Republic of Korea; (P.L.T.C.-F.); (R.A.F.); (B.T.N.); (B.A.); (W.H.K.); (S.K.)
| | - Rajkumari Mandakini Devi
- Department of Veterinary Microbiology, College of Veterinary Sciences & Animal Husbandry, Central Agricultural University (1), Jalukie 797110, India;
| | - Suk Kim
- College of Veterinary Medicine & Institute of Animal Medicine, Gyeongsang National University, Jinju 52828, Republic of Korea; (P.L.T.C.-F.); (R.A.F.); (B.T.N.); (B.A.); (W.H.K.); (S.K.)
| | - Wongi Min
- College of Veterinary Medicine & Institute of Animal Medicine, Gyeongsang National University, Jinju 52828, Republic of Korea; (P.L.T.C.-F.); (R.A.F.); (B.T.N.); (B.A.); (W.H.K.); (S.K.)
| |
Collapse
|
15
|
Flo S, Vader A, Præbel K. Brute force prey metabarcoding to explore the diets of small invertebrates. Ecol Evol 2024; 14:e11369. [PMID: 38711484 PMCID: PMC11070772 DOI: 10.1002/ece3.11369] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Revised: 04/16/2024] [Accepted: 04/17/2024] [Indexed: 05/08/2024] Open
Abstract
Prey metabarcoding has become a popular tool in molecular ecology for resolving trophic interactions at high resolution, from various sample types and animals. To date, most predator-prey studies of small-sized animals (<1 mm) have met the problem of overabundant predator DNA in dietary samples by adding blocking primers/peptide nucleic acids. These primers aim to limit the PCR amplification and detection of the predator DNA but may introduce bias to the prey composition identified by interacting with sequences that are similar to those of the predator. Here we demonstrate the use of an alternative method to explore the prey of small marine copepods using whole-body DNA extracts and deep, brute force metabarcoding of an 18S rDNA fragment. After processing and curating raw data from two sequencing runs of varying depths (0.4 and 5.4 billion raw reads), we isolated 1.3 and 52.2 million prey reads, with average depths of ~15,900 and ~120,000 prey reads per copepod individual, respectively. While data from both sequencing runs were sufficient to distinguish dietary compositions from disparate seasons, locations, and copepod species, greater sequencing depth led to better separation of clusters. As computation and sequencing are becoming ever more powerful and affordable, we expect the brute force approach to become a general standard for prey metabarcoding, as it offers a simple and affordable solution to consumers that is impractical to dissect or unknown to science.
Collapse
Affiliation(s)
- Snorre Flo
- Department of Arctic BiologyThe University Centre in SvalbardLongyearbyen, SvalbardNorway
- Faculty of Biosciences, Fisheries and EconomicsUiT The Arctic University of NorwayTromsøNorway
| | - Anna Vader
- Department of Arctic BiologyThe University Centre in SvalbardLongyearbyen, SvalbardNorway
| | - Kim Præbel
- The Norwegian College of Fishery Science (NFH)UiT The Arctic University of NorwayTromsøNorway
- Department of Forestry and Wildlife ManagementInland Norway University of Applied SciencesElverumNorway
| |
Collapse
|
16
|
Wang H, Lin K, Zhang Q, Shi J, Song X, Wu J, Zhao C, He K. HyperTMO: a trusted multi-omics integration framework based on hypergraph convolutional network for patient classification. Bioinformatics 2024; 40:btae159. [PMID: 38530977 PMCID: PMC11212491 DOI: 10.1093/bioinformatics/btae159] [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: 04/06/2023] [Revised: 02/02/2024] [Accepted: 03/24/2024] [Indexed: 03/28/2024] Open
Abstract
MOTIVATION The rapid development of high-throughput biomedical technologies can provide researchers with detailed multi-omics data. The multi-omics integrated analysis approach based on machine learning contributes a more comprehensive perspective to human disease research. However, there are still significant challenges in representing single-omics data and integrating multi-omics information. RESULTS This article presents HyperTMO, a Trusted Multi-Omics integration framework based on Hypergraph convolutional network for patient classification. HyperTMO constructs hypergraph structures to represent the association between samples in single-omics data, then evidence extraction is performed by hypergraph convolutional network, and multi-omics information is integrated at an evidence level. Last, we experimentally demonstrate that HyperTMO outperforms other state-of-the-art methods in breast cancer subtype classification and Alzheimer's disease classification tasks using multi-omics data from TCGA (BRCA) and ROSMAP datasets. Importantly, HyperTMO is the first attempt to integrate hypergraph structure, evidence theory, and multi-omics integration for patient classification. Its accurate and robust properties bring great potential for applications in clinical diagnosis. AVAILABILITY AND IMPLEMENTATION HyperTMO and datasets are publicly available at https://github.com/ippousyuga/HyperTMO.
Collapse
Affiliation(s)
- Haohua Wang
- School of Computer Science and Technology, Dalian University of Technology, Dalian, Liaoning 116024, China
| | - Kai Lin
- School of Computer Science and Technology, Dalian University of Technology, Dalian, Liaoning 116024, China
| | - Qiang Zhang
- School of Computer Science and Technology, Dalian University of Technology, Dalian, Liaoning 116024, China
| | - Jinlong Shi
- Research Center for Medical Big Data, Medical Innovation Research Division of Chinese PLA General Hospital, Beijing 100039, China
| | - Xinyu Song
- Research Center for Medical Big Data, Medical Innovation Research Division of Chinese PLA General Hospital, Beijing 100039, China
| | - Jue Wu
- Research Center for Medical Big Data, Medical Innovation Research Division of Chinese PLA General Hospital, Beijing 100039, China
| | - Chenghui Zhao
- Research Center for Medical Big Data, Medical Innovation Research Division of Chinese PLA General Hospital, Beijing 100039, China
| | - Kunlun He
- Research Center for Medical Big Data, Medical Innovation Research Division of Chinese PLA General Hospital, Beijing 100039, China
| |
Collapse
|
17
|
Wu G, Zaker A, Ebrahimi A, Tripathi S, Mer AS. Text-mining-based feature selection for anticancer drug response prediction. BIOINFORMATICS ADVANCES 2024; 4:vbae047. [PMID: 38606185 PMCID: PMC11009020 DOI: 10.1093/bioadv/vbae047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Revised: 03/09/2024] [Accepted: 03/22/2024] [Indexed: 04/13/2024]
Abstract
Motivation Predicting anticancer treatment response from baseline genomic data is a critical obstacle in personalized medicine. Machine learning methods are commonly used for predicting drug response from gene expression data. In the process of constructing these machine learning models, one of the most significant challenges is identifying appropriate features among a massive number of genes. Results In this study, we utilize features (genes) extracted using the text-mining of scientific literatures. Using two independent cancer pharmacogenomic datasets, we demonstrate that text-mining-based features outperform traditional feature selection techniques in machine learning tasks. In addition, our analysis reveals that text-mining feature-based machine learning models trained on in vitro data also perform well when predicting the response of in vivo cancer models. Our results demonstrate that text-mining-based feature selection is an easy to implement approach that is suitable for building machine learning models for anticancer drug response prediction. Availability and implementation https://github.com/merlab/text_features.
Collapse
Affiliation(s)
- Grace Wu
- Division of Engineering Science, University of Toronto, Toronto, M5S2E4, Canada
| | - Arvin Zaker
- Department of Biochemistry, Microbiology & Immunology, University of Ottawa, Ottawa, K1H8M5, Canada
- Ottawa Institute of Systems Biology, University of Ottawa, Ottawa, K1H8M5, Canada
| | - Amirhosein Ebrahimi
- Department of Biochemistry, Microbiology & Immunology, University of Ottawa, Ottawa, K1H8M5, Canada
| | - Shivanshi Tripathi
- Department of Biochemistry, Microbiology & Immunology, University of Ottawa, Ottawa, K1H8M5, Canada
- Ottawa Institute of Systems Biology, University of Ottawa, Ottawa, K1H8M5, Canada
| | - Arvind Singh Mer
- Department of Biochemistry, Microbiology & Immunology, University of Ottawa, Ottawa, K1H8M5, Canada
- Ottawa Institute of Systems Biology, University of Ottawa, Ottawa, K1H8M5, Canada
- School of Electrical Engineering & Computer Science, University of Ottawa, Ottawa, K1N6N5, Canada
| |
Collapse
|
18
|
Popova L, Carabetta VJ. The use of next-generation sequencing in personalized medicine. ARXIV 2024:arXiv:2403.03688v1. [PMID: 38495572 PMCID: PMC10942477] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Subscribe] [Scholar Register] [Indexed: 03/19/2024]
Abstract
The revolutionary progress in development of next-generation sequencing (NGS) technologies has made it possible to deliver accurate genomic information in a timely manner. Over the past several years, NGS has transformed biomedical and clinical research and found its application in the field of personalized medicine. Here we discuss the rise of personalized medicine and the history of NGS. We discuss current applications and uses of NGS in medicine, including infectious diseases, oncology, genomic medicine, and dermatology. We provide a brief discussion of selected studies where NGS was used to respond to wide variety of questions in biomedical research and clinical medicine. Finally, we discuss the challenges of implementing NGS into routine clinical use.
Collapse
Affiliation(s)
- Liya Popova
- Department of Biomedical Sciences, Cooper Medical School of Rowan University, Camden NJ, 08103
| | - Valerie J. Carabetta
- Department of Biomedical Sciences, Cooper Medical School of Rowan University, Camden NJ, 08103
| |
Collapse
|
19
|
Wang Q, Tang J, Pan L, Song A, Miao J, Zheng X, Li Z. Study on epigenotoxicity, sex hormone synthesis, and DNA damage of benzo[a]pyrene in the testis of male Ruditapes philippinarum. THE SCIENCE OF THE TOTAL ENVIRONMENT 2024; 912:169340. [PMID: 38110097 DOI: 10.1016/j.scitotenv.2023.169340] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/29/2023] [Revised: 12/10/2023] [Accepted: 12/11/2023] [Indexed: 12/20/2023]
Abstract
Research on the mechanisms of reproductive toxicity caused by persistent organic pollutants (POPs) in marine animals has received significant attention. One group of typical POPs, called polycyclic aromatic hydrocarbons (PAHs), has been found to cause various reproductive toxicities in aquatic organisms, including epigenotoxicity, reproductive endocrine disruption, DNA damage effects and other reproductive toxicity, thereby affecting gonadal development. Interestingly, male aquatic animals are more susceptible to the disturbance and toxicity of environmental pollutants. However, current studies primarily focus on vertebrates, leaving a large gap in our understanding of the reproductive toxicity and mechanisms of PAHs interference in marine invertebrates. In this study, male Ruditapes philippinarum was used as an experimental subject to investigate reproduction-related indexes in clams under the stress of benzo[a]pyrene (B[a]P) at different concentrations (0, 0.8, 4 and 20 μg/L) during the proliferative, growth, maturity, and spawning period. We analyzed the molecular mechanisms of reproductive toxicity caused by PAHs in marine bivalves, specifically epigenotoxicity, reproductive endocrine disruption, and gonadal damage-apoptotic effect. The results suggest that DNA methylation plays a crucial role in mediating B[a]P-induced reproductive toxicity in male R. philippinarum. B[a]P may affect sex hormone levels, impede spermatogenesis and testis development in clams, by inhibiting the steroid hormone synthesis pathway and downregulating genes critical for cell proliferation, testis development, and spermatid expulsion. Moreover, the spermatids of male R. philippinarum were severely impaired under the B[a]P stress, leading to reduced reproductive performance in the clams. These findings contribute to a better understanding of the reproductive toxicity response of male marine invertebrates to POPs stress.
Collapse
Affiliation(s)
- Qiaoqiao Wang
- The Key Laboratory of Mariculture, Ministry of Education, Ocean University of China, Qingdao 266003, PR China
| | - Jian Tang
- The Key Laboratory of Mariculture, Ministry of Education, Ocean University of China, Qingdao 266003, PR China
| | - Luqing Pan
- The Key Laboratory of Mariculture, Ministry of Education, Ocean University of China, Qingdao 266003, PR China.
| | - Aimin Song
- The Key Laboratory of Mariculture, Ministry of Education, Ocean University of China, Qingdao 266003, PR China
| | - Jingjing Miao
- The Key Laboratory of Mariculture, Ministry of Education, Ocean University of China, Qingdao 266003, PR China
| | - Xin Zheng
- The Key Laboratory of Mariculture, Ministry of Education, Ocean University of China, Qingdao 266003, PR China
| | - Zeyuan Li
- The Key Laboratory of Mariculture, Ministry of Education, Ocean University of China, Qingdao 266003, PR China
| |
Collapse
|
20
|
Yu X, Zhang H, Li J, Gu L, Cao L, Gong J, Xie P, Xu J. Construction of a prognostic prediction model in liver cancer based on genes involved in integrin cell surface interactions pathway by multi-omics screening. Front Cell Dev Biol 2024; 12:1237445. [PMID: 38374893 PMCID: PMC10875080 DOI: 10.3389/fcell.2024.1237445] [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: 06/22/2023] [Accepted: 01/23/2024] [Indexed: 02/21/2024] Open
Abstract
Background: Liver cancer is a common malignant tumor with an increasing incidence in recent years. We aimed to develop a model by integrating clinical information and multi-omics profiles of genes to predict survival of patients with liver cancer. Methods: The multi-omics data were integrated to identify liver cancer survival-associated signal pathways. Then, a prognostic risk score model was established based on key genes in a specific pathway, followed by the analysis of the relationship between the risk score and clinical features as well as molecular and immunologic characterization of the key genes included in the prediction model. The function experiments were performed to further elucidate the undergoing molecular mechanism. Results: Totally, 4 pathways associated with liver cancer patients' survival were identified. In the pathway of integrin cell surface interactions, low expression of COMP and SPP1, and low CNVs level of COL4A2 and ITGAV were significantly related to prognosis. Based on above 4 genes, the risk score model for prognosis was established. Risk score, ITGAV and SPP1 were the most significantly positively related to activated dendritic cell. COL4A2 and COMP were the most significantly positively associated with Type 1 T helper cell and regulatory T cell, respectively. The nomogram (involved T stage and risk score) may better predict short-term survival. The cell assay showed that overexpression of ITGAV promoted tumorigenesis. Conclusion: The risk score model constructed with four genes (COMP, SPP1, COL4A2, and ITGAV) may be used to predict survival in liver cancer patients.
Collapse
Affiliation(s)
- Xiang Yu
- Department of Radiology, Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu, China
- Department of Radiology, Chinese Academy of Sciences Sichuan Translational Medicine Research Hospital, Chengdu, China
| | - Hao Zhang
- Department of Hepatobiliary Surgery, Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu, China
- Department of Hepatobiliary Surgery, Chinese Academy of Sciences Sichuan Translational Medicine Research Hospital, Chengdu, China
| | - Jinze Li
- Department of Radiology, Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu, China
- Department of Radiology, Chinese Academy of Sciences Sichuan Translational Medicine Research Hospital, Chengdu, China
| | - Lu Gu
- Department of Radiology, Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu, China
- Department of Radiology, Chinese Academy of Sciences Sichuan Translational Medicine Research Hospital, Chengdu, China
| | - Lei Cao
- Department of Radiology, Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu, China
- Department of Radiology, Chinese Academy of Sciences Sichuan Translational Medicine Research Hospital, Chengdu, China
| | - Jun Gong
- Department of Hepatobiliary Surgery, Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu, China
- Department of Hepatobiliary Surgery, Chinese Academy of Sciences Sichuan Translational Medicine Research Hospital, Chengdu, China
| | - Ping Xie
- Department of Radiology, Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu, China
- Department of Radiology, Chinese Academy of Sciences Sichuan Translational Medicine Research Hospital, Chengdu, China
| | - Jian Xu
- Department of Hepatobiliary Surgery, Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu, China
- Department of Hepatobiliary Surgery, Chinese Academy of Sciences Sichuan Translational Medicine Research Hospital, Chengdu, China
| |
Collapse
|
21
|
Li K, Wang Z, Zhou Y, Li S. Lung adenocarcinoma identification based on hybrid feature selections and attentional convolutional neural networks. MATHEMATICAL BIOSCIENCES AND ENGINEERING : MBE 2024; 21:2991-3015. [PMID: 38454716 DOI: 10.3934/mbe.2024133] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/09/2024]
Abstract
Lung adenocarcinoma, a chronic non-small cell lung cancer, needs to be detected early. Tumor gene expression data analysis is effective for early detection, yet its challenges lie in a small sample size, high dimensionality, and multi-noise characteristics. In this study, we propose a lung adenocarcinoma convolutional neural network (LATCNN), a deep learning model tailored for accurate lung adenocarcinoma prediction and identification of key genes. During the feature selection stage, we introduce a hybrid algorithm. Initially, the fast correlation-based filter (FCBF) algorithm swiftly filters out irrelevant features, followed by applying the k-means-synthetic minority over-sampling technique (k-means-SMOTE) method to address category imbalance. Subsequently, we enhance the particle swarm optimization (PSO) algorithm by incorporating fast-decay dynamic inertia weights and utilizing the classification and regression tree (CART) as the fitness function for the second stage of feature selection, aiming to further eliminate redundant features. In the classifier construction stage, we present an attention convolutional neural network (atCNN) that incorporates an attention mechanism. This improved model conducts feature selection post lung adenocarcinoma gene expression data analysis for classification and prediction. The results show that LATCNN effectively reduces the feature dimensions and accurately identifies 12 key genes with accuracy, recall, F1 score, and MCC of 99.70%, 99.33%, 99.98%, and 98.67%, respectively. These performance metrics surpass those of other comparative models, highlighting the significance of this research for advancing lung adenocarcinoma treatment.
Collapse
Affiliation(s)
- Kunpeng Li
- School of Information Engineering, Gansu University of Chinese Medicine, Lanzhou 730000, China
| | - Zepeng Wang
- School of Information Engineering, Gansu University of Chinese Medicine, Lanzhou 730000, China
| | - Yu Zhou
- School of Information Engineering, Gansu University of Chinese Medicine, Lanzhou 730000, China
| | - Sihai Li
- School of Information Engineering, Gansu University of Chinese Medicine, Lanzhou 730000, China
| |
Collapse
|
22
|
Chai R, Zhao Y, Su Z, Liang W. Integrative analysis reveals a four-gene signature for predicting survival and immunotherapy response in colon cancer patients using bulk and single-cell RNA-seq data. Front Oncol 2023; 13:1277084. [PMID: 38023180 PMCID: PMC10644708 DOI: 10.3389/fonc.2023.1277084] [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: 08/14/2023] [Accepted: 09/25/2023] [Indexed: 12/01/2023] Open
Abstract
Background Colon cancer (CC) ranks as one of the leading causes of cancer-related mortality globally. Single-cell transcriptome sequencing (scRNA-seq) offers precise gene expression data for distinct cell types. This study aimed to utilize scRNA-seq and bulk transcriptome sequencing (bulk RNA-seq) data from CC samples to develop a novel prognostic model. Methods scRNA-seq data was downloaded from the GSE161277 database. R packages including "Seurat", "Harmony", and "singleR" were employed to categorize eight major cell types within normal and tumor tissues. By comparing tumor and normal samples, differentially expressed genes (DEGs) across these major cell types were identified. Gene Ontology (GO) enrichment analyses of DEGs for each cell type were conducted using "Metascape". DEGs-based signature construction involved Cox regression and least absolute shrinkage operator (LASSO) analyses, performed on The Cancer Genome Atlas (TCGA) training cohort. Validation occurred in the GSE39582 and GSE33382 datasets. The expression pattern of prognostic genes was verified using spatial transcriptome sequencing (ST-seq) data. Ultimately, an established prognostic nomogram based on the gene signature and age was established and calibrated. Sensitivity to chemotherapeutic drugs was predicted with the "oncoPredict" R package. Results Using scRNA-Seq data, we examined 33,213 cells, categorizing them into eight cell types within normal and tumor samples. GO enrichment analysis revealed various cancer-related pathways across DEGs in these cell types. Among the 55 DEGs identified via univariate Cox regression, four independent prognostic genes emerged: PTPN6, CXCL13, SPINK4, and NPDC1. Expression validation through ST-seq confirmed PTPN6 and CXCL13 predominance in immune cells, while SPINK4 and NPDC1 were relatively epithelial cell-specific. Creating a four-gene prognostic signature, Kaplan-Meier survival analyses emphasized higher risk scores correlating with unfavorable prognoses, confirmed across training and validation cohorts. The risk score emerged as an independent prognostic factor, supported by a reliable nomogram. Intriguingly, drug sensitivity analysis unveiled contrasting anti-cancer drug responses in the two risk groups, suggesting significant clinical implications. Conclusion We developed a novel prognostic four-gene risk model, and these genes may act as potential therapeutic targets for CC.
Collapse
Affiliation(s)
- Ruoyang Chai
- Department of General Practice, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Yajie Zhao
- Department of Geriatrics, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Zhengjia Su
- Department of Geriatrics, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Wei Liang
- Department of Geriatrics, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| |
Collapse
|
23
|
Das S, Biswas NK, Basu A. Mapinsights: deep exploration of quality issues and error profiles in high-throughput sequence data. Nucleic Acids Res 2023; 51:e75. [PMID: 37378434 PMCID: PMC10415152 DOI: 10.1093/nar/gkad539] [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: 12/09/2022] [Revised: 05/16/2023] [Accepted: 06/27/2023] [Indexed: 06/29/2023] Open
Abstract
High-throughput sequencing (HTS) has revolutionized science by enabling super-fast detection of genomic variants at base-pair resolution. Consequently, it poses the challenging problem of identification of technical artifacts, i.e. hidden non-random error patterns. Understanding the properties of sequencing artifacts holds the key in separating true variants from false positives. Here, we develop Mapinsights, a toolkit that performs quality control (QC) analysis of sequence alignment files, capable of detecting outliers based on sequencing artifacts of HTS data at a deeper resolution compared with existing methods. Mapinsights performs a cluster analysis based on novel and existing QC features derived from the sequence alignment for outlier detection. We applied Mapinsights on community standard open-source datasets and identified various quality issues including technical errors related to sequencing cycles, sequencing chemistry, sequencing libraries and across various orthogonal sequencing platforms. Mapinsights also enables identification of anomalies related to sequencing depth. A logistic regression-based model built on the features of Mapinsights shows high accuracy in detecting 'low-confidence' variant sites. Quantitative estimates and probabilistic arguments provided by Mapinsights can be utilized in identifying errors, bias and outlier samples, and also aid in improving the authenticity of variant calls.
Collapse
Affiliation(s)
- Subrata Das
- National Institute of Biomedical Genomics, Kalyani, 741251, West Bengal, India
| | - Nidhan K Biswas
- National Institute of Biomedical Genomics, Kalyani, 741251, West Bengal, India
| | - Analabha Basu
- National Institute of Biomedical Genomics, Kalyani, 741251, West Bengal, India
| |
Collapse
|
24
|
Zaytsev K, Fedorov A, Korotkov E. Classification of Promoter Sequences from Human Genome. Int J Mol Sci 2023; 24:12561. [PMID: 37628742 PMCID: PMC10454140 DOI: 10.3390/ijms241612561] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2023] [Revised: 07/28/2023] [Accepted: 08/03/2023] [Indexed: 08/27/2023] Open
Abstract
We have developed a new method for promoter sequence classification based on a genetic algorithm and the MAHDS sequence alignment method. We have created four classes of human promoters, combining 17,310 sequences out of the 29,598 present in the EPD database. We searched the human genome for potential promoter sequences (PPSs) using dynamic programming and position weight matrices representing each of the promoter sequence classes. A total of 3,065,317 potential promoter sequences were found. Only 1,241,206 of them were located in unannotated parts of the human genome. Every other PPS found intersected with either true promoters, transposable elements, or interspersed repeats. We found a strong intersection between PPSs and Alu elements as well as transcript start sites. The number of false positive PPSs is estimated to be 3 × 10-8 per nucleotide, which is several orders of magnitude lower than for any other promoter prediction method. The developed method can be used to search for PPSs in various eukaryotic genomes.
Collapse
Affiliation(s)
- Konstantin Zaytsev
- Bach Institute of Biochemistry, Federal Research Center of Biotechnology of the Russian Academy of Sciences, 119071 Moscow, Russia
| | - Alexey Fedorov
- Bach Institute of Biochemistry, Federal Research Center of Biotechnology of the Russian Academy of Sciences, 119071 Moscow, Russia
| | - Eugene Korotkov
- Institute of Bioengineering, Federal Research Center of Biotechnology of the Russian Academy of Sciences, 119071 Moscow, Russia
| |
Collapse
|
25
|
Affiliation(s)
- Bruna Gomes
- From the Departments of Medicine, Genetics, and Biomedical Data Science, Stanford University, Stanford, CA (B.G., E.A.A.); and the Department of Cardiology, Pneumology, and Angiology, Heidelberg University Hospital, Heidelberg, Germany (B.G.)
| | - Euan A Ashley
- From the Departments of Medicine, Genetics, and Biomedical Data Science, Stanford University, Stanford, CA (B.G., E.A.A.); and the Department of Cardiology, Pneumology, and Angiology, Heidelberg University Hospital, Heidelberg, Germany (B.G.)
| |
Collapse
|
26
|
Hopkins CE, McCormick K, Brock T, Wood M, Ruggiero S, Mcbride K, Kim C, Lawson JA, Helbig I, Bainbridge MN. Clinical variants in Caenorhabditis elegans expressing human STXBP1 reveal a novel class of pathogenic variants and classify variants of uncertain significance. GENETICS IN MEDICINE OPEN 2023; 1:100823. [PMID: 38827422 PMCID: PMC11141691 DOI: 10.1016/j.gimo.2023.100823] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Revised: 05/24/2023] [Accepted: 05/30/2023] [Indexed: 06/04/2024]
Abstract
Purpose Modeling disease variants in animals is useful for drug discovery, understanding disease pathology, and classifying variants of uncertain significance (VUS) as pathogenic or benign. Methods Using Clustered Regularly Interspaced Short Palindromic Repeats, we performed a Whole-gene Humanized Animal Model procedure to replace the coding sequence of the animal model's unc-18 ortholog with the coding sequence for the human STXBP1 gene. Next, we used Clustered Regularly Interspaced Short Palindromic Repeats to introduce precise point variants in the Whole-gene Humanized Animal Model-humanized STXBP1 locus from 3 clinical categories (benign, pathogenic, and VUS). Twenty-six phenotypic features extracted from video recordings were used to train machine learning classifiers on 25 pathogenic and 32 benign variants. Results Using multiple models, we were able to obtain a diagnostic sensitivity near 0.9. Twenty-three VUS were also interrogated and 8 of 23 (34.8%) were observed to be functionally abnormal. Interestingly, unsupervised clustering identified 2 distinct subsets of known pathogenic variants with distinct phenotypic features; both p.Tyr75Cys and p.Arg406Cys cluster away from other variants and show an increase in swim speed compared with hSTXBP1 worms. This leads to the hypothesis that the mechanism of disease for these 2 variants may differ from most STXBP1-mutated patients and may account for some of the clinical heterogeneity observed in the patient population. Conclusion We have demonstrated that automated analysis of a small animal system is an effective, scalable, and fast way to understand functional consequences of variants in STXBP1 and identify variant-specific intensities of aberrant activity suggesting a genotype-to-phenotype correlation is likely to occur in human clinical variations of STXBP1.
Collapse
Affiliation(s)
| | | | | | | | - Sarah Ruggiero
- Division of Neurology, Children’s Hospital of Philadelphia, Philadelphia, PA
- The Epilepsy NeuroGenetics Initiative (ENGIN), Children’s Hospital of Philadelphia, Philadelphia, PA
- Department of Biomedical and Health Informatics (DBHi), Children’s Hospital of Philadelphia, Philadelphia, PA
- University of Pennsylvania, Neuroscience Program, Philadelphia, PA
| | | | | | | | - Ingo Helbig
- Division of Neurology, Children’s Hospital of Philadelphia, Philadelphia, PA
- The Epilepsy NeuroGenetics Initiative (ENGIN), Children’s Hospital of Philadelphia, Philadelphia, PA
- Department of Biomedical and Health Informatics (DBHi), Children’s Hospital of Philadelphia, Philadelphia, PA
- University of Pennsylvania, Neuroscience Program, Philadelphia, PA
| | - Matthew N. Bainbridge
- Codified Genomics, LLC, Houston, TX
- Rady Children’s Institute for Genomic Medicine, San Diego, CA
| |
Collapse
|
27
|
Liu M, Li X, Wang J, Ji Y, Gu J, Wei Y, Peng L, Tian C, Lv P, Wang P, Liu X, Li W. Identification and validation of Rab11a in Rat orofacial inflammatory pain model induced by CFA. Neurochem Int 2023:105550. [PMID: 37268020 DOI: 10.1016/j.neuint.2023.105550] [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: 02/11/2023] [Revised: 04/29/2023] [Accepted: 05/14/2023] [Indexed: 06/04/2023]
Abstract
Orofacial pain (OFP) is a clinically very common and the most troubling condition; however, there is few effective way to relieve OFP. Rab11a, a small molecule guanosine triphosphate enzyme, is one of the Rab member family playing a vital role in intracellular endocytosis and the pain process. Therefore, we investigated the hub genes of rat OFP model induced by Complete Freund's Adjuvant (CFA) via re-analyzing microarray data (GSE111160). We found that Rab11a acted as a key hub gene in the process of OFP. During the validation of Rab11a, the OFP model was established by peripheral injection of CFA, which decreased the head withdrawal threshold (HWT) and head withdrawal lantency (HWL). Rab11a was observed in NeuN of Sp5C instead of GFAP/IBA-1, and double-IF of Rab11a and Fos positive cells were increased on the 7th day after CFA modeling statistically. Rab11a protein expression in TG and Sp5C of CFA group was also significantly increased. Interestingly, injection of Rab11a-targeted short hairpin RNA (Rab11a-shRNA) into Sp5C could reverse the decrease in HWT and HWL and reduce the expression level of Rab11a. Electrophysiological recording further demonstrated that the activity of Sp5C neuron was improved in CFA group, while Rab11a-shRNA considerably decreased the enhancement of Sp5C neuronal activity. Finally, we detected the expression level of p-PI3K, p-AKT, and p-mTOR in Sp5C of rats after injecting the Rab11a-shRNA virus. To our surprise, CFA upregulated the phosphorylation of PI3K, AKT and mTOR in Sp5C, and Rab11a-shRNA downregulated these molecules' expression. Our data suggest that CFA activates the PI3K/AKT signaling pathway through up-regulating Rab11a expression, which can induce OFP hyperalgesia development furtherly. Targeting Rab11a may be a novel treatment strategy for OFP.
Collapse
Affiliation(s)
- Miaomiao Liu
- Department of Respiratory and Critical Care Medicine, Tangdu Hospital of the Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Xin Li
- Department of Stomatology, The 960th Hospital of People's Liberation Army, Jinan, Shandong, China
| | - Jian Wang
- Department of Neurosurgery, Tangdu Hospital of the Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Yuanyuan Ji
- Department of Anatomy, School of Medicine, Northwest University, Xi'an, Shaanxi, China
| | - Junxiang Gu
- Department of Neurosurgery, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Yi Wei
- Department of Anatomy, School of Medicine, Northwest University, Xi'an, Shaanxi, China
| | - Liwei Peng
- Department of Neurosurgery, Tangdu Hospital of the Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Chao Tian
- Department of Neurosurgery, Tangdu Hospital of the Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Peiyuan Lv
- Department of Neurosurgery, Tangdu Hospital of the Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Peng Wang
- Department of Neurosurgery, Tangdu Hospital of the Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Xin Liu
- Department of Stomatology, The 960th Hospital of People's Liberation Army, Jinan, Shandong, China.
| | - Weixin Li
- Department of Neurosurgery, Tangdu Hospital of the Fourth Military Medical University, Xi'an, Shaanxi, China.
| |
Collapse
|
28
|
Zhong Y, Peng Y, Lin Y, Chen D, Zhang H, Zheng W, Chen Y, Wu C. MODILM: towards better complex diseases classification using a novel multi-omics data integration learning model. BMC Med Inform Decis Mak 2023; 23:82. [PMID: 37147619 PMCID: PMC10161645 DOI: 10.1186/s12911-023-02173-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2022] [Accepted: 04/11/2023] [Indexed: 05/07/2023] Open
Abstract
BACKGROUND Accurately classifying complex diseases is crucial for diagnosis and personalized treatment. Integrating multi-omics data has been demonstrated to enhance the accuracy of analyzing and classifying complex diseases. This can be attributed to the highly correlated nature of the data with various diseases, as well as the comprehensive and complementary information it provides. However, integrating multi-omics data for complex diseases is challenged by data characteristics such as high imbalance, scale variation, heterogeneity, and noise interference. These challenges further emphasize the importance of developing effective methods for multi-omics data integration. RESULTS We proposed a novel multi-omics data learning model called MODILM, which integrates multiple omics data to improve the classification accuracy of complex diseases by obtaining more significant and complementary information from different single-omics data. Our approach includes four key steps: 1) constructing a similarity network for each omics data using the cosine similarity measure, 2) leveraging Graph Attention Networks to learn sample-specific and intra-association features from similarity networks for single-omics data, 3) using Multilayer Perceptron networks to map learned features to a new feature space, thereby strengthening and extracting high-level omics-specific features, and 4) fusing these high-level features using a View Correlation Discovery Network to learn cross-omics features in the label space, which results in unique class-level distinctiveness for complex diseases. To demonstrate the effectiveness of MODILM, we conducted experiments on six benchmark datasets consisting of miRNA expression, mRNA, and DNA methylation data. Our results show that MODILM outperforms state-of-the-art methods, effectively improving the accuracy of complex disease classification. CONCLUSIONS Our MODILM provides a more competitive way to extract and integrate important and complementary information from multiple omics data, providing a very promising tool for supporting decision-making for clinical diagnosis.
Collapse
Affiliation(s)
- Yating Zhong
- Guangxi Key Lab of Human-Machine Interaction and Intelligent Decision, Nanning Normal University, Nanning, 530001, China
| | - Yuzhong Peng
- Guangxi Key Lab of Human-Machine Interaction and Intelligent Decision, Nanning Normal University, Nanning, 530001, China.
| | - Yanmei Lin
- School of Environment and Life Science, Nanning Normal University, Nanning, 530001, China.
| | - Dingjia Chen
- Guangxi Key Lab of Human-Machine Interaction and Intelligent Decision, Nanning Normal University, Nanning, 530001, China
| | - Hao Zhang
- School of Computer Science, Fudan University, Shanghai, 200433, China
- School of Computer, Guangdong University of Petrochemical Technology, Maoming, 525000, China
| | - Wen Zheng
- Guangxi Key Lab of Human-Machine Interaction and Intelligent Decision, Nanning Normal University, Nanning, 530001, China
| | - Yuanyuan Chen
- Guangxi Key Lab of Human-Machine Interaction and Intelligent Decision, Nanning Normal University, Nanning, 530001, China
| | - Changliang Wu
- Department of Spleen, Stomach and Liver Diseases, Guangxi International Zhuang Medical Hospital, Nanning, 530201, China
| |
Collapse
|
29
|
Gilmore LA, Parry TL, Thomas GA, Khamoui AV. Skeletal muscle omics signatures in cancer cachexia: perspectives and opportunities. J Natl Cancer Inst Monogr 2023; 2023:30-42. [PMID: 37139970 PMCID: PMC10157770 DOI: 10.1093/jncimonographs/lgad006] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Revised: 01/13/2023] [Accepted: 02/06/2023] [Indexed: 05/05/2023] Open
Abstract
Cachexia is a life-threatening complication of cancer that occurs in up to 80% of patients with advanced cancer. Cachexia reflects the systemic consequences of cancer and prominently features unintended weight loss and skeletal muscle wasting. Cachexia impairs cancer treatment tolerance, lowers quality of life, and contributes to cancer-related mortality. Effective treatments for cancer cachexia are lacking despite decades of research. High-throughput omics technologies are increasingly implemented in many fields including cancer cachexia to stimulate discovery of disease biology and inform therapy choice. In this paper, we present selected applications of omics technologies as tools to study skeletal muscle alterations in cancer cachexia. We discuss how comprehensive, omics-derived molecular profiles were used to discern muscle loss in cancer cachexia compared with other muscle-wasting conditions, to distinguish cancer cachexia from treatment-related muscle alterations, and to reveal severity-specific mechanisms during the progression of cancer cachexia from early toward severe disease.
Collapse
Affiliation(s)
- L Anne Gilmore
- Department of Clinical Nutrition, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Center for Human Nutrition, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Traci L Parry
- Department of Kinesiology, University of North Carolina Greensboro, Greensboro, NC, USA
| | - Gwendolyn A Thomas
- Department of Kinesiology, Pennsylvania State University, University Park, PA, USA
| | - Andy V Khamoui
- Department of Exercise Science and Health Promotion, Florida Atlantic University, Boca Raton, FL, USA
- Institute for Human Health and Disease Intervention, Florida Atlantic University, Jupiter, FL, USA
| |
Collapse
|
30
|
Kocere A, Lalonde RL, Mosimann C, Burger A. Lateral thinking in syndromic congenital cardiovascular disease. Dis Model Mech 2023; 16:dmm049735. [PMID: 37125615 PMCID: PMC10184679 DOI: 10.1242/dmm.049735] [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] [Indexed: 05/02/2023] Open
Abstract
Syndromic birth defects are rare diseases that can present with seemingly pleiotropic comorbidities. Prime examples are rare congenital heart and cardiovascular anomalies that can be accompanied by forelimb defects, kidney disorders and more. Whether such multi-organ defects share a developmental link remains a key question with relevance to the diagnosis, therapeutic intervention and long-term care of affected patients. The heart, endothelial and blood lineages develop together from the lateral plate mesoderm (LPM), which also harbors the progenitor cells for limb connective tissue, kidneys, mesothelia and smooth muscle. This developmental plasticity of the LPM, which founds on multi-lineage progenitor cells and shared transcription factor expression across different descendant lineages, has the potential to explain the seemingly disparate syndromic defects in rare congenital diseases. Combining patient genome-sequencing data with model organism studies has already provided a wealth of insights into complex LPM-associated birth defects, such as heart-hand syndromes. Here, we summarize developmental and known disease-causing mechanisms in early LPM patterning, address how defects in these processes drive multi-organ comorbidities, and outline how several cardiovascular and hematopoietic birth defects with complex comorbidities may be LPM-associated diseases. We also discuss strategies to integrate patient sequencing, data-aggregating resources and model organism studies to mechanistically decode congenital defects, including potentially LPM-associated orphan diseases. Eventually, linking complex congenital phenotypes to a common LPM origin provides a framework to discover developmental mechanisms and to anticipate comorbidities in congenital diseases affecting the cardiovascular system and beyond.
Collapse
Affiliation(s)
- Agnese Kocere
- University of Colorado School of Medicine, Anschutz Medical Campus, Department of Pediatrics, Section of Developmental Biology, Aurora, CO 80045, USA
- Department of Molecular Life Science, University of Zurich, 8057 Zurich, Switzerland
| | - Robert L. Lalonde
- University of Colorado School of Medicine, Anschutz Medical Campus, Department of Pediatrics, Section of Developmental Biology, Aurora, CO 80045, USA
| | - Christian Mosimann
- University of Colorado School of Medicine, Anschutz Medical Campus, Department of Pediatrics, Section of Developmental Biology, Aurora, CO 80045, USA
| | - Alexa Burger
- University of Colorado School of Medicine, Anschutz Medical Campus, Department of Pediatrics, Section of Developmental Biology, Aurora, CO 80045, USA
| |
Collapse
|
31
|
Ahmed F, Samantasinghar A, Manzoor Soomro A, Kim S, Hyun Choi K. A systematic review of computational approaches to understand cancer biology for informed drug repurposing. J Biomed Inform 2023; 142:104373. [PMID: 37120047 DOI: 10.1016/j.jbi.2023.104373] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Revised: 03/25/2023] [Accepted: 04/23/2023] [Indexed: 05/01/2023]
Abstract
Cancer is the second leading cause of death globally, trailing only heart disease. In the United States alone, 1.9 million new cancer cases and 609,360 deaths were recorded for 2022. Unfortunately, the success rate for new cancer drug development remains less than 10%, making the disease particularly challenging. This low success rate is largely attributed to the complex and poorly understood nature of cancer etiology. Therefore, it is critical to find alternative approaches to understanding cancer biology and developing effective treatments. One such approach is drug repurposing, which offers a shorter drug development timeline and lower costs while increasing the likelihood of success. In this review, we provide a comprehensive analysis of computational approaches for understanding cancer biology, including systems biology, multi-omics, and pathway analysis. Additionally, we examine the use of these methods for drug repurposing in cancer, including the databases and tools that are used for cancer research. Finally, we present case studies of drug repurposing, discussing their limitations and offering recommendations for future research in this area.
Collapse
Affiliation(s)
- Faheem Ahmed
- Department of Mechatronics Engineering, Jeju National University, Republic of Korea
| | | | | | - Sejong Kim
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, Korea; Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Korea.
| | - Kyung Hyun Choi
- Department of Mechatronics Engineering, Jeju National University, Republic of Korea.
| |
Collapse
|
32
|
Wu H, Xiong X, CUI X, Xiong J, Zhang Y, Xiang L, Xu TAO. Analysis of the influence of pyroptosis-related genes on molecular characteristics in patients with acute myocardial infarction. Medicine (Baltimore) 2023; 102:e33620. [PMID: 37083810 PMCID: PMC10118340 DOI: 10.1097/md.0000000000033620] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Accepted: 04/04/2023] [Indexed: 04/22/2023] Open
Abstract
Pyroptosis is a newly identified mode of programmed cell death, but the potential role in patients with acute myocardial infarction (AMI) remains unclear. In this study, bioinformatics methods were used to identify differentially expressed genes from peripheral blood transcriptome data between normal subjects and patients with AMI which were downloaded by the Gene Expression Omnibus database. Comparing Random Forest (RF) and Support Vector Machine (SVM) training algorithms were used to identify pyroptosis-related genes, predicting patients with AMI by nomogram based on informative genes. Moreover, clustering was used to amplify the feature of pyroptosis, in order to facilitate analysis distinct biological differences. Diversity analysis indicated that a majority of pyroptosis-related genes are expressed at higher levels in patients with AMI. The receiver operating characteristic curves show that the RF model is more responsive than the SVM machine learning model to the pyroptosis characteristics of these patients in vivo. We obtained a column line graph diagnostic model which was developed based on 19 genes established by the RF model. After the consensus clustering algorithm of single sample Gene Set Enrichment Analysis and Kyoto Encyclopedia of Genes and Genomes (KEGG) Enrichment Analysis, the results for them found that pyroptosis-related genes mediate the activation of multiple immune cells and many inflammatory pathways in the body. We used RF and SVM algorithms to determine 19 pyroptosis-related genes and evaluate their immunological effects in patients with AMI. We also constructed a series of by nomogram related to pyroptosis-related genes to predict the risk of developing AMI.
Collapse
Affiliation(s)
- Huan Wu
- School of Preclinical Medicine, Guizhou University of Traditional Chinese Medicine, Guiyang, Guizhou, China
| | - Xiaoman Xiong
- School of Preclinical Medicine, Guizhou University of Traditional Chinese Medicine, Guiyang, Guizhou, China
| | - Xueying CUI
- Qingyun County People’s Hospital, Qingyun, Shandong, China
| | - Jianlong Xiong
- School of Preclinical Medicine, Guizhou University of Traditional Chinese Medicine, Guiyang, Guizhou, China
| | - Yan Zhang
- School of Preclinical Medicine, Guizhou University of Traditional Chinese Medicine, Guiyang, Guizhou, China
| | - Liubo Xiang
- School of Preclinical Medicine, Guizhou University of Traditional Chinese Medicine, Guiyang, Guizhou, China
| | - TAO Xu
- School of Preclinical Medicine, Guizhou University of Traditional Chinese Medicine, Guiyang, Guizhou, China
| |
Collapse
|
33
|
Xuan S, Zhang J, Guo Q, Zhao L, Yao X. A Diagnostic Classifier Based on Circulating miRNA Pairs for COPD Using a Machine Learning Approach. Diagnostics (Basel) 2023; 13:diagnostics13081440. [PMID: 37189541 DOI: 10.3390/diagnostics13081440] [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: 02/16/2023] [Revised: 03/29/2023] [Accepted: 04/14/2023] [Indexed: 05/17/2023] Open
Abstract
Chronic obstructive pulmonary disease (COPD) is highly underdiagnosed, and early detection is urgent to prevent advanced progression. Circulating microRNAs (miRNAs) have been diagnostic candidates for multiple diseases. However, their diagnostic value has not yet been fully established in COPD. The purpose of this study was to develop an effective model for the diagnosis of COPD based on circulating miRNAs. We included circulating miRNA expression profiles of two independent cohorts consisting of 63 COPD and 110 normal samples, and then we constructed a miRNA pair-based matrix. Diagnostic models were developed using several machine learning algorithms. The predictive performance of the optimal model was validated in our external cohort. In this study, the diagnostic values of miRNAs based on the expression levels were unsatisfactory. We identified five key miRNA pairs and further developed seven machine learning models. The classifier based on LightGBM was selected as the final model with the area under the curve (AUC) values of 0.883 and 0.794 in test and validation datasets, respectively. We also built a web tool to assist diagnosis for clinicians. Enriched signaling pathways indicated the potential biological functions of the model. Collectively, we developed a robust machine learning model based on circulating miRNAs for COPD screening.
Collapse
Affiliation(s)
- Shurui Xuan
- Department of Respiratory & Critical Care Medicine, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing 210029, China
| | - Jiayue Zhang
- Department of Neurosurgery, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing 210029, China
| | - Qinxing Guo
- Department of Respiratory & Critical Care Medicine, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing 210029, China
| | - Liang Zhao
- Department of Neurosurgery, The Affiliated Brain Hospital of Nanjing Medical University, 264 Guangzhou Road, Nanjing 210029, China
| | - Xin Yao
- Department of Respiratory & Critical Care Medicine, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing 210029, China
| |
Collapse
|
34
|
Brockley LJ, Souza VGP, Forder A, Pewarchuk ME, Erkan M, Telkar N, Benard K, Trejo J, Stewart MD, Stewart GL, Reis PP, Lam WL, Martinez VD. Sequence-Based Platforms for Discovering Biomarkers in Liquid Biopsy of Non-Small-Cell Lung Cancer. Cancers (Basel) 2023; 15:2275. [PMID: 37190212 PMCID: PMC10136462 DOI: 10.3390/cancers15082275] [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: 03/07/2023] [Revised: 04/07/2023] [Accepted: 04/11/2023] [Indexed: 05/17/2023] Open
Abstract
Lung cancer detection and monitoring are hampered by a lack of sensitive biomarkers, which results in diagnosis at late stages and difficulty in tracking response to treatment. Recent developments have established liquid biopsies as promising non-invasive methods for detecting biomarkers in lung cancer patients. With concurrent advances in high-throughput sequencing technologies and bioinformatics tools, new approaches for biomarker discovery have emerged. In this article, we survey established and emerging biomarker discovery methods using nucleic acid materials derived from bodily fluids in the context of lung cancer. We introduce nucleic acid biomarkers extracted from liquid biopsies and outline biological sources and methods of isolation. We discuss next-generation sequencing (NGS) platforms commonly used to identify novel biomarkers and describe how these have been applied to liquid biopsy. We highlight emerging biomarker discovery methods, including applications of long-read sequencing, fragmentomics, whole-genome amplification methods for single-cell analysis, and whole-genome methylation assays. Finally, we discuss advanced bioinformatics tools, describing methods for processing NGS data, as well as recently developed software tailored for liquid biopsy biomarker detection, which holds promise for early diagnosis of lung cancer.
Collapse
Affiliation(s)
- Liam J. Brockley
- British Columbia Cancer Research Institute, Vancouver, BC V5Z 1L3, Canada; (V.G.P.S.); (A.F.); (M.E.P.); (N.T.); (K.B.); (J.T.); (M.D.S.); (G.L.S.); (W.L.L.)
| | - Vanessa G. P. Souza
- British Columbia Cancer Research Institute, Vancouver, BC V5Z 1L3, Canada; (V.G.P.S.); (A.F.); (M.E.P.); (N.T.); (K.B.); (J.T.); (M.D.S.); (G.L.S.); (W.L.L.)
- Molecular Oncology Laboratory, Experimental Research Unit, School of Medicine, São Paulo State University (UNESP), Botucatu 18618-687, SP, Brazil;
| | - Aisling Forder
- British Columbia Cancer Research Institute, Vancouver, BC V5Z 1L3, Canada; (V.G.P.S.); (A.F.); (M.E.P.); (N.T.); (K.B.); (J.T.); (M.D.S.); (G.L.S.); (W.L.L.)
| | - Michelle E. Pewarchuk
- British Columbia Cancer Research Institute, Vancouver, BC V5Z 1L3, Canada; (V.G.P.S.); (A.F.); (M.E.P.); (N.T.); (K.B.); (J.T.); (M.D.S.); (G.L.S.); (W.L.L.)
| | - Melis Erkan
- Department of Pathology and Laboratory Medicine, IWK Health Centre, Halifax, NS B3K 6R8, Canada;
- Department of Pathology, Faculty of Medicine, Dalhousie University, Halifax, NS B3K 6R8, Canada
- Beatrice Hunter Cancer Research Institute, Halifax, NS B3H 4R2, Canada
| | - Nikita Telkar
- British Columbia Cancer Research Institute, Vancouver, BC V5Z 1L3, Canada; (V.G.P.S.); (A.F.); (M.E.P.); (N.T.); (K.B.); (J.T.); (M.D.S.); (G.L.S.); (W.L.L.)
- British Columbia Children’s Hospital Research Institute, Vancouver, BC V5Z 4H4, Canada
| | - Katya Benard
- British Columbia Cancer Research Institute, Vancouver, BC V5Z 1L3, Canada; (V.G.P.S.); (A.F.); (M.E.P.); (N.T.); (K.B.); (J.T.); (M.D.S.); (G.L.S.); (W.L.L.)
| | - Jessica Trejo
- British Columbia Cancer Research Institute, Vancouver, BC V5Z 1L3, Canada; (V.G.P.S.); (A.F.); (M.E.P.); (N.T.); (K.B.); (J.T.); (M.D.S.); (G.L.S.); (W.L.L.)
| | - Matt D. Stewart
- British Columbia Cancer Research Institute, Vancouver, BC V5Z 1L3, Canada; (V.G.P.S.); (A.F.); (M.E.P.); (N.T.); (K.B.); (J.T.); (M.D.S.); (G.L.S.); (W.L.L.)
| | - Greg L. Stewart
- British Columbia Cancer Research Institute, Vancouver, BC V5Z 1L3, Canada; (V.G.P.S.); (A.F.); (M.E.P.); (N.T.); (K.B.); (J.T.); (M.D.S.); (G.L.S.); (W.L.L.)
| | - Patricia P. Reis
- Molecular Oncology Laboratory, Experimental Research Unit, School of Medicine, São Paulo State University (UNESP), Botucatu 18618-687, SP, Brazil;
- Department of Surgery and Orthopedics, Faculty of Medicine, São Paulo State University (UNESP), Botucatu 18618-687, SP, Brazil
| | - Wan L. Lam
- British Columbia Cancer Research Institute, Vancouver, BC V5Z 1L3, Canada; (V.G.P.S.); (A.F.); (M.E.P.); (N.T.); (K.B.); (J.T.); (M.D.S.); (G.L.S.); (W.L.L.)
| | - Victor D. Martinez
- Department of Pathology and Laboratory Medicine, IWK Health Centre, Halifax, NS B3K 6R8, Canada;
- Department of Pathology, Faculty of Medicine, Dalhousie University, Halifax, NS B3K 6R8, Canada
- Beatrice Hunter Cancer Research Institute, Halifax, NS B3H 4R2, Canada
| |
Collapse
|
35
|
Khadirnaikar S, Shukla S, Prasanna SRM. Machine learning based combination of multi-omics data for subgroup identification in non-small cell lung cancer. Sci Rep 2023; 13:4636. [PMID: 36944673 PMCID: PMC10030850 DOI: 10.1038/s41598-023-31426-w] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Accepted: 03/11/2023] [Indexed: 03/23/2023] Open
Abstract
Non-small Cell Lung Cancer (NSCLC) is a heterogeneous disease with a poor prognosis. Identifying novel subtypes in cancer can help classify patients with similar molecular and clinical phenotypes. This work proposes an end-to-end pipeline for subgroup identification in NSCLC. Here, we used a machine learning (ML) based approach to compress the multi-omics NSCLC data to a lower dimensional space. This data is subjected to consensus K-means clustering to identify the five novel clusters (C1-C5). Survival analysis of the resulting clusters revealed a significant difference in the overall survival of clusters (p-value: 0.019). Each cluster was then molecularly characterized to identify specific molecular characteristics. We found that cluster C3 showed minimal genetic aberration with a high prognosis. Next, classification models were developed using data from each omic level to predict the subgroup of unseen patients. Decision‑level fused classification models were then built using these classifiers, which were used to classify unseen patients into five novel clusters. We also showed that the multi-omics-based classification model outperformed single-omic-based models, and the combination of classifiers proved to be a more accurate prediction model than the individual classifiers. In summary, we have used ML models to develop a classification method and identified five novel NSCLC clusters with different genetic and clinical characteristics.
Collapse
Affiliation(s)
- Seema Khadirnaikar
- Department of Electrical Engineering, Indian Institute of Technology Dharwad, Dharwad, India
| | - Sudhanshu Shukla
- Department of Biosciences and Bioengineering, Indian Institute of Technology Dharwad, Dharwad, India.
| | - S R M Prasanna
- Department of Electrical Engineering, Indian Institute of Technology Dharwad, Dharwad, India
| |
Collapse
|
36
|
Casotti MC, Meira DD, Alves LNR, Bessa BGDO, Campanharo CV, Vicente CR, Aguiar CC, Duque DDA, Barbosa DG, dos Santos EDVW, Garcia FM, de Paula F, Santana GM, Pavan IP, Louro LS, Braga RFR, Trabach RSDR, Louro TS, de Carvalho EF, Louro ID. Translational Bioinformatics Applied to the Study of Complex Diseases. Genes (Basel) 2023; 14:419. [PMID: 36833346 PMCID: PMC9956936 DOI: 10.3390/genes14020419] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Revised: 01/29/2023] [Accepted: 01/31/2023] [Indexed: 02/10/2023] Open
Abstract
Translational Bioinformatics (TBI) is defined as the union of translational medicine and bioinformatics. It emerges as a major advance in science and technology by covering everything, from the most basic database discoveries, to the development of algorithms for molecular and cellular analysis, as well as their clinical applications. This technology makes it possible to access the knowledge of scientific evidence and apply it to clinical practice. This manuscript aims to highlight the role of TBI in the study of complex diseases, as well as its application to the understanding and treatment of cancer. An integrative literature review was carried out, obtaining articles through several websites, among them: PUBMED, Science Direct, NCBI-PMC, Scientific Electronic Library Online (SciELO), and Google Academic, published in English, Spanish, and Portuguese, indexed in the referred databases and answering the following guiding question: "How does TBI provide a scientific understanding of complex diseases?" An additional effort is aimed at the dissemination, inclusion, and perpetuation of TBI knowledge from the academic environment to society, helping the study, understanding, and elucidating of complex disease mechanics and their treatment.
Collapse
Affiliation(s)
- Matheus Correia Casotti
- Departamento de Ciências Biológicas, Universidade Federal do Espírito Santo, Vitória 29075-010, Espírito Santo, Brazil
| | - Débora Dummer Meira
- Departamento de Ciências Biológicas, Universidade Federal do Espírito Santo, Vitória 29075-010, Espírito Santo, Brazil
| | - Lyvia Neves Rebello Alves
- Departamento de Ciências Biológicas, Universidade Federal do Espírito Santo, Vitória 29075-010, Espírito Santo, Brazil
| | | | - Camilly Victória Campanharo
- Departamento de Ciências Biológicas, Universidade Federal do Espírito Santo, Vitória 29075-010, Espírito Santo, Brazil
| | - Creuza Rachel Vicente
- Departamento de Medicina Social, Universidade Federal do Espírito Santo, Vitória 29040-090, Espírito Santo, Brazil
| | - Carla Carvalho Aguiar
- Departamento de Ciências Biológicas, Universidade Federal do Espírito Santo, Vitória 29075-010, Espírito Santo, Brazil
| | - Daniel de Almeida Duque
- Departamento de Ciências Biológicas, Universidade Federal do Espírito Santo, Vitória 29075-010, Espírito Santo, Brazil
| | - Débora Gonçalves Barbosa
- Departamento de Ciências Biológicas, Universidade Federal do Espírito Santo, Vitória 29075-010, Espírito Santo, Brazil
| | | | - Fernanda Mariano Garcia
- Departamento de Ciências Biológicas, Universidade Federal do Espírito Santo, Vitória 29075-010, Espírito Santo, Brazil
| | - Flávia de Paula
- Departamento de Ciências Biológicas, Universidade Federal do Espírito Santo, Vitória 29075-010, Espírito Santo, Brazil
| | - Gabriel Mendonça Santana
- Departamento de Ciências Biológicas, Universidade Federal do Espírito Santo, Vitória 29075-010, Espírito Santo, Brazil
| | - Isabele Pagani Pavan
- Departamento de Ciências Biológicas, Universidade Federal do Espírito Santo, Vitória 29075-010, Espírito Santo, Brazil
| | - Luana Santos Louro
- Departamento de Ciências Biológicas, Universidade Federal do Espírito Santo, Vitória 29075-010, Espírito Santo, Brazil
| | - Raquel Furlani Rocon Braga
- Departamento de Ciências Biológicas, Universidade Federal do Espírito Santo, Vitória 29075-010, Espírito Santo, Brazil
| | - Raquel Silva dos Reis Trabach
- Departamento de Ciências Biológicas, Universidade Federal do Espírito Santo, Vitória 29075-010, Espírito Santo, Brazil
| | - Thomas Santos Louro
- Escola Superior de Ciências da Santa Casa de Misericórdia de Vitória (EMESCAM), Vitória 29027-502, Espírito Santo, Brazil
| | - Elizeu Fagundes de Carvalho
- Instituto de Biologia Roberto Alcantara Gomes (IBRAG), Universidade do Estado do Rio de Janeiro (UERJ), Rio de Janeiro 20551-030, Rio de Janeiro, Brazil
| | - Iúri Drumond Louro
- Departamento de Ciências Biológicas, Universidade Federal do Espírito Santo, Vitória 29075-010, Espírito Santo, Brazil
| |
Collapse
|
37
|
Feldner-Busztin D, Firbas Nisantzis P, Edmunds SJ, Boza G, Racimo F, Gopalakrishnan S, Limborg MT, Lahti L, de Polavieja GG. Dealing with dimensionality: the application of machine learning to multi-omics data. Bioinformatics 2023; 39:6986971. [PMID: 36637211 PMCID: PMC9907220 DOI: 10.1093/bioinformatics/btad021] [Citation(s) in RCA: 32] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Revised: 12/02/2022] [Accepted: 01/11/2023] [Indexed: 01/14/2023] Open
Abstract
MOTIVATION Machine learning (ML) methods are motivated by the need to automate information extraction from large datasets in order to support human users in data-driven tasks. This is an attractive approach for integrative joint analysis of vast amounts of omics data produced in next generation sequencing and other -omics assays. A systematic assessment of the current literature can help to identify key trends and potential gaps in methodology and applications. We surveyed the literature on ML multi-omic data integration and quantitatively explored the goals, techniques and data involved in this field. We were particularly interested in examining how researchers use ML to deal with the volume and complexity of these datasets. RESULTS Our main finding is that the methods used are those that address the challenges of datasets with few samples and many features. Dimensionality reduction methods are used to reduce the feature count alongside models that can also appropriately handle relatively few samples. Popular techniques include autoencoders, random forests and support vector machines. We also found that the field is heavily influenced by the use of The Cancer Genome Atlas dataset, which is accessible and contains many diverse experiments. AVAILABILITY AND IMPLEMENTATION All data and processing scripts are available at this GitLab repository: https://gitlab.com/polavieja_lab/ml_multi-omics_review/ or in Zenodo: https://doi.org/10.5281/zenodo.7361807. SUPPLEMENTARY INFORMATION Supplementary data are available at Bioinformatics online.
Collapse
Affiliation(s)
- Dylan Feldner-Busztin
- Champalimaud Centre for the Unknown, Champalimaud Foundation, 1400-038 Lisbon, Portugal
| | | | - Shelley Jane Edmunds
- Center for Evolutionary Hologenomics, GLOBE Institute, Faculty of Health and Medical Sciences, University of Copenhagen, 1353 Copenhagen, Denmark
| | - Gergely Boza
- Centre for Ecological Research, 1113 Budapest, Hungary
| | - Fernando Racimo
- Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Shyam Gopalakrishnan
- Center for Evolutionary Hologenomics, GLOBE Institute, Faculty of Health and Medical Sciences, University of Copenhagen, 1353 Copenhagen, Denmark
| | - Morten Tønsberg Limborg
- Center for Evolutionary Hologenomics, GLOBE Institute, Faculty of Health and Medical Sciences, University of Copenhagen, 1353 Copenhagen, Denmark
| | - Leo Lahti
- Department of Computing, University of Turku, 20014 Turku, Finland
| | | |
Collapse
|
38
|
Meng H, Peng Y, Li P, Su J, Jiang Y, Fu X. Global trends in research of high-throughput sequencing technology associated with chronic wounds from 2002 to 2022: A bibliometric and visualized study. Front Surg 2023; 10:1089203. [PMID: 36911623 PMCID: PMC9992981 DOI: 10.3389/fsurg.2023.1089203] [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: 11/04/2022] [Accepted: 02/06/2023] [Indexed: 02/24/2023] Open
Abstract
Background Chronic wounds are a complex medical problem. With the difficulty of skin healing, the microbial ecology of chronic wounds is an essential factor affecting wound healing. High-throughput sequencing (HTS) technology is a vital method to reveal the microbiome diversity and population structure of chronic wounds. Objective The aim of this paper was to delineate the scientific output characteristics, research trends, hotspots and frontiers of HTS technologies related to chronic wounds globally over the past 20 years. Methods We searched the Web of Science Core Collection (WoSCC) database for articles published between 2002 and 2022 and their full record information. The Bibliometrix software package was used to analyze bibliometric indicators and VOSviewer visualization analysis results. Results Ultimately, a total of 449 original articles were reviewed, and the results showed that the number of annual publications (Nps) about HTS associated with chronic wounds has steadily increased over the last 20 years. The United States and China produce the most articles and have the highest H-index, while the United States and England have the largest number of citations (Nc) in this field. The University of California, Wound Repair and Regeneration and National Institutes of Health Nih United States were the most published institutions, journals and fund resources, respectively. The global research could be divided into 3 clusters as follows: microbial infection of chronic wounds, the healing process of wounds and microscopic processes, skin repair mechanism stimulated by antimicrobial peptides and oxidative stress. In recent years, "wound healing", "infections", "expression", "inflammation", "chronic wounds", "identification" and "bacteria" "angiogenesis", "biofilms" and "diabetes" were the most frequently used keywords. In addition, research on "prevalence", "gene expression", "inflammation" and "infection" has recently become a hotspot. Conclusions This paper compares the research hotspots and directions in this field globally from the perspectives of countries, institutions and authors, analyzes the trend of international cooperation, and reveals the future development direction of the field and research hotspots of great scientific research value. Through this paper, we can further explore the value of HTS technology in chronic wounds to better solve the problem of chronic wounds.
Collapse
Affiliation(s)
- Hao Meng
- Research Center for Wound Repair and Tissue Regeneration, Medical Innovation Research Department, The PLA General Hospital, Beijing, China
| | - Yu Peng
- Research Center for Wound Repair and Tissue Regeneration, Medical Innovation Research Department, The PLA General Hospital, Beijing, China.,School of Medicine, Nankai University, Tianjin, China
| | - Pinxue Li
- School of Medicine, Nankai University, Tianjin, China
| | - Jianlong Su
- Research Center for Wound Repair and Tissue Regeneration, Medical Innovation Research Department, The PLA General Hospital, Beijing, China
| | - Yufeng Jiang
- Research Center for Wound Repair and Tissue Regeneration, Medical Innovation Research Department, The PLA General Hospital, Beijing, China
| | - Xiaobing Fu
- Research Center for Wound Repair and Tissue Regeneration, Medical Innovation Research Department, The PLA General Hospital, Beijing, China.,School of Medicine, Nankai University, Tianjin, China
| |
Collapse
|
39
|
Feng J, Fu F, Nie Y. Comprehensive genomics analysis of aging related gene signature to predict the prognosis and drug resistance of colon adenocarcinoma. Front Pharmacol 2023; 14:1121634. [PMID: 36925638 PMCID: PMC10011090 DOI: 10.3389/fphar.2023.1121634] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Accepted: 02/14/2023] [Indexed: 03/06/2023] Open
Abstract
Background: Colon adenocarcinoma (COAD) is a heterogeneous tumor and senescence is crucial in the occurrence of cancer. This study aimed to identify senescence-based subtypes and construct a prognostic signature to predict the prognosis and guide immunotherapy or chemotherapy decisions for COAD patients. Methods: Based on the single-cell RNA sequencing (scRNA-seq) data of 13 samples from the Gene Expression Omnibus (GEO) database, we assessed cellular senescence characteristics. Transcriptome data, copy number variations (CNVs) and single nucleotide variations (SNVs) data were obtained from The Cancer Genome Atlas (TCGA) database. GSE39582 and GSE17537 were used for validation. Senescence subtypes were identified using unsupervised consensus clustering analysis, and a prognostic signature was developed using univariate Cox analysis and least absolute shrinkage and selection operator (LASSO). Response of risk groups to chemotherapy was predicted using the half-maximal inhibitory concentration (IC50) values. We further analyzed the relationship between risk gene expression and methylation level. The prediction performance was assessed by nomogram. Results: Senescence-related pathways were highly enriched in malignant cells and bulk RNA-seq verified cellular senescence. Three senescence subtypes were identified, in which patients in clust3 had poorest prognosis and higher T stage, accompanied with higher tumor mutation burden (TMB) and mutations, activated inflammatory response, more immune cell infiltration, and higher immune escape tendency. A senescence-based signature using 11 genes (MFNG, GPRC5B, TNNT1, CCL22, NOXA1, PABPC1L, PCOLCE2, MID2, CPA3, HSPA1A, and CALB1) was established, and accurately predicted a lower prognosis in high risk patients. Its robustness was validated by external cohort. Low risk patients were more sensitive to small molecule drugs including Erlotinib, Sunitinib, MG-132, CGP-082996, AZ628, Sorafenib, VX-680, and Z-LLNle-CHO. Risk score was an independent prognostic factor and nomogram confirmed its reliability. Four risk genes (CALB1, CPA3, NOXA1, and TNNT1) had significant positive correlation with their methylation level, while six genes (CCL22, GPRC5B, HSPA1A, MFNG, PABPC1L, and PCOLCE2) were negatively correlated with their methylation level. Conclusion: This study provides novel understanding of heterogeneity in COAD from the perspective of senescence, and develops signatures for prognosis prediction in COAD.
Collapse
Affiliation(s)
- Jubin Feng
- The First Affiliated Hospital, Jinan University, Guangzhou, China.,Department of Gastroenterology, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Fengyihuan Fu
- The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Yuqiang Nie
- The First Affiliated Hospital, Jinan University, Guangzhou, China.,Department of Gastroenterology, School of Medicine, Guangzhou First People's Hospital, South China University of Technology, Guangzhou, China
| |
Collapse
|
40
|
Williams EC, Chazarra-Gil R, Shahsavari A, Mohorianu I. The Sum of Two Halves May Be Different from the Whole-Effects of Splitting Sequencing Samples Across Lanes. Genes (Basel) 2022; 13:genes13122265. [PMID: 36553532 PMCID: PMC9777937 DOI: 10.3390/genes13122265] [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: 11/07/2022] [Revised: 11/23/2022] [Accepted: 11/25/2022] [Indexed: 12/03/2022] Open
Abstract
The advances in high-throughput sequencing (HTS) have enabled the characterisation of biological processes at an unprecedented level of detail; most hypotheses in molecular biology rely on analyses of HTS data. However, achieving increased robustness and reproducibility of results remains a main challenge. Although variability in results may be introduced at various stages, e.g., alignment, summarisation or detection of differential expression, one source of variability was systematically omitted: the sequencing design, which propagates through analyses and may introduce an additional layer of technical variation. We illustrate qualitative and quantitative differences arising from splitting samples across lanes on bulk and single-cell sequencing. For bulk mRNAseq data, we focus on differential expression and enrichment analyses; for bulk ChIPseq data, we investigate the effect on peak calling and the peaks' properties. At the single-cell level, we concentrate on identifying cell subpopulations. We rely on markers used for assigning cell identities; both smartSeq and 10× data are presented. The observed reduction in the number of unique sequenced fragments limits the level of detail on which the different prediction approaches depend. Furthermore, the sequencing stochasticity adds in a weighting bias corroborated with variable sequencing depths and (yet unexplained) sequencing bias. Subsequently, we observe an overall reduction in sequencing complexity and a distortion in the biological signal across technologies, experimental contexts, organisms and tissues.
Collapse
Affiliation(s)
- Eleanor C. Williams
- Wellcome-MRC Cambridge Stem Cell Institute, University of Cambridge, Cambridge CB2 0AW, UK
| | - Ruben Chazarra-Gil
- Wellcome-MRC Cambridge Stem Cell Institute, University of Cambridge, Cambridge CB2 0AW, UK
- Life Sciences-Transcriptomics and Functional Genomics Lab, Barcelona Supercomputing Center (BSC-CNS), 08034 Barcelona, Spain
| | - Arash Shahsavari
- Wellcome-MRC Cambridge Stem Cell Institute, University of Cambridge, Cambridge CB2 0AW, UK
| | - Irina Mohorianu
- Wellcome-MRC Cambridge Stem Cell Institute, University of Cambridge, Cambridge CB2 0AW, UK
- Correspondence:
| |
Collapse
|
41
|
Muñoz-Barrera A, Rubio-Rodríguez LA, Díaz-de Usera A, Jáspez D, Lorenzo-Salazar JM, González-Montelongo R, García-Olivares V, Flores C. From Samples to Germline and Somatic Sequence Variation: A Focus on Next-Generation Sequencing in Melanoma Research. Life (Basel) 2022; 12:1939. [PMID: 36431075 PMCID: PMC9695713 DOI: 10.3390/life12111939] [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: 10/28/2022] [Revised: 11/12/2022] [Accepted: 11/16/2022] [Indexed: 11/24/2022] Open
Abstract
Next-generation sequencing (NGS) applications have flourished in the last decade, permitting the identification of cancer driver genes and profoundly expanding the possibilities of genomic studies of cancer, including melanoma. Here we aimed to present a technical review across many of the methodological approaches brought by the use of NGS applications with a focus on assessing germline and somatic sequence variation. We provide cautionary notes and discuss key technical details involved in library preparation, the most common problems with the samples, and guidance to circumvent them. We also provide an overview of the sequence-based methods for cancer genomics, exposing the pros and cons of targeted sequencing vs. exome or whole-genome sequencing (WGS), the fundamentals of the most common commercial platforms, and a comparison of throughputs and key applications. Details of the steps and the main software involved in the bioinformatics processing of the sequencing results, from preprocessing to variant prioritization and filtering, are also provided in the context of the full spectrum of genetic variation (SNVs, indels, CNVs, structural variation, and gene fusions). Finally, we put the emphasis on selected bioinformatic pipelines behind (a) short-read WGS identification of small germline and somatic variants, (b) detection of gene fusions from transcriptomes, and (c) de novo assembly of genomes from long-read WGS data. Overall, we provide comprehensive guidance across the main methodological procedures involved in obtaining sequencing results for the most common short- and long-read NGS platforms, highlighting key applications in melanoma research.
Collapse
Affiliation(s)
- Adrián Muñoz-Barrera
- Genomics Division, Instituto Tecnológico y de Energías Renovables (ITER), 38600 Santa Cruz de Tenerife, Spain
| | - Luis A. Rubio-Rodríguez
- Genomics Division, Instituto Tecnológico y de Energías Renovables (ITER), 38600 Santa Cruz de Tenerife, Spain
| | - Ana Díaz-de Usera
- Genomics Division, Instituto Tecnológico y de Energías Renovables (ITER), 38600 Santa Cruz de Tenerife, Spain
- Research Unit, Hospital Universitario Nuestra Señora de Candelaria, 38010 Santa Cruz de Tenerife, Spain
| | - David Jáspez
- Genomics Division, Instituto Tecnológico y de Energías Renovables (ITER), 38600 Santa Cruz de Tenerife, Spain
| | - José M. Lorenzo-Salazar
- Genomics Division, Instituto Tecnológico y de Energías Renovables (ITER), 38600 Santa Cruz de Tenerife, Spain
| | - Rafaela González-Montelongo
- Genomics Division, Instituto Tecnológico y de Energías Renovables (ITER), 38600 Santa Cruz de Tenerife, Spain
| | - Víctor García-Olivares
- Genomics Division, Instituto Tecnológico y de Energías Renovables (ITER), 38600 Santa Cruz de Tenerife, Spain
| | - Carlos Flores
- Genomics Division, Instituto Tecnológico y de Energías Renovables (ITER), 38600 Santa Cruz de Tenerife, Spain
- Research Unit, Hospital Universitario Nuestra Señora de Candelaria, 38010 Santa Cruz de Tenerife, Spain
- CIBER de Enfermedades Respiratorias, Instituto de Salud Carlos III, 28029 Madrid, Spain
- Facultad de Ciencias de la Salud, Universidad Fernando de Pessoa Canarias, 35450 Las Palmas de Gran Canaria, Spain
| |
Collapse
|
42
|
Marchetti L, Nifosì R, Martelli PL, Da Pozzo E, Cappello V, Banterle F, Trincavelli ML, Martini C, D’Elia M. Quantum computing algorithms: getting closer to critical problems in computational biology. Brief Bioinform 2022; 23:bbac437. [PMID: 36220772 PMCID: PMC9677474 DOI: 10.1093/bib/bbac437] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Revised: 08/15/2022] [Accepted: 09/08/2022] [Indexed: 12/14/2022] Open
Abstract
The recent biotechnological progress has allowed life scientists and physicians to access an unprecedented, massive amount of data at all levels (molecular, supramolecular, cellular and so on) of biological complexity. So far, mostly classical computational efforts have been dedicated to the simulation, prediction or de novo design of biomolecules, in order to improve the understanding of their function or to develop novel therapeutics. At a higher level of complexity, the progress of omics disciplines (genomics, transcriptomics, proteomics and metabolomics) has prompted researchers to develop informatics means to describe and annotate new biomolecules identified with a resolution down to the single cell, but also with a high-throughput speed. Machine learning approaches have been implemented to both the modelling studies and the handling of biomedical data. Quantum computing (QC) approaches hold the promise to resolve, speed up or refine the analysis of a wide range of these computational problems. Here, we review and comment on recently developed QC algorithms for biocomputing, with a particular focus on multi-scale modelling and genomic analyses. Indeed, differently from other computational approaches such as protein structure prediction, these problems have been shown to be adequately mapped onto quantum architectures, the main limit for their immediate use being the number of qubits and decoherence effects in the available quantum machines. Possible advantages over the classical counterparts are highlighted, along with a description of some hybrid classical/quantum approaches, which could be the closest to be realistically applied in biocomputation.
Collapse
Affiliation(s)
- Laura Marchetti
- University of Pisa, Department of Pharmacy, via Bonanno 6, 56126 Pisa Italy
| | - Riccardo Nifosì
- NEST, Istituto Nanoscienze-CNR and Scuola Normale Superiore, P.zza San Silvestro 12, 56127 Pisa Italy
| | - Pier Luigi Martelli
- University of Bologna, Department of Pharmacy and Biotechnology, via San Giacomo 9/2, 40126 Bologna Italy
| | - Eleonora Da Pozzo
- University of Pisa, Department of Pharmacy, via Bonanno 6, 56126 Pisa Italy
| | - Valentina Cappello
- Italian Institute of Technology, Center for Materials Interfaces, Viale Rinaldo Piaggio 34, 56025 Pontedera (PI), Italy
| | | | | | - Claudia Martini
- University of Pisa, Department of Pharmacy, via Bonanno 6, 56126 Pisa Italy
| | - Massimo D’Elia
- University of Pisa, Department of Physics, Largo Bruno Pontecorvo 3, 56127, Pisa Italy
- INFN, Sezione di Pisa, Largo Bruno Pontecorvo 3, I-56127 Pisa, Italy
| |
Collapse
|
43
|
Dai L, Xiong Z, Hou D, Wang Y, Li T, Long X, Chen H, Sun C. Pathogenicity and transcriptome analysis of a strain of Vibrio owensii in Fenneropenaeus merguiensis. FISH & SHELLFISH IMMUNOLOGY 2022; 130:194-205. [PMID: 36087819 DOI: 10.1016/j.fsi.2022.09.008] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/12/2022] [Revised: 08/29/2022] [Accepted: 09/02/2022] [Indexed: 06/15/2023]
Abstract
Vibrio is an important conditional pathogen in shrimp aquaculture. This research reported a dominant bacteria strain E1 isolated from a shrimp tank with the method of biofloc culture, which was further identified as Vibrio owensii. To understand the interaction between V. owensii and the host shrimp, we studied the pathogenicity of the V. owensii and the molecular mechanisms of the Fenneropenaeus merguiensis immunity during the Vibrio invasion. Drug susceptibility tests showed that V. owensii was resistant to antibiotics streptomycin oxacillin, tetracycline, minocycline, and aztreonam, but highly sensitive to cefazolin, cefotaxime, and ciprofloxacin, and moderately sensitive to cefotaxime, ampicillin, and piperacillin. Lethal concentration 50 (LC50) test was performed to evaluate the toxicity of V. owensii to F. merguiensis. The LC50 of V. owensii infected F. merguiensis after 24, 48, 72, 96, 120, 144 and 168 h were 1.21 × 107, 1.68 × 106, 6.36 × 105, 2.15 × 105, 7.58 × 104, 5.55 × 104 and 4.33 × 104 CFU/mL. In order to explore the molecular response mechanism of F. merguiensis infected with V. owensii, the hepatopancreas of F. merguiensis were sequenced at 24 hpi and 48 hpi, and a total 40,181 of unigenes were obtained. Through comparative transcriptomic analysis, 86 differentially expressed genes (DEGs) (including 38 up-regulated DEGs, and 48 down-regulated DEGs) and 305 DEGs (including 150 up-regulated DEGs, and 155 down-regulated DEGs) were identified at 24 hpi and 48 hpi, respectively. Annotation and classification analysis of these 391 DEGs showed that most of the DEGs were annotated to metableolic and immune pathways, which indicated that F. merguiensis responded to the invasion through the regulation of material metableolism and immune system genes during V. owensii infection. In the KEGG enrichment analysis, some pathways related to immune response were significantly influenced by V. owensii infection, including phagosome, MAPK signalling pathway and PI3K-Akt signalling pathway. In addition, some pathways related to the warburg effect were also significantly enriched after V. owensii infection, including pyruvate metableolism, glycolysis/gluconeogenesis, and citrate cycle (TAC cycle). Further analysis showed that C-type lectins and ficolin were also play important roles in the immune response of F. merguiensis against V. owensii infection. The current research preliminarily revealed the immune response of F. merguiensis to V. owensii infection at the molecular level, which provided valuable information to further understand the disease control and the interaction between shrimp and Vibrio.
Collapse
Affiliation(s)
- Linxin Dai
- College of Fisheries, Guangdong Ocean University, Zhanjiang, Guangdong, China
| | - Zhiwang Xiong
- College of Fisheries, Guangdong Ocean University, Zhanjiang, Guangdong, China
| | - Danqing Hou
- College of Fisheries, Guangdong Ocean University, Zhanjiang, Guangdong, China
| | - Yue Wang
- College of Fisheries, Guangdong Ocean University, Zhanjiang, Guangdong, China
| | - Ting Li
- College of Fisheries, Guangdong Ocean University, Zhanjiang, Guangdong, China
| | - Xinxin Long
- College of Fisheries, Guangdong Ocean University, Zhanjiang, Guangdong, China
| | - Haozhen Chen
- College of Fisheries, Guangdong Ocean University, Zhanjiang, Guangdong, China
| | - Chengbo Sun
- College of Fisheries, Guangdong Ocean University, Zhanjiang, Guangdong, China; Guangdong Provincial Key Laboratory of Pathogenic Biology and Epidemiology for Aquatic Economic Animals, Zhanjiang, Guangdong, China; Guangdong Provincial Laboratory of Southern Marine Science and Engineering, Zhanjiang, Guangdong, China.
| |
Collapse
|
44
|
Nogueira BMF, Krishnan S, Barreto‐Duarte B, Araújo‐Pereira M, Queiroz ATL, Ellner JJ, Salgame P, Scriba TJ, Sterling TR, Gupta A, Andrade BB. Diagnostic biomarkers for active tuberculosis: progress and challenges. EMBO Mol Med 2022; 14:e14088. [PMID: 36314872 PMCID: PMC9728055 DOI: 10.15252/emmm.202114088] [Citation(s) in RCA: 42] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Revised: 09/05/2022] [Accepted: 09/05/2022] [Indexed: 12/14/2022] Open
Abstract
Tuberculosis (TB) is a leading cause of morbidity and mortality from a single infectious agent, despite being preventable and curable. Early and accurate diagnosis of active TB is critical to both enhance patient care, improve patient outcomes, and break Mycobacterium tuberculosis (Mtb) transmission cycles. In 2020 an estimated 9.9 million people fell ill from Mtb, but only a little over half (5.8 million) received an active TB diagnosis and treatment. The World Health Organization has proposed target product profiles for biomarker- or biosignature-based diagnostics using point-of-care tests from easily accessible specimens such as urine or blood. Here we review and summarize progress made in the development of pathogen- and host-based biomarkers for active TB diagnosis. We describe several unique patient populations that have posed challenges to development of a universal diagnostic TB biomarker, such as people living with HIV, extrapulmonary TB, and children. We also review additional limitations to widespread validation and utilization of published biomarkers. We conclude with proposed solutions to enhance TB diagnostic biomarker validation and uptake.
Collapse
Affiliation(s)
- Betânia M F Nogueira
- Programa de Pós‐graduação em Ciências da SaúdeUniversidade Federal da BahiaSalvadorBrazil,Instituto Couto MaiaSalvadorBrazil,Multinational Organization Network Sponsoring Translational and Epidemiological Research (MONSTER) InitiativeSalvadorBrazil
| | - Sonya Krishnan
- Division of Infectious Diseases, Department of MedicineJohns Hopkins University School of MedicineBaltimoreMDUSA
| | - Beatriz Barreto‐Duarte
- Multinational Organization Network Sponsoring Translational and Epidemiological Research (MONSTER) InitiativeSalvadorBrazil,Curso de MedicinaUniversidade Salvador (UNIFACS)SalvadorBrazil,Programa de Pós‐Graduação em Clínica MédicaUniversidade Federal do Rio de JaneiroRio de JaneiroBrazil,Laboratório de Inflamação e Biomarcadores, Instituto Gonçalo MonizFundação Oswaldo CruzSalvadorBrazil
| | - Mariana Araújo‐Pereira
- Multinational Organization Network Sponsoring Translational and Epidemiological Research (MONSTER) InitiativeSalvadorBrazil,Laboratório de Inflamação e Biomarcadores, Instituto Gonçalo MonizFundação Oswaldo CruzSalvadorBrazil,Faculdade de MedicinaUniversidade Federal da BahiaSalvadorBrazil
| | - Artur T L Queiroz
- Instituto Couto MaiaSalvadorBrazil,Center of Data and Knowledge Integration for Health (CIDACS), Instituto Gonçalo MonizFundação Oswaldo CruzSalvadorBrazil
| | - Jerrold J Ellner
- Department of Medicine, Centre for Emerging PathogensRutgers‐New Jersey Medical SchoolNewarkNJUSA
| | - Padmini Salgame
- Department of Medicine, Centre for Emerging PathogensRutgers‐New Jersey Medical SchoolNewarkNJUSA
| | - Thomas J Scriba
- South African Tuberculosis Vaccine Initiative and Institute of Infectious Disease and Molecular Medicine, Division of Immunology, Department of PathologyUniversity of Cape TownCape TownSouth Africa
| | - Timothy R Sterling
- Division of Infectious Diseases, Department of MedicineVanderbilt University Medical CenterNashvilleTNUSA
| | - Amita Gupta
- Division of Infectious Diseases, Department of MedicineJohns Hopkins University School of MedicineBaltimoreMDUSA
| | - Bruno B Andrade
- Multinational Organization Network Sponsoring Translational and Epidemiological Research (MONSTER) InitiativeSalvadorBrazil,Curso de MedicinaUniversidade Salvador (UNIFACS)SalvadorBrazil,Laboratório de Inflamação e Biomarcadores, Instituto Gonçalo MonizFundação Oswaldo CruzSalvadorBrazil,Faculdade de MedicinaUniversidade Federal da BahiaSalvadorBrazil,Curso de MedicinaFaculdade de Tecnologia e Ciências (FTC)SalvadorBrazil,Curso de MedicinaEscola Bahiana de Medicina e Saúde Pública (EBMSP)SalvadorBrazil
| |
Collapse
|
45
|
Trego A, Keating C, Nzeteu C, Graham A, O’Flaherty V, Ijaz UZ. Beyond Basic Diversity Estimates-Analytical Tools for Mechanistic Interpretations of Amplicon Sequencing Data. Microorganisms 2022; 10:1961. [PMID: 36296237 PMCID: PMC9609705 DOI: 10.3390/microorganisms10101961] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Revised: 09/29/2022] [Accepted: 09/30/2022] [Indexed: 11/07/2022] Open
Abstract
Understanding microbial ecology through amplifying short read regions, typically 16S rRNA for prokaryotic species or 18S rRNA for eukaryotic species, remains a popular, economical choice. These methods provide relative abundances of key microbial taxa, which, depending on the experimental design, can be used to infer mechanistic ecological underpinnings. In this review, we discuss recent advancements in in situ analytical tools that have the power to elucidate ecological phenomena, unveil the metabolic potential of microbial communities, identify complex multidimensional interactions between species, and compare stability and complexity under different conditions. Additionally, we highlight methods that incorporate various modalities and additional information, which in combination with abundance data, can help us understand how microbial communities respond to change in a typical ecosystem. Whilst the field of microbial informatics continues to progress substantially, our emphasis is on popular methods that are applicable to a broad range of study designs. The application of these methods can increase our mechanistic understanding of the ongoing dynamics of complex microbial communities.
Collapse
Affiliation(s)
- Anna Trego
- Microbial Ecology Laboratory, School of Biological and Chemical Sciences and the Ryan Institute, University of Galway, University Road, H91 TK33 Galway, Ireland
| | - Ciara Keating
- Institute of Biodiversity, Animal Health & Comparative Medicine, The University of Glasgow, Oakfield Avenue, Glasgow G12 8LT, UK
| | - Corine Nzeteu
- Microbial Ecology Laboratory, School of Biological and Chemical Sciences and the Ryan Institute, University of Galway, University Road, H91 TK33 Galway, Ireland
| | - Alison Graham
- Microbial Ecology Laboratory, School of Biological and Chemical Sciences and the Ryan Institute, University of Galway, University Road, H91 TK33 Galway, Ireland
| | - Vincent O’Flaherty
- Microbial Ecology Laboratory, School of Biological and Chemical Sciences and the Ryan Institute, University of Galway, University Road, H91 TK33 Galway, Ireland
| | - Umer Zeeshan Ijaz
- Water Engineering Group, School of Engineering, The University of Glasgow, Oakfield Avenue, Glasgow G12 8LT, UK
| |
Collapse
|
46
|
Lam L, Tien T, Wildung M, White L, Sellon RK, Fidel JL, Shelden EA. Comparative whole transcriptome analysis of gene expression in three canine soft tissue sarcoma types. PLoS One 2022; 17:e0273705. [PMID: 36099287 PMCID: PMC9469979 DOI: 10.1371/journal.pone.0273705] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Accepted: 08/11/2022] [Indexed: 11/29/2022] Open
Abstract
Soft tissue sarcomas are pleiotropic tumors of mesenchymal cell origin. These tumors are rare in humans but common in veterinary practice, where they comprise up to 15% of canine skin and subcutaneous cancers. Because they present similar morphologies, primary sites, and growth characteristics, they are treated similarly, generally by surgical resection followed by radiation therapy. Previous studies have examined a variety of genetic changes as potential drivers of tumorigenesis and progression in soft tissue sarcomas as well as their use as markers for soft tissue sarcoma subtypes. However, few studies employing next generation sequencing approaches have been published. Here, we have examined gene expression patterns in canine soft tissue sarcomas using RNA-seq analysis of samples obtained from archived formalin-fixed and paraffin-embedded tumors. We provide a computational framework for using resulting data to categorize tumors, perform cross species comparisons and identify genetic changes associated with tumorigenesis. Functional overrepresentation analysis of differentially expressed genes further implicate both common and tumor-type specific transcription factors as potential mediators of tumorigenesis and aggression. Implications for tumor-type specific therapies are discussed. Our results illustrate the potential utility of this approach for the discovery of new therapeutic approaches to the management of canine soft tissue sarcomas and support the view that both common and tumor-type specific mechanisms drive the development of these tumors.
Collapse
Affiliation(s)
- Lydia Lam
- School of Molecular Biosciences, College of Veterinary Medicine, Washington State University, Pullman, WA, United States of America
| | - Tien Tien
- Veterinary Clinical Sciences, College of Veterinary Medicine, Washington State University, Pullman, WA, United States of America
| | - Mark Wildung
- School of Molecular Biosciences, College of Veterinary Medicine, Washington State University, Pullman, WA, United States of America
| | - Laura White
- Washington Animal Disease Diagnostic Laboratory, College of Veterinary Medicine, Washington State University, Pullman, WA, United States of America
| | - Rance K. Sellon
- Veterinary Clinical Sciences, College of Veterinary Medicine, Washington State University, Pullman, WA, United States of America
| | - Janean L. Fidel
- Veterinary Clinical Sciences, College of Veterinary Medicine, Washington State University, Pullman, WA, United States of America
| | - Eric A. Shelden
- School of Molecular Biosciences, College of Veterinary Medicine, Washington State University, Pullman, WA, United States of America
| |
Collapse
|
47
|
Dotolo S, Esposito Abate R, Roma C, Guido D, Preziosi A, Tropea B, Palluzzi F, Giacò L, Normanno N. Bioinformatics: From NGS Data to Biological Complexity in Variant Detection and Oncological Clinical Practice. Biomedicines 2022; 10:biomedicines10092074. [PMID: 36140175 PMCID: PMC9495893 DOI: 10.3390/biomedicines10092074] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Revised: 08/12/2022] [Accepted: 08/22/2022] [Indexed: 11/22/2022] Open
Abstract
The use of next-generation sequencing (NGS) techniques for variant detection has become increasingly important in clinical research and in clinical practice in oncology. Many cancer patients are currently being treated in clinical practice or in clinical trials with drugs directed against specific genomic alterations. In this scenario, the development of reliable and reproducible bioinformatics tools is essential to derive information on the molecular characteristics of each patient’s tumor from the NGS data. The development of bioinformatics pipelines based on the use of machine learning and statistical methods is even more relevant for the determination of complex biomarkers. In this review, we describe some important technologies, computational algorithms and models that can be applied to NGS data from Whole Genome to Targeted Sequencing, to address the problem of finding complex cancer-associated biomarkers. In addition, we explore the future perspectives and challenges faced by bioinformatics for precision medicine both at a molecular and clinical level, with a focus on an emerging complex biomarker such as homologous recombination deficiency (HRD).
Collapse
Affiliation(s)
- Serena Dotolo
- Cell Biology and Biotherapy Unit, Istituto Nazionale Tumori—IRCCS—Fondazione G. Pascale, 80131 Naples, Italy
| | - Riziero Esposito Abate
- Cell Biology and Biotherapy Unit, Istituto Nazionale Tumori—IRCCS—Fondazione G. Pascale, 80131 Naples, Italy
| | - Cristin Roma
- Cell Biology and Biotherapy Unit, Istituto Nazionale Tumori—IRCCS—Fondazione G. Pascale, 80131 Naples, Italy
| | - Davide Guido
- Bioinformatics Research Core Facility, Gemelli Science and Technology Park (GSTeP), Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Largo A. Gemelli, 8, 00168 Rome, Italy
| | - Alessia Preziosi
- Bioinformatics Research Core Facility, Gemelli Science and Technology Park (GSTeP), Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Largo A. Gemelli, 8, 00168 Rome, Italy
| | - Beatrice Tropea
- Bioinformatics Research Core Facility, Gemelli Science and Technology Park (GSTeP), Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Largo A. Gemelli, 8, 00168 Rome, Italy
| | - Fernando Palluzzi
- Bioinformatics Research Core Facility, Gemelli Science and Technology Park (GSTeP), Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Largo A. Gemelli, 8, 00168 Rome, Italy
| | - Luciano Giacò
- Bioinformatics Research Core Facility, Gemelli Science and Technology Park (GSTeP), Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Largo A. Gemelli, 8, 00168 Rome, Italy
| | - Nicola Normanno
- Cell Biology and Biotherapy Unit, Istituto Nazionale Tumori—IRCCS—Fondazione G. Pascale, 80131 Naples, Italy
- Correspondence:
| |
Collapse
|
48
|
Ntafoulis I, Koolen SLW, Leenstra S, Lamfers MLM. Drug Repurposing, a Fast-Track Approach to Develop Effective Treatments for Glioblastoma. Cancers (Basel) 2022; 14:3705. [PMID: 35954371 PMCID: PMC9367381 DOI: 10.3390/cancers14153705] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2022] [Revised: 07/25/2022] [Accepted: 07/26/2022] [Indexed: 12/10/2022] Open
Abstract
Glioblastoma (GBM) remains one of the most difficult tumors to treat. The mean overall survival rate of 15 months and the 5-year survival rate of 5% have not significantly changed for almost 2 decades. Despite progress in understanding the pathophysiology of the disease, no new effective treatments to combine with radiation therapy after surgical tumor debulking have become available since the introduction of temozolomide in 1999. One of the main reasons for this is the scarcity of compounds that cross the blood-brain barrier (BBB) and reach the brain tumor tissue in therapeutically effective concentrations. In this review, we focus on the role of the BBB and its importance in developing brain tumor treatments. Moreover, we discuss drug repurposing, a drug discovery approach to identify potential effective candidates with optimal pharmacokinetic profiles for central nervous system (CNS) penetration and that allows rapid implementation in clinical trials. Additionally, we provide an overview of repurposed candidate drug currently being investigated in GBM at the preclinical and clinical levels. Finally, we highlight the importance of phase 0 trials to confirm tumor drug exposure and we discuss emerging drug delivery technologies as an alternative route to maximize therapeutic efficacy of repurposed candidate drug.
Collapse
Affiliation(s)
- Ioannis Ntafoulis
- Brain Tumor Center, Department of Neurosurgery, Erasmus MC Cancer Institute, Erasmus University Medical Center, 3015 CN Rotterdam, The Netherlands; (I.N.); (S.L.)
| | - Stijn L. W. Koolen
- Department of Medical Oncology, Erasmus MC Cancer Institute, Erasmus University Medical Center, 3015 CN Rotterdam, The Netherlands;
- Department of Hospital Pharmacy, Erasmus University Medical Center, 3015 CN Rotterdam, The Netherlands
| | - Sieger Leenstra
- Brain Tumor Center, Department of Neurosurgery, Erasmus MC Cancer Institute, Erasmus University Medical Center, 3015 CN Rotterdam, The Netherlands; (I.N.); (S.L.)
| | - Martine L. M. Lamfers
- Brain Tumor Center, Department of Neurosurgery, Erasmus MC Cancer Institute, Erasmus University Medical Center, 3015 CN Rotterdam, The Netherlands; (I.N.); (S.L.)
| |
Collapse
|
49
|
Database of Potential Promoter Sequences in the Capsicum annuum Genome. BIOLOGY 2022; 11:biology11081117. [PMID: 35892972 PMCID: PMC9332048 DOI: 10.3390/biology11081117] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Revised: 07/19/2022] [Accepted: 07/23/2022] [Indexed: 11/16/2022]
Abstract
In this study, we used a mathematical method for the multiple alignment of highly divergent sequences (MAHDS) to create a database of potential promoter sequences (PPSs) in the Capsicum annuum genome. To search for PPSs, 20 statistically significant classes of sequences located in the range from −499 to +100 nucleotides near the annotated genes were calculated. For each class, a position–weight matrix (PWM) was computed and then used to identify PPSs in the C. annuum genome. In total, 825,136 PPSs were detected, with a false positive rate of 0.13%. The PPSs obtained with the MAHDS method were tested using TSSFinder, which detects transcription start sites. The databank of the found PPSs provides their coordinates in chromosomes, the alignment of each PPS with the PWM, and the level of statistical significance as a normal distribution argument, and can be used in genetic engineering and biotechnology.
Collapse
|
50
|
Albogami S. Comprehensive analysis of gene expression profiles to identify differential prognostic factors of primary and metastatic breast cancer. Saudi J Biol Sci 2022; 29:103318. [PMID: 35677896 PMCID: PMC9168623 DOI: 10.1016/j.sjbs.2022.103318] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Revised: 04/17/2022] [Accepted: 05/19/2022] [Indexed: 12/21/2022] Open
Abstract
Breast cancer accounts for nearly half of all cancer-related deaths in women worldwide. However, the molecular mechanisms that lead to tumour development and progression remain poorly understood and there is a need to identify candidate genes associated with primary and metastatic breast cancer progression and prognosis. In this study, candidate genes associated with prognosis of primary and metastatic breast cancer were explored through a novel bioinformatics approach. Primary and metastatic breast cancer tissues and adjacent normal breast tissues were evaluated to identify biomarkers characteristic of primary and metastatic breast cancer. The Cancer Genome Atlas-breast invasive carcinoma (TCGA-BRCA) dataset (ID: HS-01619) was downloaded using the mRNASeq platform. Genevestigator 8.3.2 was used to analyse TCGA-BRCA gene expression profiles between the sample groups and identify the differentially-expressed genes (DEGs) in each group. For each group, Gene Ontology and Kyoto Encyclopedia of Genes and Genomes pathway enrichment analyses were used to determine the function of DEGs. Networks of protein-protein interactions were constructed to identify the top hub genes with the highest degree of interaction. Additionally, the top hub genes were validated based on overall survival and immunohistochemistry using The Human Protein Atlas. Of the top 20 hub genes identified, four (KRT14, KIT, RAD51, and TTK) were considered as prognostic risk factors based on overall survival. KRT14 and KIT expression levels were upregulated while those of RAD51 and TTK were downregulated in patients with breast cancer. The four proposed candidate hub genes might aid in further understanding the molecular changes that distinguish primary breast tumours from metastatic tumours as well as help in developing novel therapeutics. Furthermore, they may serve as effective prognostic risk markers based on the strong correlation between their expression and patient overall survival.
Collapse
Key Words
- BC, breast cancer
- BP, biological process
- Breast cancer
- CC, cellular component
- CI, confidence interval
- DEG, differentially expressed gene
- Differentially expressed genes
- FDR, false discovery rate
- GEPIA, gene expression profiling interactive analysis
- GO, gene ontology
- HR, hazard ratio
- IDC, infiltrating ductal carcinoma
- KEGG, Kyoto Encyclopedia of Genes and Genomes
- MCODE, molecular complex detection
- MF, molecular function
- Metastasis
- OS, overall survival
- Overall survival
- PPI, protein-protein interaction
- Prognostic marker
- Protein-protein interaction
- RNA-Seq, RNA sequencing
- STRING, search tool for the retrieval of interacting genes
- TCGA-BRCA, The Cancer Genome Atlas-breast invasive carcinoma
Collapse
Affiliation(s)
- Sarah Albogami
- Department of Biotechnology, College of Science, Taif University, P.O. Box 11099, Taif 21944, Saudi Arabia
| |
Collapse
|