1
|
Choudhuri S, Ghosh B. Computational approach for decoding malaria drug targets from single-cell transcriptomics and finding potential drug molecule. Sci Rep 2024; 14:24064. [PMID: 39402081 PMCID: PMC11473826 DOI: 10.1038/s41598-024-72427-7] [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: 01/23/2024] [Accepted: 09/06/2024] [Indexed: 10/17/2024] Open
Abstract
Malaria is a deadly disease caused by Plasmodium parasites. While potent drugs are available in the market for malaria treatment, over the years, Plasmodium parasites have successfully developed resistance against many, if not all, front-line drugs. This poses a serious threat to global malaria eradication efforts, and the continued discovery of new drugs is necessary to tackle this debilitating disease. With recent unprecedented progress in machine learning techniques, single-cell transcriptomic in Plasmodium offers a powerful tool for identifying crucial proteins as a drug target and subsequent computational prediction of potential drugs. In this study, We have implemented a mutual-information-based feature reduction algorithm with a classification algorithm to select important proteins from transcriptomic datasets (sexual and asexual stages) for Plasmodium falciparum and then constructed the protein-protein interaction (PPI) networks of the proteins. The analysis of this PPI network revealed key proteins vital for the survival of Plasmodium falciparum. Based on the function and identification of a few strong binding sites on a couple of these key proteins, we computationally predicted a set of potential drug molecules using a deep learning-based technique. Lead drug molecules that satisfy ADMET and drug-likeliness properties are finally reported out of the generated drugs. The study offers a general computational pipeline to identify crucial proteins using scRNA-seq data sets and further development of potential new drugs.
Collapse
Affiliation(s)
- Soham Choudhuri
- Center for Computational Natural Sciences and Bioinformatics, International Institute of Information Technology, Hyderabad, 500032, India
| | - Bhaswar Ghosh
- Center for Computational Natural Sciences and Bioinformatics, International Institute of Information Technology, Hyderabad, 500032, India.
| |
Collapse
|
2
|
Collí-Dulá RC, Papatheodorou I. Single-cell RNA sequencing offers opportunities to explore the depth of physiology, adaptation, and biochemistry in non-model organisms exposed to pollution. COMPARATIVE BIOCHEMISTRY AND PHYSIOLOGY. PART D, GENOMICS & PROTEOMICS 2024; 52:101339. [PMID: 39393164 DOI: 10.1016/j.cbd.2024.101339] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Revised: 09/28/2024] [Accepted: 10/02/2024] [Indexed: 10/13/2024]
Abstract
Single-cell Sequencing technology (scSeq) has revolutionized our understanding of individual cells, uncovering unprecedented heterogeneity within tissues and cell populations, principality through single-cell RNA Sequencing (scRNA-Seq). This short review highlights the pivotal role of scRNA-Seq in elucidating genotype-phenotype relationships, particularly in biological systems. Based on published articles, our analysis involved manual curation and automated Scopus tools to illustrate recent advances in the application of scRNA-Seq. The results reveal that scRNA-Seq has been extensively utilized in various biological areas, including biochemistry, genetics, molecular biology, immunology, and microbiology, followed by health sciences covering studies related to the nervous system, immune system, human health, development, and diseases, with a particular focus on cancer research. However, the potential of scRNA-Seq extends beyond disease research, offering insights into non-model organisms' responses to environmental contaminants. By enabling the study of cellular reactions at a molecular level, scRNA-Seq provides a comprehensive understanding of intracellular heterogeneity that enhances our comprehension of physiological, biochemical, and pathological environmental impacts on non-model organisms exposed to pollution. This understanding has many practical benefits, as it can aid in regulation and conservation efforts that benefit the environment and the use of economically essential and ecologically relevant organisms.
Collapse
Affiliation(s)
- Reyna C Collí-Dulá
- Departamento de Recursos del Mar, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, 97310 Mérida, Yucatán, Mexico; Consejo Nacional de Humanidades Ciencia y Tecnología, Ciudad de México, Mexico.
| | - Irene Papatheodorou
- European Bioinformatics Institute (EMBL-EBI) European Molecular Biology Laboratory, Wellcome Trust Genome Campus, Hinxton, Cambridge CB10 1SD, United Kingdom; Earlham Institute Norwich Research Park, Norwich NR4 7UZ, UK; Medical School, University of East Anglia, Norwich Research Park, Norwich, NR4 7UA, UK.
| |
Collapse
|
3
|
Lyu Y, Lin SH, Wu H, Li Z. SCIntRuler: guiding the integration of multiple single-cell RNA-seq datasets with a novel statistical metric. Bioinformatics 2024; 40:btae537. [PMID: 39226185 DOI: 10.1093/bioinformatics/btae537] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Revised: 08/24/2024] [Accepted: 08/30/2024] [Indexed: 09/05/2024] Open
Abstract
MOTIVATION The growing number of single-cell RNA-seq (scRNA-seq) studies highlights the potential benefits of integrating multiple datasets, such as augmenting sample sizes and enhancing analytical robustness. Inherent diversity and batch discrepancies within samples or across studies continue to pose significant challenges for computational analyses. Questions persist in practice, lacking definitive answers: Should we use a specific integration method or opt for simply merging the datasets during joint analysis? Among all the existing data integration methods, which one is more suitable in specific scenarios? RESULT To fill the gap, we introduce SCIntRuler, a novel statistical metric for guiding the integration of multiple scRNA-seq datasets. SCIntRuler helps researchers make informed decisions regarding the necessity of data integration and the selection of an appropriate integration method. Our simulations and real data applications demonstrate that SCIntRuler streamlines decision-making processes and facilitates the analysis of diverse scRNA-seq datasets under varying contexts, thereby alleviating the complexities associated with the integration of heterogeneous scRNA-seq datasets. AVAILABILITY AND IMPLEMENTATION The implementation of our method is available on CRAN as an open-source R package with a user-friendly manual available: https://cloud.r-project.org/web/packages/SCIntRuler/index.html.
Collapse
Affiliation(s)
- Yue Lyu
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, United States
- Department of Biostatistics and Data Science, The University of Texas Health Science Center at Houston, Houston, TX 77030, United States
| | - Steven H Lin
- Department of Thoracic Radiation Oncology, Division of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, United States
| | - Hao Wu
- Faculty of Computer Science and Control Engineering, Shenzhen University of Advanced Technology, Shenzhen, Guangdong 518055, P.R. China
- Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong 518055, P.R. China
| | - Ziyi Li
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, United States
| |
Collapse
|
4
|
Smelik M, Zhao Y, Li X, Loscalzo J, Sysoev O, Mahmud F, Mansour Aly D, Benson M. An interactive atlas of genomic, proteomic, and metabolomic biomarkers promotes the potential of proteins to predict complex diseases. Sci Rep 2024; 14:12710. [PMID: 38830935 PMCID: PMC11148091 DOI: 10.1038/s41598-024-63399-9] [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: 02/02/2024] [Accepted: 05/28/2024] [Indexed: 06/05/2024] Open
Abstract
Multiomics analyses have identified multiple potential biomarkers of the incidence and prevalence of complex diseases. However, it is not known which type of biomarker is optimal for clinical purposes. Here, we make a systematic comparison of 90 million genetic variants, 1453 proteins, and 325 metabolites from 500,000 individuals with complex diseases from the UK Biobank. A machine learning pipeline consisting of data cleaning, data imputation, feature selection, and model training using cross-validation and comparison of the results on holdout test sets showed that proteins were most predictive, followed by metabolites, and genetic variants. Only five proteins per disease resulted in median (min-max) areas under the receiver operating characteristic curves for incidence of 0.79 (0.65-0.86) and 0.84 (0.70-0.91) for prevalence. In summary, our work suggests the potential of predicting complex diseases based on a limited number of proteins. We provide an interactive atlas (macd.shinyapps.io/ShinyApp/) to find genomic, proteomic, or metabolomic biomarkers for different complex diseases.
Collapse
Affiliation(s)
- Martin Smelik
- Medical Digital Twin Research Group, Department of Clinical Science, Intervention and Technology (CLINTEC), Karolinska Institute, Stockholm, Sweden
| | - Yelin Zhao
- Medical Digital Twin Research Group, Department of Clinical Science, Intervention and Technology (CLINTEC), Karolinska Institute, Stockholm, Sweden
| | - Xinxiu Li
- Medical Digital Twin Research Group, Department of Clinical Science, Intervention and Technology (CLINTEC), Karolinska Institute, Stockholm, Sweden
| | - Joseph Loscalzo
- Division of Cardiovascular Medicine, Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Oleg Sysoev
- Division of Statistics and Machine Learning, Department of Computer and Information Science, Linköping University, Linköping, Sweden
| | - Firoj Mahmud
- Medical Digital Twin Research Group, Department of Clinical Science, Intervention and Technology (CLINTEC), Karolinska Institute, Stockholm, Sweden
| | - Dina Mansour Aly
- Medical Digital Twin Research Group, Department of Clinical Science, Intervention and Technology (CLINTEC), Karolinska Institute, Stockholm, Sweden
| | - Mikael Benson
- Medical Digital Twin Research Group, Department of Clinical Science, Intervention and Technology (CLINTEC), Karolinska Institute, Stockholm, Sweden.
| |
Collapse
|
5
|
Zhao Y, Li X, Loscalzo J, Smelik M, Sysoev O, Wang Y, Mahmud AKMF, Mansour Aly D, Benson M. Transcript and protein signatures derived from shared molecular interactions across cancers are associated with mortality. J Transl Med 2024; 22:444. [PMID: 38734658 PMCID: PMC11088765 DOI: 10.1186/s12967-024-05268-7] [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: 02/26/2024] [Accepted: 05/01/2024] [Indexed: 05/13/2024] Open
Abstract
BACKGROUND Characterization of shared cancer mechanisms have been proposed to improve therapy strategies and prognosis. Here, we aimed to identify shared cell-cell interactions (CCIs) within the tumor microenvironment across multiple solid cancers and assess their association with cancer mortality. METHODS CCIs of each cancer were identified by NicheNet analysis of single-cell RNA sequencing data from breast, colon, liver, lung, and ovarian cancers. These CCIs were used to construct a shared multi-cellular tumor model (shared-MCTM) representing common CCIs across cancers. A gene signature was identified from the shared-MCTM and tested on the mRNA and protein level in two large independent cohorts: The Cancer Genome Atlas (TCGA, 9185 tumor samples and 727 controls across 22 cancers) and UK biobank (UKBB, 10,384 cancer patients and 5063 controls with proteomics data across 17 cancers). Cox proportional hazards models were used to evaluate the association of the signature with 10-year all-cause mortality, including sex-specific analysis. RESULTS A shared-MCTM was derived from five individual cancers. A shared gene signature was extracted from this shared-MCTM and the most prominent regulatory cell type, matrix cancer-associated fibroblast (mCAF). The signature exhibited significant expression changes in multiple cancers compared to controls at both mRNA and protein levels in two independent cohorts. Importantly, it was significantly associated with mortality in cancer patients in both cohorts. The highest hazard ratios were observed for brain cancer in TCGA (HR [95%CI] = 6.90[4.64-10.25]) and ovarian cancer in UKBB (5.53[2.08-8.80]). Sex-specific analysis revealed distinct risks, with a higher mortality risk associated with the protein signature score in males (2.41[1.97-2.96]) compared to females (1.84[1.44-2.37]). CONCLUSION We identified a gene signature from a comprehensive shared-MCTM representing common CCIs across different cancers and revealed the regulatory role of mCAF in the tumor microenvironment. The pathogenic relevance of the gene signature was supported by differential expression and association with mortality on both mRNA and protein levels in two independent cohorts.
Collapse
Affiliation(s)
- Yelin Zhao
- Medical Digital Twin Research Group, Department of Clinical Science, Intervention and Technology (CLINTEC), Karolinska Institutet, Stockholm, Sweden
| | - Xinxiu Li
- Medical Digital Twin Research Group, Department of Clinical Science, Intervention and Technology (CLINTEC), Karolinska Institutet, Stockholm, Sweden
| | - Joseph Loscalzo
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Martin Smelik
- Medical Digital Twin Research Group, Department of Clinical Science, Intervention and Technology (CLINTEC), Karolinska Institutet, Stockholm, Sweden
| | - Oleg Sysoev
- Division of Statistics and Machine Learning, Department of Computer and Information Science, Linköping University, Linköping, Sweden
| | - Yunzhang Wang
- Department of Clinical Sciences, Danderyd Hospital, Karolinska Institutet, Stockholm, Sweden
| | - A K M Firoj Mahmud
- Medical Digital Twin Research Group, Department of Clinical Science, Intervention and Technology (CLINTEC), Karolinska Institutet, Stockholm, Sweden
| | - Dina Mansour Aly
- Medical Digital Twin Research Group, Department of Clinical Science, Intervention and Technology (CLINTEC), Karolinska Institutet, Stockholm, Sweden
| | - Mikael Benson
- Medical Digital Twin Research Group, Department of Clinical Science, Intervention and Technology (CLINTEC), Karolinska Institutet, Stockholm, Sweden.
| |
Collapse
|
6
|
Katsoulakis E, Wang Q, Wu H, Shahriyari L, Fletcher R, Liu J, Achenie L, Liu H, Jackson P, Xiao Y, Syeda-Mahmood T, Tuli R, Deng J. Digital twins for health: a scoping review. NPJ Digit Med 2024; 7:77. [PMID: 38519626 PMCID: PMC10960047 DOI: 10.1038/s41746-024-01073-0] [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: 08/22/2023] [Accepted: 03/07/2024] [Indexed: 03/25/2024] Open
Abstract
The use of digital twins (DTs) has proliferated across various fields and industries, with a recent surge in the healthcare sector. The concept of digital twin for health (DT4H) holds great promise to revolutionize the entire healthcare system, including management and delivery, disease treatment and prevention, and health well-being maintenance, ultimately improving human life. The rapid growth of big data and continuous advancement in data science (DS) and artificial intelligence (AI) have the potential to significantly expedite DT research and development by providing scientific expertise, essential data, and robust cybertechnology infrastructure. Although various DT initiatives have been underway in the industry, government, and military, DT4H is still in its early stages. This paper presents an overview of the current applications of DTs in healthcare, examines consortium research centers and their limitations, and surveys the current landscape of emerging research and development opportunities in healthcare. We envision the emergence of a collaborative global effort among stakeholders to enhance healthcare and improve the quality of life for millions of individuals worldwide through pioneering research and development in the realm of DT technology.
Collapse
Affiliation(s)
- Evangelia Katsoulakis
- VA Informatics and Computing Infrastructure, Salt Lake City, UT, 84148, USA
- Department of Radiation Oncology, University of South Florida, Tampa, FL, 33606, USA
| | - Qi Wang
- Department of Mathematics, University of South Carolina, Columbia, SC, 29208, USA
| | - Huanmei Wu
- Department of Health Services Administration and Policy, Temple University, Philadelphia, PA, 19122, USA
| | - Leili Shahriyari
- Department of Mathematics and Statistics, University of Massachusetts Amherst, Amherst, MA, 01003, USA
| | - Richard Fletcher
- Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
- Department of Psychiatry, Massachusetts General Hospital, Boston, MA, 02139, USA
| | - Jinwei Liu
- Department of Computer and Information Sciences, Florida A&M University, Tallahassee, FL, 32307, USA
| | - Luke Achenie
- Department of Chemical Engineering, Virginia Polytechnic Institute and State University, Blacksburg, VA, 24060, USA
| | - Hongfang Liu
- McWilliams School of Biomedical Informatics, University of Texas Health Science Center at Houston, Houston, TX, 77030, USA
| | - Pamela Jackson
- Precision Neurotherapeutics Innovation Program & Department of Neurosurgery, Mayo Clinic, Phoenix, AZ, 85003, USA
| | - Ying Xiao
- Department of Radiation Oncology, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | | | - Richard Tuli
- Department of Radiation Oncology, University of South Florida, Tampa, FL, 33606, USA
| | - Jun Deng
- Department of Therapeutic Radiology, Yale University, New Haven, CT, 06510, USA.
| |
Collapse
|
7
|
Schäfer S, Smelik M, Sysoev O, Zhao Y, Eklund D, Lilja S, Gustafsson M, Heyn H, Julia A, Kovács IA, Loscalzo J, Marsal S, Zhang H, Li X, Gawel D, Wang H, Benson M. scDrugPrio: a framework for the analysis of single-cell transcriptomics to address multiple problems in precision medicine in immune-mediated inflammatory diseases. Genome Med 2024; 16:42. [PMID: 38509600 PMCID: PMC10956347 DOI: 10.1186/s13073-024-01314-7] [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: 03/02/2023] [Accepted: 03/12/2024] [Indexed: 03/22/2024] Open
Abstract
BACKGROUND Ineffective drug treatment is a major problem for many patients with immune-mediated inflammatory diseases (IMIDs). Important reasons are the lack of systematic solutions for drug prioritisation and repurposing based on characterisation of the complex and heterogeneous cellular and molecular changes in IMIDs. METHODS Here, we propose a computational framework, scDrugPrio, which constructs network models of inflammatory disease based on single-cell RNA sequencing (scRNA-seq) data. scDrugPrio constructs detailed network models of inflammatory diseases that integrate information on cell type-specific expression changes, altered cellular crosstalk and pharmacological properties for the selection and ranking of thousands of drugs. RESULTS scDrugPrio was developed using a mouse model of antigen-induced arthritis and validated by improved precision/recall for approved drugs, as well as extensive in vitro, in vivo, and in silico studies of drugs that were predicted, but not approved, for the studied diseases. Next, scDrugPrio was applied to multiple sclerosis, Crohn's disease, and psoriatic arthritis, further supporting scDrugPrio through prioritisation of relevant and approved drugs. However, in contrast to the mouse model of arthritis, great interindividual cellular and gene expression differences were found in patients with the same diagnosis. Such differences could explain why some patients did or did not respond to treatment. This explanation was supported by the application of scDrugPrio to scRNA-seq data from eleven individual Crohn's disease patients. The analysis showed great variations in drug predictions between patients, for example, assigning a high rank to anti-TNF treatment in a responder and a low rank in a nonresponder to that treatment. CONCLUSIONS We propose a computational framework, scDrugPrio, for drug prioritisation based on scRNA-seq of IMID disease. Application to individual patients indicates scDrugPrio's potential for personalised network-based drug screening on cellulome-, genome-, and drugome-wide scales. For this purpose, we made scDrugPrio into an easy-to-use R package ( https://github.com/SDTC-CPMed/scDrugPrio ).
Collapse
Affiliation(s)
- Samuel Schäfer
- Centre for Personalised Medicine, Linköping University, Linköping, Sweden
- Department of Gastroenterology and Hepatology, University Hospital, Linköping, Sweden
| | - Martin Smelik
- Postal Address: LIME/Medical Digital Twin Research Group, Division of ENT, CLINTEC, Karolinska Institute, Tomtebodavägen 18A. 171 65 Solna, Stockholm, Sweden
| | - Oleg Sysoev
- Division of Statistics and Machine Learning, Department of Computer and Information Science, Linkoping University, Linköping, Sweden
| | - Yelin Zhao
- Postal Address: LIME/Medical Digital Twin Research Group, Division of ENT, CLINTEC, Karolinska Institute, Tomtebodavägen 18A. 171 65 Solna, Stockholm, Sweden
| | - Desiré Eklund
- Centre for Personalised Medicine, Linköping University, Linköping, Sweden
| | - Sandra Lilja
- Centre for Personalised Medicine, Linköping University, Linköping, Sweden
- Mavatar, Inc, Stockholm, Sweden
| | - Mika Gustafsson
- Division for Bioinformatics, Department of Physics, Chemistry and Biology, Linköping University, Linköping, Sweden
| | - Holger Heyn
- CNAG-CRG, Centre for Genomic Regulation (CRG), Barcelona Institute of Science and Technology (BIST), 08028, Barcelona, Spain
- Universitat Pompeu Fabra (UPF), 08002, Barcelona, Spain
| | - Antonio Julia
- Grup de Recerca de Reumatologia, Institut de Recerca Vall d'Hebron, Barcelona, Spain
| | - István A Kovács
- Department of Physics and Astronomy, Northwestern University, Evanston, IL, 60208, USA
- Northwestern Institute On Complex Systems, Northwestern University, Evanston, IL, 60208, USA
| | - Joseph Loscalzo
- Division of Cardiovascular Medicine, Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Sara Marsal
- Grup de Recerca de Reumatologia, Institut de Recerca Vall d'Hebron, Barcelona, Spain
| | - Huan Zhang
- Centre for Personalised Medicine, Linköping University, Linköping, Sweden
| | - Xinxiu Li
- Postal Address: LIME/Medical Digital Twin Research Group, Division of ENT, CLINTEC, Karolinska Institute, Tomtebodavägen 18A. 171 65 Solna, Stockholm, Sweden
| | | | - Hui Wang
- Postal Address: LIME/Medical Digital Twin Research Group, Division of ENT, CLINTEC, Karolinska Institute, Tomtebodavägen 18A. 171 65 Solna, Stockholm, Sweden
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogenic Biology and Immunology, Xuzhou Medical University, Jiangsu, China
| | - Mikael Benson
- Postal Address: LIME/Medical Digital Twin Research Group, Division of ENT, CLINTEC, Karolinska Institute, Tomtebodavägen 18A. 171 65 Solna, Stockholm, Sweden.
| |
Collapse
|
8
|
Schäfer S, Smelik M, Sysoev O, Zhao Y, Eklund D, Lilja S, Gustafsson M, Heyn H, Julia A, Kovács IA, Loscalzo J, Marsal S, Zhang H, Li X, Gawel D, Wang H, Benson M. scDrugPrio: A framework for the analysis of single-cell transcriptomics to address multiple problems in precision medicine in immune-mediated inflammatory diseases. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.11.08.566249. [PMID: 38014022 PMCID: PMC10680570 DOI: 10.1101/2023.11.08.566249] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2023]
Abstract
Background Ineffective drug treatment is a major problem for many patients with immune-mediated inflammatory diseases (IMIDs). Important reasons are the lack of systematic solutions for drug prioritisation and repurposing based on characterisation of the complex and heterogeneous cellular and molecular changes in IMIDs. Methods Here, we propose a computational framework, scDrugPrio, which constructs network models of inflammatory disease based on single-cell RNA sequencing (scRNA-seq) data. scDrugPrio constructs detailed network models of inflammatory diseases that integrate information on cell type-specific expression changes, altered cellular crosstalk and pharmacological properties for the selection and ranking of thousands of drugs. Results scDrugPrio was developed using a mouse model of antigen-induced arthritis and validated by improved precision/recall for approved drugs, as well as extensive in vitro, in vivo, and in silico studies of drugs that were predicted, but not approved, for the studied diseases. Next, scDrugPrio was applied to multiple sclerosis, Crohn's disease, and psoriatic arthritis, further supporting scDrugPrio through prioritisation of relevant and approved drugs. However, in contrast to the mouse model of arthritis, great interindividual cellular and gene expression differences were found in patients with the same diagnosis. Such differences could explain why some patients did or did not respond to treatment. This explanation was supported by the application of scDrugPrio to scRNA-seq data from eleven individual Crohn's disease patients. The analysis showed great variations in drug predictions between patients, for example, assigning a high rank to anti-TNF treatment in a responder and a low rank in a nonresponder to that treatment. Conclusion We propose a computational framework, scDrugPrio, for drug prioritisation based on scRNA-seq of IMID disease. Application to individual patients indicates scDrugPrio's potential for personalised network-based drug screening on cellulome-, genome-, and drugome-wide scales. For this purpose, we made scDrugPrio into an easy-to-use R package (https://github.com/SDTC-CPMed/scDrugPrio).
Collapse
Affiliation(s)
- Samuel Schäfer
- Centre for Personalised Medicine, Linköping University; Linköping, Sweden
- Department of Gastroenterology and Hepatology, University Hospital, Linköping, Sweden
| | - Martin Smelik
- Centre for Personalised Medicine, Linköping University; Linköping, Sweden
- Division of ENT, CLINTEC, Karolinska Institute, Stockholm, Sweden
| | - Oleg Sysoev
- Division of Statistics and Machine Learning, Department of Computer and Information Science, Linkoping University; Linköping, Sweden
| | - Yelin Zhao
- Centre for Personalised Medicine, Linköping University; Linköping, Sweden
- Division of ENT, CLINTEC, Karolinska Institute, Stockholm, Sweden
| | - Desiré Eklund
- Centre for Personalised Medicine, Linköping University; Linköping, Sweden
| | - Sandra Lilja
- Centre for Personalised Medicine, Linköping University; Linköping, Sweden
- Mavatar, Inc., Stockholm. Sweden
| | - Mika Gustafsson
- Division for Bioinformatics, Department of Physics, Chemistry and Biology, Linköping University; Linköping, Sweden
| | - Holger Heyn
- CNAG-CRG, Centre for Genomic Regulation (CRG), Barcelona Institute of Science and Technology (BIST), 08028 Barcelona, Spain
- Universitat Pompeu Fabra (UPF), 08002 Barcelona, Spain
| | - Antonio Julia
- Grup de Recerca de Reumatologia, Institut de Recerca Vall d’Hebron, Barcelona, España
| | - István A. Kovács
- Department of Physics and Astronomy, Northwestern University, Evanston, IL 60208, USA
- Northwestern Institute on Complex Systems, Northwestern University, Evanston, IL 60208, USA
| | - Joseph Loscalzo
- Division of Cardiovascular Medicine, Channing Division of Network Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School; Boston, MA, USA
| | - Sara Marsal
- Grup de Recerca de Reumatologia, Institut de Recerca Vall d’Hebron, Barcelona, España
| | - Huan Zhang
- Centre for Personalised Medicine, Linköping University; Linköping, Sweden
| | - Xinxiu Li
- Centre for Personalised Medicine, Linköping University; Linköping, Sweden
- Division of ENT, CLINTEC, Karolinska Institute, Stockholm, Sweden
| | - Danuta Gawel
- Centre for Personalised Medicine, Linköping University; Linköping, Sweden
- Mavatar, Inc., Stockholm. Sweden
| | - Hui Wang
- Northwestern Institute on Complex Systems, Northwestern University, Evanston, IL 60208, USA
| | - Mikael Benson
- Centre for Personalised Medicine, Linköping University; Linköping, Sweden
- Division of ENT, CLINTEC, Karolinska Institute, Stockholm, Sweden
| |
Collapse
|
9
|
Tseng KC, Crump JG. Craniofacial developmental biology in the single-cell era. Development 2023; 150:dev202077. [PMID: 37812056 PMCID: PMC10617621 DOI: 10.1242/dev.202077] [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: 10/10/2023]
Abstract
The evolution of a unique craniofacial complex in vertebrates made possible new ways of breathing, eating, communicating and sensing the environment. The head and face develop through interactions of all three germ layers, the endoderm, ectoderm and mesoderm, as well as the so-called fourth germ layer, the cranial neural crest. Over a century of experimental embryology and genetics have revealed an incredible diversity of cell types derived from each germ layer, signaling pathways and genes that coordinate craniofacial development, and how changes to these underlie human disease and vertebrate evolution. Yet for many diseases and congenital anomalies, we have an incomplete picture of the causative genomic changes, in particular how alterations to the non-coding genome might affect craniofacial gene expression. Emerging genomics and single-cell technologies provide an opportunity to obtain a more holistic view of the genes and gene regulatory elements orchestrating craniofacial development across vertebrates. These single-cell studies generate novel hypotheses that can be experimentally validated in vivo. In this Review, we highlight recent advances in single-cell studies of diverse craniofacial structures, as well as potential pitfalls and the need for extensive in vivo validation. We discuss how these studies inform the developmental sources and regulation of head structures, bringing new insights into the etiology of structural birth anomalies that affect the vertebrate head.
Collapse
Affiliation(s)
- Kuo-Chang Tseng
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine of University of Southern California, Los Angeles, CA 90033, USA
| | - J. Gage Crump
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine of University of Southern California, Los Angeles, CA 90033, USA
| |
Collapse
|
10
|
Halawani R, Buchert M, Chen YPP. Deep learning exploration of single-cell and spatially resolved cancer transcriptomics to unravel tumour heterogeneity. Comput Biol Med 2023; 164:107274. [PMID: 37506451 DOI: 10.1016/j.compbiomed.2023.107274] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2023] [Revised: 07/03/2023] [Accepted: 07/16/2023] [Indexed: 07/30/2023]
Abstract
Tumour heterogeneity is one of the critical confounding aspects in decoding tumour growth. Malignant cells display variations in their gene transcription profiles and mutation spectra even when originating from a single progenitor cell. Single-cell and spatial transcriptomics sequencing have recently emerged as key technologies for unravelling tumour heterogeneity. Single-cell sequencing promotes individual cell-type identification through transcriptome-wide gene expression measurements of each cell. Spatial transcriptomics facilitates identification of cell-cell interactions and the structural organization of heterogeneous cells within a tumour tissue through associating spatial RNA abundance of cells at distinct spots in the tissue section. However, extracting features and analyzing single-cell and spatial transcriptomics data poses challenges. Single-cell transcriptome data is extremely noisy and its sparse nature and dropouts can lead to misinterpretation of gene expression and the misclassification of cell types. Deep learning predictive power can overcome data challenges, provide high-resolution analysis and enhance precision oncology applications that involve early cancer prognosis, diagnosis, patient survival estimation and anti-cancer therapy planning. In this paper, we provide a background to and review of the recent progress of deep learning frameworks to investigate tumour heterogeneity using both single-cell and spatial transcriptomics data types.
Collapse
Affiliation(s)
- Raid Halawani
- Department of Computer Science and Information Technology, La Trobe University, Melbourne, Australia
| | - Michael Buchert
- School of Cancer Medicine, La Trobe University, Melbourne, Victoria, Australia; Olivia Newton-John Cancer Research Institute, Melbourne, Victoria, Australia
| | - Yi-Ping Phoebe Chen
- Department of Computer Science and Information Technology, La Trobe University, Melbourne, Australia.
| |
Collapse
|
11
|
Van de Sande B, Lee JS, Mutasa-Gottgens E, Naughton B, Bacon W, Manning J, Wang Y, Pollard J, Mendez M, Hill J, Kumar N, Cao X, Chen X, Khaladkar M, Wen J, Leach A, Ferran E. Applications of single-cell RNA sequencing in drug discovery and development. Nat Rev Drug Discov 2023; 22:496-520. [PMID: 37117846 PMCID: PMC10141847 DOI: 10.1038/s41573-023-00688-4] [Citation(s) in RCA: 52] [Impact Index Per Article: 52.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/10/2023] [Indexed: 04/30/2023]
Abstract
Single-cell technologies, particularly single-cell RNA sequencing (scRNA-seq) methods, together with associated computational tools and the growing availability of public data resources, are transforming drug discovery and development. New opportunities are emerging in target identification owing to improved disease understanding through cell subtyping, and highly multiplexed functional genomics screens incorporating scRNA-seq are enhancing target credentialling and prioritization. ScRNA-seq is also aiding the selection of relevant preclinical disease models and providing new insights into drug mechanisms of action. In clinical development, scRNA-seq can inform decision-making via improved biomarker identification for patient stratification and more precise monitoring of drug response and disease progression. Here, we illustrate how scRNA-seq methods are being applied in key steps in drug discovery and development, and discuss ongoing challenges for their implementation in the pharmaceutical industry.
Collapse
Affiliation(s)
| | | | | | - Bart Naughton
- Computational Neurobiology, Eisai, Cambridge, MA, USA
| | - Wendi Bacon
- EMBL-EBI, Wellcome Genome Campus, Hinxton, UK
- The Open University, Milton Keynes, UK
| | | | - Yong Wang
- Precision Bioinformatics, Prometheus Biosciences, San Diego, CA, USA
| | | | - Melissa Mendez
- Genomic Sciences, GlaxoSmithKline, Collegeville, PA, USA
| | - Jon Hill
- Global Computational Biology and Digital Sciences, Boehringer Ingelheim Pharmaceuticals Inc., Ridgefield, CT, USA
| | - Namit Kumar
- Informatics & Predictive Sciences, Bristol Myers Squibb, San Diego, CA, USA
| | - Xiaohong Cao
- Genomic Research Center, AbbVie Inc., Cambridge, MA, USA
| | - Xiao Chen
- Magnet Biomedicine, Cambridge, MA, USA
| | - Mugdha Khaladkar
- Human Genetics and Computational Biology, GlaxoSmithKline, Collegeville, PA, USA
| | - Ji Wen
- Oncology Research and Development Unit, Pfizer, La Jolla, CA, USA
| | | | | |
Collapse
|
12
|
Yan D, Sun Z, Fang J, Cao S, Wang W, Chang X, Badirli S, Fu H, Liu Y. scRAA: the development of a robust and automatic annotation procedure for single-cell RNA sequencing data. J Biopharm Stat 2023:1-14. [PMID: 37162278 DOI: 10.1080/10543406.2023.2208671] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/11/2023]
Abstract
A critical task in single-cell RNA sequencing (scRNA-Seq) data analysis is to identify cell types from heterogeneous tissues. While the majority of classification methods demonstrated high performance in scRNA-Seq annotation problems, a robust and accurate solution is desired to generate reliable outcomes for downstream analyses, for instance, marker genes identification, differentially expressed genes, and pathway analysis. It is hard to establish a universally good metric. Thus, a universally good classification method for all kinds of scenarios does not exist. In addition, reference and query data in cell classification are usually from different experimental batches, and failure to consider batch effects may result in misleading conclusions. To overcome this bottleneck, we propose a robust ensemble approach to classify cells and utilize a batch correction method between reference and query data. We simulated four scenarios that comprise simple to complex batch effect and account for varying cell-type proportions. We further tested our approach on both lung and pancreas data. We found improved prediction accuracy and robust performance across simulation scenarios and real data. The incorporation of batch effect correction between reference and query, and the ensemble approach improve cell-type prediction accuracy while maintaining robustness. We demonstrated these through simulated and real scRNA-Seq data.
Collapse
Affiliation(s)
- Dongyan Yan
- Global Statistical Science, Eli Lilly & Co, Indianapolis, Indiana, USA
| | - Zhe Sun
- Global Statistical Science, Eli Lilly & Co, Indianapolis, Indiana, USA
| | - Jiyuan Fang
- Global Statistical Science, Eli Lilly & Co, Indianapolis, Indiana, USA
| | - Shanshan Cao
- Global Statistical Science, Eli Lilly & Co, Indianapolis, Indiana, USA
| | - Wenjie Wang
- Advance Analytics and Data Science, Eli Lilly & Co, Indianapolis, Indiana, USA
| | - Xinyue Chang
- Advance Analytics and Data Science, Eli Lilly & Co, Indianapolis, Indiana, USA
| | - Sarkhan Badirli
- Advance Analytics and Data Science, Eli Lilly & Co, Indianapolis, Indiana, USA
| | - Haoda Fu
- Advance Analytics and Data Science, Eli Lilly & Co, Indianapolis, Indiana, USA
| | - Yushi Liu
- Global Statistical Science, Eli Lilly & Co, Indianapolis, Indiana, USA
| |
Collapse
|
13
|
Lilja S, Li X, Smelik M, Lee EJ, Loscalzo J, Marthanda PB, Hu L, Magnusson M, Sysoev O, Zhang H, Zhao Y, Sjöwall C, Gawel D, Wang H, Benson M. Multi-organ single-cell analysis reveals an on/off switch system with potential for personalized treatment of immunological diseases. Cell Rep Med 2023; 4:100956. [PMID: 36858042 PMCID: PMC10040389 DOI: 10.1016/j.xcrm.2023.100956] [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: 08/17/2022] [Revised: 09/30/2022] [Accepted: 02/03/2023] [Indexed: 03/03/2023]
Abstract
Prioritization of disease mechanisms, biomarkers, and drug targets in immune-mediated inflammatory diseases (IMIDs) is complicated by altered interactions between thousands of genes. Our multi-organ single-cell RNA sequencing of a mouse IMID model, namely collagen-induced arthritis, shows highly complex and heterogeneous expression changes in all analyzed organs, even though only joints showed signs of inflammation. We organized those into a multi-organ multicellular disease model, which shows predicted molecular interactions within and between organs. That model supports that inflammation is switched on or off by altered balance between pro- and anti-inflammatory upstream regulators (URs) and downstream pathways. Meta-analyses of human IMIDs show a similar, but graded, on/off switch system. This system has the potential to prioritize, diagnose, and treat optimal combinations of URs on the levels of IMIDs, subgroups, and individual patients. That potential is supported by UR analyses in more than 600 sera from patients with systemic lupus erythematosus.
Collapse
Affiliation(s)
- Sandra Lilja
- Department of Pediatrics, Biomedical and Clinical Sciences, Linköping University, 58183 Linköping, Sweden; Mavatar, Inc, Vasagatan, 11120 Stockholm, Sweden
| | - Xinxiu Li
- Department of Pediatrics, Biomedical and Clinical Sciences, Linköping University, 58183 Linköping, Sweden; Medical Digital Twin Research Group, Division of Ear, Nose and Throat Diseases, Department of Clinical Science, Intervention and Technology, Karolinska Institutet, 17165 Stockholm, Sweden
| | - Martin Smelik
- Department of Pediatrics, Biomedical and Clinical Sciences, Linköping University, 58183 Linköping, Sweden; Medical Digital Twin Research Group, Division of Ear, Nose and Throat Diseases, Department of Clinical Science, Intervention and Technology, Karolinska Institutet, 17165 Stockholm, Sweden
| | - Eun Jung Lee
- Department of Otorhinolaryngology, Yonsei University Wonju College of Medicine, Wonju, Ganwong 26460, Korea
| | - Joseph Loscalzo
- Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA; Channing Division of Network Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Pratheek Bellur Marthanda
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, 1425 Madison Avenue, New York, NY 10029, USA
| | - Lang Hu
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogenic Biology and Immunology, Xuzhou Medical University, Xuzhou, Jiangsu 221000, China
| | - Mattias Magnusson
- The National Board of Health and Welfare, Socialstyrelsen, 11259 Stockholm, Sweden
| | - Oleg Sysoev
- Department of Computer and Information Science, Linköping University, 58183 Linköping, Sweden
| | - Huan Zhang
- Department of Pediatrics, Biomedical and Clinical Sciences, Linköping University, 58183 Linköping, Sweden
| | - Yelin Zhao
- Department of Pediatrics, Biomedical and Clinical Sciences, Linköping University, 58183 Linköping, Sweden; Medical Digital Twin Research Group, Division of Ear, Nose and Throat Diseases, Department of Clinical Science, Intervention and Technology, Karolinska Institutet, 17165 Stockholm, Sweden
| | - Christopher Sjöwall
- Biomedical and Clinical Sciences, Division of Inflammation and Infection/Rheumatology, Linköping University, 58183 Linköping, Sweden
| | - Danuta Gawel
- Mavatar, Inc, Vasagatan, 11120 Stockholm, Sweden
| | - Hui Wang
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogenic Biology and Immunology, Xuzhou Medical University, Xuzhou, Jiangsu 221000, China
| | - Mikael Benson
- Department of Pediatrics, Biomedical and Clinical Sciences, Linköping University, 58183 Linköping, Sweden; Medical Digital Twin Research Group, Division of Ear, Nose and Throat Diseases, Department of Clinical Science, Intervention and Technology, Karolinska Institutet, 17165 Stockholm, Sweden.
| |
Collapse
|
14
|
Huang Z, Wang J, Lu X, Mohd Zain A, Yu G. scGGAN: single-cell RNA-seq imputation by graph-based generative adversarial network. Brief Bioinform 2023; 24:7024714. [PMID: 36733262 DOI: 10.1093/bib/bbad040] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Revised: 12/21/2022] [Accepted: 01/18/2023] [Indexed: 02/04/2023] Open
Abstract
Single-cell RNA sequencing (scRNA-seq) data are typically with a large number of missing values, which often results in the loss of critical gene signaling information and seriously limit the downstream analysis. Deep learning-based imputation methods often can better handle scRNA-seq data than shallow ones, but most of them do not consider the inherent relations between genes, and the expression of a gene is often regulated by other genes. Therefore, it is essential to impute scRNA-seq data by considering the regional gene-to-gene relations. We propose a novel model (named scGGAN) to impute scRNA-seq data that learns the gene-to-gene relations by Graph Convolutional Networks (GCN) and global scRNA-seq data distribution by Generative Adversarial Networks (GAN). scGGAN first leverages single-cell and bulk genomics data to explore inherent relations between genes and builds a more compact gene relation network to jointly capture the homogeneous and heterogeneous information. Then, it constructs a GCN-based GAN model to integrate the scRNA-seq, gene sequencing data and gene relation network for generating scRNA-seq data, and trains the model through adversarial learning. Finally, it utilizes data generated by the trained GCN-based GAN model to impute scRNA-seq data. Experiments on simulated and real scRNA-seq datasets show that scGGAN can effectively identify dropout events, recover the biologically meaningful expressions, determine subcellular states and types, improve the differential expression analysis and temporal dynamics analysis. Ablation experiments confirm that both the gene relation network and gene sequence data help the imputation of scRNA-seq data.
Collapse
Affiliation(s)
- Zimo Huang
- MEng student at School of Software, Shandong University, China
| | - Jun Wang
- Joint SDU-NTU Centre for Artificial Intelligence Research (C-FAIR), Shandong University, China
| | - Xudong Lu
- Joint SDU-NTU Centre for Artificial Intelligence Research (C-FAIR), Shandong University, China
| | | | - Guoxian Yu
- School of Software, Shandong University, China
| |
Collapse
|
15
|
Becker LM, Chen SH, Rodor J, de Rooij LPMH, Baker AH, Carmeliet P. Deciphering endothelial heterogeneity in health and disease at single-cell resolution: progress and perspectives. Cardiovasc Res 2023; 119:6-27. [PMID: 35179567 PMCID: PMC10022871 DOI: 10.1093/cvr/cvac018] [Citation(s) in RCA: 20] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Revised: 12/16/2021] [Accepted: 02/16/2022] [Indexed: 11/14/2022] Open
Abstract
Endothelial cells (ECs) constitute the inner lining of vascular beds in mammals and are crucial for homeostatic regulation of blood vessel physiology, but also play a key role in pathogenesis of many diseases, thereby representing realistic therapeutic targets. However, it has become evident that ECs are heterogeneous, encompassing several subtypes with distinct functions, which makes EC targeting and modulation in diseases challenging. The rise of the new single-cell era has led to an emergence of studies aimed at interrogating transcriptome diversity along the vascular tree, and has revolutionized our understanding of EC heterogeneity from both a physiological and pathophysiological context. Here, we discuss recent landmark studies aimed at teasing apart the heterogeneous nature of ECs. We cover driving (epi)genetic, transcriptomic, and metabolic forces underlying EC heterogeneity in health and disease, as well as current strategies used to combat disease-enriched EC phenotypes, and propose strategies to transcend largely descriptive heterogeneity towards prioritization and functional validation of therapeutically targetable drivers of EC diversity. Lastly, we provide an overview of the most recent advances and hurdles in single EC OMICs.
Collapse
Affiliation(s)
| | | | | | | | - Andrew H Baker
- Corresponding authors. Tel: +32 16 32 62 47, E-mail: (P.C.); Tel: +44 (0)131 242 6774, E-mail: (A.H.B.)
| | - Peter Carmeliet
- Corresponding authors. Tel: +32 16 32 62 47, E-mail: (P.C.); Tel: +44 (0)131 242 6774, E-mail: (A.H.B.)
| |
Collapse
|
16
|
Lee S, Vu HM, Lee JH, Lim H, Kim MS. Advances in Mass Spectrometry-Based Single Cell Analysis. BIOLOGY 2023; 12:395. [PMID: 36979087 PMCID: PMC10045136 DOI: 10.3390/biology12030395] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Revised: 02/27/2023] [Accepted: 03/01/2023] [Indexed: 03/06/2023]
Abstract
Technological developments and improvements in single-cell isolation and analytical platforms allow for advanced molecular profiling at the single-cell level, which reveals cell-to-cell variation within the admixture cells in complex biological or clinical systems. This helps to understand the cellular heterogeneity of normal or diseased tissues and organs. However, most studies focused on the analysis of nucleic acids (e.g., DNA and RNA) and mass spectrometry (MS)-based analysis for proteins and metabolites of a single cell lagged until recently. Undoubtedly, MS-based single-cell analysis will provide a deeper insight into cellular mechanisms related to health and disease. This review summarizes recent advances in MS-based single-cell analysis methods and their applications in biology and medicine.
Collapse
Affiliation(s)
- Siheun Lee
- School of Undergraduate Studies, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu 42988, Republic of Korea
| | - Hung M. Vu
- Department of New Biology, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu 42988, Republic of Korea
| | - Jung-Hyun Lee
- Department of New Biology, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu 42988, Republic of Korea
| | - Heejin Lim
- Center for Scientific Instrumentation, Korea Basic Science Institute (KBSI), Cheongju 28119, Republic of Korea
| | - Min-Sik Kim
- Department of New Biology, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu 42988, Republic of Korea
- New Biology Research Center, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu 42988, Republic of Korea
- Center for Cell Fate Reprogramming and Control, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu 42988, Republic of Korea
| |
Collapse
|
17
|
Olguin JE, Mendoza-Rodriguez MG, Sanchez-Barrera CA, Terrazas LI. Is the combination of immunotherapy with conventional chemotherapy the key to increase the efficacy of colorectal cancer treatment? World J Gastrointest Oncol 2023; 15:251-267. [PMID: 36908325 PMCID: PMC9994043 DOI: 10.4251/wjgo.v15.i2.251] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/24/2022] [Revised: 11/03/2022] [Accepted: 01/10/2023] [Indexed: 02/14/2023] Open
Abstract
Colorectal cancer (CRC) is among the most prevalent and deadly neoplasms worldwide. According to GLOBOCAN predictions, its incidence will increase from 1.15 million CRC cases in 2020 to 1.92 million cases in 2040. Therefore, a better understanding of the mechanisms involved in CRC development is necessary to improve strategies focused on reducing the incidence, prevalence, and mortality of this oncological pathology. Surgery, chemotherapy, and radiotherapy are the main strategies for treating CRC. The conventional chemotherapeutic agent utilized throughout the last four decades is 5-fluorouracil, notwithstanding its low efficiency as a single therapy. In contrast, combining 5-fluorouracil therapy with leucovorin and oxaliplatin or irinotecan increases its efficiency. However, these treatments have limited and temporary solutions and aggressive side effects. Additionally, most patients treated with these regimens develop drug resistance, which leads to disease progression. The immune response is considered a hallmark of cancer; thus, the use of new strategies and methodologies involving immune molecules, cells, and transcription factors has been suggested for CRC patients diagnosed in stages III and IV. Despite the critical advances in immunotherapy, the development and impact of immune checkpoint inhibitors on CRC is still under investigation because less than 25% of CRC patients display an increased 5-year survival. The causes of CRC are diverse and include modifiable environmental factors (smoking, diet, obesity, and alcoholism), individual genetic mutations, and inflammation-associated bowel diseases. Due to these diverse causes, the solutions likely cannot be generalized. Interestingly, new strategies, such as single-cell multiomics, proteomics, genomics, flow cytometry, and massive sequencing for tumor microenvironment analysis, are beginning to clarify the way forward. Thus, the individual mechanisms involved in developing the CRC microenvironment, their causes, and their consequences need to be understood from a genetic and immunological perspective. This review highlighted the importance of altering the immune response in CRC. It focused on drugs that may modulate the immune response and show specific efficacy and contrasted with evidence that immunosuppression or the promotion of the immune response is the answer to generating effective treatments with combined chemotherapeutic drugs.
Collapse
Affiliation(s)
- Jonadab E Olguin
- Laboratorio Nacional en Salud, Diagnóstico Molecular y Efecto Ambiental en Enfermedades Crónico-degenerativas, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de México, Tlalnepantla 54090, Estado de Mexico, Mexico
| | - Monica G Mendoza-Rodriguez
- Unidad de Biomedicina, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de México, Tlalnepantla 54090, Estado de Mexico, Mexico
| | - C Angel Sanchez-Barrera
- Unidad de Biomedicina, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de México, Tlalnepantla 54090, Estado de Mexico, Mexico
| | - Luis I Terrazas
- Laboratorio Nacional en Salud, Diagnóstico Molecular y Efecto Ambiental en Enfermedades Crónico-degenerativas, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de México, Tlalnepantla 54090, Estado de Mexico, Mexico
- Unidad de Biomedicina, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de México, Tlalnepantla 54090, Estado de Mexico, Mexico
| |
Collapse
|
18
|
Feng S, Li J, Tian J, Lu S, Zhao Y. Application of Single-Cell and Spatial Omics in Musculoskeletal Disorder Research. Int J Mol Sci 2023; 24:2271. [PMID: 36768592 PMCID: PMC9917071 DOI: 10.3390/ijms24032271] [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: 11/30/2022] [Revised: 01/16/2023] [Accepted: 01/19/2023] [Indexed: 01/26/2023] Open
Abstract
Musculoskeletal disorders, including fractures, scoliosis, heterotopic ossification, osteoporosis, osteoarthritis, disc degeneration, and muscular injury, etc., can occur at any stage of human life. Understanding the occurrence and development mechanism of musculoskeletal disorders, as well as the changes in tissues and cells during therapy, might help us find targeted treatment methods. Single-cell techniques provide excellent tools for studying alterations at the cellular level of disorders. However, the application of these techniques in research on musculoskeletal disorders is still limited. This review summarizes the current single-cell and spatial omics used in musculoskeletal disorders. Cell isolation, experimental methods, and feasible experimental designs for single-cell studies of musculoskeletal system diseases have been reviewed based on tissue characteristics. Then, the paper summarizes the latest findings of single-cell studies in musculoskeletal disorders from three aspects: bone and ossification, joint, and muscle and tendon disorders. Recent discoveries about the cell populations involved in these diseases are highlighted. Furthermore, the therapeutic responses of musculoskeletal disorders, especially single-cell changes after the treatments of implants, stem cell therapies, and drugs are described. Finally, the application potential and future development directions of single-cell and spatial omics in research on musculoskeletal diseases are discussed.
Collapse
Affiliation(s)
- Site Feng
- School of Medicine, Tsinghua University, Beijing 100084, China
| | - Jiahao Li
- Department of Orthopaedic Surgery, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Science, Beijing 100730, China
| | - Jingjing Tian
- Medical Science Research Center, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing 100730, China
| | - Sheng Lu
- The Key Laboratory of Digital Orthopaedics of Yunnan Provincial, Department of Orthopedic Surgery, The First People’s Hospital of Yunnan Province, The Affiliated Hospital of Kunming University of Science and Technology, Kunming 650032, China
| | - Yu Zhao
- Department of Orthopaedic Surgery, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Science, Beijing 100730, China
| |
Collapse
|
19
|
Ding L, Li X, Zhu H, Luo H. Single-Cell Sequencing in Rheumatic Diseases: New Insights from the Perspective of the Cell Type. Aging Dis 2022; 13:1633-1651. [PMID: 36465169 PMCID: PMC9662270 DOI: 10.14336/ad.2022.0323] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Accepted: 03/23/2022] [Indexed: 11/02/2023] Open
Abstract
Rheumatic diseases are a group of highly heterogeneous autoimmune and inflammatory disorders involving multiple systems. Dysfunction of immune and non-immune cells participates in the complex pathogenesis of rheumatic diseases. Therefore, studies on the abnormal activation of cell subtypes provided a specific basis for understanding the pathogenesis of rheumatic diseases, which promoted the accuracy of disease diagnosis and the effectiveness of various treatments. However, there was still a far way to achieve individualized precision medicine as the result of heterogeneity among cell subtypes. To obtain the biological information of cell subtypes, single-cell sequencing, a cutting-edge technology, is used for analyzing their genomes, transcriptomes, epigenetics, and proteomics. Novel results identified multiple cell subtypes in tissues of patients with rheumatic diseases by single-cell sequencing. Consequently, we provide an overview of recent applications of single-cell sequencing in rheumatic disease and cross-tissue to understand the cell subtypes and functions.
Collapse
Affiliation(s)
- Liqing Ding
- The Department of Rheumatology and Immunology, Xiangya Hospital of Central South University, Changsha, Hunan, China.
| | - Xiaojing Li
- The Department of Rheumatology and Immunology, Xiangya Hospital of Central South University, Changsha, Hunan, China.
| | - Honglin Zhu
- The Department of Rheumatology and Immunology, Xiangya Hospital of Central South University, Changsha, Hunan, China.
- Provincial Clinical Research Center for Rheumatic and Immunologic Diseases, Xiangya Hospital, Changsha, Hunan, China.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Changsha, Hunan, China.
| | - Hui Luo
- The Department of Rheumatology and Immunology, Xiangya Hospital of Central South University, Changsha, Hunan, China.
- Provincial Clinical Research Center for Rheumatic and Immunologic Diseases, Xiangya Hospital, Changsha, Hunan, China.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Changsha, Hunan, China.
| |
Collapse
|
20
|
Hasanaj E, Alavi A, Gupta A, Póczos B, Bar-Joseph Z. Multiset multicover methods for discriminative marker selection. CELL REPORTS METHODS 2022; 2:100332. [PMID: 36452867 PMCID: PMC9701606 DOI: 10.1016/j.crmeth.2022.100332] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 08/12/2022] [Accepted: 10/18/2022] [Indexed: 06/17/2023]
Abstract
Markers are increasingly being used for several high-throughput data analysis and experimental design tasks. Examples include the use of markers for assigning cell types in scRNA-seq studies, for deconvolving bulk gene expression data, and for selecting marker proteins in single-cell spatial proteomics studies. Most marker selection methods focus on differential expression (DE) analysis. Although such methods work well for data with a few non-overlapping marker sets, they are not appropriate for large atlas-size datasets where several cell types and tissues are considered. To address this, we define the phenotype cover (PC) problem for marker selection and present algorithms that can improve the discriminative power of marker sets. Analysis of these sets on several marker-selection tasks suggests that these methods can lead to solutions that accurately distinguish different phenotypes in the data.
Collapse
Affiliation(s)
- Euxhen Hasanaj
- Machine Learning Department, School of Computer Science, Carnegie Mellon University, Pittsburgh, PA 15213, USA
| | - Amir Alavi
- Computational Biology Department, School of Computer Science, Carnegie Mellon University, Pittsburgh, PA 15213, USA
| | - Anupam Gupta
- Computer Science Department, School of Computer Science, Carnegie Mellon University, Pittsburgh, PA 15213, USA
| | - Barnabás Póczos
- Machine Learning Department, School of Computer Science, Carnegie Mellon University, Pittsburgh, PA 15213, USA
| | - Ziv Bar-Joseph
- Machine Learning Department, School of Computer Science, Carnegie Mellon University, Pittsburgh, PA 15213, USA
- Computational Biology Department, School of Computer Science, Carnegie Mellon University, Pittsburgh, PA 15213, USA
| |
Collapse
|
21
|
Magnusson R, Rundquist O, Kim MJ, Hellberg S, Na CH, Benson M, Gomez-Cabrero D, Kockum I, Tegnér JN, Piehl F, Jagodic M, Mellergård J, Altafini C, Ernerudh J, Jenmalm MC, Nestor CE, Kim MS, Gustafsson M. RNA-sequencing and mass-spectrometry proteomic time-series analysis of T-cell differentiation identified multiple splice variants models that predicted validated protein biomarkers in inflammatory diseases. Front Mol Biosci 2022; 9:916128. [PMID: 36106020 PMCID: PMC9465313 DOI: 10.3389/fmolb.2022.916128] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Accepted: 07/25/2022] [Indexed: 12/18/2022] Open
Abstract
Profiling of mRNA expression is an important method to identify biomarkers but complicated by limited correlations between mRNA expression and protein abundance. We hypothesised that these correlations could be improved by mathematical models based on measuring splice variants and time delay in protein translation. We characterised time-series of primary human naïve CD4+ T cells during early T helper type 1 differentiation with RNA-sequencing and mass-spectrometry proteomics. We performed computational time-series analysis in this system and in two other key human and murine immune cell types. Linear mathematical mixed time delayed splice variant models were used to predict protein abundances, and the models were validated using out-of-sample predictions. Lastly, we re-analysed RNA-seq datasets to evaluate biomarker discovery in five T-cell associated diseases, further validating the findings for multiple sclerosis (MS) and asthma. The new models significantly out-performing models not including the usage of multiple splice variants and time delays, as shown in cross-validation tests. Our mathematical models provided more differentially expressed proteins between patients and controls in all five diseases. Moreover, analysis of these proteins in asthma and MS supported their relevance. One marker, sCD27, was validated in MS using two independent cohorts for evaluating response to treatment and disease prognosis. In summary, our splice variant and time delay models substantially improved the prediction of protein abundance from mRNA expression in three different immune cell types. The models provided valuable biomarker candidates, which were further validated in MS and asthma.
Collapse
Affiliation(s)
- Rasmus Magnusson
- Bioinformatics, Department of Physics, Chemistry and Biology, Linköping University, Linköping, Sweden
| | - Olof Rundquist
- Bioinformatics, Department of Physics, Chemistry and Biology, Linköping University, Linköping, Sweden
| | - Min Jung Kim
- Department of Applied Chemistry, College of Applied Sciences, Kyung Hee University, Yong-in, South Korea
| | - Sandra Hellberg
- Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| | - Chan Hyun Na
- Department of Neurology, Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Mikael Benson
- Centre for Personalised Medicine, Linköping University, Linköping, Sweden
| | - David Gomez-Cabrero
- Navarrabiomed, Complejo Hospitalario de Navarra, Universidad Pública de Navarra, IdiSNA, Pamplona, Spain
| | - Ingrid Kockum
- Department of Clinical Neuroscience, Center for Molecular Medicine, Karolinska Institute, Stockholm, Sweden
| | - Jesper N. Tegnér
- Biological and Environmental Sciences and Engineering Division, Computer, Electrical and Mathematical Sciences and Engineering Division, King Abdullah University of Science and Technology (KAUST), Thuwal, Saudi Arabia
- Unit of Computational Medicine, Department of Medicine, Solna, Center for Molecular Medicine, Karolinska Institutet, Solna, Sweden
- Science for Life Laboratory, Solna, Sweden
| | - Fredrik Piehl
- Department of Clinical Neuroscience, Center for Molecular Medicine, Karolinska Institute, Stockholm, Sweden
| | - Maja Jagodic
- Department of Clinical Neuroscience, Center for Molecular Medicine, Karolinska Institute, Stockholm, Sweden
| | - Johan Mellergård
- Department of Neurology, Linköping University, Linköping, Sweden
- Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| | - Claudio Altafini
- Department of Automatic Control, Linköping University, Linköping, Sweden
| | - Jan Ernerudh
- Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
- Department of Clinical Immunology and Transfusion Medicine, Linköping University, Linköping, Sweden
| | - Maria C. Jenmalm
- Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
- *Correspondence: Maria C. Jenmalm, ; Mika Gustafsson,
| | - Colm E. Nestor
- Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| | - Min-Sik Kim
- Department of New Biology, Daegu Gyeongbuk Institute of Science and Technology, Daegu, South Korea
| | - Mika Gustafsson
- Bioinformatics, Department of Physics, Chemistry and Biology, Linköping University, Linköping, Sweden
- *Correspondence: Maria C. Jenmalm, ; Mika Gustafsson,
| |
Collapse
|
22
|
Shilts J, Severin Y, Galaway F, Müller-Sienerth N, Chong ZS, Pritchard S, Teichmann S, Vento-Tormo R, Snijder B, Wright GJ. A physical wiring diagram for the human immune system. Nature 2022; 608:397-404. [PMID: 35922511 PMCID: PMC9365698 DOI: 10.1038/s41586-022-05028-x] [Citation(s) in RCA: 42] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Accepted: 06/28/2022] [Indexed: 12/14/2022]
Abstract
The human immune system is composed of a distributed network of cells circulating throughout the body, which must dynamically form physical associations and communicate using interactions between their cell-surface proteomes1. Despite their therapeutic potential2, our map of these surface interactions remains incomplete3,4. Here, using a high-throughput surface receptor screening method, we systematically mapped the direct protein interactions across a recombinant library that encompasses most of the surface proteins that are detectable on human leukocytes. We independently validated and determined the biophysical parameters of each novel interaction, resulting in a high-confidence and quantitative view of the receptor wiring that connects human immune cells. By integrating our interactome with expression data, we identified trends in the dynamics of immune interactions and constructed a reductionist mathematical model that predicts cellular connectivity from basic principles. We also developed an interactive multi-tissue single-cell atlas that infers immune interactions throughout the body, revealing potential functional contexts for new interactions and hubs in multicellular networks. Finally, we combined targeted protein stimulation of human leukocytes with multiplex high-content microscopy to link our receptor interactions to functional roles, in terms of both modulating immune responses and maintaining normal patterns of intercellular associations. Together, our work provides a systematic perspective on the intercellular wiring of the human immune system that extends from systems-level principles of immune cell connectivity down to mechanistic characterization of individual receptors, which could offer opportunities for therapeutic intervention.
Collapse
Affiliation(s)
- Jarrod Shilts
- Cell Surface Signalling Laboratory, Wellcome Sanger Institute, Cambridge, UK.
| | - Yannik Severin
- Institute of Molecular Systems Biology, ETH Zurich, Zurich, Switzerland
| | - Francis Galaway
- Cell Surface Signalling Laboratory, Wellcome Sanger Institute, Cambridge, UK
| | | | - Zheng-Shan Chong
- Cell Surface Signalling Laboratory, Wellcome Sanger Institute, Cambridge, UK
| | - Sophie Pritchard
- Cellular Genetics Programme, Wellcome Sanger Institute, Cambridge, UK
| | - Sarah Teichmann
- Cellular Genetics Programme, Wellcome Sanger Institute, Cambridge, UK
| | - Roser Vento-Tormo
- Cellular Genetics Programme, Wellcome Sanger Institute, Cambridge, UK
| | - Berend Snijder
- Institute of Molecular Systems Biology, ETH Zurich, Zurich, Switzerland
| | - Gavin J Wright
- Cell Surface Signalling Laboratory, Wellcome Sanger Institute, Cambridge, UK.
- Department of Biology, Hull York Medical School, York Biomedical Research Institute, University of York, York, UK.
| |
Collapse
|
23
|
Becker J, Fakhiri J, Grimm D. Fantastic AAV Gene Therapy Vectors and How to Find Them—Random Diversification, Rational Design and Machine Learning. Pathogens 2022; 11:pathogens11070756. [PMID: 35890005 PMCID: PMC9318892 DOI: 10.3390/pathogens11070756] [Citation(s) in RCA: 40] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Revised: 06/30/2022] [Accepted: 07/01/2022] [Indexed: 11/16/2022] Open
Abstract
Parvoviruses are a diverse family of small, non-enveloped DNA viruses that infect a wide variety of species, tissues and cell types. For over half a century, their intriguing biology and pathophysiology has fueled intensive research aimed at dissecting the underlying viral and cellular mechanisms. Concurrently, their broad host specificity (tropism) has motivated efforts to develop parvoviruses as gene delivery vectors for human cancer or gene therapy applications. While the sum of preclinical and clinical data consistently demonstrates the great potential of these vectors, these findings also illustrate the importance of enhancing and restricting in vivo transgene expression in desired cell types. To this end, major progress has been made especially with vectors based on Adeno-associated virus (AAV), whose capsid is highly amenable to bioengineering, repurposing and expansion of its natural tropism. Here, we provide an overview of the state-of-the-art approaches to create new AAV variants with higher specificity and efficiency of gene transfer in on-target cells. We first review traditional and novel directed evolution approaches, including high-throughput screening of AAV capsid libraries. Next, we discuss programmable receptor-mediated targeting with a focus on two recent technologies that utilize high-affinity binders. Finally, we highlight one of the latest stratagems for rational AAV vector characterization and optimization, namely, machine learning, which promises to facilitate and accelerate the identification of next-generation, safe and precise gene delivery vehicles.
Collapse
Affiliation(s)
- Jonas Becker
- Department of Infectious Diseases/Virology, Medical Faculty, University of Heidelberg, Center for Integrative Infectious Diseases Research (CIID), BioQuant, 69120 Heidelberg, Germany;
- Faculty of Biosciences, University of Heidelberg, 69120 Heidelberg, Germany
| | - Julia Fakhiri
- Roche Pharma Research and Early Development, Therapeutic Modalities, Roche Innovation Center Munich, Roche Diagnostics GmbH, Nonnenwald 2, 82377 Penzberg, Germany
- Correspondence: (J.F.); (D.G.); Tel.: +49-174-3486203 (J.F.); +49-6221-5451331 (D.G.)
| | - Dirk Grimm
- Department of Infectious Diseases/Virology, Medical Faculty, University of Heidelberg, Center for Integrative Infectious Diseases Research (CIID), BioQuant, 69120 Heidelberg, Germany;
- German Center for Infection Research (DZIF), Partner Site Heidelberg, 69120 Heidelberg, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Heidelberg, 69120 Heidelberg, Germany
- Correspondence: (J.F.); (D.G.); Tel.: +49-174-3486203 (J.F.); +49-6221-5451331 (D.G.)
| |
Collapse
|
24
|
Feleke M, Feng W, Rothzerg E, Song D, Wei Q, Kõks S, Wood D, Liu Y, Xu J. Single-cell RNA-seq identification of four differentially expressed survival-related genes by a TARGET: Osteosarcoma database analysis. Exp Biol Med (Maywood) 2022; 247:921-930. [PMID: 35285281 PMCID: PMC9189571 DOI: 10.1177/15353702221080131] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Accepted: 01/23/2022] [Indexed: 09/05/2023] Open
Abstract
Osteosarcoma (OS) differentially expressed genes (DEGs) have been predicted using the data portal of the Therapeutically Applicable Research to Generate Effective Treatments (TARGET). In this study, we sought to identify cell types that specially express key DEGs (MUC1, COL13A1, JAG2, and KAZALD1) in each of the nine identified cell populations derived from tissues of OS tumors with single-cell RNA-sequencing data. Gene expression levels were pairwise compared between cell clusters and a p value < 0.05 was considered differentially expressed. It was revealed that MUC1 is expressed at high levels in osteoblastic OS cells followed by carcinoma-associated fibroblasts (CAFs) and plasmocytes, respectively. COL13A1 is highly expressed in osteoblastic OS cells, CAFs, and endothelial cells (ECs), respectively. The KAZALD1 gene is expressed in CAFs and osteoblastic OS cells at high levels, but at very low levels in plasmocytes, osteoclasts, NK/T, myeloid cells 1, myeloid cells 2, ECs, and B cells. JAG2 is expressed at significantly high levels in ECs and osteoblastic OS cells, and at relatively lower levels in all other cell types. Interestingly, LSAMP, as an established gene in the development of OS shows high expression in osteoblastic OS cells and CAFs but low in other cells such as osteoclasts. Our findings here highlight the heterogeneity of OS cells and cell-type-dependent DEGs which have potential as therapeutic targets in OS.
Collapse
Affiliation(s)
- Mesalie Feleke
- School of Biomedical Sciences, The University of Western Australia, Perth, WA 6009, Australia
| | - Wenyu Feng
- Department of Orthopaedics, First Affiliated Hospital of Guangxi Medical University, Nanning 530021, China
| | - Emel Rothzerg
- School of Biomedical Sciences, The University of Western Australia, Perth, WA 6009, Australia
| | - Dezhi Song
- School of Biomedical Sciences, The University of Western Australia, Perth, WA 6009, Australia
- Research Centre for Regenerative Medicine, Guangxi Medical University, Nanning 530021, China
| | - Qingjun Wei
- Department of Orthopaedics, First Affiliated Hospital of Guangxi Medical University, Nanning 530021, China
| | - Sulev Kõks
- Perron Institute for Neurological and Translational Science, Queen Elizabeth II Medical Centre, Nedlands, WA 6009, Australia
- Centre for Molecular Medicine and Innovative Therapeutics, Murdoch University, Murdoch, WA 6150, Australia
| | - David Wood
- Medical School, The University of Western Australia, Perth, WA 6009, Australia
| | - Yun Liu
- School of Biomedical Sciences, The University of Western Australia, Perth, WA 6009, Australia
- Department of Orthopaedics, First Affiliated Hospital of Guangxi Medical University, Nanning 530021, China
| | - Jiake Xu
- School of Biomedical Sciences, The University of Western Australia, Perth, WA 6009, Australia
| |
Collapse
|
25
|
Fiocchi C, Iliopoulos D. Inflammatory Bowel Disease Therapy: Beyond the Immunome. Front Immunol 2022; 13:864762. [PMID: 35615360 PMCID: PMC9124778 DOI: 10.3389/fimmu.2022.864762] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Accepted: 04/11/2022] [Indexed: 12/19/2022] Open
Affiliation(s)
- Claudio Fiocchi
- Department of Inflammation & Immunity, Lerner Research Institute Cleveland, Cleveland, OH, United States
- Department of Gastroenterology, Hepatology & Nutrition, Digestive Disease and Surgery Institute, Cleveland Clinic, Cleveland, OH, United States
- *Correspondence: Claudio Fiocchi,
| | | |
Collapse
|
26
|
Li X, Lee EJ, Lilja S, Loscalzo J, Schäfer S, Smelik M, Strobl MR, Sysoev O, Wang H, Zhang H, Zhao Y, Gawel DR, Bohle B, Benson M. A dynamic single cell-based framework for digital twins to prioritize disease genes and drug targets. Genome Med 2022; 14:48. [PMID: 35513850 PMCID: PMC9074288 DOI: 10.1186/s13073-022-01048-4] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Accepted: 04/13/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Medical digital twins are computational disease models for drug discovery and treatment. Unresolved problems include how to organize and prioritize between disease-associated changes in digital twins, on cellulome- and genome-wide scales. We present a dynamic framework that can be used to model such changes and thereby prioritize upstream regulators (URs) for biomarker- and drug discovery. METHODS We started with seasonal allergic rhinitis (SAR) as a disease model, by analyses of in vitro allergen-stimulated peripheral blood mononuclear cells (PBMC) from SAR patients. Time-series a single-cell RNA-sequencing (scRNA-seq) data of these cells were used to construct multicellular network models (MNMs) at each time point of molecular interactions between cell types. We hypothesized that predicted molecular interactions between cell types in the MNMs could be traced to find an UR gene, at an early time point. We performed bioinformatic and functional studies of the MNMs to develop a scalable framework to prioritize UR genes. This framework was tested on a single-cell and bulk-profiling data from SAR and other inflammatory diseases. RESULTS Our scRNA-seq-based time-series MNMs of SAR showed thousands of differentially expressed genes (DEGs) across multiple cell types, which varied between time points. Instead of a single-UR gene in each MNM, we found multiple URs dispersed across the cell types. Thus, at each time point, the MNMs formed multi-directional networks. The absence of linear hierarchies and time-dependent variations in MNMs complicated the prioritization of URs. For example, the expression and functions of Th2 cytokines, which are approved drug targets in allergies, varied across cell types, and time points. Our analyses of bulk- and single-cell data from other inflammatory diseases also revealed multi-directional networks that showed stage-dependent variations. We therefore developed a quantitative approach to prioritize URs: we ranked the URs based on their predicted effects on downstream target cells. Experimental and bioinformatic analyses supported that this kind of ranking is a tractable approach for prioritizing URs. CONCLUSIONS We present a scalable framework for modeling dynamic changes in digital twins, on cellulome- and genome-wide scales, to prioritize UR genes for biomarker and drug discovery.
Collapse
Affiliation(s)
- Xinxiu Li
- Centre for Personalized Medicine, Linköping University, Linköping, Sweden
| | - Eun Jung Lee
- Centre for Personalized Medicine, Linköping University, Linköping, Sweden
- Department of Otorhinolaryngology, Yonsei University Wonju College of Medicine, Wonju, Korea
| | - Sandra Lilja
- Centre for Personalized Medicine, Linköping University, Linköping, Sweden
| | - Joseph Loscalzo
- Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
- Channing Division of Network Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Samuel Schäfer
- Centre for Personalized Medicine, Linköping University, Linköping, Sweden
| | - Martin Smelik
- Centre for Personalized Medicine, Linköping University, Linköping, Sweden
| | - Maria Regina Strobl
- Department of Pathophysiology and Allergy Research, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Oleg Sysoev
- Division of Statistics and Machine Learning, Department of Computer and Information Science, Linkoping University, Linköping, Sweden
| | - Hui Wang
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogenic Biology and Immunology, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Huan Zhang
- Centre for Personalized Medicine, Linköping University, Linköping, Sweden
| | - Yelin Zhao
- Centre for Personalized Medicine, Linköping University, Linköping, Sweden
| | - Danuta R Gawel
- Centre for Personalized Medicine, Linköping University, Linköping, Sweden
| | - Barbara Bohle
- Department of Pathophysiology and Allergy Research, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Mikael Benson
- Centre for Personalized Medicine, Linköping University, Linköping, Sweden.
- Crown Princess Victoria Children's Hospital, Linköping University Hospital, Linköping, Sweden.
- Division of ENT Diseases, Department of Clinical Sciences, Intervention and Technology, Karolinska Institutet, Stockholm, Sweden.
| |
Collapse
|
27
|
Liu HP, Wang D, Lai HM. Can we infer tumor presence of single cell transcriptomes and their tumor of origin from bulk transcriptomes by machine learning? Comput Struct Biotechnol J 2022; 20:2672-2679. [PMID: 35685355 PMCID: PMC9162953 DOI: 10.1016/j.csbj.2022.05.035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Revised: 05/09/2022] [Accepted: 05/19/2022] [Indexed: 11/17/2022] Open
Abstract
There is a growing need to build a model that uses single cell RNA-seq (scRNA-seq) to separate malignant cells from nonmalignant cells and to identify tumor of origin of single cells and/or circulating tumor cells (CTCs). Currently, it is infeasible to build a tumor of origin model learnt from scRNA-seq by machine learning (ML). We then wondered if an ML model learnt from bulk transcriptomes is applicable to scRNA-seq to infer single cells’ tumor presence and further indicate their tumor of origin. We used k-nearest neighbors, one-versus-all support vector machine, one-versus-one support vector machine, random forest and introduced scTumorTrace to conduct a pioneering experiment containing leukocytes and seven major cancer types where bulk RNA-seq and scRNA-seq data were available. 13 ML models learnt from bulk RNA-seq were all reliable to use (F-score > 96%) shown by a validation set of bulk transcriptomes, but none of them was applicable to scRNA-seq except scTumorTrace. Making inferences from bulk RNA-seq to scRNA-seq was impaired by feature selection and improved by log2-transformed TPM units. scTumorTrace with transcriptome-wide 2-tuples showed F-score beyond 98.74 and 94.29% in inferring tumor presence and tumor of origin at single-cell resolution and correctly identified 45 single candidate prostate CTCs but lineage-confirmed non-CTCs as leukocytes. We concluded that modern ML techniques are quantitative and could hardly address the raised questions. scTumorTrace with transcriptome-wide 2-tuples is qualitative, standardization-free and not subject to log2-transformed quantities, enabling us to infer tumor presence of single cell transcriptomes and their tumor of origin from bulk transcriptomes.
Collapse
Affiliation(s)
- Hua-Ping Liu
- National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital & Shenzhen Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shenzhen 518116, China
| | - Dongwen Wang
- National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital & Shenzhen Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shenzhen 518116, China
| | - Hung-Ming Lai
- Aiphaqua Genomics Research Unit, Taipei 111, Taiwan
- Corresponding author.
| |
Collapse
|
28
|
López-Cortés A, Guerrero S, Ortiz-Prado E, Yumiceba V, Vera-Guapi A, León Cáceres Á, Simbaña-Rivera K, Gómez-Jaramillo AM, Echeverría-Garcés G, García-Cárdenas JM, Guevara-Ramírez P, Cabrera-Andrade A, Puig San Andrés L, Cevallos-Robalino D, Bautista J, Armendáriz-Castillo I, Pérez-Villa A, Abad-Sojos A, Ramos-Medina MJ, León-Sosa A, Abarca E, Pérez-Meza ÁA, Nieto-Jaramillo K, Jácome AV, Morillo A, Arias-Erazo F, Fuenmayor-González L, Quiñones LA, Kyriakidis NC. Pulmonary Inflammatory Response in Lethal COVID-19 Reveals Potential Therapeutic Targets and Drugs in Phases III/IV Clinical Trials. Front Pharmacol 2022; 13:833174. [PMID: 35422702 PMCID: PMC9002106 DOI: 10.3389/fphar.2022.833174] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Accepted: 03/07/2022] [Indexed: 12/26/2022] Open
Abstract
Background: It is imperative to identify drugs that allow treating symptoms of severe COVID-19. Respiratory failure is the main cause of death in severe COVID-19 patients, and the host inflammatory response at the lungs remains poorly understood. Methods: Therefore, we retrieved data from post-mortem lungs from COVID-19 patients and performed in-depth in silico analyses of single-nucleus RNA sequencing data, inflammatory protein interactome network, and shortest pathways to physiological phenotypes to reveal potential therapeutic targets and drugs in advanced-stage COVID-19 clinical trials. Results: Herein, we analyzed transcriptomics data of 719 inflammatory response genes across 19 cell types (116,313 nuclei) from lung autopsies. The functional enrichment analysis of the 233 significantly expressed genes showed that the most relevant biological annotations were inflammatory response, innate immune response, cytokine production, interferon production, macrophage activation, blood coagulation, NLRP3 inflammasome complex, and the TLR, JAK-STAT, NF-κB, TNF, oncostatin M signaling pathways. Subsequently, we identified 34 essential inflammatory proteins with both high-confidence protein interactions and shortest pathways to inflammation, cell death, glycolysis, and angiogenesis. Conclusion: We propose three small molecules (baricitinib, eritoran, and montelukast) that can be considered for treating severe COVID-19 symptoms after being thoroughly evaluated in COVID-19 clinical trials.
Collapse
Affiliation(s)
- Andrés López-Cortés
- Programa de Investigación en Salud Global, Facultad de Ciencias de la Salud, Universidad Internacional SEK, Quito, Ecuador.,Latin American Network for the Implementation and Validation of Clinical Pharmacogenomics Guidelines (RELIVAF-CYTED), Madrid, Spain
| | - Santiago Guerrero
- Escuela de Medicina, Facultad de Ciencias Médicas de la Salud y de la Vida, Universidad Internacional del Ecuador, Quito, Ecuador
| | - Esteban Ortiz-Prado
- One Health Research Group, Faculty of Medicine, Universidad de Las Américas, Quito, Ecuador
| | - Verónica Yumiceba
- Institut für Humangenetik Lübeck, Universität zu Lübeck, Lübeck, Germany
| | - Antonella Vera-Guapi
- Integrated Research and Treatment Center, Center for Sepsis Control and Care (CSCC), Jena University Hospital, Jena, Germany
| | - Ángela León Cáceres
- Heidelberg Institute of Global Health, Faculty of Medicine, University of Heidelberg, Heidelberg, Germany
| | - Katherine Simbaña-Rivera
- One Health Research Group, Faculty of Medicine, Universidad de Las Américas, Quito, Ecuador.,Latin American Network for Cancer Research (LAN-CANCER), Lima, Peru
| | - Ana María Gómez-Jaramillo
- Centro de Investigación para la Salud en América Latina (CISeAL), Pontificia Universidad Católica del Ecuador, Quito, Ecuador
| | - Gabriela Echeverría-Garcés
- Latin American Network for the Implementation and Validation of Clinical Pharmacogenomics Guidelines (RELIVAF-CYTED), Madrid, Spain
| | - Jennyfer M García-Cárdenas
- Escuela de Medicina, Facultad de Ciencias Médicas de la Salud y de la Vida, Universidad Internacional del Ecuador, Quito, Ecuador
| | - Patricia Guevara-Ramírez
- Latin American Network for the Implementation and Validation of Clinical Pharmacogenomics Guidelines (RELIVAF-CYTED), Madrid, Spain
| | | | | | | | | | - Isaac Armendáriz-Castillo
- Facultade de Ciencias, Universidade da Coruña, A Coruña, Spain.,Instituto Nacional de Investigación en Salud Pública, Quito, Ecuador
| | - Andy Pérez-Villa
- Latin American Network for the Implementation and Validation of Clinical Pharmacogenomics Guidelines (RELIVAF-CYTED), Madrid, Spain
| | | | | | | | | | - Álvaro A Pérez-Meza
- Biotechnology Engineering Career, Faculty of Life Sciences, Universidad Regional Amazónica Ikiam, Tena, Ecuador
| | | | - Andrea V Jácome
- Faculty of Medicine, Universidad de Las Américas, Quito, Ecuador
| | | | | | | | - Luis Abel Quiñones
- Latin American Network for the Implementation and Validation of Clinical Pharmacogenomics Guidelines (RELIVAF-CYTED), Madrid, Spain.,Laboratory of Chemical Carcinogenesis and Pharmacogenetics, Department of Basic-Clinical Oncology, Faculty of Medicine, University of Chile, Santiago, Chile
| | - Nikolaos C Kyriakidis
- One Health Research Group, Faculty of Medicine, Universidad de Las Américas, Quito, Ecuador
| |
Collapse
|
29
|
Integrated bioinformatic analysis of gene expression profiling data to identify combinatorial biomarkers in inflammatory skin disease. Sci Rep 2022; 12:5889. [PMID: 35393522 PMCID: PMC8989986 DOI: 10.1038/s41598-022-09840-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2021] [Accepted: 03/28/2022] [Indexed: 11/18/2022] Open
Abstract
Selection of appropriate biomarker to identify inflammatory skin diseases is complicated by the involvement of thousands of differentially expressed genes (DEGs) across multiple cell types and organs. This study aimed to identify combinatorial biomarkers in inflammatory skin diseases. From one gene expression microarray profiling dataset, we performed bioinformatic analyses on dataset from lesional skin biopsies of patients with inflammatory skin diseases (atopic dermatitis [AD], contact eczema [KE], lichen planus [Li], psoriasis vulgaris [Pso]) and healthy controls to identify the involved pathways, predict upstream regulators, and potential measurable extracellular biomarkers. Overall, 434, 629, 581, and 738 DEGs were mapped in AD, KE, Li, and Pso, respectively; 238 identified DEGs were shared among four different inflammatory skin diseases. Bioinformatic analysis on four inflammatory skin diseases showed significant activation of pathways with known pathogenic relevance. Common upstream regulators, with upregulated predicted activity, identified were CNR1 and BMP4. We found the following common serum biomarkers: ACR, APOE, ASIP, CRISP1, DKK1, IL12B, IL9, MANF, MDK, NRTN, PCSK5, and VEGFC. Considerable differences of gene expression changes, involved pathways, upstream regulators, and biomarkers were found in different inflammatory skin diseases. Integrated bioinformatic analysis identified 12 potential common biomarkers of inflammatory skin diseases requiring further evaluation.
Collapse
|
30
|
Muench DE, Sun Z, Sharma A, Tang C, Crampton JS, Lao C, Kersjes K, Chang W, Na S. A Pathogenic Th17/CD38 + Macrophage Feedback Loop Drives Inflammatory Arthritis through TNF-α. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2022; 208:1315-1328. [PMID: 35197330 DOI: 10.4049/jimmunol.2101025] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Accepted: 01/04/2022] [Indexed: 12/29/2022]
Abstract
The pathobiology of rheumatoid inflammatory diseases, including rheumatoid arthritis (RA) and psoriatic arthritis, involves the interplay between innate and adaptive immune components and resident synoviocytes. Single-cell analyses of patient samples and relevant mouse models have characterized many cellular subsets in RA. However, the impact of interactions between cell types is not fully understood. In this study, we temporally profiled murine arthritic synovial isolates at the single-cell level to identify perturbations similar to those found in human RA. Notably, murine macrophage subtypes like those found in RA patients were expanded in arthritis and linked to promoting the function of Th17 cells in the joint. In vitro experiments identified a capacity for murine macrophages to maintain the functionality and expansion of Th17 cells. Reciprocally, murine Th17 cell-derived TNF-α induced CD38+ macrophages that enhanced Th17 functionality. Murine synovial CD38+ macrophages were expanded during arthritis, and their depletion or blockade via TNF-α neutralization alleviated disease while reducing IL-17A-producing cells. These findings identify a cellular feedback loop that promotes Th17 cell pathogenicity through TNF-α to drive inflammatory arthritis.
Collapse
Affiliation(s)
- David E Muench
- Immunology Discovery Research, Lilly Research Laboratories, Eli Lilly and Company, San Diego, CA
| | - Zhe Sun
- Discovery Research, Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, IN; and
| | - Anchal Sharma
- Research Information and Digital Solutions, Lilly Research Laboratories, Eli Lilly and Company, New York, NY
| | - Crystal Tang
- Immunology Discovery Research, Lilly Research Laboratories, Eli Lilly and Company, San Diego, CA
| | - Jordan S Crampton
- Immunology Discovery Research, Lilly Research Laboratories, Eli Lilly and Company, San Diego, CA
| | - Christopher Lao
- Immunology Discovery Research, Lilly Research Laboratories, Eli Lilly and Company, San Diego, CA
| | - Kara Kersjes
- Immunology Discovery Research, Lilly Research Laboratories, Eli Lilly and Company, San Diego, CA
| | - William Chang
- Immunology Discovery Research, Lilly Research Laboratories, Eli Lilly and Company, San Diego, CA
| | - Songqing Na
- Immunology Discovery Research, Lilly Research Laboratories, Eli Lilly and Company, San Diego, CA;
| |
Collapse
|
31
|
Xiang J, Zhang J, Zhao Y, Wu FX, Li M. Biomedical data, computational methods and tools for evaluating disease-disease associations. Brief Bioinform 2022; 23:6522999. [PMID: 35136949 DOI: 10.1093/bib/bbac006] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Revised: 01/04/2022] [Accepted: 01/05/2022] [Indexed: 12/12/2022] Open
Abstract
In recent decades, exploring potential relationships between diseases has been an active research field. With the rapid accumulation of disease-related biomedical data, a lot of computational methods and tools/platforms have been developed to reveal intrinsic relationship between diseases, which can provide useful insights to the study of complex diseases, e.g. understanding molecular mechanisms of diseases and discovering new treatment of diseases. Human complex diseases involve both external phenotypic abnormalities and complex internal molecular mechanisms in organisms. Computational methods with different types of biomedical data from phenotype to genotype can evaluate disease-disease associations at different levels, providing a comprehensive perspective for understanding diseases. In this review, available biomedical data and databases for evaluating disease-disease associations are first summarized. Then, existing computational methods for disease-disease associations are reviewed and classified into five groups in terms of the usages of biomedical data, including disease semantic-based, phenotype-based, function-based, representation learning-based and text mining-based methods. Further, we summarize software tools/platforms for computation and analysis of disease-disease associations. Finally, we give a discussion and summary on the research of disease-disease associations. This review provides a systematic overview for current disease association research, which could promote the development and applications of computational methods and tools/platforms for disease-disease associations.
Collapse
Affiliation(s)
- Ju Xiang
- School of Computer Science and Engineering, Central South University, China
| | - Jiashuai Zhang
- Hunan Provincial Key Lab on Bioinformatics, School of Computer Science and Engineering, Central South University, Changsha, Hunan 410083, China
| | - Yichao Zhao
- School of Computer Science and Engineering, Central South University, China
| | - Fang-Xiang Wu
- Hunan Provincial Key Lab on Bioinformatics, School of Computer Science and Engineering, Central South University, Changsha, Hunan 410083, China
| | - Min Li
- Division of Biomedical Engineering and Department of Mechanical Engineering at University of Saskatchewan, Saskatoon, Canada
| |
Collapse
|
32
|
Johnson TS, Yu CY, Huang Z, Xu S, Wang T, Dong C, Shao W, Zaid MA, Huang X, Wang Y, Bartlett C, Zhang Y, Walker BA, Liu Y, Huang K, Zhang J. Diagnostic Evidence GAuge of Single cells (DEGAS): a flexible deep transfer learning framework for prioritizing cells in relation to disease. Genome Med 2022; 14:11. [PMID: 35105355 PMCID: PMC8808996 DOI: 10.1186/s13073-022-01012-2] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Accepted: 01/07/2022] [Indexed: 12/13/2022] Open
Abstract
We propose DEGAS (Diagnostic Evidence GAuge of Single cells), a novel deep transfer learning framework, to transfer disease information from patients to cells. We call such transferrable information "impressions," which allow individual cells to be associated with disease attributes like diagnosis, prognosis, and response to therapy. Using simulated data and ten diverse single-cell and patient bulk tissue transcriptomic datasets from glioblastoma multiforme (GBM), Alzheimer's disease (AD), and multiple myeloma (MM), we demonstrate the feasibility, flexibility, and broad applications of the DEGAS framework. DEGAS analysis on myeloma single-cell transcriptomics identified PHF19high myeloma cells associated with progression. Availability: https://github.com/tsteelejohnson91/DEGAS .
Collapse
Affiliation(s)
- Travis S Johnson
- Department of Medicine, Indiana University School of Medicine, 535 Barnhill Dr, Indianapolis, IN, 46202, USA
- Department of Biomedical Informatics, The Ohio State University College of Medicine, 370 W 9th Ave, Columbus, OH, 43210, USA
- Department of Biostatistics and Health Data Science, Indiana University School of Medicine, 410 W 10th St, Suite 3000, Indianapolis, IN, 46202, USA
| | - Christina Y Yu
- Department of Medicine, Indiana University School of Medicine, 535 Barnhill Dr, Indianapolis, IN, 46202, USA
- Department of Biomedical Informatics, The Ohio State University College of Medicine, 370 W 9th Ave, Columbus, OH, 43210, USA
| | - Zhi Huang
- School of Electrical and Computer Engineering, Purdue University, 465 Northwestern Ave, West Lafayette, IN, 47907, USA
| | - Siwen Xu
- Center for Computational Biology and Bioinformatics, Indiana University School of Medicine, 410 W. 10th St, Suite 5000, Indianapolis, IN, 46202, USA
| | - Tongxin Wang
- Department of Computer Science, Indiana University, 150 S Woodlawn Ave, Bloomington, IN, 47405, USA
| | - Chuanpeng Dong
- Center for Computational Biology and Bioinformatics, Indiana University School of Medicine, 410 W. 10th St, Suite 5000, Indianapolis, IN, 46202, USA
| | - Wei Shao
- Department of Medicine, Indiana University School of Medicine, 535 Barnhill Dr, Indianapolis, IN, 46202, USA
| | - Mohammad Abu Zaid
- Department of Medicine, Indiana University School of Medicine, 535 Barnhill Dr, Indianapolis, IN, 46202, USA
| | - Xiaoqing Huang
- Department of Biostatistics and Health Data Science, Indiana University School of Medicine, 410 W 10th St, Suite 3000, Indianapolis, IN, 46202, USA
| | - Yijie Wang
- Department of Computer Science, Indiana University, 150 S Woodlawn Ave, Bloomington, IN, 47405, USA
| | - Christopher Bartlett
- Battelle Center for Mathematical Medicine, Nationwide Children's Hospital, 575 Children's Crossroad, Columbus, OH, 43215, USA
| | - Yan Zhang
- Department of Biomedical Informatics, The Ohio State University College of Medicine, 370 W 9th Ave, Columbus, OH, 43210, USA
- The Ohio State University Comprehensive Cancer Center (OSUCCC - James), Starling-Loving Hall, 320 W 10th Ave, Columbus, OH, 43210, USA
| | - Brian A Walker
- Division of Hematology Oncology, Indiana University Melvin and Bren Simon Comprehensive Cancer Center, 535 Barnhill Dr, Indianapolis, IN, 46202, USA
| | - Yunlong Liu
- Center for Computational Biology and Bioinformatics, Indiana University School of Medicine, 410 W. 10th St, Suite 5000, Indianapolis, IN, 46202, USA
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, 410 W 10th St, Suite 4000, Indianapolis, IN, 46202, USA
| | - Kun Huang
- Department of Medicine, Indiana University School of Medicine, 535 Barnhill Dr, Indianapolis, IN, 46202, USA.
- Department of Biostatistics and Health Data Science, Indiana University School of Medicine, 410 W 10th St, Suite 3000, Indianapolis, IN, 46202, USA.
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, 410 W 10th St, Suite 4000, Indianapolis, IN, 46202, USA.
- Regenstrief Institute, 1101 W 10th St, Indianapolis, IN, 46202, USA.
| | - Jie Zhang
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, 410 W 10th St, Suite 4000, Indianapolis, IN, 46202, USA.
| |
Collapse
|
33
|
Huang PH, Kim KH, Schermer M. Ethical Issues of Digital Twins for Personalized Health Care Service: Preliminary Mapping Study. J Med Internet Res 2022; 24:e33081. [PMID: 35099399 PMCID: PMC8844982 DOI: 10.2196/33081] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Revised: 10/27/2021] [Accepted: 11/16/2021] [Indexed: 01/16/2023] Open
Abstract
BACKGROUND The concept of digital twins has great potential for transforming the existing health care system by making it more personalized. As a convergence of health care, artificial intelligence, and information and communication technologies, personalized health care services that are developed under the concept of digital twins raise a myriad of ethical issues. Although some of the ethical issues are known to researchers working on digital health and personalized medicine, currently, there is no comprehensive review that maps the major ethical risks of digital twins for personalized health care services. OBJECTIVE This study aims to fill the research gap by identifying the major ethical risks of digital twins for personalized health care services. We first propose a working definition for digital twins for personalized health care services to facilitate future discussions on the ethical issues related to these emerging digital health services. We then develop a process-oriented ethical map to identify the major ethical risks in each of the different data processing phases. METHODS We resorted to the literature on eHealth, personalized medicine, precision medicine, and information engineering to identify potential issues and developed a process-oriented ethical map to structure the inquiry in a more systematic way. The ethical map allows us to see how each of the major ethical concerns emerges during the process of transforming raw data into valuable information. Developers of a digital twin for personalized health care service may use this map to identify ethical risks during the development stage in a more systematic way and can proactively address them. RESULTS This paper provides a working definition of digital twins for personalized health care services by identifying 3 features that distinguish the new application from other eHealth services. On the basis of the working definition, this paper further layouts 10 major operational problems and the corresponding ethical risks. CONCLUSIONS It is challenging to address all the major ethical risks that a digital twin for a personalized health care service might encounter proactively without a conceptual map at hand. The process-oriented ethical map we propose here can assist the developers of digital twins for personalized health care services in analyzing ethical risks in a more systematic manner.
Collapse
Affiliation(s)
- Pei-Hua Huang
- Department of Medical Ethics, Philosophy and History of Medicine, Erasmus MC University Medical Center, Rotterdam, Netherlands
| | - Ki-Hun Kim
- Department of Industrial Engineering, Pusan National University, Busan, Republic of Korea
| | - Maartje Schermer
- Department of Medical Ethics, Philosophy and History of Medicine, Erasmus MC University Medical Center, Rotterdam, Netherlands
| |
Collapse
|
34
|
Bahonar S, Montazeri H. Somatic Single-Nucleotide Variant Calling from Single-Cell DNA Sequencing Data Using SCAN-SNV. Methods Mol Biol 2022; 2493:267-277. [PMID: 35751821 DOI: 10.1007/978-1-0716-2293-3_17] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
SCAN-SNV is a recent computational tool for somatic single-nucleotide variant (SNV) identification from the single-cell DNA sequencing data. The workflow of the SCAN-SNV package is as follows. First, candidate somatic SNVs and credible heterozygous single-nucleotide polymorphisms (hSNP) are obtained by analyzing single-cell and matched bulk sequencing data, respectively. Subsequently, SCAN-SNV estimates genome-wide allele-specific amplification balance (AB) at any position of DNA sequencing data using a probabilistic spatial statistical model. Finally, candidate somatic SNVs that are likely artifacts according to the AB predictions are further removed to obtain putative mutations. This chapter provides a step-by-step practical guide of the package by explaining how to install and use the variance caller in a real-world example.
Collapse
Affiliation(s)
- Sajedeh Bahonar
- Department of Bioinformatics, Institute of Biochemistry and Biophysics (IBB), University of Tehran, Tehran, Iran
| | - Hesam Montazeri
- Department of Bioinformatics, Institute of Biochemistry and Biophysics (IBB), University of Tehran, Tehran, Iran.
| |
Collapse
|
35
|
Dubay R, Urban JN, Darling EM. Single-Cell Microgels for Diagnostics and Therapeutics. ADVANCED FUNCTIONAL MATERIALS 2021; 31:2009946. [PMID: 36329867 PMCID: PMC9629779 DOI: 10.1002/adfm.202009946] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Indexed: 05/14/2023]
Abstract
Cell encapsulation within hydrogel droplets is transforming what is feasible in multiple fields of biomedical science such as tissue engineering and regenerative medicine, in vitro modeling, and cell-based therapies. Recent advances have allowed researchers to miniaturize material encapsulation complexes down to single-cell scales, where each complex, termed a single-cell microgel, contains only one cell surrounded by a hydrogel matrix while remaining <100 μm in size. With this achievement, studies requiring single-cell resolution are now possible, similar to those done using liquid droplet encapsulation. Of particular note, applications involving long-term in vitro cultures, modular bioinks, high-throughput screenings, and formation of 3D cellular microenvironments can be tuned independently to suit the needs of individual cells and experimental goals. In this progress report, an overview of established materials and techniques used to fabricate single-cell microgels, as well as insight into potential alternatives is provided. This focused review is concluded by discussing applications that have already benefited from single-cell microgel technologies, as well as prospective applications on the cusp of achieving important new capabilities.
Collapse
Affiliation(s)
- Ryan Dubay
- Center for Biomedical Engineering, Brown University, 175 Meeting St., Providence, RI 02912, USA
- Draper, 555 Technology Sq., Cambridge, MA 02139, USA
| | - Joseph N Urban
- Center for Biomedical Engineering, Brown University, 175 Meeting St., Providence, RI 02912, USA
| | - Eric M Darling
- Department of Molecular Pharmacology, Physiology, and Biotechnology, Center for Biomedical Engineering, School of Engineering, Department of Orthopaedics, Brown University, 175 Meeting St., Providence, RI 02912, USA
| |
Collapse
|
36
|
Huang W, Wang D, Yao YF. Understanding the pathogenesis of infectious diseases by single-cell RNA sequencing. MICROBIAL CELL 2021; 8:208-222. [PMID: 34527720 PMCID: PMC8404151 DOI: 10.15698/mic2021.09.759] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Revised: 07/13/2021] [Accepted: 07/21/2021] [Indexed: 12/17/2022]
Abstract
Infections are highly orchestrated and dynamic processes, which involve both pathogen and host. Transcriptional profiling at the single-cell level enables the analysis of cell diversity, heterogeneity of the immune response, and detailed molecular mechanisms underlying infectious diseases caused by bacteria, viruses, fungi, and parasites. Herein, we highlight recent remarkable advances in single-cell RNA sequencing (scRNA-seq) technologies and their applications in the investigation of host-pathogen interactions, current challenges and potential prospects for disease treatment are discussed as well. We propose that with the aid of scRNA-seq, the mechanism of infectious diseases will be further revealed thus inspiring the development of novel interventions and therapies.
Collapse
Affiliation(s)
- Wanqiu Huang
- Laboratory of Bacterial Pathogenesis, Department of Microbiology and Immunology, Institutes of Medical Sciences, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Danni Wang
- Laboratory of Bacterial Pathogenesis, Department of Microbiology and Immunology, Institutes of Medical Sciences, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Yu-Feng Yao
- Laboratory of Bacterial Pathogenesis, Department of Microbiology and Immunology, Institutes of Medical Sciences, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China.,Department of Infectious Diseases, Shanghai Ruijin Hospital, Shanghai 200025, China
| |
Collapse
|
37
|
Franks PW, Melén E, Friedman M, Sundström J, Kockum I, Klareskog L, Almqvist C, Bergen SE, Czene K, Hägg S, Hall P, Johnell K, Malarstig A, Catrina A, Hagström H, Benson M, Gustav Smith J, Gomez MF, Orho-Melander M, Jacobsson B, Halfvarson J, Repsilber D, Oresic M, Jern C, Melin B, Ohlsson C, Fall T, Rönnblom L, Wadelius M, Nordmark G, Johansson Å, Rosenquist R, Sullivan PF. Technological readiness and implementation of genomic-driven precision medicine for complex diseases. J Intern Med 2021; 290:602-620. [PMID: 34213793 DOI: 10.1111/joim.13330] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Revised: 03/21/2021] [Accepted: 04/12/2021] [Indexed: 12/20/2022]
Abstract
The fields of human genetics and genomics have generated considerable knowledge about the mechanistic basis of many diseases. Genomic approaches to diagnosis, prognostication, prevention and treatment - genomic-driven precision medicine (GDPM) - may help optimize medical practice. Here, we provide a comprehensive review of GDPM of complex diseases across major medical specialties. We focus on technological readiness: how rapidly a test can be implemented into health care. Although these areas of medicine are diverse, key similarities exist across almost all areas. Many medical areas have, within their standards of care, at least one GDPM test for a genetic variant of strong effect that aids the identification/diagnosis of a more homogeneous subset within a larger disease group or identifies a subset with different therapeutic requirements. However, for almost all complex diseases, the majority of patients do not carry established single-gene mutations with large effects. Thus, research is underway that seeks to determine the polygenic basis of many complex diseases. Nevertheless, most complex diseases are caused by the interplay of genetic, behavioural and environmental risk factors, which will likely necessitate models for prediction and diagnosis that incorporate genetic and non-genetic data.
Collapse
Affiliation(s)
- P W Franks
- From the, Department of Clinical Sciences, Lund University Diabetes Center, Lund University, Malmö, Sweden.,Department of Nutrition, Harvard School of Public Health, Boston, MA, USA
| | - E Melén
- Department of Clinical Science and Education Södersjukhuset, Karolinska Institutet, Stockholm, Sweden
| | - M Friedman
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
| | - J Sundström
- Department of Cardiology, Akademiska Sjukhuset, Uppsala, Sweden.,George Institute for Global Health, Camperdown, NSW, Australia.,Medical Sciences, Uppsala University, Uppsala, Sweden
| | - I Kockum
- Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden.,Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - L Klareskog
- Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden.,Department of Rheumatology, Karolinska Institutet, Stockholm, Sweden
| | - C Almqvist
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - S E Bergen
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - K Czene
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - S Hägg
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - P Hall
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden.,Department of Oncology, Södersjukhuset, Stockholm, Sweden
| | - K Johnell
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - A Malarstig
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden.,Pfizer, Worldwide Research and Development, Stockholm, Sweden
| | - A Catrina
- Department of Medicine, Karolinska Institutet, Stockholm, Sweden
| | - H Hagström
- Department of Medicine, Karolinska Institutet, Stockholm, Sweden.,Division of Hepatology, Department of Upper GI, Karolinska University Hospital, Stockholm, Sweden
| | - M Benson
- Department of Pediatrics, Linkopings Universitet, Linkoping, Sweden.,Division of Ear, Nose and Throat Diseases, Department of Clinical Sciences, Intervention and Technology (CLINTEC), Karolinska Institutet, Stockholm, Sweden
| | - J Gustav Smith
- Department of Cardiology and Wallenberg Center for Molecular Medicine, Clinical Sciences, Lund University and Skåne University Hospital, Lund, Sweden.,Department of Molecular and Clinical Medicine, Institute of Medicine, Gothenburg University and Sahlgrenska University Hospital, Gothenburg, Sweden
| | - M F Gomez
- From the, Department of Clinical Sciences, Lund University Diabetes Center, Lund University, Malmö, Sweden
| | - M Orho-Melander
- From the, Department of Clinical Sciences, Lund University Diabetes Center, Lund University, Malmö, Sweden
| | - B Jacobsson
- Division of Health Data and Digitalisation, Norwegian Institute of Public Health, Genetics and Bioinformatics, Oslo, Norway.,Department of Obstetrics and Gynecology, Region Västra Götaland, Sahlgrenska University Hospital, Gothenburg, Sweden.,Department of Obstetrics and Gynecology, Institute of Clinical Sciences, University of Gothenburg, Gothenburg, Sweden
| | - J Halfvarson
- School of Medical Sciences, Örebro University, Örebro, Sweden
| | - D Repsilber
- Functional Bioinformatics, Örebro University, Örebro, Sweden
| | - M Oresic
- School of Medical Sciences, Örebro University, Örebro, Sweden.,Turku Bioscience Centre, University of Turku and Åbo Akademi University, Turku, FI, Finland
| | - C Jern
- Department of Clinical Genetics and Genomics, Region Västra Götaland, Sahlgrenska University Hospital, Gothenburg, Sweden.,Department of Laboratory Medicine, Institute of Biomedicine, University of Gothenburg, Gothenburg, Sweden
| | - B Melin
- Department of Radiation Sciences, Oncology, Umeå Universitet, Umeå, Sweden
| | - C Ohlsson
- Department of Internal Medicine and Clinical Nutrition, Institute of Medicine, Sahlgrenska Osteoporosis Centre, CBAR, University of Gothenburg, Gothenburg, Sweden.,Department of Drug Treatment, Region Västra Götaland, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - T Fall
- Department of Medical Sciences, Molecular Epidemiology, Uppsala University, Uppsala, Sweden
| | - L Rönnblom
- Department of Medical Sciences, Rheumatology & Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - M Wadelius
- Department of Medical Sciences, Clinical Pharmacogenomics & Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - G Nordmark
- Department of Medical Sciences, Rheumatology & Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Å Johansson
- Institute for Immunology, Genetics, and Pathology, Uppsala University, Uppsala, Sweden
| | - R Rosenquist
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
| | - P F Sullivan
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden.,Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.,Department of Psychiatry, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| |
Collapse
|
38
|
Zhou A, Wu B, Yu H, Tang Y, Liu J, Jia Y, Yang X, Xiang L. Current Understanding of Osteoimmunology in Certain Osteoimmune Diseases. Front Cell Dev Biol 2021; 9:698068. [PMID: 34485284 PMCID: PMC8416088 DOI: 10.3389/fcell.2021.698068] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Accepted: 07/15/2021] [Indexed: 02/05/2023] Open
Abstract
The skeletal system and immune system seem to be two independent systems. However, there in fact are extensive and multiple crosstalk between them. The concept of osteoimmunology was created to describe those interdisciplinary events, but it has been constantly updated over time. In this review, we summarize the interactions between the skeletal and immune systems in the co-development of the two systems and the progress of certain typical bone abnormalities and bone regeneration on the cellular and molecular levels according to the mainstream novel study. At the end of the review, we also highlighted the possibility of extending the research scope of osteoimmunology to other systemic diseases. In conclusion, we propose that osteoimmunology is a promising perspective to uncover the mechanism of related diseases; meanwhile, a study from the point of view of osteoimmunology may also provide innovative ideas and resolutions to achieve the balance of internal homeostasis.
Collapse
Affiliation(s)
- Anqi Zhou
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Bingfeng Wu
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Hui Yu
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- Department of Oral Implantology, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Yufei Tang
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Jiayi Liu
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Yinan Jia
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Xiaoyu Yang
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Lin Xiang
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- Department of Oral Implantology, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| |
Collapse
|
39
|
Zielinski JM, Luke JJ, Guglietta S, Krieg C. High Throughput Multi-Omics Approaches for Clinical Trial Evaluation and Drug Discovery. Front Immunol 2021; 12:590742. [PMID: 33868223 PMCID: PMC8044891 DOI: 10.3389/fimmu.2021.590742] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2020] [Accepted: 03/01/2021] [Indexed: 12/12/2022] Open
Abstract
High throughput single cell multi-omics platforms, such as mass cytometry (cytometry by time-of-flight; CyTOF), high dimensional imaging (>6 marker; Hyperion, MIBIscope, CODEX, MACSima) and the recently evolved genomic cytometry (Citeseq or REAPseq) have enabled unprecedented insights into many biological and clinical questions, such as hematopoiesis, transplantation, cancer, and autoimmunity. In synergy with constantly adapting new single-cell analysis approaches and subsequent accumulating big data collections from these platforms, whole atlases of cell types and cellular and sub-cellular interaction networks are created. These atlases build an ideal scientific discovery environment for reference and data mining approaches, which often times reveals new cellular disease networks. In this review we will discuss how combinations and fusions of different -omic workflows on a single cell level can be used to examine cellular phenotypes, immune effector functions, and even dynamic changes, such as metabolomic state of different cells in a sample or even in a defined tissue location. We will touch on how pre-print platforms help in optimization and reproducibility of workflows, as well as community outreach. We will also shortly discuss how leveraging single cell multi-omic approaches can be used to accelerate cellular biomarker discovery during clinical trials to predict response to therapy, follow responsive cell types, and define novel druggable target pathways. Single cell proteome approaches already have changed how we explore cellular mechanism in disease and during therapy. Current challenges in the field are how we share these disruptive technologies to the scientific communities while still including new approaches, such as genomic cytometry and single cell metabolomics.
Collapse
Affiliation(s)
- Jessica M. Zielinski
- Hollings Cancer Center, Medical University of South Carolina (MUSC), Charleston, SC, United States
| | - Jason J. Luke
- Hillman Cancer Center, Department of Medicine, Division of Hematology/Oncology, University of Pittsburgh Medical Center, Pittsburgh, PA, United States
| | - Silvia Guglietta
- Hollings Cancer Center, Medical University of South Carolina (MUSC), Charleston, SC, United States
| | - Carsten Krieg
- Hollings Cancer Center, Medical University of South Carolina (MUSC), Charleston, SC, United States
| |
Collapse
|
40
|
scGNN is a novel graph neural network framework for single-cell RNA-Seq analyses. Nat Commun 2021; 12:1882. [PMID: 33767197 PMCID: PMC7994447 DOI: 10.1038/s41467-021-22197-x] [Citation(s) in RCA: 112] [Impact Index Per Article: 37.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Accepted: 02/24/2021] [Indexed: 12/22/2022] Open
Abstract
Single-cell RNA-sequencing (scRNA-Seq) is widely used to reveal the heterogeneity and dynamics of tissues, organisms, and complex diseases, but its analyses still suffer from multiple grand challenges, including the sequencing sparsity and complex differential patterns in gene expression. We introduce the scGNN (single-cell graph neural network) to provide a hypothesis-free deep learning framework for scRNA-Seq analyses. This framework formulates and aggregates cell–cell relationships with graph neural networks and models heterogeneous gene expression patterns using a left-truncated mixture Gaussian model. scGNN integrates three iterative multi-modal autoencoders and outperforms existing tools for gene imputation and cell clustering on four benchmark scRNA-Seq datasets. In an Alzheimer’s disease study with 13,214 single nuclei from postmortem brain tissues, scGNN successfully illustrated disease-related neural development and the differential mechanism. scGNN provides an effective representation of gene expression and cell–cell relationships. It is also a powerful framework that can be applied to general scRNA-Seq analyses. Single-cell RNA-Seq suffers from heterogeneity in sequencing sparsity and complex differential patterns in gene expression. Here, the authors introduce a graph neural network based on a hypothesis-free deep learning framework as an effective representation of gene expression and cell–cell relationships.
Collapse
|
41
|
Epigenomic and transcriptomic analysis of chronic inflammatory diseases. Genes Genomics 2021; 43:227-236. [PMID: 33638813 DOI: 10.1007/s13258-021-01045-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Accepted: 01/06/2021] [Indexed: 12/13/2022]
Abstract
Chronic inflammatory diseases (CIDs) have complex pathologies that result from aberrant and persistent immune responses. However, the precise triggers and mechanisms remain elusive. An important aspect of CID research focuses on epigenetics modifications, which regulate gene expression and provide a dynamic transcriptional response to inflammation. In recent years, mounting evidence has demonstrated an association between epigenomic and transcriptomic dysregulation and the phenotypes of CIDs. In particular, epigenetic changes at cis-regulatory elements have provided new insights for immune cell-specific alterations that contribute to disease etiology. Furthermore, the advancements in single-cell genomics provide novel solutions to cell type heterogeneity, which has long posed challenges for CID diagnosis and treatment. In this review, we discuss the current state of epigenomics research of CID and the insights derived from single-cell transcriptomic and epigenomic studies.
Collapse
|
42
|
López-Cortés A, Guevara-Ramírez P, Kyriakidis NC, Barba-Ostria C, León Cáceres Á, Guerrero S, Ortiz-Prado E, Munteanu CR, Tejera E, Cevallos-Robalino D, Gómez-Jaramillo AM, Simbaña-Rivera K, Granizo-Martínez A, Pérez-M G, Moreno S, García-Cárdenas JM, Zambrano AK, Pérez-Castillo Y, Cabrera-Andrade A, Puig San Andrés L, Proaño-Castro C, Bautista J, Quevedo A, Varela N, Quiñones LA, Paz-y-Miño C. In silico Analyses of Immune System Protein Interactome Network, Single-Cell RNA Sequencing of Human Tissues, and Artificial Neural Networks Reveal Potential Therapeutic Targets for Drug Repurposing Against COVID-19. Front Pharmacol 2021; 12:598925. [PMID: 33716737 PMCID: PMC7952300 DOI: 10.3389/fphar.2021.598925] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Accepted: 01/11/2021] [Indexed: 12/15/2022] Open
Abstract
Background: There is pressing urgency to identify therapeutic targets and drugs that allow treating COVID-19 patients effectively. Methods: We performed in silico analyses of immune system protein interactome network, single-cell RNA sequencing of human tissues, and artificial neural networks to reveal potential therapeutic targets for drug repurposing against COVID-19. Results: We screened 1,584 high-confidence immune system proteins in ACE2 and TMPRSS2 co-expressing cells, finding 25 potential therapeutic targets significantly overexpressed in nasal goblet secretory cells, lung type II pneumocytes, and ileal absorptive enterocytes of patients with several immunopathologies. Then, we performed fully connected deep neural networks to find the best multitask classification model to predict the activity of 10,672 drugs, obtaining several approved drugs, compounds under investigation, and experimental compounds with the highest area under the receiver operating characteristics. Conclusion: After being effectively analyzed in clinical trials, these drugs can be considered for treatment of severe COVID-19 patients. Scripts can be downloaded at https://github.com/muntisa/immuno-drug-repurposing-COVID-19.
Collapse
Affiliation(s)
- Andrés López-Cortés
- Centro de Investigación Genética y Genómica, Facultad de Ciencias de la Salud Eugenio Espejo, Universidad UTE, Quito, Ecuador
- RNASA-IMEDIR, Computer Science Faculty, University of A Coruna, A Coruña, Spain
- Latin American Network for the Implementation and Validation of Clinical Pharmacogenomics Guidelines (RELIVAF-CYTED), Madrid, Spain
| | - Patricia Guevara-Ramírez
- Centro de Investigación Genética y Genómica, Facultad de Ciencias de la Salud Eugenio Espejo, Universidad UTE, Quito, Ecuador
| | - Nikolaos C. Kyriakidis
- One Health Research Group, Faculty of Medicine, Universidad de Las Américas (UDLA), Quito, Ecuador
| | - Carlos Barba-Ostria
- One Health Research Group, Faculty of Medicine, Universidad de Las Américas (UDLA), Quito, Ecuador
| | - Ángela León Cáceres
- Heidelberg Institute of Global Health, Faculty of Medicine, Heidelberg University, Heidelberg, Germany
- Instituto de Salud Pública, Facultad de Medicina, Pontificia Universidad Católica del Ecuador, Quito, Ecuador
- Tropical Herping, Quito, Ecuador
| | - Santiago Guerrero
- Centro de Investigación Genética y Genómica, Facultad de Ciencias de la Salud Eugenio Espejo, Universidad UTE, Quito, Ecuador
| | - Esteban Ortiz-Prado
- One Health Research Group, Faculty of Medicine, Universidad de Las Américas (UDLA), Quito, Ecuador
| | - Cristian R. Munteanu
- RNASA-IMEDIR, Computer Science Faculty, University of A Coruna, A Coruña, Spain
- Biomedical Research Institute of A Coruna (INIBIC), University Hospital Complex of A Coruna (CHUAC), A Coruña, Spain
- Centro de Información en Tecnologías de la Información y las Comunicaciones (CITIC), A Coruña, Spain
| | - Eduardo Tejera
- Grupo de Bio-Quimioinformática, Universidad de Las Américas (UDLA), Quito, Ecuador
| | | | | | - Katherine Simbaña-Rivera
- One Health Research Group, Faculty of Medicine, Universidad de Las Américas (UDLA), Quito, Ecuador
| | - Adriana Granizo-Martínez
- Carrera de Medicina, Facultad de Ciencias de la Salud Eugenio Espejo, Universidad UTE, Quito, Ecuador
| | - Gabriela Pérez-M
- Centro Clínico Quirúrgico Ambulatorio Hospital del Día El Batán, Instituto Ecuatoriano de Seguridad Social, Quito, Ecuador
| | - Silvana Moreno
- Department of Plant Biology, Faculty of Natural Resources and Agricultural Sciences, Swedish University of Agricultural Sciences, Uppsala, Sweden
| | - Jennyfer M. García-Cárdenas
- Centro de Investigación Genética y Genómica, Facultad de Ciencias de la Salud Eugenio Espejo, Universidad UTE, Quito, Ecuador
| | - Ana Karina Zambrano
- Centro de Investigación Genética y Genómica, Facultad de Ciencias de la Salud Eugenio Espejo, Universidad UTE, Quito, Ecuador
- Biomedical Research Institute of A Coruna (INIBIC), University Hospital Complex of A Coruna (CHUAC), A Coruña, Spain
| | | | - Alejandro Cabrera-Andrade
- RNASA-IMEDIR, Computer Science Faculty, University of A Coruna, A Coruña, Spain
- Grupo de Bio-Quimioinformática, Universidad de Las Américas (UDLA), Quito, Ecuador
| | - Lourdes Puig San Andrés
- Centro de Investigación Genética y Genómica, Facultad de Ciencias de la Salud Eugenio Espejo, Universidad UTE, Quito, Ecuador
| | | | - Jhommara Bautista
- Facultad de Ingeniería y Ciencias Aplicadas-Biotecnología, Universidad de Las Américas, Quito, Ecuador
| | - Andreina Quevedo
- Centro de Investigación Genética y Genómica, Facultad de Ciencias de la Salud Eugenio Espejo, Universidad UTE, Quito, Ecuador
| | - Nelson Varela
- Latin American Network for the Implementation and Validation of Clinical Pharmacogenomics Guidelines (RELIVAF-CYTED), Madrid, Spain
- Laboratory of Chemical Carcinogenesis and Pharmacogenetics, Department of Basic-Clinical Oncology, Faculty of Medicine, University of Chile, Santiago, Chile
| | - Luis Abel Quiñones
- Latin American Network for the Implementation and Validation of Clinical Pharmacogenomics Guidelines (RELIVAF-CYTED), Madrid, Spain
- Laboratory of Chemical Carcinogenesis and Pharmacogenetics, Department of Basic-Clinical Oncology, Faculty of Medicine, University of Chile, Santiago, Chile
| | - César Paz-y-Miño
- Centro de Investigación Genética y Genómica, Facultad de Ciencias de la Salud Eugenio Espejo, Universidad UTE, Quito, Ecuador
| |
Collapse
|
43
|
Nayak R, Hasija Y. A hitchhiker's guide to single-cell transcriptomics and data analysis pipelines. Genomics 2021; 113:606-619. [PMID: 33485955 DOI: 10.1016/j.ygeno.2021.01.007] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2020] [Revised: 12/30/2020] [Accepted: 01/18/2021] [Indexed: 12/20/2022]
Abstract
Single-cell transcriptomics (SCT) is a tour de force in the era of big omics data that has led to the accumulation of massive cellular transcription data at an astounding resolution of single cells. It provides valuable insights into cells previously unachieved by bulk cell analysis and is proving crucial in uncovering cellular heterogeneity, identifying rare cell populations, distinct cell-lineage trajectories, and mechanisms involved in complex cellular processes. SCT data is highly complex and necessitates advanced statistical and computational methods for analysis. This review provides a comprehensive overview of the steps in a typical SCT workflow, starting from experimental protocol to data analysis, deliberating various pipelines used. We discuss recent trends, challenges, machine learning methods for data analysis, and future prospects. We conclude by listing the multitude of scRNA-seq data applications and how it shall revolutionize our understanding of cellular biology and diseases.
Collapse
Affiliation(s)
- Richa Nayak
- Department of Biotechnology, Delhi Technological University, Delhi 110042, India
| | - Yasha Hasija
- Department of Biotechnology, Delhi Technological University, Delhi 110042, India.
| |
Collapse
|
44
|
Shukla R, Henkel ND, Alganem K, Hamoud AR, Reigle J, Alnafisah RS, Eby HM, Imami AS, Creeden JF, Miruzzi SA, Meller J, Mccullumsmith RE. Signature-based approaches for informed drug repurposing: targeting CNS disorders. Neuropsychopharmacology 2021; 46:116-130. [PMID: 32604402 PMCID: PMC7688959 DOI: 10.1038/s41386-020-0752-6] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/08/2020] [Revised: 05/30/2020] [Accepted: 06/22/2020] [Indexed: 12/15/2022]
Abstract
CNS disorders, and in particular psychiatric illnesses, lack definitive disease-altering therapeutics. The limited understanding of the mechanisms driving these illnesses with the slow pace and high cost of drug development exacerbates this issue. For these reasons, drug repurposing - both a less expensive and time-efficient practice compared to de novo drug development - has been a promising strategy to overcome the paucity of treatments available for these debilitating disorders. While empirical drug-repurposing has been a routine practice in clinical psychiatry, innovative, informed, and cost-effective repurposing efforts using big data ("omics") have been designed to characterize drugs by structural and transcriptomic signatures. These strategies, in conjunction with ontological integration, provide an important opportunity to address knowledge-based challenges associated with drug development for CNS disorders. In this review, we discuss various signature-based in silico approaches to drug repurposing, its integration with multiple omics platforms, and how this data can be used for clinically relevant, evidence-based drug repurposing. These tools provide an exciting translational avenue to merge omics-based drug discovery platforms with patient-specific disease signatures, ultimately facilitating the identification of new therapies for numerous psychiatric disorders.
Collapse
Affiliation(s)
- Rammohan Shukla
- Department of Neurosciences, University of Toledo, Toledo, OH, USA.
| | | | - Khaled Alganem
- Department of Neurosciences, University of Toledo, Toledo, OH, USA
| | | | - James Reigle
- Division of Biomedical Informatics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | | | - Hunter M Eby
- Department of Neurosciences, University of Toledo, Toledo, OH, USA
| | - Ali S Imami
- Department of Neurosciences, University of Toledo, Toledo, OH, USA
| | - Justin F Creeden
- Department of Neurosciences, University of Toledo, Toledo, OH, USA
| | - Scott A Miruzzi
- Department of Neurosciences, University of Toledo, Toledo, OH, USA
| | - Jaroslaw Meller
- Division of Biomedical Informatics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
- Department of Cancer Biology, University of Cincinnati College of Medicine, Cincinnati, OH, 45267, USA
- Department of Environmental Health, University of Cincinnati College of Medicine, Cincinnati, OH, USA
- Department of Electrical Engineering and Computing Systems, University of Cincinnati College of Medicine, Cincinnati, OH, USA
- Department of Informatics, Nicolaus Copernicus University, Torun, Poland
| | - Robert E Mccullumsmith
- Department of Neurosciences, University of Toledo, Toledo, OH, USA
- Neurosciences Institute, ProMedica, Toledo, OH, USA
| |
Collapse
|
45
|
de Weerd HA, Badam TVS, Martínez-Enguita D, Åkesson J, Muthas D, Gustafsson M, Lubovac-Pilav Z. MODifieR: an Ensemble R Package for Inference of Disease Modules from Transcriptomics Networks. Bioinformatics 2020; 36:3918-3919. [PMID: 32271876 DOI: 10.1093/bioinformatics/btaa235] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2019] [Revised: 03/27/2020] [Accepted: 04/02/2020] [Indexed: 12/24/2022] Open
Abstract
MOTIVATION Complex diseases are due to the dense interactions of many disease-associated factors that dysregulate genes that in turn form the so-called disease modules, which have shown to be a powerful concept for understanding pathological mechanisms. There exist many disease module inference methods that rely on somewhat different assumptions, but there is still no gold standard or best-performing method. Hence, there is a need for combining these methods to generate robust disease modules. RESULTS We developed MODule IdentiFIER (MODifieR), an ensemble R package of nine disease module inference methods from transcriptomics networks. MODifieR uses standardized input and output allowing the possibility to combine individual modules generated from these methods into more robust disease-specific modules, contributing to a better understanding of complex diseases. AVAILABILITY AND IMPLEMENTATION MODifieR is available under the GNU GPL license and can be freely downloaded from https://gitlab.com/Gustafsson-lab/MODifieR and as a Docker image from https://hub.docker.com/r/ddeweerd/modifier. SUPPLEMENTARY INFORMATION Supplementary data are available at Bioinformatics online.
Collapse
Affiliation(s)
- Hendrik A de Weerd
- School of Bioscience, Systems Biology Research Center, Skövde 541 45, Sweden.,Department of Physics, Chemistry and Biology, Linköping University, Linköping 581 83, Sweden
| | - Tejaswi V S Badam
- School of Bioscience, Systems Biology Research Center, Skövde 541 45, Sweden.,Department of Physics, Chemistry and Biology, Linköping University, Linköping 581 83, Sweden
| | - David Martínez-Enguita
- Department of Physics, Chemistry and Biology, Linköping University, Linköping 581 83, Sweden
| | - Julia Åkesson
- School of Bioscience, Systems Biology Research Center, Skövde 541 45, Sweden.,Department of Physics, Chemistry and Biology, Linköping University, Linköping 581 83, Sweden
| | - Daniel Muthas
- Translational Science and Experimental Medicine, Early Respiratory, Inflammation and Autoimmunity, BioPharmaceuticals R&D, AstraZeneca, Mölndal 43183, Sweden
| | - Mika Gustafsson
- Department of Physics, Chemistry and Biology, Linköping University, Linköping 581 83, Sweden
| | | |
Collapse
|
46
|
Li J, Jiang W, Han H, Liu J, Liu B, Wang Y. ScGSLC: An unsupervised graph similarity learning framework for single-cell RNA-seq data clustering. Comput Biol Chem 2020; 90:107415. [PMID: 33307360 DOI: 10.1016/j.compbiolchem.2020.107415] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Revised: 09/30/2020] [Accepted: 10/06/2020] [Indexed: 01/18/2023]
Abstract
Accurate clustering of cells from single-cell RNA sequencing (scRNA-seq) data is an essential step for biological analysis such as putative cell type identification. However, scRNA-seq data has high dimension and high sparsity, which makes traditional clustering methods less effective to reflect the similarity between cells. Since genetic network fundamentally defines the functions of cell and deep learning shows strong advantages in network representation learning, we propose a novel scRNA-seq clustering framework ScGSLC based on graph similarity learning. ScGSLC effectively integrates scRNA-seq data and protein-protein interaction network to a graph. Then graph convolution network is employed by ScGSLC to embedding graph and clustering the cells by the calculated similarity between graphs. Unsupervised clustering results of nine public data sets demonstrate that ScGSLC shows better performance than the state-of-the-art methods.
Collapse
Affiliation(s)
- Junyi Li
- School of Computer Science and Technology, Harbin Institute of Technology (Shenzhen), Shenzhen, Guangdong 518055, China.
| | - Wei Jiang
- School of Computer Science and Technology, Harbin Institute of Technology (Shenzhen), Shenzhen, Guangdong 518055, China
| | - Henry Han
- Department of Computer and Information Science, Fordham University, New York, NY 10023, USA; School of Computer Science, Qinghai Normal University, Xining 810008, China
| | - Jing Liu
- South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, Guangdong 510530, China
| | - Bo Liu
- Center for Bioinformatics, School of Computer Science and Technology, Harbin Institute of Technology, Harbin, Heilongjiang 150001, China
| | - Yadong Wang
- School of Computer Science and Technology, Harbin Institute of Technology (Shenzhen), Shenzhen, Guangdong 518055, China; Center for Bioinformatics, School of Computer Science and Technology, Harbin Institute of Technology, Harbin, Heilongjiang 150001, China.
| |
Collapse
|
47
|
González-Serna D, Villanueva-Martin G, Acosta-Herrera M, Márquez A, Martín J. Approaching Shared Pathophysiology in Immune-Mediated Diseases through Functional Genomics. Genes (Basel) 2020; 11:E1482. [PMID: 33317201 PMCID: PMC7762979 DOI: 10.3390/genes11121482] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Revised: 12/01/2020] [Accepted: 12/04/2020] [Indexed: 12/14/2022] Open
Abstract
Immune-mediated diseases (IMDs) are complex pathologies that are strongly influenced by environmental and genetic factors. Associations between genetic loci and susceptibility to these diseases have been widely studied, and hundreds of risk variants have emerged during the last two decades, with researchers observing a shared genetic pattern among them. Nevertheless, the pathological mechanism behind these associations remains a challenge that has just started to be understood thanks to functional genomic approaches. Transcriptomics, regulatory elements, chromatin interactome, as well as the experimental characterization of genomic findings, constitute key elements in the emerging understandings of how genetics affects the etiopathogenesis of IMDs. In this review, we will focus on the latest advances in the field of functional genomics, centering our attention on systemic rheumatic IMDs.
Collapse
Affiliation(s)
- David González-Serna
- Institute of Parasitology and Biomedicine López-Neyra, Consejo Superior de Investigaciones Científicas (IPBLN-CSIC), 18016 Granada, Spain; (D.G.-S.); (G.V.-M.); (M.A.-H.); (A.M.)
| | - Gonzalo Villanueva-Martin
- Institute of Parasitology and Biomedicine López-Neyra, Consejo Superior de Investigaciones Científicas (IPBLN-CSIC), 18016 Granada, Spain; (D.G.-S.); (G.V.-M.); (M.A.-H.); (A.M.)
| | - Marialbert Acosta-Herrera
- Institute of Parasitology and Biomedicine López-Neyra, Consejo Superior de Investigaciones Científicas (IPBLN-CSIC), 18016 Granada, Spain; (D.G.-S.); (G.V.-M.); (M.A.-H.); (A.M.)
| | - Ana Márquez
- Institute of Parasitology and Biomedicine López-Neyra, Consejo Superior de Investigaciones Científicas (IPBLN-CSIC), 18016 Granada, Spain; (D.G.-S.); (G.V.-M.); (M.A.-H.); (A.M.)
- Systemic Autoimmune Disease Unit, Hospital Clínico San Cecilio, Instituto de Investigación Biosanitaria ibs.GRANADA, 18016 Granada, Spain
| | - Javier Martín
- Institute of Parasitology and Biomedicine López-Neyra, Consejo Superior de Investigaciones Científicas (IPBLN-CSIC), 18016 Granada, Spain; (D.G.-S.); (G.V.-M.); (M.A.-H.); (A.M.)
| |
Collapse
|
48
|
Uyar B, Palmer D, Kowald A, Murua Escobar H, Barrantes I, Möller S, Akalin A, Fuellen G. Single-cell analyses of aging, inflammation and senescence. Ageing Res Rev 2020; 64:101156. [PMID: 32949770 PMCID: PMC7493798 DOI: 10.1016/j.arr.2020.101156] [Citation(s) in RCA: 93] [Impact Index Per Article: 23.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2020] [Revised: 08/23/2020] [Accepted: 08/25/2020] [Indexed: 02/09/2023]
Abstract
Single-cell gene expression (transcriptomics) data are becoming robust and abundant, and are increasingly used to track organisms along their life-course. This allows investigation into how aging affects cellular transcriptomes, and how changes in transcriptomes may underlie aging, including chronic inflammation (inflammaging), immunosenescence and cellular senescence. We compiled and tabulated aging-related single-cell datasets published to date, collected and discussed relevant findings, and inspected some of these datasets ourselves. We specifically note insights that cannot (or not easily) be based on bulk data. For example, in some datasets, the fraction of cells expressing p16 (CDKN2A), one of the most prominent markers of cellular senescence, was reported to increase, in addition to its upregulated mean expression over all cells. Moreover, we found evidence for inflammatory processes in most datasets, some of these driven by specific cells of the immune system. Further, single-cell data are specifically useful to investigate whether transcriptional heterogeneity (also called noise or variability) increases with age, and many (but not all) studies in our review report an increase in such heterogeneity. Finally, we demonstrate some stability of marker gene expression patterns across closely similar studies and suggest that single-cell experiments may hold the key to provide detailed insights whenever interventions (countering aging, inflammation, senescence, disease, etc.) are affecting cells depending on cell type.
Collapse
Affiliation(s)
- Bora Uyar
- Bioinformatics and Omics Data Science Platform, Berlin Institute for Medical Systems Biology, Max Delbrück Center for Molecular Medicine, Berlin, Germany
| | - Daniel Palmer
- Rostock University Medical Center, Institute for Biostatistics and Informatics in Medicine and Aging Research, Rostock, Germany
| | - Axel Kowald
- Rostock University Medical Center, Institute for Biostatistics and Informatics in Medicine and Aging Research, Rostock, Germany
| | - Hugo Murua Escobar
- Rostock University Medical Center, Department of Hematology, Oncology and Palliative Medicine, Department of Medicine III, Rostock, Germany
| | - Israel Barrantes
- Rostock University Medical Center, Institute for Biostatistics and Informatics in Medicine and Aging Research, Rostock, Germany
| | - Steffen Möller
- Rostock University Medical Center, Institute for Biostatistics and Informatics in Medicine and Aging Research, Rostock, Germany
| | - Altuna Akalin
- Bioinformatics and Omics Data Science Platform, Berlin Institute for Medical Systems Biology, Max Delbrück Center for Molecular Medicine, Berlin, Germany
| | - Georg Fuellen
- Rostock University Medical Center, Institute for Biostatistics and Informatics in Medicine and Aging Research, Rostock, Germany.
| |
Collapse
|
49
|
Zhao M, Jiang J, Zhao M, Chang C, Wu H, Lu Q. The Application of Single-Cell RNA Sequencing in Studies of Autoimmune Diseases: a Comprehensive Review. Clin Rev Allergy Immunol 2020; 60:68-86. [PMID: 33236283 DOI: 10.1007/s12016-020-08813-6] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/21/2020] [Indexed: 01/15/2023]
Abstract
Complex composition is one of the most important features of the immune system, involving many types of organs, tissues, cells, and molecules that perform immune functions. The normal function of each component of the immune system is the guarantee for maintaining the relatively stable immune function of the body. When the self-immune tolerance mechanism of the body is unregulated or destroyed, the immune system reacts to autoantigens, resulting in damage to self-tissues and organs or an immunopathological state with abnormal functions. Autoimmune diseases are diverse, and their pathogenesis is complicated. Various immune cells and their interactions play significant roles in the occurrence and development of diseases. The solution to heterogeneity of immune cells is the basic science and translational understanding of how genes and the environment interact to induce disease so that we can develop personalized medicine, a goal that has to this point eluded scientists. Single-cell RNA sequencing (scRNA-Seq) refers to a new technique allowing high-throughput sequencing analysis of the whole transcriptome to reveal the gene expression status of individual cells. It has emerged as an indispensable tool in the field of life science research, and can help identify the complex mechanism of cell heterogeneity, discover new cell subsets, and help uncover the molecular mechanisms of pathogenesis, the evolution of disorders, and drug resistance. This information can provide us with new strategies for diagnosis and prognostic evaluation, as well as monitoring treatment responses. In this review, we summarize the crucial experimental procedures used for single-cell RNA sequencing, and the current applications of this technique to study autoimmune diseases are described in detail. This technique will be widely used in more in-depth studies of autoimmune diseases and will contribute to the diagnosis and therapies of these disorders.
Collapse
Affiliation(s)
- Mingming Zhao
- Department of Dermatology, the Second Xiangya Hospital, Central South University, 410011, Changsha, Hunan, People's Republic of China.,Hunan Key Laboratory of Medical Epigenomics, 410011, Changsha, Hunan, People's Republic of China
| | - Jiao Jiang
- Department of Dermatology, the Second Xiangya Hospital, Central South University, 410011, Changsha, Hunan, People's Republic of China.,Hunan Key Laboratory of Medical Epigenomics, 410011, Changsha, Hunan, People's Republic of China
| | - Ming Zhao
- Department of Dermatology, the Second Xiangya Hospital, Central South University, 410011, Changsha, Hunan, People's Republic of China.,Hunan Key Laboratory of Medical Epigenomics, 410011, Changsha, Hunan, People's Republic of China
| | - Christopher Chang
- Division of Pediatric Immunology and Allergy, Joe DiMaggio Children's Hospital, Hollywood, FL, 33021, USA.,Division of Rheumatology, Allergy and Clinical Immunology, University of California, Davis, CA, 95616, USA
| | - Haijing Wu
- Department of Dermatology, the Second Xiangya Hospital, Central South University, 410011, Changsha, Hunan, People's Republic of China. .,Hunan Key Laboratory of Medical Epigenomics, 410011, Changsha, Hunan, People's Republic of China.
| | - Qianjin Lu
- Department of Dermatology, the Second Xiangya Hospital, Central South University, 410011, Changsha, Hunan, People's Republic of China. .,Hunan Key Laboratory of Medical Epigenomics, 410011, Changsha, Hunan, People's Republic of China.
| |
Collapse
|
50
|
Rahem SM, Epsi NJ, Coffman FD, Mitrofanova A. Genome-wide analysis of therapeutic response uncovers molecular pathways governing tamoxifen resistance in ER+ breast cancer. EBioMedicine 2020; 61:103047. [PMID: 33099086 PMCID: PMC7585053 DOI: 10.1016/j.ebiom.2020.103047] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2020] [Revised: 09/02/2020] [Accepted: 09/18/2020] [Indexed: 01/10/2023] Open
Abstract
Background Prioritization of breast cancer patients based on the risk of resistance to tamoxifen plays a significant role in personalized therapeutic planning and improving disease course and outcomes. Methods In this work, we demonstrate that a genome-wide pathway-centric computational framework elucidates molecular pathways as markers of tamoxifen resistance in ER+ breast cancer patients. In particular, we associated activity levels of molecular pathways with a wide spectrum of response to tamoxifen, which defined markers of tamoxifen resistance in patients with ER+ breast cancer. Findings We identified five biological pathways as markers of tamoxifen failure and demonstrated their ability to predict the risk of tamoxifen resistance in two independent patient cohorts (Test cohort1: log-rank p-value = 0.02, adjusted HR = 3.11; Test cohort2: log-rank p-value = 0.01, adjusted HR = 4.24). We have shown that these pathways are not markers of aggressiveness and outperform known markers of tamoxifen response. Furthermore, for adoption into clinic, we derived a list of pathway read-out genes and their associated scoring system, which assigns a risk of tamoxifen resistance for new incoming patients. Interpretation We propose that the identified pathways and their read-out genes can be utilized to prioritize patients who would benefit from tamoxifen treatment and patients at risk of tamoxifen resistance that should be offered alternative regimens. Funding This work was supported by the Rutgers SHP Dean's research grant, Rutgers start-up funds, Libyan Ministry of Higher Education and Scientific Research, and Katrina Kehlet Graduate Award from The NJ Chapter of the Healthcare Information Management Systems Society.
Collapse
Affiliation(s)
- Sarra M Rahem
- Department of Biomedical and Health Informatics, Rutgers School of Health Professions, Rutgers Biomedical and Health Sciences, USA
| | - Nusrat J Epsi
- Department of Biomedical and Health Informatics, Rutgers School of Health Professions, Rutgers Biomedical and Health Sciences, USA
| | - Frederick D Coffman
- Department of Biomedical and Health Informatics, Rutgers School of Health Professions, Rutgers Biomedical and Health Sciences, USA; Department of Physician Assistant Studies and Practice, USA; Department of Pathology & Laboratory Medicine, New Jersey Medical School, Newark, New Jersey 07107, USA
| | - Antonina Mitrofanova
- Department of Biomedical and Health Informatics, Rutgers School of Health Professions, Rutgers Biomedical and Health Sciences, USA; Rutgers Cancer Institute of New Jersey, Rutgers, The State University of New Jersey, New Brunswick, New Jersey 08901, USA.
| |
Collapse
|