1
|
Liu T, Liu C, Li Q, Zheng X, Zou F. ARTdeConv: adaptive regularized tri-factor non-negative matrix factorization for cell type deconvolution. NAR Genom Bioinform 2025; 7:lqaf046. [PMID: 40290316 PMCID: PMC12034106 DOI: 10.1093/nargab/lqaf046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Revised: 03/02/2025] [Accepted: 04/14/2025] [Indexed: 04/30/2025] Open
Abstract
Accurate deconvolution of cell types from bulk gene expression is crucial for understanding cellular compositions and uncovering cell-type specific differential expression and physiological states of diseased tissues. Existing deconvolution methods have limitations, such as requiring complete cellular gene expression signatures or neglecting partial biological information. Moreover, these methods often overlook varying cell-type messenger RNA amounts, leading to biased proportion estimates. Additionally, they do not effectively utilize valuable reference information from external studies, such as means and ranges of population cell-type proportions. To address these challenges, we introduce an adaptive regularized tri-factor non-negative matrix factorization approach for deconvolution (ARTdeConv). We rigorously establish the numerical convergence of our algorithm. Through benchmark simulations, we demonstrate the superior performance of ARTdeConv compared to state-of-the-art semi-reference-based and reference-free methods as well as its robustness under challenges to its assumptions. In a real-world application to a dataset from a trivalent influenza vaccine study, our method accurately estimates cellular proportions, as evidenced by the nearly perfect Pearson's correlation between ARTdeConv estimates and flow cytometry measurements. Moreover, our analysis of ARTdeConv estimates in COVID-19 patients reveals patterns consistent with important immunological phenomena observed in other studies. The proposed method, ARTdeConv, is implemented as an R package and can be accessed on GitHub for researchers and practitioners.
Collapse
Affiliation(s)
- Tianyi Liu
- Department of Biostatistics, The University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Chuwen Liu
- Department of Biostatistics, The University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Quefeng Li
- Department of Biostatistics, The University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Xiaojing Zheng
- Department of Biostatistics, The University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
- Department of Pediatrics, The University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Fei Zou
- Department of Biostatistics, The University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
- Department of Genetics, The University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| |
Collapse
|
2
|
Huang J, Du Y, Kelly KR, Lv J, Fan Y, Zhong JF, Sun F. DeepDeconUQ estimates malignant cell fraction prediction intervals in bulk RNA-seq tissue. PLoS Comput Biol 2025; 21:e1013133. [PMID: 40465796 PMCID: PMC12162100 DOI: 10.1371/journal.pcbi.1013133] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2024] [Revised: 06/12/2025] [Accepted: 05/13/2025] [Indexed: 06/16/2025] Open
Abstract
Accurate estimation of malignant cell fractions in tissues plays a critical role in cancer diagnosis, prognosis, and subsequent treatment decisions. However, most currently available methods provide only point estimates, neglecting the quantification of uncertainties, which is essential for both clinical and research applications. This study introduces DeepDeconUQ, a deep neural network model developed to estimate prediction intervals for malignant cell fractions based on bulk RNA-seq data. This approach addresses limitations in current malignant cell fraction estimation methods by integrating uncertainty quantification into predictions of cancer cell fractions. DeepDeconUQ leverages single-cell RNA sequencing (scRNA-seq) data in conjunction with conformalized quantile regression to produce reliable prediction intervals. The model trains a quantile regression neural network to establish upper and lower bounds for cancer cell proportions, followed by a calibration step that refines these intervals to ensure both statistical validity (coverage probability) and discrimination (narrow intervals). Benchmark analyses indicate that DeepDeconUQ consistently surpasses existing methods, achieving high coverage accuracy with tight prediction intervals across simulated and real cancer datasets. The robustness of DeepDeconUQ is further demonstrated by its resilience to various gene expression perturbations. The DeepDeconUQ method is publicly accessible at https://github.com/jiaweih14/DeepDeconUQ.
Collapse
Affiliation(s)
- Jiawei Huang
- Department of Quantitative and Computational Biology, University of Southern California, Los Angeles, California, United States of America
| | - Yuxuan Du
- Department of Quantitative and Computational Biology, University of Southern California, Los Angeles, California, United States of America
- Department of Electrical and Computer Engineering, University of Texas at San Antonio, San Antonio, Texas, United States of America
| | - Kevin R. Kelly
- Division of Hematology, University of Southern California, Los Angeles, California, United States of America
| | - Jinchi Lv
- Data Sciences and Operations Department, University of Southern California, Los Angeles, California, United States of America
| | - Yingying Fan
- Data Sciences and Operations Department, University of Southern California, Los Angeles, California, United States of America
| | - Jiang F. Zhong
- Department of Basic Sciences, School of Medicine, Loma Linda University, Loma Linda, California, United States of America
| | - Fengzhu Sun
- Department of Quantitative and Computational Biology, University of Southern California, Los Angeles, California, United States of America
| |
Collapse
|
3
|
Li J, Yang Y, Yi Z, Zhu Y, Yang H, Chen B, Lobie PE, Ma S. Microdroplet-Engineered Skeletal Muscle Organoids from Primary Tissue Recapitulate Parental Physiology with High Reproducibility. RESEARCH (WASHINGTON, D.C.) 2025; 8:0699. [PMID: 40375923 PMCID: PMC12078942 DOI: 10.34133/research.0699] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/12/2025] [Revised: 04/14/2025] [Accepted: 04/18/2025] [Indexed: 05/18/2025]
Abstract
Achieving high maturity and functionality in in vitro skeletal muscle models is essential for advancing our understanding of muscle biology, disease mechanisms, and drug discovery. However, current models struggle to fully recapitulate key features such as sarcomere structure, muscle fiber composition, and contractile function while also ensuring consistency and rapid production. Adult stem cells residing in muscle tissue are known for their powerful regenerative potential, yet tissue-derived skeletal muscle organoids have not been established. In this study, we introduce droplet-engineered skeletal muscle organoids derived from primary tissue using cascade-tubing microfluidics. These droplet-engineered organoids (DEOs) exhibit high maturity, including well-developed striated sarcomeres, spontaneous and stimulated contractions, and recapitulation of parental muscle fiber types. Notably, DEOs are produced in just 8 d without the need for primary cell culture-substantially accelerating the 50- to 60-d process required by classical organoid models. Additionally, the cascade-tubing microfluidics platform enables high-throughput production of hundreds of uniform DEO replicates from a small tissue sample, providing a scalable and reproducible solution for skeletal muscle research and drug screening.
Collapse
Affiliation(s)
- Jiawei Li
- Tsinghua Shenzhen International Graduate School (SIGS),
Tsinghua University, Shenzhen 518055, China
- Key Laboratory of Industrial Biocatalysis, Ministry of Education,
Tsinghua University, Beijing 100084, China
- Meatoid Biotechnology Limited, Shenzhen 518107, China
| | - Yiming Yang
- Tsinghua Shenzhen International Graduate School (SIGS),
Tsinghua University, Shenzhen 518055, China
| | - Ziqi Yi
- Tsinghua Shenzhen International Graduate School (SIGS),
Tsinghua University, Shenzhen 518055, China
- Key Laboratory of Industrial Biocatalysis, Ministry of Education,
Tsinghua University, Beijing 100084, China
| | - Yu Zhu
- Tsinghua Shenzhen International Graduate School (SIGS),
Tsinghua University, Shenzhen 518055, China
- Key Laboratory of Industrial Biocatalysis, Ministry of Education,
Tsinghua University, Beijing 100084, China
| | - Haowei Yang
- Tsinghua Shenzhen International Graduate School (SIGS),
Tsinghua University, Shenzhen 518055, China
- Key Laboratory of Industrial Biocatalysis, Ministry of Education,
Tsinghua University, Beijing 100084, China
| | - Baiming Chen
- School of Medicine,
The Chinese University of Hong Kong, Shenzhen 518172, China
| | - Peter E. Lobie
- Tsinghua Shenzhen International Graduate School (SIGS),
Tsinghua University, Shenzhen 518055, China
| | - Shaohua Ma
- Tsinghua Shenzhen International Graduate School (SIGS),
Tsinghua University, Shenzhen 518055, China
- Key Laboratory of Industrial Biocatalysis, Ministry of Education,
Tsinghua University, Beijing 100084, China
- Key Lab of Active Proteins and Peptides Green Biomanufacturing of Guangdong Higher Education Institutes,
Tsinghua Shenzhen International Graduate School, Shenzhen 518055, China
| |
Collapse
|
4
|
Chen SH, Yu BY, Kuo WY, Lin YB, Su SY, Chuang WH, Lu IH, Lin CY. Unveiling the immune microenvironment of complex tissues and tumors in transcriptomics through a deconvolution approach. BMC Cancer 2025; 25:733. [PMID: 40307726 PMCID: PMC12044707 DOI: 10.1186/s12885-025-14089-w] [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: 11/15/2022] [Accepted: 03/28/2025] [Indexed: 05/02/2025] Open
Abstract
Accurately resolving the composition of tumor-infiltrating leukocytes is pivotal for advancing cancer immunotherapy strategies. Despite the success of some clinical trials, applying these strategies remains limited due to the challenges in deciphering the immune microenvironment. In this study, we developed a streamlined, two-step workflow to address the complexity of bioinformatics processes involved in analyzing immune cell composition from transcriptomics data. Our dockerized toolkit, DOCexpress_fastqc, integrates the hisat2-stringtie pipeline with customized scripts within Galaxy/Docker environments, facilitating RNA sequencing (RNA-seq) gene expression profiling. The output from DOCexpress_fastqc is seamlessly formatted with mySORT, a web application that employs a deconvolution algorithm to determine the immune content across 21 cell subclasses. We validated mySORT using synthetic pseudo-bulk data derived from single-cell RNA sequencing (scRNA-seq) datasets. Our predictions exhibit strong concordance with the ground-truth immune cell composition, achieving Pearson's correlation coefficients of 0.871 in melanoma patients and 0.775 in head and neck cancer patients. Additionally, mySORT outperforms existing methods like CIBERSORT in accuracy and provides a wide range of data visualization features, such as hierarchical clustering and cell complexity plots. The toolkit and web application are freely available for the research community, providing enhanced resolution for conventional bulk RNA sequencing data and facilitating the analysis of immune microenvironment responses in immunotherapy. The mySORT demo website and Docker image are free at https://mysort.iis.sinica.edu.tw and https://hub.docker.com/r/lsbnb/mysort_2022 .
Collapse
Affiliation(s)
- Shu-Hwa Chen
- TMU Research Center of Cancer Translational Medicine, Taipei Medical University, 250 Wu-Xing Street, Taipei, Taiwan
| | - Bo-Yi Yu
- Research Center for Advanced Science and Technology, the University of Tokyo, 4-6-1 Komaba, Meguro-Ku, Tokyo, 153-8904, Japan
| | - Wen-Yu Kuo
- Institute of Information Science, Academia Sinica, 128 Academia Road, Section 2, Nankang, Taipei, 115, Taiwan
| | - Ya-Bo Lin
- Institute of Information Science, Academia Sinica, 128 Academia Road, Section 2, Nankang, Taipei, 115, Taiwan
| | - Sheng-Yao Su
- Department of Smart Computing and Applied Mathematics, Tunghai University, Taichung City, 407224, Taiwan
| | - Wei-Hsuan Chuang
- Institute of Information Science, Academia Sinica, 128 Academia Road, Section 2, Nankang, Taipei, 115, Taiwan
| | - I-Hsuan Lu
- Institute of Information Science, Academia Sinica, 128 Academia Road, Section 2, Nankang, Taipei, 115, Taiwan
| | - Chung-Yen Lin
- Institute of Information Science, Academia Sinica, 128 Academia Road, Section 2, Nankang, Taipei, 115, Taiwan.
- Institute of Fisheries Science, National Taiwan University, Taipei, Taiwan.
- Genome and Systems Biology Degree Program, National Taiwan University, Taipei, Taiwan.
| |
Collapse
|
5
|
Ahn C, Divoux A, Zhou M, Seldin MM, Sparks LM, Whytock KL. Optimized RNA sequencing deconvolution illustrates the impact of obesity and weight loss on cell composition of human adipose tissue. Obesity (Silver Spring) 2025; 33:936-948. [PMID: 40176378 PMCID: PMC12018139 DOI: 10.1002/oby.24264] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Revised: 01/24/2025] [Accepted: 01/27/2025] [Indexed: 04/04/2025]
Abstract
OBJECTIVE Cellular heterogeneity of human adipose tissue is linked to the pathophysiology of obesity and may impact the response to energy restriction and changes in fat mass. Herein, we provide an optimized pipeline to estimate cellular composition in human abdominal subcutaneous adipose tissue (ASAT) bulk RNA sequencing (RNA-seq) datasets using a single-nuclei RNA-seq signature matrix. METHODS A deconvolution pipeline for ASAT was optimized by benchmarking publicly available algorithms using a signature matrix derived from ASAT single-nuclei RNA-seq data from 20 adults and then applied to estimate ASAT cell-type proportions in publicly available obesity and weight loss studies. RESULTS Individuals with obesity had greater proportions of macrophages and lower proportions of adipocyte subpopulations and vascular cells compared with lean individuals. Two months of diet-induced weight loss increased the estimated proportions of macrophages; however, 2 years of diet-induced weight loss reduced the estimated proportions of macrophages, thereby suggesting a biphasic nature of cellular remodeling of ASAT during weight loss. CONCLUSIONS Our optimized high-throughput pipeline facilitates the assessment of composition changes of highly characterized cell types in large numbers of ASAT samples using low-cost bulk RNA-seq. Our data reveal novel changes in cellular heterogeneity and its association with cardiometabolic health in humans with obesity and following weight loss.
Collapse
Affiliation(s)
- Cheehoon Ahn
- Translational Research Institute, AdventHealth, Orlando, Florida, USA
| | - Adeline Divoux
- Translational Research Institute, AdventHealth, Orlando, Florida, USA
| | - Mingqi Zhou
- Department of Biological Chemistry and Center for Epigenetics and Metabolism, University of California, Irvine, California, USA
| | - Marcus M Seldin
- Department of Biological Chemistry and Center for Epigenetics and Metabolism, University of California, Irvine, California, USA
| | - Lauren M Sparks
- Translational Research Institute, AdventHealth, Orlando, Florida, USA
| | - Katie L Whytock
- Translational Research Institute, AdventHealth, Orlando, Florida, USA
| |
Collapse
|
6
|
Wang J, Ye F, Chai H, Jiang Y, Wang T, Ran X, Xia Q, Xu Z, Fu Y, Zhang G, Wu H, Guo G, Guo H, Ruan Y, Wang Y, Xing D, Xu X, Zhang Z. Advances and applications in single-cell and spatial genomics. SCIENCE CHINA. LIFE SCIENCES 2025; 68:1226-1282. [PMID: 39792333 DOI: 10.1007/s11427-024-2770-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Accepted: 10/10/2024] [Indexed: 01/12/2025]
Abstract
The applications of single-cell and spatial technologies in recent times have revolutionized the present understanding of cellular states and the cellular heterogeneity inherent in complex biological systems. These advancements offer unprecedented resolution in the examination of the functional genomics of individual cells and their spatial context within tissues. In this review, we have comprehensively discussed the historical development and recent progress in the field of single-cell and spatial genomics. We have reviewed the breakthroughs in single-cell multi-omics technologies, spatial genomics methods, and the computational strategies employed toward the analyses of single-cell atlas data. Furthermore, we have highlighted the advances made in constructing cellular atlases and their clinical applications, particularly in the context of disease. Finally, we have discussed the emerging trends, challenges, and opportunities in this rapidly evolving field.
Collapse
Affiliation(s)
- Jingjing Wang
- Bone Marrow Transplantation Center of the First Affiliated Hospital & Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou, 310058, China
| | - Fang Ye
- Bone Marrow Transplantation Center of the First Affiliated Hospital & Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou, 310058, China
| | - Haoxi Chai
- Life Sciences Institute and The Second Affiliated Hospital, Zhejiang University, Hangzhou, 310058, China
| | - Yujia Jiang
- BGI Research, Shenzhen, 518083, China
- BGI Research, Hangzhou, 310030, China
| | - Teng Wang
- Biomedical Pioneering Innovation Center (BIOPIC) and School of Life Sciences, Peking University, Beijing, 100871, China
- Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, 100871, China
| | - Xia Ran
- Bone Marrow Transplantation Center of the First Affiliated Hospital & Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou, 310058, China
- Institute of Hematology, Zhejiang University, Hangzhou, 310000, China
| | - Qimin Xia
- Biomedical Pioneering Innovation Center (BIOPIC) and School of Life Sciences, Peking University, Beijing, 100871, China
| | - Ziye Xu
- Department of Laboratory Medicine of The First Affiliated Hospital & Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou, 310058, China
| | - Yuting Fu
- Bone Marrow Transplantation Center of the First Affiliated Hospital & Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou, 310058, China
- Center for Stem Cell and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou, 310058, China
| | - Guodong Zhang
- Bone Marrow Transplantation Center of the First Affiliated Hospital & Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou, 310058, China
- Center for Stem Cell and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou, 310058, China
| | - Hanyu Wu
- Bone Marrow Transplantation Center of the First Affiliated Hospital & Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou, 310058, China
- Center for Stem Cell and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou, 310058, China
| | - Guoji Guo
- Bone Marrow Transplantation Center of the First Affiliated Hospital & Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou, 310058, China.
- Center for Stem Cell and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou, 310058, China.
- Zhejiang Provincial Key Lab for Tissue Engineering and Regenerative Medicine, Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cell and Regenerative Medicine, Hangzhou, 310058, China.
- Institute of Hematology, Zhejiang University, Hangzhou, 310000, China.
| | - Hongshan Guo
- Bone Marrow Transplantation Center of the First Affiliated Hospital & Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou, 310058, China.
- Institute of Hematology, Zhejiang University, Hangzhou, 310000, China.
| | - Yijun Ruan
- Life Sciences Institute and The Second Affiliated Hospital, Zhejiang University, Hangzhou, 310058, China.
| | - Yongcheng Wang
- Department of Laboratory Medicine of The First Affiliated Hospital & Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou, 310058, China.
| | - Dong Xing
- Biomedical Pioneering Innovation Center (BIOPIC) and School of Life Sciences, Peking University, Beijing, 100871, China.
- Beijing Advanced Innovation Center for Genomics (ICG), Peking University, Beijing, 100871, China.
| | - Xun Xu
- BGI Research, Shenzhen, 518083, China.
- BGI Research, Hangzhou, 310030, China.
- Guangdong Provincial Key Laboratory of Genome Read and Write, BGI Research, Shenzhen, 518083, China.
| | - Zemin Zhang
- Biomedical Pioneering Innovation Center (BIOPIC) and School of Life Sciences, Peking University, Beijing, 100871, China.
| |
Collapse
|
7
|
Yang T, Qin Y, Yan S, Guo S, Sun J, Huang J, Li J, Zhou Q, Jin X, Wang WJ. Comprehensive evaluation of methods for identifying tissues or cell types of origin of the plasma cell-free transcriptome. PeerJ 2025; 13:e19241. [PMID: 40256737 PMCID: PMC12009560 DOI: 10.7717/peerj.19241] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2024] [Accepted: 03/11/2025] [Indexed: 04/22/2025] Open
Abstract
Plasma cell-free RNA (cfRNA) is derived from cells in various tissues and organs throughout the body and reflects the physiological and pathological conditions. Identifying the origins of cfRNA is essential for comprehending its variations. Only a few tools are designed for cfRNA deconvolution, and most studies have relied on traditional bulk RNA methods. In this study, we employed human tissue and cell transcriptomic data as reference sets and evaluated the performance of seven deconvolution methods on cfRNA. We compared the analysis results of cell types and tissues of origin of plasma cfRNA and chose to use single-cell RNA sequencing (scRNA-seq) data as reference to conduct further evaluation of deconvolution methods. Subsequently, we assessed the accuracy and robustness of the methods by utilizing simulated cfRNA data generated from scRNA-seq. We also evaluated the methods' accuracy on real plasma cfRNA data by analyzing the correlation between the predicted cell proportions and the corresponding clinical indicators. Moreover, we compared the methods' effectiveness in revealing the impacts of diseases on cells and evaluated the performance of cancer classification models based on the cell origin data they provided. In summary, our study provides valuable insights into cfRNA origin analysis, enhancing its potential in biomedical research.
Collapse
Affiliation(s)
- Tingyu Yang
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China
- BGI Research, Shenzhen, China
| | - Yulong Qin
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China
- BGI Research, Shenzhen, China
| | - Shuo Yan
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China
- BGI Research, Shenzhen, China
| | - Sijia Guo
- BGI Research, Shenzhen, China
- College of Life Sciences, Northwest University, Xi’an, China
| | | | - Jiayi Huang
- BGI Research, Shenzhen, China
- College of Life Sciences and Oceanography, Shenzhen University, Shenzhen, China
| | - Jiayi Li
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China
- BGI Research, Shenzhen, China
| | | | - Xin Jin
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China
- BGI Research, Shenzhen, China
- Shenzhen Key Laboratory of Transomics Biotechnologies, BGI Research, Shenzhen, China
- The Innovation Centre of Ministry of Education for Development and Diseases, School of Medicine, South China University of Technology, Guangzhou, China
| | | |
Collapse
|
8
|
D'Sa K, Choi ML, Wagen AZ, Setó-Salvia N, Kopach O, Evans JR, Rodrigues M, Lopez-Garcia P, Lachica J, Clarke BE, Singh J, Ghareeb A, Bayne J, Grant-Peters M, Garcia-Ruiz S, Chen Z, Rodriques S, Athauda D, Gustavsson EK, Gagliano Taliun SA, Toomey C, Reynolds RH, Young G, Strohbuecker S, Warner T, Rusakov DA, Patani R, Bryant C, Klenerman DA, Gandhi S, Ryten M. Astrocytic RNA editing regulates the host immune response to alpha-synuclein. SCIENCE ADVANCES 2025; 11:eadp8504. [PMID: 40215316 PMCID: PMC11988446 DOI: 10.1126/sciadv.adp8504] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Accepted: 03/07/2025] [Indexed: 04/14/2025]
Abstract
RNA editing is a posttranscriptional mechanism that targets changes in RNA transcripts to modulate innate immune responses. We report the role of astrocyte-specific, ADAR1-mediated RNA editing in neuroinflammation in Parkinson's disease (PD). We generated human induced pluripotent stem cell-derived astrocytes, neurons and cocultures and exposed them to small soluble alpha-synuclein aggregates. Oligomeric alpha-synuclein triggered an inflammatory glial state associated with Toll-like receptor activation, viral responses, and cytokine secretion. This reactive state resulted in loss of neurosupportive functions and the induction of neuronal toxicity. Notably, interferon response pathways were activated leading to up-regulation and isoform switching of the RNA deaminase enzyme, ADAR1. ADAR1 mediates A-to-I RNA editing, and increases in RNA editing were observed in inflammatory pathways in cells, as well as in postmortem human PD brain. Aberrant, or dysregulated, ADAR1 responses and RNA editing may lead to sustained inflammatory reactive states in astrocytes triggered by alpha-synuclein aggregation, and this may drive the neuroinflammatory cascade in Parkinson's.
Collapse
Affiliation(s)
- Karishma D'Sa
- Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, Queen Square, London WC1N 3BG, UK
- The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD 20815, USA
| | - Minee L. Choi
- Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, Queen Square, London WC1N 3BG, UK
- The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD 20815, USA
- Department of Brain & Cognitive Sciences, KAIST, 921 Dehak-ro, Daejeon, Republic of Korea
| | - Aaron Z. Wagen
- Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, Queen Square, London WC1N 3BG, UK
- The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD 20815, USA
- Department of Genetics and Genomic Medicine, Great Ormond Street Institute of Child Health, University College London, London WC1N 1EH, UK
| | - Núria Setó-Salvia
- Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, Queen Square, London WC1N 3BG, UK
- Reta Lila Weston Institute, UCL Queen Square Institute of Neurology, London WC1N 3BG, UK
| | - Olga Kopach
- Department of Clinical and Experimental Epilepsy, UCL Queen Square Institute of Neurology, London WC1N 3BG, UK
- Neuroscience and Cell Biology Research Institute, City St George’s, University of London, Cranmer Terrace, London SW17 0RE, UK
| | - James R. Evans
- Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, Queen Square, London WC1N 3BG, UK
- The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD 20815, USA
| | - Margarida Rodrigues
- Department of Chemistry, University of Cambridge, Cambridge CB2 1EW, UK
- UK Dementia Research Institute at The University of Cambridge, Cambridge CB2 0AH, UK
| | - Patricia Lopez-Garcia
- Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, Queen Square, London WC1N 3BG, UK
- The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD 20815, USA
| | - Joanne Lachica
- Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, Queen Square, London WC1N 3BG, UK
- The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD 20815, USA
- UCL Queen Square Institute of Neurology, University College London, London WC1N 3BG, UK
| | - Benjamin E. Clarke
- The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
- Department of Neuromuscular Disease, UCL Queen Square Institute of Neurology, Queen Square, London WC1N 3BG, UK
| | - Jaijeet Singh
- Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, Queen Square, London WC1N 3BG, UK
- The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
| | - Ali Ghareeb
- Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, Queen Square, London WC1N 3BG, UK
- Applied Biotechnology Lab, The Francis Crick Institute, London NW1 1AT, UK
| | - James Bayne
- Applied Biotechnology Lab, The Francis Crick Institute, London NW1 1AT, UK
- Nuffield Department of Orthopaedics, Rheumatology, and Musculoskeletal Sciences, University of Oxford, Oxford OX3 7LD, UK
| | - Melissa Grant-Peters
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD 20815, USA
- Department of Genetics and Genomic Medicine, Great Ormond Street Institute of Child Health, University College London, London WC1N 1EH, UK
| | - Sonia Garcia-Ruiz
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD 20815, USA
- Department of Genetics and Genomic Medicine, Great Ormond Street Institute of Child Health, University College London, London WC1N 1EH, UK
| | - Zhongbo Chen
- Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, Queen Square, London WC1N 3BG, UK
- The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
| | - Samuel Rodriques
- Applied Biotechnology Lab, The Francis Crick Institute, London NW1 1AT, UK
- FutureHouse, 1405 Minnesota Street, San Francisco, CA 94107, USA
| | - Dilan Athauda
- Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, Queen Square, London WC1N 3BG, UK
- The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
| | - Emil K. Gustavsson
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD 20815, USA
- Department of Genetics and Genomic Medicine, Great Ormond Street Institute of Child Health, University College London, London WC1N 1EH, UK
- UCL Queen Square Institute of Neurology, University College London, London WC1N 3BG, UK
| | - Sarah A. Gagliano Taliun
- Montréal Heart Institute, Montréal, QC, Canada
- Department of Medicine and Department of Neurosciences, Université de Montréal, Montréal, QC, Canada
| | - Christina Toomey
- Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, Queen Square, London WC1N 3BG, UK
- The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD 20815, USA
- UCL Queen Square Institute of Neurology, University College London, London WC1N 3BG, UK
| | - Regina H. Reynolds
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD 20815, USA
- Department of Genetics and Genomic Medicine, Great Ormond Street Institute of Child Health, University College London, London WC1N 1EH, UK
| | - George Young
- The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
- MRC Laboratory of Medical Sciences, London W12 0HS, UK
| | - Stephanie Strohbuecker
- The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD 20815, USA
| | - Thomas Warner
- Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, Queen Square, London WC1N 3BG, UK
- Reta Lila Weston Institute, UCL Queen Square Institute of Neurology, London WC1N 3BG, UK
| | - Dmitri A. Rusakov
- Department of Clinical and Experimental Epilepsy, UCL Queen Square Institute of Neurology, London WC1N 3BG, UK
| | - Rickie Patani
- The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
- Department of Neuromuscular Disease, UCL Queen Square Institute of Neurology, Queen Square, London WC1N 3BG, UK
| | - Clare Bryant
- Department of Veterinary Medicine, University of Cambridge, Cambridge CB3 0ES, UK
| | - David A. Klenerman
- Department of Chemistry, University of Cambridge, Cambridge CB2 1EW, UK
- UK Dementia Research Institute at The University of Cambridge, Cambridge CB2 0AH, UK
| | - Sonia Gandhi
- Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, Queen Square, London WC1N 3BG, UK
- The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD 20815, USA
| | - Mina Ryten
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD 20815, USA
- Department of Genetics and Genomic Medicine, Great Ormond Street Institute of Child Health, University College London, London WC1N 1EH, UK
- UK Dementia Research Institute at The University of Cambridge, Cambridge CB2 0AH, UK
- Department of Clinical Neurosciences, School of Clinical Medicine, University of Cambridge, Cambridge CB2 0SP, UK
- Department of Genetics, University of Cambridge, Cambridge CB2 3EH, UK
| |
Collapse
|
9
|
Wang J, Guay H, Chang D. Crohn's Disease and Ulcerative Colitis Share 2 Molecular Subtypes With Different Mechanisms and Drug Responses. J Crohns Colitis 2025; 19:jjae152. [PMID: 39361323 DOI: 10.1093/ecco-jcc/jjae152] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Revised: 09/04/2024] [Accepted: 10/02/2024] [Indexed: 03/29/2025]
Abstract
BACKGROUND AND AIMS Several therapies have been approved to treat Crohn's disease (CD) and ulcerative colitis (UC), indicating that both diseases may share the same molecular subtypes. The aim of this study is to identify shared patient subtypes with common molecular drivers of disease. METHODS Five public datasets with 406 CD and 421 UC samples were integrated to identify molecular subtypes. Then, the patient labels from 6 independent datasets and 8 treatment datasets were predicted for validating subtypes and identifying the relationship with response status of corticosteroids, infliximab, vedolizumab, and ustekinumab. RESULTS Two molecular subtypes were identified from the training datasets, in which CD and UC patients were relatively evenly represented in each subtype. We found 6 S1-specific gene modules related to innate/adaptive immune responses and tissue remodeling and 9 S1-specific cell types (cycling T cells, Tregs, CD8+ lamina propria, follicular B cells, cycling B cells, plasma cells, inflammatory monocytes, inflammatory fibroblasts, and postcapillary venules). Subtype S2 was associated with 3 modules related to metabolism functions and 4 cell types (immature enterocytes, transit amplifying cells, immature goblet cells, and WNT5B+ cells). The subtypes can be replicated in 6 independent datasets based on a 20-gene classifier. Furthermore, response rates to 4 treatments in subtype S2 were significantly higher than those in subtype S1. CONCLUSIONS This study discovered and validated a robust transcriptome-based molecular classification shared by CD and UC and built a 20-gene classifier. Because 2 subtypes have different molecular mechanisms and drug response, our classification may aid interpretation of heterogeneous molecular and clinical information in inflammatory bowel disease patients.
Collapse
Affiliation(s)
- Jing Wang
- Genomic Research Center, AbbVie Inc., Cambridge, MA, USA
| | - Heath Guay
- AbbVie Bioresearch Center, Worcester, MA, USA
| | - Dan Chang
- Genomic Research Center, AbbVie Inc., Cambridge, MA, USA
| |
Collapse
|
10
|
Thomas MPH, Ajaib S, Tanner G, Bulpitt AJ, Stead LF. GBMPurity: A Machine Learning Tool for Estimating Glioblastoma Tumour Purity from Bulk RNA-seq Data. Neuro Oncol 2025:noaf026. [PMID: 39891579 DOI: 10.1093/neuonc/noaf026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2024] [Indexed: 02/03/2025] Open
Abstract
BACKGROUND Glioblastoma (GBM) presents a significant clinical challenge due to its aggressive nature and extensive heterogeneity. Tumour purity, the proportion of malignant cells within a tumour, is an important covariate for understanding the disease, having direct clinical relevance or obscuring signal of the malignant portion in molecular analyses of bulk samples. However, current methods for estimating tumour purity are non-specific and technically demanding. Therefore, we aimed to build a reliable and accessible purity estimator for GBM. METHODS We developed GBMPurity, a deep-learning model specifically designed to estimate the purity of IDH-wildtype primary GBM from bulk RNA-seq data. The model was trained using simulated pseudobulk tumours of known purity from labelled single-cell data acquired from the GBmap resource. The performance of GBMPurity was evaluated and compared to several existing tools using independent datasets. RESULTS GBMPurity outperformed existing tools, achieving a mean absolute error of 0.15 and a concordance correlation coefficient of 0.88 on validation datasets. We demonstrate the utility of GBMPurity through inference on bulk RNA-seq samples and observe reduced purity of the Proneural molecular subtype relative to the Classical, attributed to the increased presence of healthy brain cells. CONCLUSIONS GBMPurity provides a reliable and accessible tool for estimating tumour purity from bulk RNA-seq data, enhancing the interpretation of bulk RNA-seq data and offering valuable insights into GBM biology. To facilitate the use of this model by the wider research community, GBMPurity is available as a web-based tool at: https://gbmdeconvoluter.leeds.ac.uk/.
Collapse
Affiliation(s)
- Morgan P H Thomas
- School of Computer Science, University of Leeds, UK
- Leeds Institute of Medical Research at St James's, University of Leeds, Leeds, UK
| | - Shoaib Ajaib
- Leeds Institute of Medical Research at St James's, University of Leeds, Leeds, UK
| | - Georgette Tanner
- Leeds Institute of Medical Research at St James's, University of Leeds, Leeds, UK
| | | | - Lucy F Stead
- Leeds Institute of Medical Research at St James's, University of Leeds, Leeds, UK
| |
Collapse
|
11
|
Guo S, Liu X, Cheng X, Jiang Y, Ji S, Liang Q, Koval A, Li Y, Owen LA, Kim IK, Aparicio A, Lee S, Sood AK, Kopetz S, Shen JP, Weinstein JN, DeAngelis MM, Chen R, Wang W. A deconvolution framework that uses single-cell sequencing plus a small benchmark data set for accurate analysis of cell type ratios in complex tissue samples. Genome Res 2025; 35:147-161. [PMID: 39586714 PMCID: PMC11789644 DOI: 10.1101/gr.278822.123] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Accepted: 11/19/2024] [Indexed: 11/27/2024]
Abstract
Bulk deconvolution with single-cell/nucleus RNA-seq data is critical for understanding heterogeneity in complex biological samples, yet the technological discrepancy across sequencing platforms limits deconvolution accuracy. To address this, we utilize an experimental design to match inter-platform biological signals, hence revealing the technological discrepancy, and then develop a deconvolution framework called DeMixSC using this well-matched, that is, benchmark, data. Built upon a novel weighted nonnegative least-squares framework, DeMixSC identifies and adjusts genes with high technological discrepancy and aligns the benchmark data with large patient cohorts of matched-tissue-type for large-scale deconvolution. Our results using two benchmark data sets of healthy retinas and ovarian cancer tissues suggest much-improved deconvolution accuracy. Leveraging tissue-specific benchmark data sets, we applied DeMixSC to a large cohort of 453 age-related macular degeneration patients and a cohort of 30 ovarian cancer patients with various responses to neoadjuvant chemotherapy. Only DeMixSC successfully unveiled biologically meaningful differences across patient groups, demonstrating its broad applicability in diverse real-world clinical scenarios. Our findings reveal the impact of technological discrepancy on deconvolution performance and underscore the importance of a well-matched data set to resolve this challenge. The developed DeMixSC framework is generally applicable for accurately deconvolving large cohorts of disease tissues, including cancers, when a well-matched benchmark data set is available.
Collapse
Affiliation(s)
- Shuai Guo
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030, USA
| | - Xiaoqian Liu
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030, USA
| | - Xuesen Cheng
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas 77030, USA
| | - Yujie Jiang
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030, USA
- Department of Statistics, Rice University, Houston, Texas 77005, USA
| | - Shuangxi Ji
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030, USA
| | - Qingnan Liang
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas 77030, USA
| | - Andrew Koval
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030, USA
- Department of Statistics, Rice University, Houston, Texas 77005, USA
| | - Yumei Li
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas 77030, USA
| | - Leah A Owen
- Department of Ophthalmology, Jacobs School of Medicine and Biomedical Engineering, SUNY University at Buffalo, Buffalo, New York 14209, USA
- Department of Population Health Sciences, University of Utah School of Medicine, Salt Lake City, Utah 84108, USA
- Department of Ophthalmology and Visual Sciences, University of Utah School of Medicine, Salt Lake City, Utah 84132, USA
| | - Ivana K Kim
- USA Retina Service, Harvard Medical School, Massachusetts Eye and Ear, Boston, Massachusetts 02114, USA
| | - Ana Aparicio
- Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas 77230, USA
| | - Sanghoon Lee
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas 77230, USA
| | - Anil K Sood
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas 77230, USA
| | - Scott Kopetz
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030, USA
| | - John Paul Shen
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030, USA
| | - John N Weinstein
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030, USA
- Department of Systems Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030, USA
| | - Margaret M DeAngelis
- Department of Ophthalmology, Jacobs School of Medicine and Biomedical Engineering, SUNY University at Buffalo, Buffalo, New York 14209, USA
- Department of Population Health Sciences, University of Utah School of Medicine, Salt Lake City, Utah 84108, USA
- Department of Ophthalmology and Visual Sciences, University of Utah School of Medicine, Salt Lake City, Utah 84132, USA
- VA Western New York Healthcare System, Buffalo, New York 14215, USA
| | - Rui Chen
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas 77030, USA
| | - Wenyi Wang
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030, USA;
| |
Collapse
|
12
|
Fantecelle CH, Polaco Covre L, Lopes PO, Sarmento IV, Decote-Ricardo D, Geraldo Freire-de-Lima C, de Matos Guedes HL, Pimentel MIF, Conceição-Silva F, Maretti-Mira AC, Borges VM, Pedreira de Carvalho L, de Carvalho EM, Mosser D, Falqueto A, Akbar AN, Gomes DCO. Senescence-related genes are associated with the immunopathology signature of American tegumentary leishmaniasis lesions and may predict progression to mucosal leishmaniasis. Clin Exp Immunol 2025; 219:uxae088. [PMID: 39428748 PMCID: PMC11771187 DOI: 10.1093/cei/uxae088] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Revised: 08/27/2024] [Accepted: 10/17/2024] [Indexed: 10/22/2024] Open
Abstract
The American tegumentary leishmaniasis (ATL) is caused by protozoans of the genus Leishmania and varies from mild localized cutaneous leishmaniasis (LCL) form to more severe manifestations such as the diffuse cutaneous leishmaniasis (DCL) form and the mucosal leishmaniasis (ML) form. Previously, we demonstrated the accumulation of senescent cells in skin lesions of patients with LCL. Moreover, lesional transcriptomic analyses revealed a robust co-induction of senescence and pro-inflammatory gene signatures, highlighting the critical role of senescent T cells in orchestrating pathology. In this work we hypothesized that senescent cells might operate differently among the ATL spectrum, potentially influencing immunopathological mechanisms and clinical outcome. We analysed previously published RNA-Seq datasets of skin biopsies of healthy subjects and lesional skin from DCL patients, LCL patients, and LCL patients that, after treatment, progressed to mucosal leishmaniasis (MLP). Our findings demonstrate a robust presence of a CD8 T-cell signature associated with both LCL and MLP lesions. Moreover, both inflammatory and cytotoxic signatures were significantly upregulated, showing a strong increase in MLP and LCL groups, but not DCL. The senescence signature was elevated between LCL and MLP groups, representing the only distinguishable signature of immunopathology between them. Interestingly, our analyses further revealed the senescence signature's capacity to predict progression from LCL to mucosal forms, which was not observed with other signatures. Both the senescence-signature score and specific senescence-associated genes demonstrated an increased capacity to predict mucosal progression, with correct predictions exceeding 97% of cases. Collectively, our findings contribute to a comprehensive understanding of immunosenescence in ATL and suggest that senescence may represent the latest and most important signature of the immunopathogenisis. This highlights its potential value in predicting disease severity.
Collapse
Affiliation(s)
| | - Luciana Polaco Covre
- Núcleo de Doenças Infecciosas, Universidade Federal do Espírito Santo, Vitória, Brazil
- Division of Medicine, University College London, London, UK
| | - Paola Oliveira Lopes
- Núcleo de Biotecnologia, Universidade Federal do Espírito Santo, Vitória, Brazil
| | | | - Debora Decote-Ricardo
- Departamento de Veterinária, Universidade Federal Rural do Rio de Janeiro, Rio de Janeiro, Brazil
| | | | - Herbert Leonel de Matos Guedes
- Instituto de Microbiologia Professor Paulo de Goes, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
- Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro, Brazil
| | | | | | - Ana C Maretti-Mira
- USC Research Center for Liver Diseases, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Valéria M Borges
- Instituto Gonçalo Muniz, Fundação Oswaldo Cruz, Salvador, Brazil
| | | | | | - David Mosser
- Department of Cell Biology and Molecular Genetics, University of Maryland, College Park, Maryland, USA
| | - Aloisio Falqueto
- Departamento de Medicina Social, Universidade Federal do Espírito Santo, Vitoria, Brazil
| | - Arne N Akbar
- Division of Medicine, University College London, London, UK
| | - Daniel Claudio Oliveira Gomes
- Núcleo de Doenças Infecciosas, Universidade Federal do Espírito Santo, Vitória, Brazil
- Núcleo de Biotecnologia, Universidade Federal do Espírito Santo, Vitória, Brazil
| |
Collapse
|
13
|
Davidson NR, Zhang F, Greene CS. BuDDI: Bulk Deconvolution with Domain Invariance to predict cell-type-specific perturbations from bulk. PLoS Comput Biol 2025; 21:e1012742. [PMID: 39823522 PMCID: PMC11790236 DOI: 10.1371/journal.pcbi.1012742] [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: 06/20/2024] [Revised: 02/03/2025] [Accepted: 12/20/2024] [Indexed: 01/19/2025] Open
Abstract
While single-cell experiments provide deep cellular resolution within a single sample, some single-cell experiments are inherently more challenging than bulk experiments due to dissociation difficulties, cost, or limited tissue availability. This creates a situation where we have deep cellular profiles of one sample or condition, and bulk profiles across multiple samples and conditions. To bridge this gap, we propose BuDDI (BUlk Deconvolution with Domain Invariance). BuDDI utilizes domain adaptation techniques to effectively integrate available corpora of case-control bulk and reference scRNA-seq observations to infer cell-type-specific perturbation effects. BuDDI achieves this by learning independent latent spaces within a single variational autoencoder (VAE) encompassing at least four sources of variability: 1) cell type proportion, 2) perturbation effect, 3) structured experimental variability, and 4) remaining variability. Since each latent space is encouraged to be independent, we simulate perturbation responses by independently composing each latent space to simulate cell-type-specific perturbation responses. We evaluated BuDDI's performance on simulated and real data with experimental designs of increasing complexity. We first validated that BuDDI could learn domain invariant latent spaces on data with matched samples across each source of variability. Then we validated that BuDDI could accurately predict cell-type-specific perturbation response when no single-cell perturbed profiles were used during training; instead, only bulk samples had both perturbed and non-perturbed observations. Finally, we validated BuDDI on predicting sex-specific differences, an experimental design where it is not possible to have matched samples. In each experiment, BuDDI outperformed all other comparative methods and baselines. As more reference atlases are completed, BuDDI provides a path to combine these resources with bulk-profiled treatment or disease signatures to study perturbations, sex differences, or other factors at single-cell resolution.
Collapse
Affiliation(s)
- Natalie R. Davidson
- Department of Biomedical Informatics, University of Colorado Anschutz School of Medicine, Aurora, Colorado, United States of America
| | - Fan Zhang
- Department of Biomedical Informatics, University of Colorado Anschutz School of Medicine, Aurora, Colorado, United States of America
- Department of Medicine Rheumatology, University of Colorado Anschutz School of Medicine, Aurora, Colorado, United States of America
| | - Casey S. Greene
- Department of Biomedical Informatics, University of Colorado Anschutz School of Medicine, Aurora, Colorado, United States of America
| |
Collapse
|
14
|
Gulati GS, D'Silva JP, Liu Y, Wang L, Newman AM. Profiling cell identity and tissue architecture with single-cell and spatial transcriptomics. Nat Rev Mol Cell Biol 2025; 26:11-31. [PMID: 39169166 DOI: 10.1038/s41580-024-00768-2] [Citation(s) in RCA: 25] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/16/2024] [Indexed: 08/23/2024]
Abstract
Single-cell transcriptomics has broadened our understanding of cellular diversity and gene expression dynamics in healthy and diseased tissues. Recently, spatial transcriptomics has emerged as a tool to contextualize single cells in multicellular neighbourhoods and to identify spatially recurrent phenotypes, or ecotypes. These technologies have generated vast datasets with targeted-transcriptome and whole-transcriptome profiles of hundreds to millions of cells. Such data have provided new insights into developmental hierarchies, cellular plasticity and diverse tissue microenvironments, and spurred a burst of innovation in computational methods for single-cell analysis. In this Review, we discuss recent advancements, ongoing challenges and prospects in identifying and characterizing cell states and multicellular neighbourhoods. We discuss recent progress in sample processing, data integration, identification of subtle cell states, trajectory modelling, deconvolution and spatial analysis. Furthermore, we discuss the increasing application of deep learning, including foundation models, in analysing single-cell and spatial transcriptomics data. Finally, we discuss recent applications of these tools in the fields of stem cell biology, immunology, and tumour biology, and the future of single-cell and spatial transcriptomics in biological research and its translation to the clinic.
Collapse
Affiliation(s)
- Gunsagar S Gulati
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | | | - Yunhe Liu
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Linghua Wang
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- The University of Texas MD Anderson Cancer Center UTHealth Houston Graduate School of Biomedical Sciences, Houston, TX, USA
| | - Aaron M Newman
- Department of Biomedical Data Science, Stanford University, Stanford, CA, USA.
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University, Stanford, CA, USA.
- Stanford Cancer Institute, Stanford University, Stanford, CA, USA.
- Chan Zuckerberg Biohub - San Francisco, San Francisco, CA, USA.
| |
Collapse
|
15
|
Maurizio A, Tascini AS, Morelli MJ. SurfR: Riding the wave of RNA-seq data with a comprehensive bioconductor package to identify surface protein-coding genes. BIOINFORMATICS ADVANCES 2024; 5:vbae201. [PMID: 39735574 PMCID: PMC11671034 DOI: 10.1093/bioadv/vbae201] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Revised: 11/26/2024] [Accepted: 12/12/2024] [Indexed: 12/31/2024]
Abstract
Motivation Proteins at the cell surface connect signaling networks and largely determine a cell's capacity to communicate and interact with its environment. In particular, variations in transcriptomic profiles are often observed between healthy and diseased cells, leading to distinct sets of cell-surface proteins. For these reasons, cell-surface proteins may act as biomarkers for the detection of cells of interest in tissues or body fluids, are often the target of pharmaceutical agents, and hold significant promise in the clinical practice for diagnosis, prognosis, treatment development, and evaluation of therapy response. Therefore, implementing robust methods to identify condition-specific cell-surface proteins is of pivotal importance to advance biomedical research. Results We developed SurfR, an R/Bioconductor package providing a streamlined end-to-end workflow for computationally identifying surface protein-coding genes from expression data. Our user-friendly, comprehensive workflow performs systematic expression data retrieval from public databases, differential gene expression across conditions, integration of datasets, enrichment analysis, identification of targetable proteins on a condition of interest, and data visualization. Availability and implementation SurfR is released under GNU-GPL-v3.0 License. Source code, documentation, examples, and tutorials are available through Bioconductor (http://www.bioconductor.org/packages/SurfR). RMD notebooks with the use cases code described in the manuscript can be found on GitHub (https://github.com/auroramaurizio/SurfR_UseCases).
Collapse
Affiliation(s)
- Aurora Maurizio
- Center for Omics Sciences, IRCCS San Raffaele Scientific Institute, Milan 20132, Italy
| | - Anna Sofia Tascini
- Center for Omics Sciences, IRCCS San Raffaele Scientific Institute, Milan 20132, Italy
- Universita‘Vita-Salute San Raffaele, Milan 20132, Italy
| | - Marco J Morelli
- Center for Omics Sciences, IRCCS San Raffaele Scientific Institute, Milan 20132, Italy
- Universita‘Vita-Salute San Raffaele, Milan 20132, Italy
| |
Collapse
|
16
|
de Sena Costa de Oliveira D, Cunha JLS. Comment on: Salivary DNA methylation derived estimates of biological aging, cellular frequency, and protein expression as predictors of oral mucositis severity and survival in head and neck cancer patients. Oral Oncol 2024; 159:107097. [PMID: 39488167 DOI: 10.1016/j.oraloncology.2024.107097] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2024] [Accepted: 10/28/2024] [Indexed: 11/04/2024]
Affiliation(s)
| | - John Lennon Silva Cunha
- Center for Biological and Health Sciences, Federal University of Western Bahia (UFOB), Barreiras, BA, Brazil.
| |
Collapse
|
17
|
Xiong X, Liu Y, Pu D, Yang Z, Bi Z, Tian L, Li X. DeSide: A unified deep learning approach for cellular deconvolution of tumor microenvironment. Proc Natl Acad Sci U S A 2024; 121:e2407096121. [PMID: 39514318 PMCID: PMC11573681 DOI: 10.1073/pnas.2407096121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Accepted: 09/23/2024] [Indexed: 11/16/2024] Open
Abstract
Cellular deconvolution via bulk RNA sequencing (RNA-seq) presents a cost-effective and efficient alternative to experimental methods such as flow cytometry and single-cell RNA-seq (scRNA-seq) for analyzing the complex cellular composition of tumor microenvironments. Despite challenges due to heterogeneity within and among tumors, our innovative deep learning-based approach, DeSide, shows exceptional accuracy in estimating the proportions of 16 distinct cell types and subtypes within solid tumors. DeSide integrates biological pathways and assesses noncancerous cell types first, effectively sidestepping the issue of highly variable gene expression profiles (GEPs) associated with cancer cells. By leveraging scRNA-seq data from six cancer types and 185 cancer cell lines across 22 cancer types as references, our method introduces distinctive sampling and filtering techniques to generate a high-quality training set that closely replicates real tumor GEPs, based on The Cancer Genome Atlas (TCGA) bulk RNA-seq data. With this model and high-quality training set, DeSide outperforms existing methods in estimating tumor purity and the proportions of noncancerous cells within solid tumors. Our model precisely predicts cellular compositions across 19 cancer types from TCGA and proves its effectiveness with multiple additional external datasets. Crucially, DeSide enables the identification and analysis of combinatorial cell type pairs, facilitating the stratification of cancer patients into prognostically significant groups. This approach not only provides deeper insights into the dynamics of tumor biology but also highlights potential therapeutic targets by underscoring the importance of specific cell type or subtype interactions.
Collapse
Affiliation(s)
- Xin Xiong
- Department of Physics, Hong Kong Baptist University, Hong Kong, China
| | - Yerong Liu
- Key Laboratory of Quantitative Synthetic Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Dandan Pu
- Key Laboratory of Quantitative Synthetic Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Zhu Yang
- State Key Laboratory of Environmental and Biological Analysis, Hong Kong Baptist University, Hong Kong, China
| | - Zedong Bi
- Lingang Laboratory, Shanghai 200031, China
| | - Liang Tian
- Department of Physics, Hong Kong Baptist University, Hong Kong, China
- State Key Laboratory of Environmental and Biological Analysis, Hong Kong Baptist University, Hong Kong, China
- Institute of Computational and Theoretical Studies, Hong Kong Baptist University, Hong Kong, China
- Institute of Systems Medicine and Health Sciences, Hong Kong Baptist University, Hong Kong, China
| | - Xuefei Li
- Key Laboratory of Quantitative Synthetic Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| |
Collapse
|
18
|
Zhao H, Cheng Y, Li J, Zhou J, Yang H, Yu F, Yu F, Khutsishvili D, Wang Z, Jiang S, Tan K, Kuang Y, Xing X, Ma S. Droplet-engineered organoids recapitulate parental tissue transcriptome with inter-organoid homogeneity and inter-tumor cell heterogeneity. FUNDAMENTAL RESEARCH 2024; 4:1506-1514. [PMID: 39734523 PMCID: PMC11670719 DOI: 10.1016/j.fmre.2022.05.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Revised: 05/03/2022] [Accepted: 05/20/2022] [Indexed: 11/19/2022] Open
Abstract
Organoids are expected to function as effective human organ models for precision cancer studies and drug development. Currently, primary tissue-derived organoids, termed non-engineered organoids (NEOs), are produced by manual pipetting or liquid handling that compromises organoid-organoid homogeneity and organoid-tissue consistency. Droplet-based microfluidics enables automated organoid production with high organoid-organoid homogeneity, organoid-tissue consistency, and a significantly improved production spectrum. It takes advantage of droplet-encapsulation of defined populations of cells and droplet-rendered microstructures that guide cell self-organization. Herein, we studied the droplet-engineered organoids (DEOs), derived from mouse liver tissues and human liver tumors, by using transcriptional analysis and cellular deconvolution on bulk RNA-seq data. The characteristics of DEOs are compared with the parental liver tissues (or tumors) and NEOs. The DEOs are proven higher reproducibility and consistency with the parental tissues, have a high production spectrum and shortened modeling time, and possess inter-organoid homogeneity and inter-tumor cell heterogeneity.
Collapse
Affiliation(s)
- Haoran Zhao
- Tsinghua Shenzhen International Graduate School (SIGS), Tsinghua University, Shenzhen 518055, China
- Tsinghua-Berkeley Shenzhen Institute (TBSI), Shenzhen 518055, China
| | - Yifan Cheng
- Tsinghua Shenzhen International Graduate School (SIGS), Tsinghua University, Shenzhen 518055, China
- Tsinghua-Berkeley Shenzhen Institute (TBSI), Shenzhen 518055, China
| | - Jiawei Li
- Tsinghua Shenzhen International Graduate School (SIGS), Tsinghua University, Shenzhen 518055, China
- Tsinghua-Berkeley Shenzhen Institute (TBSI), Shenzhen 518055, China
| | - Jiaqi Zhou
- Tsinghua Shenzhen International Graduate School (SIGS), Tsinghua University, Shenzhen 518055, China
- Tsinghua-Berkeley Shenzhen Institute (TBSI), Shenzhen 518055, China
| | - Haowei Yang
- Tsinghua Shenzhen International Graduate School (SIGS), Tsinghua University, Shenzhen 518055, China
- Tsinghua-Berkeley Shenzhen Institute (TBSI), Shenzhen 518055, China
| | - Feng Yu
- Tsinghua Shenzhen International Graduate School (SIGS), Tsinghua University, Shenzhen 518055, China
- Tsinghua-Berkeley Shenzhen Institute (TBSI), Shenzhen 518055, China
| | - Feihong Yu
- Tsinghua Shenzhen International Graduate School (SIGS), Tsinghua University, Shenzhen 518055, China
| | - Davit Khutsishvili
- Tsinghua Shenzhen International Graduate School (SIGS), Tsinghua University, Shenzhen 518055, China
- Tsinghua-Berkeley Shenzhen Institute (TBSI), Shenzhen 518055, China
| | - Zitian Wang
- Tsinghua Shenzhen International Graduate School (SIGS), Tsinghua University, Shenzhen 518055, China
- Tsinghua-Berkeley Shenzhen Institute (TBSI), Shenzhen 518055, China
| | - Shengwei Jiang
- Tsinghua Shenzhen International Graduate School (SIGS), Tsinghua University, Shenzhen 518055, China
- Tsinghua-Berkeley Shenzhen Institute (TBSI), Shenzhen 518055, China
| | - Kaixin Tan
- Department of Chemical and Biological Engineering, Hong Kong University of Science and Technology, Hong Kong 999077, China
| | - Yi Kuang
- Department of Chemical and Biological Engineering, Hong Kong University of Science and Technology, Hong Kong 999077, China
- HKUST Shenzhen Research Institute, Shenzhen 518057, China
| | - Xinhui Xing
- Tsinghua Shenzhen International Graduate School (SIGS), Tsinghua University, Shenzhen 518055, China
| | - Shaohua Ma
- Tsinghua Shenzhen International Graduate School (SIGS), Tsinghua University, Shenzhen 518055, China
- Tsinghua-Berkeley Shenzhen Institute (TBSI), Shenzhen 518055, China
| |
Collapse
|
19
|
D’Orsi L, Capasso B, Lamacchia G, Pizzichini P, Ferranti S, Liverani A, Fontana C, Panunzi S, De Gaetano A, Lo Presti E. Recent Advances in Artificial Intelligence to Improve Immunotherapy and the Use of Digital Twins to Identify Prognosis of Patients with Solid Tumors. Int J Mol Sci 2024; 25:11588. [PMID: 39519142 PMCID: PMC11546512 DOI: 10.3390/ijms252111588] [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: 08/08/2024] [Revised: 10/21/2024] [Accepted: 10/23/2024] [Indexed: 11/16/2024] Open
Abstract
To date, the public health system has been impacted by the increasing costs of many diagnostic and therapeutic pathways due to limited resources. At the same time, we are constantly seeking to improve these paths through approaches aimed at personalized medicine. To achieve the required levels of diagnostic and therapeutic precision, it is necessary to integrate data from different sources and simulation platforms. Today, artificial intelligence (AI), machine learning (ML), and predictive computer models are more efficient at guiding decisions regarding better therapies and medical procedures. The evolution of these multiparametric and multimodal systems has led to the creation of digital twins (DTs). The goal of our review is to summarize AI applications in discovering new immunotherapies and developing predictive models for more precise immunotherapeutic decision-making. The findings from this literature review highlight that DTs, particularly predictive mathematical models, will be pivotal in advancing healthcare outcomes. Over time, DTs will indeed bring the benefits of diagnostic precision and personalized treatment to a broader spectrum of patients.
Collapse
Affiliation(s)
- Laura D’Orsi
- National Research Council of Italy, Institute for Systems Analysis and Computer Science “A. Ruberti”, BioMatLab, Via dei Taurini, 19, 00185 Rome, RM, Italy; (L.D.); (S.P.); (A.D.G.)
| | - Biagio Capasso
- Department of General Surgery, Policlinico Militare di Roma “Celio”, Piazza Celimontana, 50, 00184 Rome, RM, Italy; (B.C.); (S.F.)
| | - Giuseppe Lamacchia
- General Surgery Unit, Regina Apostolorum Hospital, Via S. Francesco d’Assisi, 50, 00041 Albano Laziale, RM, Italy; (G.L.); (A.L.)
| | - Paolo Pizzichini
- Department of Intensive Care Unit, Policlinico Militare di Roma “Celio”, Piazza Celimontana, 50, 00184 Rome, RM, Italy; (P.P.); (C.F.)
| | - Sergio Ferranti
- Department of General Surgery, Policlinico Militare di Roma “Celio”, Piazza Celimontana, 50, 00184 Rome, RM, Italy; (B.C.); (S.F.)
| | - Andrea Liverani
- General Surgery Unit, Regina Apostolorum Hospital, Via S. Francesco d’Assisi, 50, 00041 Albano Laziale, RM, Italy; (G.L.); (A.L.)
| | - Costantino Fontana
- Department of Intensive Care Unit, Policlinico Militare di Roma “Celio”, Piazza Celimontana, 50, 00184 Rome, RM, Italy; (P.P.); (C.F.)
| | - Simona Panunzi
- National Research Council of Italy, Institute for Systems Analysis and Computer Science “A. Ruberti”, BioMatLab, Via dei Taurini, 19, 00185 Rome, RM, Italy; (L.D.); (S.P.); (A.D.G.)
| | - Andrea De Gaetano
- National Research Council of Italy, Institute for Systems Analysis and Computer Science “A. Ruberti”, BioMatLab, Via dei Taurini, 19, 00185 Rome, RM, Italy; (L.D.); (S.P.); (A.D.G.)
- National Research Council of Italy, Institute for Biomedical Research and Innovation (CNR-IRIB), Via Ugo La Malfa, 153, 90146 Palermo, PA, Italy
- Department of Biomatics, Óbuda University, Bécsi Road 96/B, H-1034 Budapest, Hungary
| | - Elena Lo Presti
- National Research Council of Italy, Institute for Biomedical Research and Innovation (CNR-IRIB), Via Ugo La Malfa, 153, 90146 Palermo, PA, Italy
| |
Collapse
|
20
|
Zhang T, Rui W, Sun Y, Tian Y, Li Q, Zhang Q, Zhao Y, Liu Z, Wang T. Identification of nitric oxide-mediated necroptosis as the predominant death route in Parkinson's disease. MOLECULAR BIOMEDICINE 2024; 5:44. [PMID: 39443410 PMCID: PMC11499487 DOI: 10.1186/s43556-024-00213-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Accepted: 09/20/2024] [Indexed: 10/25/2024] Open
Abstract
Parkinson's disease (PD) involves multiple forms of neuronal cell death, but the dominant pathway involved in disease progression remains unclear. This study employed RNA sequencing (RNA-seq) of brain tissue to explore gene expression patterns across different stages of PD. Using the Scaden deep learning algorithm, we predicted neurocyte subtypes and modelled dynamic interactions for five classic cell death pathways to identify the predominant routes of neuronal death during PD progression. Our cell type-specific analysis revealed an increasing shift towards necroptosis, which was strongly correlated with nitric oxide synthase (NOS) expression across most neuronal subtypes. In vitro experiments confirmed that nitric oxide (NO) is a key mediator of necroptosis, leading to nuclear shrinkage and decreased mitochondrial membrane potential via phosphorylation of the PIP1/PIP3/MLKL signalling cascade. Importantly, specific necroptosis inhibitors significantly mitigated neuronal damage in both in vitro and in vivo PD models. Further analysis revealed that NO-mediated necroptosis is prevalent in early-onset Alzheimer's disease (AD) and amyotrophic lateral sclerosis (ALS) and across multiple brain regions but not in brain tumours. Our findings suggest that NO-mediated necroptosis is a critical pathway in PD and other neurodegenerative disorders, providing potential targets for therapeutic intervention.
Collapse
Affiliation(s)
- Ting Zhang
- School of Medicine, Shihezi University, Shihezi, 832000, China
- Key Laboratory of Xinjiang Endemic and Ethnic Diseases, School of Medicine, Shihezi University, Shihezi, 832000, China
| | - Wenjing Rui
- Changping Laboratory, Beijing, 102206, China
| | - Yue Sun
- School of Medicine, Shihezi University, Shihezi, 832000, China
- Key Laboratory of Xinjiang Endemic and Ethnic Diseases, School of Medicine, Shihezi University, Shihezi, 832000, China
- Prenatal Diagnosis Center of Urumqi Maternal and Child Health Hospital, Urumuqi, 830000, Xinjiang, China
| | - Yunyun Tian
- School of Medicine, Shihezi University, Shihezi, 832000, China
- Key Laboratory of Xinjiang Endemic and Ethnic Diseases, School of Medicine, Shihezi University, Shihezi, 832000, China
| | - Qiaoyan Li
- School of Medicine, Shihezi University, Shihezi, 832000, China
- Key Laboratory of Xinjiang Endemic and Ethnic Diseases, School of Medicine, Shihezi University, Shihezi, 832000, China
| | - Qian Zhang
- School of Medicine, Shihezi University, Shihezi, 832000, China
- Key Laboratory of Xinjiang Endemic and Ethnic Diseases, School of Medicine, Shihezi University, Shihezi, 832000, China
| | - Yanchun Zhao
- School of Medicine, Shihezi University, Shihezi, 832000, China
- Key Laboratory of Xinjiang Endemic and Ethnic Diseases, School of Medicine, Shihezi University, Shihezi, 832000, China
| | - Zongzhi Liu
- Changping Laboratory, Beijing, 102206, China.
- Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing, 100101, China.
| | - Tiepeng Wang
- School of Medicine, Shihezi University, Shihezi, 832000, China.
- Key Laboratory of Xinjiang Endemic and Ethnic Diseases, School of Medicine, Shihezi University, Shihezi, 832000, China.
- Key Laboratory of Biomacromolecules (CAS), National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China.
| |
Collapse
|
21
|
Covre LP, Fantecelle CH, Garcia de Moura R, Oliveira Lopes P, Sarmento IV, Freire-de-Lima CG, Decote-Ricardo D, de Matos Guedes HL, da Fonsceca-Martins AM, de Carvalho LP, de Carvalho EM, Mosser DM, Falqueto A, Akbar AN, Gomes DCO. Lesional senescent CD4 + T cells mediate bystander cytolysis and contribute to the skin pathology of human cutaneous leishmaniasis. Front Immunol 2024; 15:1475146. [PMID: 39497830 PMCID: PMC11532160 DOI: 10.3389/fimmu.2024.1475146] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Accepted: 09/23/2024] [Indexed: 11/07/2024] Open
Abstract
Cytotoxic activity is a hallmark of the immunopathogenesis in human cutaneous leishmaniasis (CL). In this study, we identified accumulation of CD4+ granzyme B producing T cells with increased cytotoxic capacity in CL lesions. These cells showed enhanced expression of activating NK receptors (NKG2D and NKG2C), diminished expression of inhibitory NKG2A, along with the upregulation of the senescence marker CD57. Notably, CD4+ T cells freshly isolated from CL lesions demonstrated remarkable capacity to mediate NL-like bystander cytolysis. Phenotypic analyses revealed that lesional CD4+ T cells are mainly composed of late-differentiated effector (CD27-CD45RA-) and terminally differentiated (senescent) TEMRA (CD27-CD45RA+) subsets. Interestingly, the TEMRA CD4+ T cells exhibited higher expression of granzyme B and CD107a. Collectively, our results provide the first evidence that senescent cytotoxic CD4+ T cells may support the skin pathology of human cutaneous leishmaniasis and, together with our previous findings, support the notion that multiple subsets of cytotoxic senescent cells may be involved in inducing the skin lesions in these patients.
Collapse
Affiliation(s)
- Luciana Polaco Covre
- Núcleo de Doenças Infecciosas, Universidade Federal do Espírito Santo, Vitória, Brazil
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
- Division of Medicine, University College London, London, United Kingdom
| | | | | | - Paola Oliveira Lopes
- Núcleo de Biotecnologia, Universidade Federal do Espírito Santo, Vitória, Brazil
| | | | | | - Debora Decote-Ricardo
- Departamento de Veterinária, Universidade Federal Rural do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Herbert Leonel de Matos Guedes
- Instituto de Microbiologia Professor Paulo de Goes, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
- Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro, Brazil
| | | | | | | | - David M. Mosser
- Department of Cell Biology and Molecular Genetics, University of Maryland, College Park, MD, United States
| | - Aloisio Falqueto
- Departamento de Medicina Social, Universidade Federal do Espírito Santo, Vitória, Brazil
| | - Arne N. Akbar
- Division of Medicine, University College London, London, United Kingdom
| | - Daniel Claudio Oliveira Gomes
- Núcleo de Doenças Infecciosas, Universidade Federal do Espírito Santo, Vitória, Brazil
- Núcleo de Biotecnologia, Universidade Federal do Espírito Santo, Vitória, Brazil
| |
Collapse
|
22
|
Gao Y, Li J, Cheng W, Diao T, Liu H, Bo Y, Liu C, Zhou W, Chen M, Zhang Y, Liu Z, Han W, Chen R, Peng J, Zhu L, Hou W, Zhang Z. Cross-tissue human fibroblast atlas reveals myofibroblast subtypes with distinct roles in immune modulation. Cancer Cell 2024; 42:1764-1783.e10. [PMID: 39303725 DOI: 10.1016/j.ccell.2024.08.020] [Citation(s) in RCA: 32] [Impact Index Per Article: 32.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/28/2023] [Revised: 07/28/2024] [Accepted: 08/28/2024] [Indexed: 09/22/2024]
Abstract
Fibroblasts, known for their functional diversity, play crucial roles in inflammation and cancer. In this study, we conduct comprehensive single-cell RNA sequencing analyses on fibroblast cells from 517 human samples, spanning 11 tissue types and diverse pathological states. We identify distinct fibroblast subpopulations with universal and tissue-specific characteristics. Pathological conditions lead to significant shifts in fibroblast compositions, including the expansion of immune-modulating fibroblasts during inflammation and tissue-remodeling myofibroblasts in cancer. Within the myofibroblast category, we identify four transcriptionally distinct subpopulations originating from different developmental origins, with LRRC15+ myofibroblasts displaying terminally differentiated features. Both LRRC15+ and MMP1+ myofibroblasts demonstrate pro-tumor potential that contribute to the immune-excluded and immune-suppressive tumor microenvironments (TMEs), whereas PI16+ fibroblasts show potential anti-tumor functions in adjacent non-cancerous regions. Fibroblast-subtype compositions define patient subtypes with distinct clinical outcomes. This study advances our understanding of fibroblast biology and suggests potential therapeutic strategies for targeting specific fibroblast subsets in cancer treatment.
Collapse
Affiliation(s)
- Yang Gao
- School of Chemical Biology and Biotechnology, Shenzhen Graduate School, Peking University, Shenzhen 518055, China; Institute of Cancer Research, Shenzhen Bay Laboratory, Shenzhen 518132, China
| | - Jianan Li
- Changping Laboratory, Beijing 102206, China
| | - Wenfeng Cheng
- Biomedical Pioneering Innovation Center (BIOPIC), Academy for Advanced Interdisciplinary Studies, and School of Life Sciences, Peking University, Beijing 100871, China
| | - Tian Diao
- Biomedical Pioneering Innovation Center (BIOPIC), Academy for Advanced Interdisciplinary Studies, and School of Life Sciences, Peking University, Beijing 100871, China
| | - Huilan Liu
- Biomedical Pioneering Innovation Center (BIOPIC), Academy for Advanced Interdisciplinary Studies, and School of Life Sciences, Peking University, Beijing 100871, China
| | - Yufei Bo
- Biomedical Pioneering Innovation Center (BIOPIC), Academy for Advanced Interdisciplinary Studies, and School of Life Sciences, Peking University, Beijing 100871, China
| | - Chang Liu
- Biomedical Pioneering Innovation Center (BIOPIC), Academy for Advanced Interdisciplinary Studies, and School of Life Sciences, Peking University, Beijing 100871, China
| | - Wei Zhou
- Institute of Cancer Research, Shenzhen Bay Laboratory, Shenzhen 518132, China
| | - Minmin Chen
- Institute of Cancer Research, Shenzhen Bay Laboratory, Shenzhen 518132, China
| | - Yuanyuan Zhang
- Biomedical Pioneering Innovation Center (BIOPIC), Academy for Advanced Interdisciplinary Studies, and School of Life Sciences, Peking University, Beijing 100871, China; State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Zhihua Liu
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Weidong Han
- Department of Bio-therapeutic, the First Medical Center, Chinese PLA General Hospital, Beijing 100853, China
| | - Rufu Chen
- Department of Pancreatic Surgery, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou 510180, China
| | - Jirun Peng
- Department of Surgery, Beijing Shijitan Hospital, Capital Medical University, Beijing 100038, China; Ninth School of Clinical Medicine, Peking University, Beijing 100038, China
| | - Linnan Zhu
- Biomedical Pioneering Innovation Center (BIOPIC), Academy for Advanced Interdisciplinary Studies, and School of Life Sciences, Peking University, Beijing 100871, China
| | - Wenhong Hou
- The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan 523710, China.
| | - Zemin Zhang
- Biomedical Pioneering Innovation Center (BIOPIC), Academy for Advanced Interdisciplinary Studies, and School of Life Sciences, Peking University, Beijing 100871, China.
| |
Collapse
|
23
|
Todhunter ME, Jubair S, Verma R, Saqe R, Shen K, Duffy B. Artificial intelligence and machine learning applications for cultured meat. Front Artif Intell 2024; 7:1424012. [PMID: 39381621 PMCID: PMC11460582 DOI: 10.3389/frai.2024.1424012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Accepted: 08/21/2024] [Indexed: 10/10/2024] Open
Abstract
Cultured meat has the potential to provide a complementary meat industry with reduced environmental, ethical, and health impacts. However, major technological challenges remain which require time-and resource-intensive research and development efforts. Machine learning has the potential to accelerate cultured meat technology by streamlining experiments, predicting optimal results, and reducing experimentation time and resources. However, the use of machine learning in cultured meat is in its infancy. This review covers the work available to date on the use of machine learning in cultured meat and explores future possibilities. We address four major areas of cultured meat research and development: establishing cell lines, cell culture media design, microscopy and image analysis, and bioprocessing and food processing optimization. In addition, we have included a survey of datasets relevant to CM research. This review aims to provide the foundation necessary for both cultured meat and machine learning scientists to identify research opportunities at the intersection between cultured meat and machine learning.
Collapse
Affiliation(s)
| | - Sheikh Jubair
- Alberta Machine Intelligence Institute, Edmonton, AB, Canada
| | - Ruchika Verma
- Alberta Machine Intelligence Institute, Edmonton, AB, Canada
| | - Rikard Saqe
- Department of Biology, University of Waterloo, Waterloo, ON, Canada
| | - Kevin Shen
- Department of Mathematics, University of Waterloo, Waterloo, ON, Canada
| | | |
Collapse
|
24
|
Yasumizu Y, Kinoshita M, Zhang MJ, Motooka D, Suzuki K, Nojima S, Koizumi N, Okuzaki D, Funaki S, Shintani Y, Ohkura N, Morii E, Okuno T, Mochizuki H. Spatial transcriptomics elucidates medulla niche supporting germinal center response in myasthenia gravis-associated thymoma. Cell Rep 2024; 43:114677. [PMID: 39180749 DOI: 10.1016/j.celrep.2024.114677] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Revised: 07/30/2024] [Accepted: 08/08/2024] [Indexed: 08/26/2024] Open
Abstract
Myasthenia gravis (MG) is etiologically associated with thymus abnormalities, but its pathology in the thymus remains unclear. In this study, we attempt to narrow down the features associated with MG using spatial transcriptome analysis of thymoma and thymic hyperplasia samples. We find that the majority of thymomas are constituted by the cortical region. However, the small medullary region is enlarged in seropositive thymomas and contains polygenic enrichment and MG-specific germinal center structures. Neuromuscular medullary thymic epithelial cells, previously identified as MG-specific autoantigen-producing cells, are enriched in the cortico-medullary junction. The medulla is characterized by a specific chemokine pattern and immune cell composition, including migratory dendritic cells and effector regulatory T cells. Similar germinal center structures and immune microenvironments are also observed in the thymic hyperplasia medulla. This study shows that the medulla and junction areas are linked to MG pathology and provides insights into future MG research.
Collapse
Affiliation(s)
- Yoshiaki Yasumizu
- Department of Neurology, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan; Department of Experimental Immunology, Immunology Frontier Research Center, Osaka University, Suita, Osaka, Japan; Integrated Frontier Research for Medical Science Division, Institute for Open and Transdisciplinary Research Initiatives (OTRI), Osaka University, Suita, Osaka, Japan; Department of Neurology, Yale School of Medicine, New Haven, CT, USA.
| | - Makoto Kinoshita
- Department of Neurology, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
| | - Martin Jinye Zhang
- Ray and Stephanie Lane Computational Biology Department, School of Computer Science, Carnegie Mellon University, Pittsburgh, PA, USA; Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Daisuke Motooka
- Integrated Frontier Research for Medical Science Division, Institute for Open and Transdisciplinary Research Initiatives (OTRI), Osaka University, Suita, Osaka, Japan; Genome Information Research Center, Research Institute for Microbial Diseases, Osaka University, Suita, Osaka, Japan
| | - Koichiro Suzuki
- BIKEN-RIMD NGS Laboratory, Research Institute for Microbial Diseases, Osaka University, Suita, Japan; Biomedical Science Center, The Research Foundation for Microbial Diseases of Osaka University (BIKEN), Suita, Japan
| | - Satoshi Nojima
- Department of Pathology, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
| | - Naoshi Koizumi
- Department of Neurology, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
| | - Daisuke Okuzaki
- Integrated Frontier Research for Medical Science Division, Institute for Open and Transdisciplinary Research Initiatives (OTRI), Osaka University, Suita, Osaka, Japan; Genome Information Research Center, Research Institute for Microbial Diseases, Osaka University, Suita, Osaka, Japan
| | - Soichiro Funaki
- Department of General Thoracic Surgery, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
| | - Yasushi Shintani
- Department of General Thoracic Surgery, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
| | - Naganari Ohkura
- Department of Experimental Immunology, Immunology Frontier Research Center, Osaka University, Suita, Osaka, Japan; Department of Frontier Research in Tumor Immunology, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
| | - Eiichi Morii
- Department of Pathology, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
| | - Tatsusada Okuno
- Department of Neurology, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan.
| | - Hideki Mochizuki
- Department of Neurology, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan; Integrated Frontier Research for Medical Science Division, Institute for Open and Transdisciplinary Research Initiatives (OTRI), Osaka University, Suita, Osaka, Japan
| |
Collapse
|
25
|
Cheng Y, Xu SM, Santucci K, Lindner G, Janitz M. Machine learning and related approaches in transcriptomics. Biochem Biophys Res Commun 2024; 724:150225. [PMID: 38852503 DOI: 10.1016/j.bbrc.2024.150225] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2024] [Revised: 05/18/2024] [Accepted: 06/03/2024] [Indexed: 06/11/2024]
Abstract
Data acquisition for transcriptomic studies used to be the bottleneck in the transcriptomic analytical pipeline. However, recent developments in transcriptome profiling technologies have increased researchers' ability to obtain data, resulting in a shift in focus to data analysis. Incorporating machine learning to traditional analytical methods allows the possibility of handling larger volumes of complex data more efficiently. Many bioinformaticians, especially those unfamiliar with ML in the study of human transcriptomics and complex biological systems, face a significant barrier stemming from their limited awareness of the current landscape of ML utilisation in this field. To address this gap, this review endeavours to introduce those individuals to the general types of ML, followed by a comprehensive range of more specific techniques, demonstrated through examples of their incorporation into analytical pipelines for human transcriptome investigations. Important computational aspects such as data pre-processing, task formulation, results (performance of ML models), and validation methods are encompassed. In hope of better practical relevance, there is a strong focus on studies published within the last five years, almost exclusively examining human transcriptomes, with outcomes compared with standard non-ML tools.
Collapse
Affiliation(s)
- Yuning Cheng
- School of Biotechnology and Biomolecular Sciences, University of New South Wales, Sydney, NSW, 2052, Australia
| | - Si-Mei Xu
- School of Biotechnology and Biomolecular Sciences, University of New South Wales, Sydney, NSW, 2052, Australia
| | - Kristina Santucci
- School of Biotechnology and Biomolecular Sciences, University of New South Wales, Sydney, NSW, 2052, Australia
| | - Grace Lindner
- School of Biotechnology and Biomolecular Sciences, University of New South Wales, Sydney, NSW, 2052, Australia
| | - Michael Janitz
- School of Biotechnology and Biomolecular Sciences, University of New South Wales, Sydney, NSW, 2052, Australia.
| |
Collapse
|
26
|
Liu Y, Vierkant R, Bhagwate A, Jons W, Stallings-Mann M, McCauley B, Carter J, Stephens M, Pfrender M, Littlepage L, Radisky D, Cunningham J, Degnim A, Winham S, Wang C. Evaluating cell type deconvolution in FFPE breast tissue: application to benign breast disease. NAR Genom Bioinform 2024; 6:lqae098. [PMID: 40162103 PMCID: PMC11952925 DOI: 10.1093/nargab/lqae098] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Revised: 07/11/2024] [Accepted: 07/23/2024] [Indexed: 04/02/2025] Open
Abstract
Transcriptome profiling using RNA sequencing (RNA-seq) of bulk formalin-fixed paraffin-embedded (FFPE) tissue blocks is a standard method in biomedical research. However, when used on tissues with diverse cell type compositions, it yields averaged gene expression profiles, complicating biomarker identification due to variations in cell proportions. To address the need for optimized strategies for defining individual cell type compositions from bulk FFPE samples, we constructed single-cell RNA-seq reference data for breast tissue and tested cell type deconvolution methods. Initial simulation experiments showed similar performances across multiple commonly used deconvolution methods. However, the introduction of FFPE artifacts significantly impacted their performances, with a root mean squared error (RMSE) ranging between 0.04 and 0.17. Scaden, a deep learning-based method, consistently outperformed the others, demonstrating robustness against FFPE artifacts. Testing these methods on our 62-sample RNA-seq benign breast disease cohort in which cell type composition was estimated using digital pathology approaches, we found that pre-filtering of the reference data enhanced the accuracy of most methods, realizing up to a 32% reduction in RMSE. To support further research efforts in this domain, we introduce SCdeconR, an R package designed for streamlined cell type deconvolution assessments and downstream analyses.
Collapse
Affiliation(s)
- Yuanhang Liu
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN 55905, USA
| | - Robert A Vierkant
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN 55905, USA
| | - Aditya Bhagwate
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN 55905, USA
| | - William A Jons
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN 55905, USA
- Biomedical Engineering and Physiology Graduate Program, Mayo Clinic Graduate School of Biomedical Sciences, Rochester, MN 55905, USA
| | | | - Bryan M McCauley
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN 55905, USA
| | - Jodi M Carter
- Department of Laboratory Medicine & Pathology, University of Alberta, Edmonton, AB T6G 2R3, Canada
| | - Melissa T Stephens
- Genomics and Bioinformatics Core Facility, University of Notre Dame, Notre Dame, IN 46556, USA
| | - Michael E Pfrender
- Department of Biological Sciences, 109B Galvin Life Science Center, University of Notre Dame, Notre Dame, IN 46556, USA
| | - Laurie E Littlepage
- Department of Chemistry and Biochemistry, Harper Cancer Research Institute, University of Notre Dame, Notre Dame, IN 46556, USA
| | - Derek C Radisky
- Department of Cancer Biology, Mayo Clinic, 4500 San Pablo Road, Jacksonville, FL 32224, USA
| | - Julie M Cunningham
- Department of Laboratory Medicine and Pathology, Mayo Clinic, 200 1st Street SW, Rochester, MN 55905, USA
| | - Amy C Degnim
- Department of Surgery, Mayo Clinic, 200 1st Street SW, Rochester, MN 55905, USA
| | - Stacey J Winham
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN 55905, USA
| | - Chen Wang
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN 55905, USA
| |
Collapse
|
27
|
Li J, Wang D, Tang F, Ling X, Zhang W, Zhang Z. Pan-cancer integrative analyses dissect the remodeling of endothelial cells in human cancers. Natl Sci Rev 2024; 11:nwae231. [PMID: 39345334 PMCID: PMC11429526 DOI: 10.1093/nsr/nwae231] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Revised: 06/04/2024] [Accepted: 06/23/2024] [Indexed: 10/01/2024] Open
Abstract
Therapeutics targeting tumor endothelial cells (TECs) have been explored for decades, with only suboptimal efficacy achieved, partly due to an insufficient understanding of the TEC heterogeneity across cancer patients. We integrated single-cell RNA-seq data of 575 cancer patients from 19 solid tumor types, comprehensively charting the TEC phenotypic diversities. Our analyses uncovered underappreciated compositional and functional heterogeneity in TECs from a pan-cancer perspective. Two subsets, CXCR4 + tip cells and SELE + veins, represented the prominent angiogenic and proinflammatory phenotypes of TECs, respectively. They exhibited distinct spatial organization patterns, and compared to adjacent non-tumor tissues, tumor tissue showed an increased prevalence of CXCR4 + tip cells, yet with SELE + veins depleted. Such functional and spatial characteristics underlie their differential associations with the response of anti-angiogenic therapies and immunotherapies. Our integrative resources and findings open new avenues to understand and clinically intervene in the tumor vasculature.
Collapse
Affiliation(s)
- Jinhu Li
- Biomedical Pioneering Innovation Center (BIOPIC), School of Life Sciences, Peking University, Beijing 100871, China
| | - Dongfang Wang
- Biomedical Pioneering Innovation Center (BIOPIC), School of Life Sciences, Peking University, Beijing 100871, China
| | - Fei Tang
- Biomedical Pioneering Innovation Center (BIOPIC), School of Life Sciences, Peking University, Beijing 100871, China
- Academy for Advanced Interdisciplinary Studies, Peking University, Beijing 100871, China
| | - Xinnan Ling
- Biomedical Pioneering Innovation Center (BIOPIC), School of Life Sciences, Peking University, Beijing 100871, China
| | - Wenjie Zhang
- Biomedical Pioneering Innovation Center (BIOPIC), School of Life Sciences, Peking University, Beijing 100871, China
| | - Zemin Zhang
- Biomedical Pioneering Innovation Center (BIOPIC), School of Life Sciences, Peking University, Beijing 100871, China
- Academy for Advanced Interdisciplinary Studies, Peking University, Beijing 100871, China
| |
Collapse
|
28
|
White BS, de Reyniès A, Newman AM, Waterfall JJ, Lamb A, Petitprez F, Lin Y, Yu R, Guerrero-Gimenez ME, Domanskyi S, Monaco G, Chung V, Banerjee J, Derrick D, Valdeolivas A, Li H, Xiao X, Wang S, Zheng F, Yang W, Catania CA, Lang BJ, Bertus TJ, Piermarocchi C, Caruso FP, Ceccarelli M, Yu T, Guo X, Bletz J, Coller J, Maecker H, Duault C, Shokoohi V, Patel S, Liliental JE, Simon S, Saez-Rodriguez J, Heiser LM, Guinney J, Gentles AJ. Community assessment of methods to deconvolve cellular composition from bulk gene expression. Nat Commun 2024; 15:7362. [PMID: 39191725 PMCID: PMC11350143 DOI: 10.1038/s41467-024-50618-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Accepted: 07/11/2024] [Indexed: 08/29/2024] Open
Abstract
We evaluate deconvolution methods, which infer levels of immune infiltration from bulk expression of tumor samples, through a community-wide DREAM Challenge. We assess six published and 22 community-contributed methods using in vitro and in silico transcriptional profiles of admixed cancer and healthy immune cells. Several published methods predict most cell types well, though they either were not trained to evaluate all functional CD8+ T cell states or do so with low accuracy. Several community-contributed methods address this gap, including a deep learning-based approach, whose strong performance establishes the applicability of this paradigm to deconvolution. Despite being developed largely using immune cells from healthy tissues, deconvolution methods predict levels of tumor-derived immune cells well. Our admixed and purified transcriptional profiles will be a valuable resource for developing deconvolution methods, including in response to common challenges we observe across methods, such as sensitive identification of functional CD4+ T cell states.
Collapse
Affiliation(s)
- Brian S White
- Sage Bionetworks, Seattle, WA, USA
- The Jackson Laboratory for Genomic Medicine, Farmington, CT, USA
| | - Aurélien de Reyniès
- Centre de Recherche des Cordeliers, INSERM U1138, Université Paris Cité, Paris, France
| | - Aaron M Newman
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University, Stanford, CA, USA
- Department of Biomedical Data Science, Stanford University, Stanford, CA, USA
| | - Joshua J Waterfall
- INSERM U830 and Translational Research Department, Institut Curie, PSL Research University, Paris, France
| | | | - Florent Petitprez
- Programme Cartes d'Identité des Tumeurs, Ligue Nationale Contre le Cancer, Paris, France
- MRC Centre for Reproductive Health, the Queen's Medical Research Institute, University of Edinburgh, Edinburgh, UK
| | - Yating Lin
- Xiamen University, Xiamen, Fujian, China
| | | | - Martin E Guerrero-Gimenez
- Institute of Biochemistry and Biotechnology, School of Medicine, National University of Cuyo, Mendoza, Argentina
| | | | - Gianni Monaco
- BIOGEM Institute of Molecular Biology and Genetics, Ariano Irpino, AV, Italy
| | | | | | - Daniel Derrick
- Department of Biomedical Engineering, Knight Cancer Institute, Oregon Health & Science University, Portland, OR, USA
| | - Alberto Valdeolivas
- Heidelberg University, Faculty of Medicine, and Heidelberg University Hospital, Institute for Computational Biomedicine, Bioquant, Heidelberg, Germany
| | - Haojun Li
- Xiamen University, Xiamen, Fujian, China
| | - Xu Xiao
- Xiamen University, Xiamen, Fujian, China
| | - Shun Wang
- Department of Pathology, Cancer Hospital, Chinese Aacdemy of Medical Science, Beijing, China
| | | | | | - Carlos A Catania
- Laboratory of Intelligent Systems (LABSIN), Engineering School, National University of Cuyo, Mendoza, Argentina
| | - Benjamin J Lang
- Department of Radiation Oncology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | | | | | - Francesca P Caruso
- BIOGEM Institute of Molecular Biology and Genetics, Ariano Irpino, AV, Italy
| | - Michele Ceccarelli
- BIOGEM Institute of Molecular Biology and Genetics, Ariano Irpino, AV, Italy
- Sylvester Comprehensive Cancer Center, Department of Public Health Sciences, University of Miami Miller School of Medicine, Miami, Florida, USA
| | | | | | | | - John Coller
- Stanford Functional Genomics Facility, Stanford University School of Medicine, Stanford, CA, USA
| | - Holden Maecker
- Institute for Immunity, Transplantation, and Infection, Stanford University School of Medicine, Stanford, CA, USA
| | - Caroline Duault
- Institute for Immunity, Transplantation, and Infection, Stanford University School of Medicine, Stanford, CA, USA
| | - Vida Shokoohi
- Stanford Functional Genomics Facility, Stanford University School of Medicine, Stanford, CA, USA
| | - Shailja Patel
- Translational Applications Service Center, Stanford University School of Medicine, Stanford, CA, USA
| | - Joanna E Liliental
- Translational Applications Service Center, Stanford University School of Medicine, Stanford, CA, USA
| | | | - Julio Saez-Rodriguez
- Heidelberg University, Faculty of Medicine, and Heidelberg University Hospital, Institute for Computational Biomedicine, Bioquant, Heidelberg, Germany
| | - Laura M Heiser
- Department of Biomedical Engineering, Knight Cancer Institute, Oregon Health & Science University, Portland, OR, USA
| | | | - Andrew J Gentles
- Department of Biomedical Data Science, Stanford University, Stanford, CA, USA.
- Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA.
- Department of Pathology, Stanford University, Stanford, CA, USA.
| |
Collapse
|
29
|
Sorek G, Haim Y, Chalifa-Caspi V, Lazarescu O, Ziv-Agam M, Hagemann T, Nono Nankam PA, Blüher M, Liberty IF, Dukhno O, Kukeev I, Yeger-Lotem E, Rudich A, Levin L. sNucConv: A bulk RNA-seq deconvolution method trained on single-nucleus RNA-seq data to estimate cell-type composition of human adipose tissues. iScience 2024; 27:110368. [PMID: 39071890 PMCID: PMC11277759 DOI: 10.1016/j.isci.2024.110368] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Revised: 02/27/2024] [Accepted: 06/21/2024] [Indexed: 07/30/2024] Open
Abstract
Deconvolution algorithms mostly rely on single-cell RNA-sequencing (scRNA-seq) data applied onto bulk RNA-sequencing (bulk RNA-seq) to estimate tissues' cell-type composition, with performance accuracy validated on deposited databases. Adipose tissues' cellular composition is highly variable, and adipocytes can only be captured by single-nucleus RNA-sequencing (snRNA-seq). Here we report the development of sNucConv, a Scaden deep-learning-based deconvolution tool, trained using 5 hSAT and 7 hVAT snRNA-seq-based data corrected by (i) snRNA-seq/bulk RNA-seq highly correlated genes and (ii) individual cell-type regression models. Applying sNucConv on our bulk RNA-seq data resulted in cell-type proportion estimation of 15 and 13 cell types, with accuracy of R = 0.93 (range: 0.76-0.97) and R = 0.95 (range: 0.92-0.98) for hVAT and hSAT, respectively. This performance level was further validated on an independent set of samples (5 hSAT; 5 hVAT). The resulting model was depot specific, reflecting depot differences in gene expression patterns. Jointly, sNucConv provides proof-of-concept for producing validated deconvolution models for tissues un-amenable to scRNA-seq.
Collapse
Affiliation(s)
- Gil Sorek
- Bioinformatics Core Facility, llse Katz Institute for Nanoscale Science and Technology, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Yulia Haim
- Department of Clinical Biochemistry and Pharmacology, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Vered Chalifa-Caspi
- Bioinformatics Core Facility, llse Katz Institute for Nanoscale Science and Technology, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Or Lazarescu
- Department of Clinical Biochemistry and Pharmacology, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Maya Ziv-Agam
- Department of Clinical Biochemistry and Pharmacology, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Tobias Hagemann
- Helmholtz Institute for Metabolic, Obesity and Vascular Research (HI-MAG) of the Helmholtz Zentrum München at the University of Leipzig and University Hospital Leipzig, Leipzig, Germany
| | - Pamela Arielle Nono Nankam
- Helmholtz Institute for Metabolic, Obesity and Vascular Research (HI-MAG) of the Helmholtz Zentrum München at the University of Leipzig and University Hospital Leipzig, Leipzig, Germany
| | - Matthias Blüher
- Helmholtz Institute for Metabolic, Obesity and Vascular Research (HI-MAG) of the Helmholtz Zentrum München at the University of Leipzig and University Hospital Leipzig, Leipzig, Germany
| | - Idit F. Liberty
- Soroka University Medical Center, and the Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Oleg Dukhno
- Soroka University Medical Center, and the Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Ivan Kukeev
- Soroka University Medical Center, and the Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Esti Yeger-Lotem
- Department of Clinical Biochemistry and Pharmacology, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Assaf Rudich
- Department of Clinical Biochemistry and Pharmacology, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Liron Levin
- Bioinformatics Core Facility, llse Katz Institute for Nanoscale Science and Technology, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| |
Collapse
|
30
|
Wang J, Fonseca GJ, Ding J. scSemiProfiler: Advancing large-scale single-cell studies through semi-profiling with deep generative models and active learning. Nat Commun 2024; 15:5989. [PMID: 39013867 PMCID: PMC11252419 DOI: 10.1038/s41467-024-50150-1] [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: 11/29/2023] [Accepted: 06/28/2024] [Indexed: 07/18/2024] Open
Abstract
Single-cell sequencing is a crucial tool for dissecting the cellular intricacies of complex diseases. Its prohibitive cost, however, hampers its application in expansive biomedical studies. Traditional cellular deconvolution approaches can infer cell type proportions from more affordable bulk sequencing data, yet they fall short in providing the detailed resolution required for single-cell-level analyses. To overcome this challenge, we introduce "scSemiProfiler", an innovative computational framework that marries deep generative models with active learning strategies. This method adeptly infers single-cell profiles across large cohorts by fusing bulk sequencing data with targeted single-cell sequencing from a few rigorously chosen representatives. Extensive validation across heterogeneous datasets verifies the precision of our semi-profiling approach, aligning closely with true single-cell profiling data and empowering refined cellular analyses. Originally developed for extensive disease cohorts, "scSemiProfiler" is adaptable for broad applications. It provides a scalable, cost-effective solution for single-cell profiling, facilitating in-depth cellular investigation in various biological domains.
Collapse
Affiliation(s)
- Jingtao Wang
- Meakins-Christe Laboratories, Research Institute of McGill University Health Centre, 1001 Decarie Blvd, Montreal, H4A 3J1, Quebec, Canada
- Department of Medicine, Division of Experimental Medicine, McGill University, 1001 Decarie Blvd, Montreal, H4A 3J1, Quebec, Canada
| | - Gregory J Fonseca
- Meakins-Christe Laboratories, Research Institute of McGill University Health Centre, 1001 Decarie Blvd, Montreal, H4A 3J1, Quebec, Canada
- Department of Medicine, Division of Experimental Medicine, McGill University, 1001 Decarie Blvd, Montreal, H4A 3J1, Quebec, Canada
- Quantitative Life Sciences, McGill University, 845 Rue Sherbrooke Ouest, Montreal, H3A 0G4, Quebec, Canada
| | - Jun Ding
- Meakins-Christe Laboratories, Research Institute of McGill University Health Centre, 1001 Decarie Blvd, Montreal, H4A 3J1, Quebec, Canada.
- Department of Medicine, Division of Experimental Medicine, McGill University, 1001 Decarie Blvd, Montreal, H4A 3J1, Quebec, Canada.
- Quantitative Life Sciences, McGill University, 845 Rue Sherbrooke Ouest, Montreal, H3A 0G4, Quebec, Canada.
- School of Computer Science, McGill University, 3480 Rue University, Montreal, H3A 2A7, Quebec, Canada.
- Mila-Quebec AI Institute, 6666 Rue Saint-Urbain, Montreal, H2S 3H1, Quebec, Canada.
| |
Collapse
|
31
|
Fu Y, Tao J, Liu T, Liu Y, Qiu J, Su D, Wang R, Luo W, Cao Z, Weng G, Zhang T, Zhao Y. Unbiasedly decoding the tumor microenvironment with single-cell multiomics analysis in pancreatic cancer. Mol Cancer 2024; 23:140. [PMID: 38982491 PMCID: PMC11232163 DOI: 10.1186/s12943-024-02050-7] [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: 05/07/2024] [Accepted: 06/21/2024] [Indexed: 07/11/2024] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a highly aggressive malignancy with a poor prognosis and limited therapeutic options. Research on the tumor microenvironment (TME) of PDAC has propelled the development of immunotherapeutic and targeted therapeutic strategies with a promising future. The emergence of single-cell sequencing and mass spectrometry technologies, coupled with spatial omics, has collectively revealed the heterogeneity of the TME from a multiomics perspective, outlined the development trajectories of cell lineages, and revealed important functions of previously underrated myeloid cells and tumor stroma cells. Concurrently, these findings necessitated more refined annotations of biological functions at the cell cluster or single-cell level. Precise identification of all cell clusters is urgently needed to determine whether they have been investigated adequately and to identify target cell clusters with antitumor potential, design compatible treatment strategies, and determine treatment resistance. Here, we summarize recent research on the PDAC TME at the single-cell multiomics level, with an unbiased focus on the functions and potential classification bases of every cellular component within the TME, and look forward to the prospects of integrating single-cell multiomics data and retrospectively reusing bulk sequencing data, hoping to provide new insights into the PDAC TME.
Collapse
Affiliation(s)
- Yifan Fu
- General Surgery Department, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
- 4+4 Medical Doctor Program, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| | - Jinxin Tao
- General Surgery Department, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| | - Tao Liu
- General Surgery Department, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| | - Yueze Liu
- General Surgery Department, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| | - Jiangdong Qiu
- General Surgery Department, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| | - Dan Su
- General Surgery Department, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| | - Ruobing Wang
- General Surgery Department, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| | - Wenhao Luo
- General Surgery Department, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| | - Zhe Cao
- General Surgery Department, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| | - Guihu Weng
- General Surgery Department, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| | - Taiping Zhang
- General Surgery Department, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China.
- Clinical Immunology Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China.
| | - Yupei Zhao
- General Surgery Department, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China.
| |
Collapse
|
32
|
Hu M, Chikina M. Heterogeneous pseudobulk simulation enables realistic benchmarking of cell-type deconvolution methods. Genome Biol 2024; 25:169. [PMID: 38956606 PMCID: PMC11218230 DOI: 10.1186/s13059-024-03292-w] [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: 01/20/2023] [Accepted: 05/29/2024] [Indexed: 07/04/2024] Open
Abstract
BACKGROUND Computational cell type deconvolution enables the estimation of cell type abundance from bulk tissues and is important for understanding tissue microenviroment, especially in tumor tissues. With rapid development of deconvolution methods, many benchmarking studies have been published aiming for a comprehensive evaluation for these methods. Benchmarking studies rely on cell-type resolved single-cell RNA-seq data to create simulated pseudobulk datasets by adding individual cells-types in controlled proportions. RESULTS In our work, we show that the standard application of this approach, which uses randomly selected single cells, regardless of the intrinsic difference between them, generates synthetic bulk expression values that lack appropriate biological variance. We demonstrate why and how the current bulk simulation pipeline with random cells is unrealistic and propose a heterogeneous simulation strategy as a solution. The heterogeneously simulated bulk samples match up with the variance observed in real bulk datasets and therefore provide concrete benefits for benchmarking in several ways. We demonstrate that conceptual classes of deconvolution methods differ dramatically in their robustness to heterogeneity with reference-free methods performing particularly poorly. For regression-based methods, the heterogeneous simulation provides an explicit framework to disentangle the contributions of reference construction and regression methods to performance. Finally, we perform an extensive benchmark of diverse methods across eight different datasets and find BayesPrism and a hybrid MuSiC/CIBERSORTx approach to be the top performers. CONCLUSIONS Our heterogeneous bulk simulation method and the entire benchmarking framework is implemented in a user friendly package https://github.com/humengying0907/deconvBenchmarking and https://doi.org/10.5281/zenodo.8206516 , enabling further developments in deconvolution methods.
Collapse
Affiliation(s)
- Mengying Hu
- Department of Computational and Systems Biology, University of Pittsburgh, Pittsburgh, USA
- Joint Carnegie Mellon - University of Pittsburgh Computational Biology PhD Program, University of Pittsburgh, Pittsburgh, USA
| | - Maria Chikina
- Department of Computational and Systems Biology, University of Pittsburgh, Pittsburgh, USA.
- Joint Carnegie Mellon - University of Pittsburgh Computational Biology PhD Program, University of Pittsburgh, Pittsburgh, USA.
| |
Collapse
|
33
|
Zhang X, Chen R, Huo Z, Li W, Jiang M, Su G, Liu Y, Cai Y, Huang W, Xiong Y, Wang S. Blood-based molecular and cellular biomarkers of early response to neoadjuvant PD-1 blockade in patients with non-small cell lung cancer. Cancer Cell Int 2024; 24:225. [PMID: 38951894 PMCID: PMC11218110 DOI: 10.1186/s12935-024-03412-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2024] [Accepted: 06/22/2024] [Indexed: 07/03/2024] Open
Abstract
BACKGROUND Despite the improved survival observed in PD-1/PD-L1 blockade therapy, a substantial proportion of cancer patients, including those with non-small cell lung cancer (NSCLC), still lack a response. METHODS Transcriptomic profiling was conducted on a discovery cohort comprising 100 whole blood samples, as collected multiple times from 48 healthy controls (including 43 published data) and 31 NSCLC patients that under treatment with a combination of anti-PD-1 Tislelizumab and chemotherapy. Differentially expressed genes (DEGs), simulated immune cell subsets, and germline DNA mutational markers were identified from patients achieved a pathological complete response during the early treatment cycles. The predictive values of mutational markers were further validated in an independent immunotherapy cohort of 1661 subjects, and then confirmed in genetically matched lung cancer cell lines by a co-culturing model. RESULTS The gene expression of hundreds of DEGs (FDR p < 0.05, fold change < -2 or > 2) distinguished responders from healthy controls, indicating the potential to stratify patients utilizing early on-treatment features from blood. PD-1-mediated cell abundance changes in memory CD4 + and regulatory T cell subset were more significant or exclusively observed in responders. A panel of top-ranked genetic alterations showed significant associations with improved survival (p < 0.05) and heightened responsiveness to anti-PD-1 treatment in patient cohort and co-cultured cell lines. CONCLUSION This study discovered and validated peripheral blood-based biomarkers with evident predictive efficacy for early therapy response and patient stratification before treatment for neoadjuvant PD-1 blockade in NSCLC patients.
Collapse
Affiliation(s)
- Xi Zhang
- School of Life Science, Northwest University, Xi'an, Shaanxi, 710069, China.
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, School of Medicine, Northwest University, 710069, Shaanxi, Xi'an, China.
| | - Rui Chen
- School of Life Science, Northwest University, Xi'an, Shaanxi, 710069, China
| | - Zirong Huo
- School of Life Science, Northwest University, Xi'an, Shaanxi, 710069, China
| | - Wenqing Li
- School of Life Science, Northwest University, Xi'an, Shaanxi, 710069, China
| | - Mengju Jiang
- School of Life Science, Northwest University, Xi'an, Shaanxi, 710069, China
| | - Guodong Su
- School of Life Science, Northwest University, Xi'an, Shaanxi, 710069, China
| | - Yuru Liu
- School of Life Science, Northwest University, Xi'an, Shaanxi, 710069, China
| | - Yu Cai
- School of Life Science, Northwest University, Xi'an, Shaanxi, 710069, China
| | - Wuhao Huang
- Department of Lung Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin Lung Cancer Center, Tianjin, 300060, China
| | - Yuyan Xiong
- School of Life Science, Northwest University, Xi'an, Shaanxi, 710069, China
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, School of Medicine, Northwest University, 710069, Shaanxi, Xi'an, China
| | - Shengguang Wang
- Department of Lung Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin Lung Cancer Center, Tianjin, 300060, China.
| |
Collapse
|
34
|
Görtler F, Mensching-Buhr M, Skaar Ø, Schrod S, Sterr T, Schäfer A, Beißbarth T, Joshi A, Zacharias HU, Grellscheid SN, Altenbuchinger M. Adaptive digital tissue deconvolution. Bioinformatics 2024; 40:i100-i109. [PMID: 38940181 PMCID: PMC11256946 DOI: 10.1093/bioinformatics/btae263] [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] [Indexed: 06/29/2024] Open
Abstract
MOTIVATION The inference of cellular compositions from bulk and spatial transcriptomics data increasingly complements data analyses. Multiple computational approaches were suggested and recently, machine learning techniques were developed to systematically improve estimates. Such approaches allow to infer additional, less abundant cell types. However, they rely on training data which do not capture the full biological diversity encountered in transcriptomics analyses; data can contain cellular contributions not seen in the training data and as such, analyses can be biased or blurred. Thus, computational approaches have to deal with unknown, hidden contributions. Moreover, most methods are based on cellular archetypes which serve as a reference; e.g. a generic T-cell profile is used to infer the proportion of T-cells. It is well known that cells adapt their molecular phenotype to the environment and that pre-specified cell archetypes can distort the inference of cellular compositions. RESULTS We propose Adaptive Digital Tissue Deconvolution (ADTD) to estimate cellular proportions of pre-selected cell types together with possibly unknown and hidden background contributions. Moreover, ADTD adapts prototypic reference profiles to the molecular environment of the cells, which further resolves cell-type specific gene regulation from bulk transcriptomics data. We verify this in simulation studies and demonstrate that ADTD improves existing approaches in estimating cellular compositions. In an application to bulk transcriptomics data from breast cancer patients, we demonstrate that ADTD provides insights into cell-type specific molecular differences between breast cancer subtypes. AVAILABILITY AND IMPLEMENTATION A python implementation of ADTD and a tutorial are available at Gitlab and zenodo (doi:10.5281/zenodo.7548362).
Collapse
Affiliation(s)
- Franziska Görtler
- Computational Biology Unit, Department of Biological Sciences, University of Bergen, N-5008 Bergen, Norway
- Department of Oncology and Medical Physics, Haukeland University Hospital, 5021 Bergen, Norway
| | - Malte Mensching-Buhr
- Department of Medical Bioinformatics, University Medical Center Göttingen, 37075 Göttingen, Germany
| | - Ørjan Skaar
- Department of Informatics, Computational Biology Unit, University of Bergen, N-5008 Bergen, Norway
| | - Stefan Schrod
- Department of Medical Bioinformatics, University Medical Center Göttingen, 37075 Göttingen, Germany
| | - Thomas Sterr
- Institute of Theoretical Physics, University of Regensburg, 93053 Regensburg, Germany
| | - Andreas Schäfer
- Institute of Theoretical Physics, University of Regensburg, 93053 Regensburg, Germany
| | - Tim Beißbarth
- Department of Medical Bioinformatics, University Medical Center Göttingen, 37075 Göttingen, Germany
| | - Anagha Joshi
- Department of Clinical Science, Computational Biology Unit, University of Bergen, N-5008 Bergen, Norway
| | - Helena U Zacharias
- Peter L. Reichertz Institute for Medical Informatics of TU Braunschweig and Hannover Medical School, Hannover Medical School, 30625 Hannover, Germany
| | | | - Michael Altenbuchinger
- Department of Medical Bioinformatics, University Medical Center Göttingen, 37075 Göttingen, Germany
| |
Collapse
|
35
|
Jin YW, Hu P, Liu Q. NNICE: a deep quantile neural network algorithm for expression deconvolution. Sci Rep 2024; 14:14040. [PMID: 38890415 PMCID: PMC11189483 DOI: 10.1038/s41598-024-65053-w] [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: 01/06/2024] [Accepted: 06/17/2024] [Indexed: 06/20/2024] Open
Abstract
The composition of cell-type is a key indicator of health. Advancements in bulk gene expression data curation, single cell RNA-sequencing technologies, and computational deconvolution approaches offer a new perspective to learn about the composition of different cell types in a quick and affordable way. In this study, we developed a quantile regression and deep learning-based method called Neural Network Immune Contexture Estimator (NNICE) to estimate the cell type abundance and its uncertainty by automatically deconvolving bulk RNA-seq data. The proposed NNICE model was able to successfully recover ground-truth cell type fraction values given unseen bulk mixture gene expression profiles from the same dataset it was trained on. Compared with baseline methods, NNICE achieved better performance on deconvolve both pseudo-bulk gene expressions (Pearson correlation R = 0.9) and real bulk gene expression data (Pearson correlation R = 0.9) across all cell types. In conclusion, NNICE combines statistic inference with deep learning to provide accurate and interpretable cell type deconvolution from bulk gene expression.
Collapse
Affiliation(s)
- Yong Won Jin
- Department of Biochemistry & Medical Genetics, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB, R3E 0J9, Canada
| | - Pingzhao Hu
- Department of Biochemistry & Medical Genetics, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB, R3E 0J9, Canada
- Department of Biochemistry, Schulich School of Medicine & Dentistry, Western University, London, ON, N6A 5C1, Canada
| | - Qian Liu
- Department of Applied Computer Science, University of Winnipeg, Winnipeg, MB, R3B 2E9, Canada.
| |
Collapse
|
36
|
Tiong KL, Luzhbin D, Yeang CH. Assessing transcriptomic heterogeneity of single-cell RNASeq data by bulk-level gene expression data. BMC Bioinformatics 2024; 25:209. [PMID: 38867193 PMCID: PMC11167951 DOI: 10.1186/s12859-024-05825-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Accepted: 06/03/2024] [Indexed: 06/14/2024] Open
Abstract
BACKGROUND Single-cell RNA sequencing (sc-RNASeq) data illuminate transcriptomic heterogeneity but also possess a high level of noise, abundant missing entries and sometimes inadequate or no cell type annotations at all. Bulk-level gene expression data lack direct information of cell population composition but are more robust and complete and often better annotated. We propose a modeling framework to integrate bulk-level and single-cell RNASeq data to address the deficiencies and leverage the mutual strengths of each type of data and enable a more comprehensive inference of their transcriptomic heterogeneity. Contrary to the standard approaches of factorizing the bulk-level data with one algorithm and (for some methods) treating single-cell RNASeq data as references to decompose bulk-level data, we employed multiple deconvolution algorithms to factorize the bulk-level data, constructed the probabilistic graphical models of cell-level gene expressions from the decomposition outcomes, and compared the log-likelihood scores of these models in single-cell data. We term this framework backward deconvolution as inference operates from coarse-grained bulk-level data to fine-grained single-cell data. As the abundant missing entries in sc-RNASeq data have a significant effect on log-likelihood scores, we also developed a criterion for inclusion or exclusion of zero entries in log-likelihood score computation. RESULTS We selected nine deconvolution algorithms and validated backward deconvolution in five datasets. In the in-silico mixtures of mouse sc-RNASeq data, the log-likelihood scores of the deconvolution algorithms were strongly anticorrelated with their errors of mixture coefficients and cell type specific gene expression signatures. In the true bulk-level mouse data, the sample mixture coefficients were unknown but the log-likelihood scores were strongly correlated with accuracy rates of inferred cell types. In the data of autism spectrum disorder (ASD) and normal controls, we found that ASD brains possessed higher fractions of astrocytes and lower fractions of NRGN-expressing neurons than normal controls. In datasets of breast cancer and low-grade gliomas (LGG), we compared the log-likelihood scores of three simple hypotheses about the gene expression patterns of the cell types underlying the tumor subtypes. The model that tumors of each subtype were dominated by one cell type persistently outperformed an alternative model that each cell type had elevated expression in one gene group and tumors were mixtures of those cell types. Superiority of the former model is also supported by comparing the real breast cancer sc-RNASeq clusters with those generated by simulated sc-RNASeq data. CONCLUSIONS The results indicate that backward deconvolution serves as a sensible model selection tool for deconvolution algorithms and facilitates discerning hypotheses about cell type compositions underlying heterogeneous specimens such as tumors.
Collapse
Affiliation(s)
- Khong-Loon Tiong
- Institute of Statistical Science, Academia Sinica, Taipei, Taiwan
| | - Dmytro Luzhbin
- Institute of Statistical Science, Academia Sinica, Taipei, Taiwan
| | | |
Collapse
|
37
|
Caldi Gomes L, Hänzelmann S, Hausmann F, Khatri R, Oller S, Parvaz M, Tzeplaeff L, Pasetto L, Gebelin M, Ebbing M, Holzapfel C, Columbro SF, Scozzari S, Knöferle J, Cordts I, Demleitner AF, Deschauer M, Dufke C, Sturm M, Zhou Q, Zelina P, Sudria-Lopez E, Haack TB, Streb S, Kuzma-Kozakiewicz M, Edbauer D, Pasterkamp RJ, Laczko E, Rehrauer H, Schlapbach R, Carapito C, Bonetto V, Bonn S, Lingor P. Multiomic ALS signatures highlight subclusters and sex differences suggesting the MAPK pathway as therapeutic target. Nat Commun 2024; 15:4893. [PMID: 38849340 PMCID: PMC11161513 DOI: 10.1038/s41467-024-49196-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Accepted: 05/28/2024] [Indexed: 06/09/2024] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a debilitating motor neuron disease and lacks effective disease-modifying treatments. This study utilizes a comprehensive multiomic approach to investigate the early and sex-specific molecular mechanisms underlying ALS. By analyzing the prefrontal cortex of 51 patients with sporadic ALS and 50 control subjects, alongside four transgenic mouse models (C9orf72-, SOD1-, TDP-43-, and FUS-ALS), we have uncovered significant molecular alterations associated with the disease. Here, we show that males exhibit more pronounced changes in molecular pathways compared to females. Our integrated analysis of transcriptomes, (phospho)proteomes, and miRNAomes also identified distinct ALS subclusters in humans, characterized by variations in immune response, extracellular matrix composition, mitochondrial function, and RNA processing. The molecular signatures of human subclusters were reflected in specific mouse models. Our study highlighted the mitogen-activated protein kinase (MAPK) pathway as an early disease mechanism. We further demonstrate that trametinib, a MAPK inhibitor, has potential therapeutic benefits in vitro and in vivo, particularly in females, suggesting a direction for developing targeted ALS treatments.
Collapse
Affiliation(s)
- Lucas Caldi Gomes
- Technical University of Munich, School of Medicine, rechts der Isar Hospital, Clinical Department of Neurology, Munich, Germany
| | - Sonja Hänzelmann
- III. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Center for Biomedical AI, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Institute of Medical Systems Biology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Fabian Hausmann
- Center for Biomedical AI, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Institute of Medical Systems Biology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Robin Khatri
- Center for Biomedical AI, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Institute of Medical Systems Biology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Sergio Oller
- Center for Biomedical AI, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Institute of Medical Systems Biology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Mojan Parvaz
- Technical University of Munich, School of Medicine, rechts der Isar Hospital, Clinical Department of Neurology, Munich, Germany
| | - Laura Tzeplaeff
- Technical University of Munich, School of Medicine, rechts der Isar Hospital, Clinical Department of Neurology, Munich, Germany
| | - Laura Pasetto
- Research Center for ALS, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | - Marie Gebelin
- Laboratoire de Spectrométrie de Masse Bio-Organique, Université de Strasbourg, Infrastructure Nationale de Protéomique, Strasbourg, France
| | - Melanie Ebbing
- Center for Biomedical AI, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Institute of Medical Systems Biology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Constantin Holzapfel
- Center for Biomedical AI, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Institute of Medical Systems Biology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | | | - Serena Scozzari
- Research Center for ALS, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | - Johanna Knöferle
- Technical University of Munich, School of Medicine, rechts der Isar Hospital, Clinical Department of Neurology, Munich, Germany
| | - Isabell Cordts
- Technical University of Munich, School of Medicine, rechts der Isar Hospital, Clinical Department of Neurology, Munich, Germany
| | - Antonia F Demleitner
- Technical University of Munich, School of Medicine, rechts der Isar Hospital, Clinical Department of Neurology, Munich, Germany
| | - Marcus Deschauer
- Technical University of Munich, School of Medicine, rechts der Isar Hospital, Clinical Department of Neurology, Munich, Germany
| | - Claudia Dufke
- Institute of Medical Genetics and Applied Genomics, University of Tübingen, Tübingen, Germany
| | - Marc Sturm
- Institute of Medical Genetics and Applied Genomics, University of Tübingen, Tübingen, Germany
| | - Qihui Zhou
- German Center for Neurodegenerative Diseases (DZNE), München, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Pavol Zelina
- Department of Translational Neuroscience, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Emma Sudria-Lopez
- Department of Translational Neuroscience, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Tobias B Haack
- Institute of Medical Genetics and Applied Genomics, University of Tübingen, Tübingen, Germany
- Center for Rare Diseases, University of Tübingen, Tübingen, Germany
| | - Sebastian Streb
- Functional Genomics Center Zürich, ETH Zürich and University of Zürich, Zürich, Switzerland
| | | | - Dieter Edbauer
- German Center for Neurodegenerative Diseases (DZNE), München, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - R Jeroen Pasterkamp
- Department of Translational Neuroscience, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Endre Laczko
- Functional Genomics Center Zürich, ETH Zürich and University of Zürich, Zürich, Switzerland
| | - Hubert Rehrauer
- Functional Genomics Center Zürich, ETH Zürich and University of Zürich, Zürich, Switzerland
| | - Ralph Schlapbach
- Functional Genomics Center Zürich, ETH Zürich and University of Zürich, Zürich, Switzerland
| | - Christine Carapito
- Laboratoire de Spectrométrie de Masse Bio-Organique, Université de Strasbourg, Infrastructure Nationale de Protéomique, Strasbourg, France
| | - Valentina Bonetto
- Research Center for ALS, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | - Stefan Bonn
- Center for Biomedical AI, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.
- Institute of Medical Systems Biology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.
| | - Paul Lingor
- Technical University of Munich, School of Medicine, rechts der Isar Hospital, Clinical Department of Neurology, Munich, Germany.
- German Center for Neurodegenerative Diseases (DZNE), München, Germany.
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany.
| |
Collapse
|
38
|
Yasumizu Y, Hagiwara M, Umezu Y, Fuji H, Iwaisako K, Asagiri M, Uemoto S, Nakamura Y, Thul S, Ueyama A, Yokoi K, Tanemura A, Nose Y, Saito T, Wada H, Kakuda M, Kohara M, Nojima S, Morii E, Doki Y, Sakaguchi S, Ohkura N. Neural-net-based cell deconvolution from DNA methylation reveals tumor microenvironment associated with cancer prognosis. NAR Cancer 2024; 6:zcae022. [PMID: 38751935 PMCID: PMC11094754 DOI: 10.1093/narcan/zcae022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Revised: 04/18/2024] [Accepted: 05/01/2024] [Indexed: 05/18/2024] Open
Abstract
DNA methylation is a pivotal epigenetic modification that defines cellular identity. While cell deconvolution utilizing this information is considered useful for clinical practice, current methods for deconvolution are limited in their accuracy and resolution. In this study, we collected DNA methylation data from 945 human samples derived from various tissues and tumor-infiltrating immune cells and trained a neural network model with them. The model, termed MEnet, predicted abundance of cell population together with the detailed immune cell status from bulk DNA methylation data, and showed consistency to those of flow cytometry and histochemistry. MEnet was superior to the existing methods in the accuracy, speed, and detectable cell diversity, and could be applicable for peripheral blood, tumors, cell-free DNA, and formalin-fixed paraffin-embedded sections. Furthermore, by applying MEnet to 72 intrahepatic cholangiocarcinoma samples, we identified immune cell profiles associated with cancer prognosis. We believe that cell deconvolution by MEnet has the potential for use in clinical settings.
Collapse
Affiliation(s)
- Yoshiaki Yasumizu
- Department of Experimental Immunology, Immunology Frontier Research Center, Osaka University, Suita, Osaka, Japan
- Integrated Frontier Research for Medical Science Division, Institute for Open and Transdisciplinary Research Initiatives (OTRI), Osaka University, Suita, Osaka, Japan
| | - Masaki Hagiwara
- Department of Experimental Immunology, Immunology Frontier Research Center, Osaka University, Suita, Osaka, Japan
- Department of Basic Research in Tumor Immunology, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
- Pharmaceutical Research Division, Shionogi & Co., Ltd., Toyonaka, Osaka, Japan
| | - Yuto Umezu
- Faculty of Medicine, Osaka University, Suita, Osaka, Japan
| | - Hiroaki Fuji
- Department of Hepato-Biliary-Pancreatic Surgery, Hyogo Medical University, Nishinomiya, Hyogo, Japan
- Division of Hepato-Biliary-Pancreatic Surgery and Transplantation, Department of Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Kyoto, Japan
| | - Keiko Iwaisako
- Division of Hepato-Biliary-Pancreatic Surgery and Transplantation, Department of Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Kyoto, Japan
- Faculty of Life and Medical Sciences, Doshisha University, Kyotanabe, Kyoto, Japan
| | - Masataka Asagiri
- Department of Pharmacology, Yamaguchi University Graduate School of Medicine, Ube, Yamaguchi, Japan
| | - Shinji Uemoto
- Shiga University Medical Science, Otsu, Shiga, Japan
| | - Yamami Nakamura
- Department of Experimental Immunology, Immunology Frontier Research Center, Osaka University, Suita, Osaka, Japan
| | - Sophia Thul
- Department of Experimental Immunology, Immunology Frontier Research Center, Osaka University, Suita, Osaka, Japan
| | - Azumi Ueyama
- Pharmaceutical Research Division, Shionogi & Co., Ltd., Toyonaka, Osaka, Japan
- Department of Clinical Research in Tumor Immunology, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
| | - Kazunori Yokoi
- Department of Dermatology, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
| | - Atsushi Tanemura
- Department of Dermatology, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
| | - Yohei Nose
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
| | - Takuro Saito
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
| | - Hisashi Wada
- Department of Clinical Research in Tumor Immunology, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
| | - Mamoru Kakuda
- Department of Obstetrics and Gynecology, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
| | - Masaharu Kohara
- Department of Pathology, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
| | - Satoshi Nojima
- Department of Pathology, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
| | - Eiichi Morii
- Department of Pathology, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
| | - Yuichiro Doki
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
| | - Shimon Sakaguchi
- Department of Experimental Immunology, Immunology Frontier Research Center, Osaka University, Suita, Osaka, Japan
- Department of Experimental Immunology, Institute for Life and Medical Sciences, Kyoto University, Kyoto, Kyoto, Japan
| | - Naganari Ohkura
- Department of Experimental Immunology, Immunology Frontier Research Center, Osaka University, Suita, Osaka, Japan
- Department of Basic Research in Tumor Immunology, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
| |
Collapse
|
39
|
Nguyen H, Nguyen H, Tran D, Draghici S, Nguyen T. Fourteen years of cellular deconvolution: methodology, applications, technical evaluation and outstanding challenges. Nucleic Acids Res 2024; 52:4761-4783. [PMID: 38619038 PMCID: PMC11109966 DOI: 10.1093/nar/gkae267] [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: 10/26/2023] [Revised: 03/01/2024] [Accepted: 04/02/2024] [Indexed: 04/16/2024] Open
Abstract
Single-cell RNA sequencing (scRNA-Seq) is a recent technology that allows for the measurement of the expression of all genes in each individual cell contained in a sample. Information at the single-cell level has been shown to be extremely useful in many areas. However, performing single-cell experiments is expensive. Although cellular deconvolution cannot provide the same comprehensive information as single-cell experiments, it can extract cell-type information from bulk RNA data, and therefore it allows researchers to conduct studies at cell-type resolution from existing bulk datasets. For these reasons, a great effort has been made to develop such methods for cellular deconvolution. The large number of methods available, the requirement of coding skills, inadequate documentation, and lack of performance assessment all make it extremely difficult for life scientists to choose a suitable method for their experiment. This paper aims to fill this gap by providing a comprehensive review of 53 deconvolution methods regarding their methodology, applications, performance, and outstanding challenges. More importantly, the article presents a benchmarking of all these 53 methods using 283 cell types from 30 tissues of 63 individuals. We also provide an R package named DeconBenchmark that allows readers to execute and benchmark the reviewed methods (https://github.com/tinnlab/DeconBenchmark).
Collapse
Affiliation(s)
- Hung Nguyen
- Department of Computer Science and Software Engineering, Auburn University, Auburn, AL, USA
| | - Ha Nguyen
- Department of Computer Science and Software Engineering, Auburn University, Auburn, AL, USA
| | - Duc Tran
- Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Sorin Draghici
- Department of Computer Science, Wayne State University, Detroit, MI, USA
- Advaita Bioinformatics, Ann Arbor, MI, USA
| | - Tin Nguyen
- Department of Computer Science and Software Engineering, Auburn University, Auburn, AL, USA
| |
Collapse
|
40
|
Huang J, Du Y, Stucky A, Kelly KR, Zhong JF, Sun F. DeepDecon accurately estimates cancer cell fractions in bulk RNA-seq data. PATTERNS (NEW YORK, N.Y.) 2024; 5:100969. [PMID: 38800361 PMCID: PMC11117059 DOI: 10.1016/j.patter.2024.100969] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Revised: 01/15/2024] [Accepted: 03/21/2024] [Indexed: 05/29/2024]
Abstract
Understanding the cellular composition of a disease-related tissue is important in disease diagnosis, prognosis, and downstream treatment. Recent advances in single-cell RNA-sequencing (scRNA-seq) technique have allowed the measurement of gene expression profiles for individual cells. However, scRNA-seq is still too expensive to be used for large-scale population studies, and bulk RNA-seq is still widely used in such situations. An essential challenge is to deconvolve cellular composition for bulk RNA-seq data based on scRNA-seq data. Here, we present DeepDecon, a deep neural network model that leverages single-cell gene expression information to accurately predict the fraction of cancer cells in bulk tissues. It provides a refining strategy in which the cancer cell fraction is iteratively estimated by a set of trained models. When applied to simulated and real cancer data, DeepDecon exhibits superior performance compared to existing decomposition methods in terms of accuracy.
Collapse
Affiliation(s)
- Jiawei Huang
- Department of Quantitative and Computational Biology, University of Southern California, Los Angeles, CA 90089, USA
| | - Yuxuan Du
- Department of Quantitative and Computational Biology, University of Southern California, Los Angeles, CA 90089, USA
| | - Andres Stucky
- Department of Basic Sciences, School of Medicine, Loma Linda University, Loma Linda, CA 92350, USA
| | - Kevin R. Kelly
- Division of Hematology, University of Southern California, Los Angeles, CA 90089, USA
| | - Jiang F. Zhong
- Department of Basic Sciences, School of Medicine, Loma Linda University, Loma Linda, CA 92350, USA
| | - Fengzhu Sun
- Department of Quantitative and Computational Biology, University of Southern California, Los Angeles, CA 90089, USA
| |
Collapse
|
41
|
Jin L, Macoritto M, Wang J, Bi Y, Wang F, Suarez-Fueyo A, Paez-Cortez J, Hu C, Knight H, Mascanfroni I, Staron MM, Schwartz Sterman A, Houghton JM, Westmoreland S, Tian Y. Multi-Omics Characterization of Colon Mucosa and Submucosa/Wall from Crohn's Disease Patients. Int J Mol Sci 2024; 25:5108. [PMID: 38791146 PMCID: PMC11121447 DOI: 10.3390/ijms25105108] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2024] [Revised: 03/17/2024] [Accepted: 04/25/2024] [Indexed: 05/26/2024] Open
Abstract
Crohn's disease (CD) is a subtype of inflammatory bowel disease (IBD) characterized by transmural disease. The concept of transmural healing (TH) has been proposed as an indicator of deep clinical remission of CD and as a predictor of favorable treatment endpoints. Understanding the pathophysiology involved in transmural disease is critical to achieving these endpoints. However, most studies have focused on the intestinal mucosa, overlooking the contribution of the intestinal wall in Crohn's disease. Multi-omics approaches have provided new avenues for exploring the pathogenesis of Crohn's disease and identifying potential biomarkers. We aimed to use transcriptomic and proteomic technologies to compare immune and mesenchymal cell profiles and pathways in the mucosal and submucosa/wall compartments to better understand chronic refractory disease elements to achieve transmural healing. The results revealed similarities and differences in gene and protein expression profiles, metabolic mechanisms, and immune and non-immune pathways between these two compartments. Additionally, the identification of protein isoforms highlights the complex molecular mechanisms underlying this disease, such as decreased RTN4 isoforms (RTN4B2 and RTN4C) in the submucosa/wall, which may be related to the dysregulation of enteric neural processes. These findings have the potential to inform the development of novel therapeutic strategies to achieve TH.
Collapse
Affiliation(s)
- Liang Jin
- AbbVie Bioresearch Center, Worcester, MA 01605, USA; (L.J.)
| | | | - Jing Wang
- Immunology Research, AbbVie, Cambridge, MA 02139, USA (A.S.-F.)
| | - Yingtao Bi
- AbbVie Bioresearch Center, Worcester, MA 01605, USA; (L.J.)
| | - Fei Wang
- AbbVie Bioresearch Center, Worcester, MA 01605, USA; (L.J.)
| | | | | | - Chenqi Hu
- Alnylam Pharmaceuticals, Cambridge, MA 02139, USA
| | - Heather Knight
- AbbVie Bioresearch Center, Worcester, MA 01605, USA; (L.J.)
| | | | | | | | - Jean Marie Houghton
- Division of Gastroenterology, Department of Medicine, UMass Chan Medical School, Worcester, MA 01655, USA;
| | | | - Yu Tian
- AbbVie Bioresearch Center, Worcester, MA 01605, USA; (L.J.)
| |
Collapse
|
42
|
Vornholt E, Liharska LE, Cheng E, Hashemi A, Park YJ, Ziafat K, Wilkins L, Silk H, Linares LM, Thompson RC, Sullivan B, Moya E, Nadkarni GN, Sebra R, Schadt EE, Kopell BH, Charney AW, Beckmann ND. Characterizing cell type specific transcriptional differences between the living and postmortem human brain. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.05.01.24306590. [PMID: 38746297 PMCID: PMC11092720 DOI: 10.1101/2024.05.01.24306590] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/16/2024]
Abstract
Single-nucleus RNA sequencing (snRNA-seq) is often used to define gene expression patterns characteristic of brain cell types as well as to identify cell type specific gene expression signatures of neurological and mental illnesses in postmortem human brains. As methods to obtain brain tissue from living individuals emerge, it is essential to characterize gene expression differences associated with tissue originating from either living or postmortem subjects using snRNA-seq, and to assess whether and how such differences may impact snRNA-seq studies of brain tissue. To address this, human prefrontal cortex single nuclei gene expression was generated and compared between 31 samples from living individuals and 21 postmortem samples. The same cell types were consistently identified in living and postmortem nuclei, though for each cell type, a large proportion of genes were differentially expressed between samples from postmortem and living individuals. Notably, estimation of cell type proportions by cell type deconvolution of pseudo-bulk data was found to be more accurate in samples from living individuals. To allow for future integration of living and postmortem brain gene expression, a model was developed that quantifies from gene expression data the probability a human brain tissue sample was obtained postmortem. These probabilities are established as a means to statistically account for the gene expression differences between samples from living and postmortem individuals. Together, the results presented here provide a deep characterization of both differences between snRNA-seq derived from samples from living and postmortem individuals, as well as qualify and account for their effect on common analyses performed on this type of data.
Collapse
|
43
|
Khatri R, Machart P, Bonn S. DISSECT: deep semi-supervised consistency regularization for accurate cell type fraction and gene expression estimation. Genome Biol 2024; 25:112. [PMID: 38689377 PMCID: PMC11061925 DOI: 10.1186/s13059-024-03251-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Accepted: 04/17/2024] [Indexed: 05/02/2024] Open
Abstract
Cell deconvolution is the estimation of cell type fractions and cell type-specific gene expression from mixed data. An unmet challenge in cell deconvolution is the scarcity of realistic training data and the domain shift often observed in synthetic training data. Here, we show that two novel deep neural networks with simultaneous consistency regularization of the target and training domains significantly improve deconvolution performance. Our algorithm, DISSECT, outperforms competing algorithms in cell fraction and gene expression estimation by up to 14 percentage points. DISSECT can be easily adapted to other biomedical data types, as exemplified by our proteomic deconvolution experiments.
Collapse
Affiliation(s)
- Robin Khatri
- Institute of Medical Systems Biology, Center for Molecular Neurobiology, Center for Biomedical AI, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Pierre Machart
- Institute of Medical Systems Biology, Center for Molecular Neurobiology, Center for Biomedical AI, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Stefan Bonn
- Institute of Medical Systems Biology, Center for Molecular Neurobiology, Center for Biomedical AI, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.
| |
Collapse
|
44
|
Meng G, Pan Y, Tang W, Zhang L, Cui Y, Schumacher FR, Wang M, Wang R, He S, Krischer J, Li Q, Feng H. imply: improving cell-type deconvolution accuracy using personalized reference profiles. Genome Med 2024; 16:65. [PMID: 38685057 PMCID: PMC11057104 DOI: 10.1186/s13073-024-01338-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Accepted: 04/18/2024] [Indexed: 05/02/2024] Open
Abstract
Using computational tools, bulk transcriptomics can be deconvoluted to estimate the abundance of constituent cell types. However, existing deconvolution methods are conditioned on the assumption that the whole study population is served by a single reference panel, ignoring person-to-person heterogeneity. Here, we present imply, a novel algorithm to deconvolute cell type proportions using personalized reference panels. Simulation studies demonstrate reduced bias compared with existing methods. Real data analyses on longitudinal consortia show disparities in cell type proportions are associated with several disease phenotypes in Type 1 diabetes and Parkinson's disease. imply is available through the R/Bioconductor package ISLET at https://bioconductor.org/packages/ISLET/ .
Collapse
Affiliation(s)
- Guanqun Meng
- Department of Population and Quantitative Health Sciences, Case Western Reserve University, Cleveland, 44106, OH, USA
| | - Yue Pan
- Department of Biostatistics, St. Jude Children's Research Hospital, Memphis, 38105, TN, USA
| | - Wen Tang
- Department of Population and Quantitative Health Sciences, Case Western Reserve University, Cleveland, 44106, OH, USA
| | - Lijun Zhang
- Department of Population and Quantitative Health Sciences, Case Western Reserve University, Cleveland, 44106, OH, USA
| | - Ying Cui
- Department of Biomedical Data Science, Stanford University, Stanford, 94305, CA, USA
| | - Fredrick R Schumacher
- Department of Population and Quantitative Health Sciences, Case Western Reserve University, Cleveland, 44106, OH, USA
| | - Ming Wang
- Department of Population and Quantitative Health Sciences, Case Western Reserve University, Cleveland, 44106, OH, USA
| | - Rui Wang
- Department of Surgery, Division of Surgical Oncology, University Hospitals Cleveland Medical Center, Cleveland, 44106, OH, USA
| | - Sijia He
- Department of Biostatistics, University of Michigan, Ann Arbor, 48109, MI, USA
| | - Jeffrey Krischer
- Health Informatics Institute, University of South Florida, Tampa, 38105, FL, USA
| | - Qian Li
- Department of Biostatistics, St. Jude Children's Research Hospital, Memphis, 38105, TN, USA.
| | - Hao Feng
- Department of Population and Quantitative Health Sciences, Case Western Reserve University, Cleveland, 44106, OH, USA.
| |
Collapse
|
45
|
Ning L, Quan C, Wang Y, Wu Z, Yuan P, Xie N. scRNA-seq characterizing the heterogeneity of fibroblasts in breast cancer reveals a novel subtype SFRP4 + CAF that inhibits migration and predicts prognosis. Front Oncol 2024; 14:1348299. [PMID: 38686196 PMCID: PMC11056562 DOI: 10.3389/fonc.2024.1348299] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2023] [Accepted: 03/27/2024] [Indexed: 05/02/2024] Open
Abstract
Introduction Cancer-associated fibroblasts (CAFs) are a diverse group of cells that significantly impact the tumor microenvironment and therapeutic responses in breast cancer (BC). Despite their importance, the comprehensive profile of CAFs in BC remains to be fully elucidated. Methods To address this gap, we utilized single-cell RNA sequencing (scRNA-seq) to delineate the CAF landscape within 14 BC normal-tumor paired samples. We further corroborated our findings by analyzing several public datasets, thereby validating the newly identified CAF subtype. Additionally, we conducted coculture experiments with BC cells to assess the functional implications of this CAF subtype. Results Our scRNA-seq analysis unveiled eight distinct CAF subtypes across five tumor and six adjacent normal tissue samples. Notably, we discovered a novel subtype, designated as SFRP4+ CAFs, which was predominantly observed in normal tissues. The presence of SFRP4+ CAFs was substantiated by two independent scRNA-seq datasets and a spatial transcriptomics dataset. Functionally, SFRP4+ CAFs were found to impede BC cell migration and the epithelial-mesenchymal transition (EMT) process by secreting SFRP4, thereby modulating the WNT signaling pathway. Furthermore, we established that elevated expression levels of SFRP4+ CAF markers correlate with improved survival outcomes in BC patients, yet paradoxically, they predict a diminished response to neoadjuvant chemotherapy in cases of triple-negative breast cancer. Conclusion This investigation sheds light on the heterogeneity of CAFs in BC and introduces a novel SFRP4+ CAF subtype that hinders BC cell migration. This discovery holds promise as a potential biomarker for refined prognostic assessment and therapeutic intervention in BC.
Collapse
Affiliation(s)
- Lvwen Ning
- Biobank, Shenzhen Second People’s Hospital, First Affiliated Hospital of Shenzhen University, Health Science Center, Shenzhen University, Shenzhen, China
- Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Chuntao Quan
- Biobank, Shenzhen Second People’s Hospital, First Affiliated Hospital of Shenzhen University, Health Science Center, Shenzhen University, Shenzhen, China
| | - Yue Wang
- Biobank, Shenzhen Second People’s Hospital, First Affiliated Hospital of Shenzhen University, Health Science Center, Shenzhen University, Shenzhen, China
| | - Zhijie Wu
- Biobank, Shenzhen Second People’s Hospital, First Affiliated Hospital of Shenzhen University, Health Science Center, Shenzhen University, Shenzhen, China
| | - Peixiu Yuan
- College of Materials and Energy, South China Agricultural University, Guangzhou, China
| | - Ni Xie
- Biobank, Shenzhen Second People’s Hospital, First Affiliated Hospital of Shenzhen University, Health Science Center, Shenzhen University, Shenzhen, China
| |
Collapse
|
46
|
Hsu YC, Chiu YC, Lu TP, Hsiao TH, Chen Y. Predicting drug response through tumor deconvolution by cancer cell lines. PATTERNS (NEW YORK, N.Y.) 2024; 5:100949. [PMID: 38645769 PMCID: PMC11026976 DOI: 10.1016/j.patter.2024.100949] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/04/2023] [Revised: 02/07/2024] [Accepted: 02/12/2024] [Indexed: 04/23/2024]
Abstract
Large-scale cancer drug sensitivity data have become available for a collection of cancer cell lines, but only limited drug response data from patients are available. Bridging the gap in pharmacogenomics knowledge between in vitro and in vivo datasets remains challenging. In this study, we trained a deep learning model, Scaden-CA, for deconvoluting tumor data into proportions of cancer-type-specific cell lines. Then, we developed a drug response prediction method using the deconvoluted proportions and the drug sensitivity data from cell lines. The Scaden-CA model showed excellent performance in terms of concordance correlation coefficients (>0.9 for model testing) and the correctly deconvoluted rate (>70% across most cancers) for model validation using Cancer Cell Line Encyclopedia (CCLE) bulk RNA data. We applied the model to tumors in The Cancer Genome Atlas (TCGA) dataset and examined associations between predicted cell viability and mutation status or gene expression levels to understand underlying mechanisms of potential value for drug repurposing.
Collapse
Affiliation(s)
- Yu-Ching Hsu
- Bioinformatics Program, Taiwan International Graduate Program, National Taiwan University, Taipei 115, Taiwan
- Bioinformatics Program, Institute of Statistical Science, Taiwan International Graduate Program, Academia Sinica, Taipei 115, Taiwan
- Institute of Health Data Analytics and Statistics, Department of Public Health, College of Public Health, National Taiwan University, Taipei 100, Taiwan
- Greehey Children’s Cancer Research Institute, University of Texas Health San Antonio, San Antonio, TX 78229, USA
| | - Yu-Chiao Chiu
- Department of Medicine, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15261, USA
- UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh, PA 15232, USA
| | - Tzu-Pin Lu
- Institute of Health Data Analytics and Statistics, Department of Public Health, College of Public Health, National Taiwan University, Taipei 100, Taiwan
| | - Tzu-Hung Hsiao
- Department of Medical Research, Taichung Veterans General Hospital, Taichung 40705, Taiwan
| | - Yidong Chen
- Greehey Children’s Cancer Research Institute, University of Texas Health San Antonio, San Antonio, TX 78229, USA
- Department of Population Health Sciences, University of Texas Health San Antonio, San Antonio, TX 78229, USA
| |
Collapse
|
47
|
Davidson NR, Zhang F, Greene CS. BuDDI: BulkDeconvolution withDomainInvariance to predict cell-type-specific perturbations from bulk. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.07.20.549951. [PMID: 37503097 PMCID: PMC10370205 DOI: 10.1101/2023.07.20.549951] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/29/2023]
Abstract
While single-cell experiments provide deep cellular resolution within a single sample, some single-cell experiments are inherently more challenging than bulk experiments due to dissociation difficulties, cost, or limited tissue availability. This creates a situation where we have deep cellular profiles of one sample or condition, and bulk profiles across multiple samples and conditions. To bridge this gap, we propose BuDDI (BUlk Deconvolution with Domain Invariance). BuDDI utilizes domain adaptation techniques to effectively integrate available corpora of case-control bulk and reference scRNA-seq observations to infer cell-type-specific perturbation effects. BuDDI achieves this by learning independent latent spaces within a single variational autoencoder (VAE) encompassing at least four sources of variability: 1) cell type proportion, 2) perturbation effect, 3) structured experimental variability, and 4) remaining variability. Since each latent space is encouraged to be independent, we simulate perturbation responses by independently composing each latent space to simulate cell-type-specific perturbation responses. We evaluated BuDDI's performance on simulated and real data with experimental designs of increasing complexity. We first validated that BuDDI could learn domain invariant latent spaces on data with matched samples across each source of variability. Then we validated that BuDDI could accurately predict cell-type-specific perturbation response when no single-cell perturbed profiles were used during training; instead, only bulk samples had both perturbed and non-perturbed observations. Finally, we validated BuDDI on predicting sex-specific differences, an experimental design where it is not possible to have matched samples. In each experiment, BuDDI outperformed all other comparative methods and baselines. As more reference atlases are completed, BuDDI provides a path to combine these resources with bulk-profiled treatment or disease signatures to study perturbations, sex differences, or other factors at single-cell resolution.
Collapse
Affiliation(s)
- Natalie R Davidson
- Department of Biomedical Informatics, University of Colorado Anschutz School of Medicine, Aurora, Colorado, United States of America · Funded by the Gordon and Betty Moore Foundation (GBMF 4552), NHGRI of the National Institutes of Health (K99HG012945), NCI of the National Institutes of Health (R01CA237170, R01CA243188, R01CA200854)
| | - Fan Zhang
- Department of Medicine Rheumatology, University of Colorado Anschutz School of Medicine, Aurora, Colorado, United States of America; Department of Biomedical Informatics, University of Colorado Anschutz School of Medicine, Aurora, Colorado, United States of America · Funded by the Arthritis National Research Foundation Award, the PhRMA foundation, and the University of Colorado Translational Research Scholars Program Award
| | - Casey S Greene
- Department of Biomedical Informatics, University of Colorado Anschutz School of Medicine, Aurora, Colorado, United States of America · Funded by the Gordon and Betty Moore Foundation (GBMF 4552), NCI of the National Institutes of Health (R01CA237170, R01CA243188, R01CA200854)
| |
Collapse
|
48
|
Avram V, Yadav S, Sahasrabudhe P, Chang D, Wang J. IBDTransDB: a manually curated transcriptomic database for inflammatory bowel disease. Database (Oxford) 2024; 2024:baae026. [PMID: 38564306 PMCID: PMC10986744 DOI: 10.1093/database/baae026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2023] [Revised: 01/05/2024] [Accepted: 03/15/2024] [Indexed: 04/04/2024]
Abstract
Inflammatory Bowel Disease (IBD) therapies are ineffective in at least 40% patients, and transcriptomic datasets have been widely used to reveal the pathogenesis and to identify the novel drug targets for these patients. Although public IBD transcriptomic datasets are available from many web-based tools/databases, due to the unstructured metadata and data description of these public datasets, most of these tools/databases do not allow querying datasets based on multiple keywords (e.g. colon and infliximab). Furthermore, few tools/databases can compare and integrate the datasets from the query results. To fill these gaps, we have developed IBDTransDB (https://abbviegrc.shinyapps.io/ibdtransdb/), a manually curated transcriptomic database for IBD. IBDTransDB includes a manually curated database with 34 transcriptomic datasets (2932 samples, 122 differential comparisons) and a query system supporting 35 keywords from 5 attributes (e.g. tissue and treatment). IBDTransDB also provides three modules for data analyses and integration. IBDExplore allows interactive visualization of differential gene list, pathway enrichment, gene signature and cell deconvolution analyses from a single dataset. IBDCompare supports comparisons of selected genes or pathways from multiple datasets across different conditions. IBDIntegrate performs meta-analysis to prioritize a list of genes/pathways based on user-selected datasets and conditions. Using two case studies related to infliximab treatment, we demonstrated that IBDTransDB provides a unique platform for biologists and clinicians to reveal IBD pathogenesis and identify the novel targets by integrating with other omics data. Database URL: https://abbviegrc.shinyapps.io/ibdtransdb/.
Collapse
Affiliation(s)
- Victor Avram
- Genomics Research Center, AbbVie Inc, 200 Sidney Street, Cambridge, MA 02139, USA
| | - Shweta Yadav
- Genomics Research Center, AbbVie Inc, 200 Sidney Street, Cambridge, MA 02139, USA
| | - Pranav Sahasrabudhe
- Genomics Research Center, AbbVie Inc, 200 Sidney Street, Cambridge, MA 02139, USA
| | - Dan Chang
- Genomics Research Center, AbbVie Inc, 200 Sidney Street, Cambridge, MA 02139, USA
| | - Jing Wang
- Genomics Research Center, AbbVie Inc, 200 Sidney Street, Cambridge, MA 02139, USA
| |
Collapse
|
49
|
Vathrakokoili Pournara A, Miao Z, Beker OY, Nolte N, Brazma A, Papatheodorou I. CATD: a reproducible pipeline for selecting cell-type deconvolution methods across tissues. BIOINFORMATICS ADVANCES 2024; 4:vbae048. [PMID: 38638280 PMCID: PMC11023940 DOI: 10.1093/bioadv/vbae048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Revised: 02/20/2024] [Accepted: 03/21/2024] [Indexed: 04/20/2024]
Abstract
Motivation Cell-type deconvolution methods aim to infer cell composition from bulk transcriptomic data. The proliferation of developed methods coupled with inconsistent results obtained in many cases, highlights the pressing need for guidance in the selection of appropriate methods. Additionally, the growing accessibility of single-cell RNA sequencing datasets, often accompanied by bulk expression from related samples enable the benchmark of existing methods. Results In this study, we conduct a comprehensive assessment of 31 methods, utilizing single-cell RNA-sequencing data from diverse human and mouse tissues. Employing various simulation scenarios, we reveal the efficacy of regression-based deconvolution methods, highlighting their sensitivity to reference choices. We investigate the impact of bulk-reference differences, incorporating variables such as sample, study and technology. We provide validation using a gold standard dataset from mononuclear cells and suggest a consensus prediction of proportions when ground truth is not available. We validated the consensus method on data from the stomach and studied its spillover effect. Importantly, we propose the use of the critical assessment of transcriptomic deconvolution (CATD) pipeline which encompasses functionalities for generating references and pseudo-bulks and running implemented deconvolution methods. CATD streamlines simultaneous deconvolution of numerous bulk samples, providing a practical solution for speeding up the evaluation of newly developed methods. Availability and implementation https://github.com/Papatheodorou-Group/CATD_snakemake.
Collapse
Affiliation(s)
- Anna Vathrakokoili Pournara
- European Molecular Biology Laboratory, European Bioinformatics Institute (EMBL-EBI), Wellcome Genome Campus, Hinxton, Cambridge, CB10 1SD, United Kingdom
| | - Zhichao Miao
- European Molecular Biology Laboratory, European Bioinformatics Institute (EMBL-EBI), Wellcome Genome Campus, Hinxton, Cambridge, CB10 1SD, United Kingdom
- Open Targets, Wellcome Genome Campus, Hinxton CB10 1SD, United Kingdom
- GMU-GIBH Joint School of Life Sciences, Guangzhou Laboratory, Guangzhou Medical University, Guangzhou, 511436, China
| | - Ozgur Yilimaz Beker
- European Molecular Biology Laboratory, European Bioinformatics Institute (EMBL-EBI), Wellcome Genome Campus, Hinxton, Cambridge, CB10 1SD, United Kingdom
- Faculty of Engineering and Natural Sciences, Sabanci University, Tuzla 34956, Turkey
| | - Nadja Nolte
- European Molecular Biology Laboratory, European Bioinformatics Institute (EMBL-EBI), Wellcome Genome Campus, Hinxton, Cambridge, CB10 1SD, United Kingdom
- Department of Biotechnology and Systems Biology, National Institute of Biology, Ljubljana, 121-1000, Slovenia
| | - Alvis Brazma
- European Molecular Biology Laboratory, European Bioinformatics Institute (EMBL-EBI), Wellcome Genome Campus, Hinxton, Cambridge, CB10 1SD, United Kingdom
| | - Irene Papatheodorou
- European Molecular Biology Laboratory, European Bioinformatics Institute (EMBL-EBI), Wellcome Genome Campus, Hinxton, Cambridge, CB10 1SD, United Kingdom
- Open Targets, Wellcome Genome Campus, Hinxton CB10 1SD, United Kingdom
- Earlham Institute, Norwich Research Park, Norwich NR4 7UZ, United Kingdom
| |
Collapse
|
50
|
Nechanitzky R, Ramachandran P, Nechanitzky D, Li WY, Wakeham AC, Haight J, Saunders ME, Epelman S, Mak TW. CaSSiDI: novel single-cell "Cluster Similarity Scoring and Distinction Index" reveals critical functions for PirB and context-dependent Cebpb repression. Cell Death Differ 2024; 31:265-279. [PMID: 38383888 PMCID: PMC10923835 DOI: 10.1038/s41418-024-01268-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Revised: 01/15/2024] [Accepted: 02/07/2024] [Indexed: 02/23/2024] Open
Abstract
PirB is an inhibitory cell surface receptor particularly prominent on myeloid cells. PirB curtails the phenotypes of activated macrophages during inflammation or tumorigenesis, but its functions in macrophage homeostasis are obscure. To elucidate PirB-related functions in macrophages at steady-state, we generated and compared single-cell RNA-sequencing (scRNAseq) datasets obtained from myeloid cell subsets of wild type (WT) and PirB-deficient knockout (PirB KO) mice. To facilitate this analysis, we developed a novel approach to clustering parameter optimization called "Cluster Similarity Scoring and Distinction Index" (CaSSiDI). We demonstrate that CaSSiDI is an adaptable computational framework that facilitates tandem analysis of two scRNAseq datasets by optimizing clustering parameters. We further show that CaSSiDI offers more advantages than a standard Seurat analysis because it allows direct comparison of two or more independently clustered datasets, thereby alleviating the need for batch-correction while identifying the most similar and different clusters. Using CaSSiDI, we found that PirB is a novel regulator of Cebpb expression that controls the generation of Ly6Clo patrolling monocytes and the expansion properties of peritoneal macrophages. PirB's effect on Cebpb is tissue-specific since it was not observed in splenic red pulp macrophages (RPMs). However, CaSSiDI revealed a segregation of the WT RPM population into a CD68loIrf8+ "neuronal-primed" subset and an CD68hiFtl1+ "iron-loaded" subset. Our results establish the utility of CaSSiDI for single-cell assay analyses and the determination of optimal clustering parameters. Our application of CaSSiDI in this study has revealed previously unknown roles for PirB in myeloid cell populations. In particular, we have discovered homeostatic functions for PirB that are related to Cebpb expression in distinct macrophage subsets.
Collapse
Affiliation(s)
- Robert Nechanitzky
- Princess Margaret Cancer Centre, Ontario Cancer Institute, University Health Network, Toronto, ON, Canada.
- Providence Therapeutics Holdings Inc., Calgary, AB, Canada.
| | - Parameswaran Ramachandran
- Princess Margaret Cancer Centre, Ontario Cancer Institute, University Health Network, Toronto, ON, Canada
| | - Duygu Nechanitzky
- Princess Margaret Cancer Centre, Ontario Cancer Institute, University Health Network, Toronto, ON, Canada
| | - Wanda Y Li
- Centre for Oncology and Immunology, Hong Kong Science Park, Hong Kong SAR, China
| | - Andrew C Wakeham
- Princess Margaret Cancer Centre, Ontario Cancer Institute, University Health Network, Toronto, ON, Canada
| | - Jillian Haight
- Princess Margaret Cancer Centre, Ontario Cancer Institute, University Health Network, Toronto, ON, Canada
| | - Mary E Saunders
- Princess Margaret Cancer Centre, Ontario Cancer Institute, University Health Network, Toronto, ON, Canada
| | - Slava Epelman
- Toronto General Hospital Research Institute, University Health Network, Toronto, ON, Canada
- Ted Rogers Centre for Heart Research, Translational Biology and Engineering Program, Toronto, ON, Canada
- Peter Munk Cardiac Centre, UHN, Toronto, ON, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
- Departments of Immunology and Medical Biophysics, University of Toronto, Toronto, ON, Canada
| | - Tak W Mak
- Princess Margaret Cancer Centre, Ontario Cancer Institute, University Health Network, Toronto, ON, Canada.
- Centre for Oncology and Immunology, Hong Kong Science Park, Hong Kong SAR, China.
- Department of Pathology Department of Pathology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China.
| |
Collapse
|