1
|
Zhang C, Wang L, Shi Q. Computational modeling for deciphering tissue microenvironment heterogeneity from spatially resolved transcriptomics. Comput Struct Biotechnol J 2024; 23:2109-2115. [PMID: 38800634 PMCID: PMC11126885 DOI: 10.1016/j.csbj.2024.05.028] [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: 03/30/2024] [Revised: 05/15/2024] [Accepted: 05/16/2024] [Indexed: 05/29/2024] Open
Abstract
Spatial transcriptomics techniques, while measuring gene expression, retain spatial location information, aiding in situ studies of organismal tissue architecture and the progression of pathological processes. These techniques generate vast amounts of omics data, necessitating the development of computational methods to reveal the underlying tissue microenvironment heterogeneity. The main directions in spatial transcriptomics data analysis are spatial domain detection and spatial deconvolution, which can identify spatial functional regions and parse the distribution of cell types in spatial transcriptomics data by integrating single-cell transcriptomics data. In these two research directions, many computational methods have been successively proposed. This article will categorize them into three types: machine learning-based methods, probabilistic models-based methods, and deep learning-based methods. It will list and discuss the representative algorithms of each type along with their advantages and disadvantages and describe the datasets and evaluation metrics used to assess these computational methods, facilitating researchers in selecting suitable computational methods according to their research needs. Finally, combining the latest technological developments and the advantages and disadvantages of current algorithms, this article will look forward to the future directions of computational method development.
Collapse
Affiliation(s)
- Chuanchao Zhang
- Key Laboratory of Systems Health Science of Zhejiang Province, School of Life Science, Hangzhou Institute for Advanced Study, Hangzhou 310024; University of Chinese Academy of Sciences, China
| | - Lequn Wang
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai 200031, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Qianqian Shi
- Hubei Key Laboratory of Agricultural Bioinformatics, College of Informatics, Huazhong Agricultural University, Wuhan 430070, China
- Hubei Engineering Technology Research Center of Agricultural Big Data, Huazhong Agricultural University, Wuhan 430070, Hubei, China
| |
Collapse
|
2
|
Wang Z, Li Z, Luan T, Cui G, Shu S, Liang Y, Zhang K, Xiao J, Yu W, Cui J, Li A, Peng G, Fang Y. A spatiotemporal molecular atlas of mouse spinal cord injury identifies a distinct astrocyte subpopulation and therapeutic potential of IGFBP2. Dev Cell 2024; 59:2787-2803.e8. [PMID: 39029468 DOI: 10.1016/j.devcel.2024.06.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Revised: 03/26/2024] [Accepted: 06/20/2024] [Indexed: 07/21/2024]
Abstract
Spinal cord injury (SCI) triggers a cascade of intricate molecular and cellular changes that determine the outcome. In this study, we resolve the spatiotemporal organization of the injured mouse spinal cord and quantitatively assess in situ cell-cell communication following SCI. By analyzing existing single-cell RNA sequencing datasets alongside our spatial data, we delineate a subpopulation of Igfbp2-expressing astrocytes that migrate from the white matter (WM) to gray matter (GM) and become reactive upon SCI, termed Astro-GMii. Further, Igfbp2 upregulation promotes astrocyte migration, proliferation, and reactivity, and the secreted IGFBP2 protein fosters neurite outgrowth. Finally, we show that IGFBP2 significantly reduces neuronal loss and remarkably improves the functional recovery in a mouse model of SCI in vivo. Together, this study not only provides a comprehensive molecular atlas of SCI but also exemplifies how this rich resource can be applied to endow cells and genes with functional insight and therapeutic potential.
Collapse
Affiliation(s)
- Zeqing Wang
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai 201210, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Zhuxia Li
- Center for Cell Lineage and Development, CAS Key Laboratory of Regenerative Biology, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, GIBH-CUHK Joint Research Laboratory on Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Tianle Luan
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai 201210, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Guizhong Cui
- Guangzhou National Laboratory, Guangzhou 510005, China
| | - Shunpan Shu
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai 201210, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yiyao Liang
- Key Laboratory of CNS Regeneration (Ministry of Education), Guangdong Key Laboratory of Non-human Primate Research, GHM Institute of CNS Regeneration, Jinan University, Guangzhou 510632, China
| | - Kai Zhang
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai 201210, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Jingshu Xiao
- Guangzhou National Laboratory, Guangzhou 510005, China
| | - Wei Yu
- Key Laboratory of CNS Regeneration (Ministry of Education), Guangdong Key Laboratory of Non-human Primate Research, GHM Institute of CNS Regeneration, Jinan University, Guangzhou 510632, China
| | - Jihong Cui
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai 201210, China
| | - Ang Li
- Key Laboratory of CNS Regeneration (Ministry of Education), Guangdong Key Laboratory of Non-human Primate Research, GHM Institute of CNS Regeneration, Jinan University, Guangzhou 510632, China.
| | - Guangdun Peng
- Center for Cell Lineage and Development, CAS Key Laboratory of Regenerative Biology, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, GIBH-CUHK Joint Research Laboratory on Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China; University of Chinese Academy of Sciences, Beijing 100049, China.
| | - Yanshan Fang
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai 201210, China; University of Chinese Academy of Sciences, Beijing 100049, China.
| |
Collapse
|
3
|
Chen H, Lee YJ, Ovando-Ricardez JA, Rosas L, Rojas M, Mora AL, Bar-Joseph Z, Lugo-Martinez J. Recovering single-cell expression profiles from spatial transcriptomics with scResolve. CELL REPORTS METHODS 2024; 4:100864. [PMID: 39326411 DOI: 10.1016/j.crmeth.2024.100864] [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: 01/04/2024] [Revised: 06/14/2024] [Accepted: 08/30/2024] [Indexed: 09/28/2024]
Abstract
Many popular spatial transcriptomics techniques lack single-cell resolution. Instead, these methods measure the collective gene expression for each location from a mixture of cells, potentially containing multiple cell types. Here, we developed scResolve, a method for recovering single-cell expression profiles from spatial transcriptomics measurements at multi-cellular resolution. scResolve accurately restores expression profiles of individual cells at their locations, which is unattainable with cell type deconvolution. Applications of scResolve on human breast cancer data and human lung disease data demonstrate that scResolve enables cell-type-specific differential gene expression analysis between different tissue contexts and accurate identification of rare cell populations. The spatially resolved cellular-level expression profiles obtained through scResolve facilitate more flexible and precise spatial analysis that complements raw multi-cellular level analysis.
Collapse
Affiliation(s)
- Hao Chen
- Ray and Stephanie Lane Computational Biology Department, School of Computer Science, Carnegie Mellon University, Pittsburgh, PA 15213, USA
| | - Young Je Lee
- Ray and Stephanie Lane Computational Biology Department, School of Computer Science, Carnegie Mellon University, Pittsburgh, PA 15213, USA
| | - Jose A Ovando-Ricardez
- Dorothy M. Davis Heart and Lung Research Institute, Division of Pulmonary, Critical Care and Sleep Medicine, Department of Internal Medicine, The Ohio State University, Columbus, OH 43210, USA
| | - Lorena Rosas
- Dorothy M. Davis Heart and Lung Research Institute, Division of Pulmonary, Critical Care and Sleep Medicine, Department of Internal Medicine, The Ohio State University, Columbus, OH 43210, USA
| | - Mauricio Rojas
- Dorothy M. Davis Heart and Lung Research Institute, Division of Pulmonary, Critical Care and Sleep Medicine, Department of Internal Medicine, The Ohio State University, Columbus, OH 43210, USA
| | - Ana L Mora
- Dorothy M. Davis Heart and Lung Research Institute, Division of Pulmonary, Critical Care and Sleep Medicine, Department of Internal Medicine, The Ohio State University, Columbus, OH 43210, USA
| | - Ziv Bar-Joseph
- Ray and Stephanie Lane Computational Biology Department, School of Computer Science, Carnegie Mellon University, Pittsburgh, PA 15213, USA; Machine Learning Department, School of Computer Science, Carnegie Mellon University, Pittsburgh, PA 15213, USA
| | - Jose Lugo-Martinez
- Ray and Stephanie Lane Computational Biology Department, School of Computer Science, Carnegie Mellon University, Pittsburgh, PA 15213, USA.
| |
Collapse
|
4
|
Yan G, Hua SH, Li JJ. Categorization of 33 computational methods to detect spatially variable genes from spatially resolved transcriptomics data. ARXIV 2024:arXiv:2405.18779v4. [PMID: 38855546 PMCID: PMC11160866] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 06/11/2024]
Abstract
In the analysis of spatially resolved transcriptomics data, detecting spatially variable genes (SVGs) is crucial. Numerous computational methods exist, but varying SVG definitions and methodologies lead to incomparable results. We review 33 state-of-the-art methods, categorizing SVGs into three types: overall, cell-type-specific, and spatial-domain-marker SVGs. Our review explains the intuitions underlying these methods, summarizes their applications, and categorizes the hypothesis tests they use in the trade-off between generality and specificity for SVG detection. We discuss challenges in SVG detection and propose future directions for improvement. Our review offers insights for method developers and users, advocating for category-specific benchmarking.
Collapse
Affiliation(s)
- Guanao Yan
- Department of Statistics, University of California, Los Angeles, CA 90095-1554
| | - Shuo Harper Hua
- Department of Biomedical Data Science, Stanford University, Stanford, CA 94305
| | - Jingyi Jessica Li
- Department of Statistics, University of California, Los Angeles, CA 90095-1554
- Department of Human Genetics, University of California, Los Angeles, CA 90095-7088
- Department of Computational Medicine, University of California, Los Angeles, CA 90095-1766
- Department of Biostatistics, University of California, Los Angeles, CA 90095-1772
- Radcliffe Institute for Advanced Study, Harvard University, Cambridge, MA 02138
| |
Collapse
|
5
|
Deng SL, Fu Q, Liu Q, Huang FJ, Zhang M, Zhou X. Modulating endothelial cell dynamics in fat grafting: the impact of DLL4 siRNA via adipose stem cell extracellular vesicles. Am J Physiol Cell Physiol 2024; 327:C929-C945. [PMID: 39099421 PMCID: PMC11481985 DOI: 10.1152/ajpcell.00186.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Revised: 07/01/2024] [Accepted: 07/19/2024] [Indexed: 08/06/2024]
Abstract
In the context of improving the efficacy of autologous fat grafts (AFGs) in reconstructive surgery, this study delineates the novel use of adipose-derived mesenchymal stem cells (ADSCs) and their extracellular vesicles (EVs) as vehicles for delivering delta-like ligand 4 (DLL4) siRNA. The aim was to inhibit DLL4, a gene identified through transcriptome analysis as a critical player in the vascular endothelial cells of AFG tissues, thereby negatively affecting endothelial cell functions and graft survival through the Notch signaling pathway. By engineering ADSC EVs to carry DLL4 siRNA (ADSC EVs-siDLL4), the research demonstrated a marked improvement in endothelial cell proliferation, migration, and lumen formation, and enhanced angiogenesis in vivo, leading to a significant increase in the survival rate of AFGs. This approach presents a significant advancement in the field of tissue engineering and regenerative medicine, offering a potential method to overcome the limitations of current fat grafting techniques.NEW & NOTEWORTHY This study introduces a groundbreaking method for enhancing autologous fat graft survival using adipose-derived stem cell extracellular vesicles (ADSC EVs) to deliver DLL4 siRNA. By targeting the delta-like ligand 4 (DLL4) gene, crucial in endothelial cell dynamics, this innovative approach significantly improves endothelial cell functions and angiogenesis, marking a substantial advancement in tissue engineering and regenerative medicine.
Collapse
Affiliation(s)
- Sen-Lin Deng
- Plastic and Aesthetic Department, People's Hospital of Chongqing Banan District, Chongqing, People's Republic of China
| | - Qiang Fu
- Department of Dermatology and Cosmetology, Chongqing Hospital of Traditional Chinese Medicine, Chongqing, People's Republic of China
| | - Qing Liu
- Banan District Center for Disease Control and Prevention, Chongqing, People's Republic of China
| | - Fu-Jun Huang
- Department of Dermatology and Cosmetology, Chongqing Hospital of Traditional Chinese Medicine, Chongqing, People's Republic of China
| | - Miao Zhang
- Department of Dermatology and Cosmetology, Chongqing Hospital of Traditional Chinese Medicine, Chongqing, People's Republic of China
| | - Xun Zhou
- Department of Dermatology and Cosmetology, Chongqing Hospital of Traditional Chinese Medicine, Chongqing, People's Republic of China
| |
Collapse
|
6
|
Kim H, Kim KE, Madan E, Martin P, Gogna R, Rhee HW, Won KJ. Unveiling contact-mediated cellular crosstalk. Trends Genet 2024; 40:868-879. [PMID: 38906738 DOI: 10.1016/j.tig.2024.05.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Revised: 05/28/2024] [Accepted: 05/29/2024] [Indexed: 06/23/2024]
Abstract
Cell-cell interactions orchestrate complex functions in multicellular organisms, forming a regulatory network for diverse biological processes. Their disruption leads to disease states. Recent advancements - including single-cell sequencing and spatial transcriptomics, coupled with powerful bioengineering and molecular tools - have revolutionized our understanding of how cells respond to each other. Notably, spatial transcriptomics allows us to analyze gene expression changes based on cell proximity, offering a unique window into the impact of cell-cell contact. Additionally, computational approaches are being developed to decipher how cell contact governs the symphony of cellular responses. This review explores these cutting-edge approaches, providing valuable insights into deciphering the intricate cellular changes influenced by cell-cell communication.
Collapse
Affiliation(s)
- Hyobin Kim
- Department of Computational Biomedicine, Cedars-Sinai Medical Center, West, Hollywood, CA, USA
| | - Kwang-Eun Kim
- Department of Convergence Medicine, Yonsei University Wonju College of Medicine, Wonju, South Korea; Department of Chemistry, Seoul National University, Seoul, South Korea
| | - Esha Madan
- Massey Cancer Center, Virginia Commonwealth University, Richmond, VA, USA; School of Medicine, Institute of Molecular Medicine, Virginia Commonwealth University, Richmond, VA, USA; Department of Human and Molecular Genetics, School of Medicine, Virginia Commonwealth University, Richmond, VA, USA
| | - Patrick Martin
- Department of Computational Biomedicine, Cedars-Sinai Medical Center, West, Hollywood, CA, USA
| | - Rajan Gogna
- Massey Cancer Center, Virginia Commonwealth University, Richmond, VA, USA; School of Medicine, Institute of Molecular Medicine, Virginia Commonwealth University, Richmond, VA, USA; Department of Human and Molecular Genetics, School of Medicine, Virginia Commonwealth University, Richmond, VA, USA
| | - Hyun-Woo Rhee
- Department of Chemistry, Seoul National University, Seoul, South Korea.
| | - Kyoung-Jae Won
- Department of Computational Biomedicine, Cedars-Sinai Medical Center, West, Hollywood, CA, USA.
| |
Collapse
|
7
|
Li W, Zhang H, Wang L, Wang P, Yu K. STGAT: Graph attention networks for deconvolving spatial transcriptomics data. COMPUTER METHODS AND PROGRAMS IN BIOMEDICINE 2024; 257:108431. [PMID: 39461117 DOI: 10.1016/j.cmpb.2024.108431] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Revised: 09/03/2024] [Accepted: 09/16/2024] [Indexed: 10/29/2024]
Abstract
BACKGROUND AND OBJECTIVE Spatially resolved gene expression profiles are crucial for understanding tissue structure and function. However, the lack of single-cell resolution in these profiles demands their integration with single-cell RNA sequencing data for accurate dataset deconvolution. We propose STGAT, an innovative deconvolution method that leverages graph attention networks to enhance spatial transcriptomic (ST) data analysis. METHODS STGAT generates pseudo-ST data that more comprehensively represents the cell-type composition within real-ST data by using three different sampling probabilities. A comprehensive combined graph is then constructed to capture the complex relationships both across pseudo- and real-ST data and within each dataset. Moreover, integrating a graph attention network further enables STGAT to dynamically assign the weights to the connections between spots, significantly enhancing the accuracy of cell-type composition predictions. RESULTS Extensive comparative experiments on simulated and real-world datasets, demonstrate the superior performance of STGAT for cell-type deconvolution. The method outperforms six established methods and is robust across various biological contexts. CONCLUSION STGAT exhibits more precise results in cell-type composition inference that are more consistent with the known knowledge, suggesting its potential utility in improving the resolution and accuracy of spatial transcriptomics data analysis.
Collapse
Affiliation(s)
- Wei Li
- Key Laboratory of Intelligent Computing in Medical Image (MIIC), Northeastern University, Shenyang, 110000, Liaoning, China; National Frontiers Science Center for Industrial Intelligence and Systems Optimization, Shenyang, 110819, Liaoning, China
| | - Huixia Zhang
- School of Computer Science and Engineering, Northeastern University, Shenyang, 110819, Liaoning, China.
| | - Linjie Wang
- School of Computer Science and Engineering, Northeastern University, Shenyang, 110819, Liaoning, China.
| | - Pengyun Wang
- School of Computer Science and Engineering, Northeastern University, Shenyang, 110819, Liaoning, China
| | - Kun Yu
- College of Medicine and Bioinformation Engineering, Northeastern University, Shenyang, 110819, Liaoning, China
| |
Collapse
|
8
|
Ludin A, Stirtz GL, Tal A, Nirmal AJ, Besson N, Jones SM, Pfaff KL, Manos M, Liu S, Barrera I, Gong Q, Rodrigues CP, Sahu A, Jerison E, Alessi JV, Ricciuti B, Richardson DS, Weiss JD, Moreau HM, Stanhope ME, Afeyan AB, Sefton J, McCall WD, Formato E, Yang S, Zhou Y, van Konijnenburg DPH, Cole HL, Cordova M, Deng L, Rajadhyaksha M, Quake SR, Awad MM, Chen F, Sorger PK, Hodi FS, Rodig SJ, Murphy GF, Zon LI. Craters on the melanoma surface facilitate tumor-immune interactions and demonstrate pathologic response to checkpoint blockade in humans. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.18.613595. [PMID: 39345527 PMCID: PMC11429731 DOI: 10.1101/2024.09.18.613595] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 10/01/2024]
Abstract
Immunotherapy leads to cancer eradication despite the tumor's immunosuppressive environment. Here, we used extended long-term in-vivo imaging and high-resolution spatial transcriptomics of endogenous melanoma in zebrafish, and multiplex imaging of human melanoma, to identify domains that facilitate immune response during immunotherapy. We identified crater-shaped pockets at the margins of zebrafish and human melanoma, rich with beta-2 microglobulin (B2M) and antigen recognition molecules. The craters harbor the highest density of CD8+ T cells in the tumor. In zebrafish, CD8+ T cells formed prolonged interactions with melanoma cells within craters, characteristic of antigen recognition. Following immunostimulatory treatment, the craters enlarged and became the major site of activated CD8+ T cell accumulation and tumor killing that was B2M dependent. In humans, craters predicted immune response to ICB therapy, showing response better than high T cell infiltration. This marks craters as potential new diagnostic tool for immunotherapy success and targets to enhance ICB response.
Collapse
Affiliation(s)
- Aya Ludin
- Harvard Stem Cell and Regenerative Biology Department, Harvard University; Boston, MA, USA
- Stem Cell Program and Division of Hematology/Oncology, Boston Children’s Hospital and Dana-Farber Cancer Institute; Boston, MA, USA
- These authors contributed equally
| | - Georgia L. Stirtz
- Harvard Stem Cell and Regenerative Biology Department, Harvard University; Boston, MA, USA
- These authors contributed equally
| | - Asaf Tal
- Harvard Stem Cell and Regenerative Biology Department, Harvard University; Boston, MA, USA
| | - Ajit J. Nirmal
- Department of Dermatology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
- Ludwig Center at Harvard; Boston, MA, USA
- Laboratory of Systems Pharmacology, Department of Systems Biology, Harvard Medical School, Boston, MA, 02115, USA
- Department of Systems Biology, Harvard Medical School; Boston, MA, USA
| | - Naomi Besson
- Center for Immuno-Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Stephanie M. Jones
- Center for Immuno-Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Kathleen L. Pfaff
- Center for Immuno-Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Michael Manos
- Center for Immuno-Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Sophia Liu
- Biophysics Program, Harvard University, Boston, MA 02115, USA
- Harvard-MIT Division of Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA 02139, USA
| | - Irving Barrera
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Qiyu Gong
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Cecilia Pessoa Rodrigues
- Harvard Stem Cell and Regenerative Biology Department, Harvard University; Boston, MA, USA
- Stem Cell Program and Division of Hematology/Oncology, Boston Children’s Hospital and Dana-Farber Cancer Institute; Boston, MA, USA
| | - Aditi Sahu
- Dermatology Service, Memorial Sloan Kettering Cancer Center; New York, NY, USA.|
| | - Elizabeth Jerison
- Department of Physics, University of Chicago, Chicago, IL 60637, USA, Institute for Biophysical Dynamics, and James Franck Institute
| | - Joao V. Alessi
- Lowe Center for Thoracic Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Biagio Ricciuti
- Lowe Center for Thoracic Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Douglas S. Richardson
- Harvard Center for Biological Imaging, Department of Molecular and Cellular Biology, Harvard University; Cambridge, MA, USA
| | - Jodi D. Weiss
- Harvard Stem Cell and Regenerative Biology Department, Harvard University; Boston, MA, USA
| | - Hadley M. Moreau
- Harvard Stem Cell and Regenerative Biology Department, Harvard University; Boston, MA, USA
| | - Meredith E. Stanhope
- Harvard Stem Cell and Regenerative Biology Department, Harvard University; Boston, MA, USA
| | - Alexander B. Afeyan
- Harvard Stem Cell and Regenerative Biology Department, Harvard University; Boston, MA, USA
| | - James Sefton
- Harvard Stem Cell and Regenerative Biology Department, Harvard University; Boston, MA, USA
| | - Wyatt D. McCall
- Harvard Stem Cell and Regenerative Biology Department, Harvard University; Boston, MA, USA
| | - Emily Formato
- Harvard Stem Cell and Regenerative Biology Department, Harvard University; Boston, MA, USA
- Stem Cell Program and Division of Hematology/Oncology, Boston Children’s Hospital and Dana-Farber Cancer Institute; Boston, MA, USA
| | - Song Yang
- Stem Cell Program and Division of Hematology/Oncology, Boston Children’s Hospital and Dana-Farber Cancer Institute; Boston, MA, USA
| | - Yi Zhou
- Stem Cell Program and Division of Hematology/Oncology, Boston Children’s Hospital and Dana-Farber Cancer Institute; Boston, MA, USA
| | | | - Hannah L. Cole
- Harvard Stem Cell and Regenerative Biology Department, Harvard University; Boston, MA, USA
| | - Miguel Cordova
- Dermatology Service, Memorial Sloan Kettering Cancer Center; New York, NY, USA.|
| | - Liang Deng
- Dermatology Service, Memorial Sloan Kettering Cancer Center; New York, NY, USA.|
| | - Milind Rajadhyaksha
- Dermatology Service, Memorial Sloan Kettering Cancer Center; New York, NY, USA.|
| | - Stephen R. Quake
- Department of Bioengineering and Applied sciences, Stanford University, Stanford, CA, USA
- Chan Zuckerberg Biohub, San Francisco, CA, USA
| | - Mark M. Awad
- Lowe Center for Thoracic Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Fei Chen
- Harvard Stem Cell and Regenerative Biology Department, Harvard University; Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Peter K. Sorger
- Ludwig Center at Harvard; Boston, MA, USA
- Laboratory of Systems Pharmacology, Department of Systems Biology, Harvard Medical School, Boston, MA, 02115, USA
- Department of Systems Biology, Harvard Medical School; Boston, MA, USA
| | - F. Stephen Hodi
- Center for Immuno-Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Department of medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215
- Parker Institute for Cancer Immunotherapy
| | - Scott J. Rodig
- Center for Immuno-Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Department of Pathology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - George F. Murphy
- Department of Pathology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Leonard I. Zon
- Harvard Stem Cell and Regenerative Biology Department, Harvard University; Boston, MA, USA
- Stem Cell Program and Division of Hematology/Oncology, Boston Children’s Hospital and Dana-Farber Cancer Institute; Boston, MA, USA
- Howard Hughes Medical Institute, Harvard medical school; Boston MA, USA
| |
Collapse
|
9
|
Hu C, Borji M, Marrero GJ, Kumar V, Weir JA, Kammula SV, Macosko EZ, Chen F. Scalable imaging-free spatial genomics through computational reconstruction. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.05.606465. [PMID: 39149311 PMCID: PMC11326198 DOI: 10.1101/2024.08.05.606465] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 08/17/2024]
Abstract
Tissue organization arises from the coordinated molecular programs of cells. Spatial genomics maps cells and their molecular programs within the spatial context of tissues. However, current methods measure spatial information through imaging or direct registration, which often require specialized equipment and are limited in scale. Here, we developed an imaging-free spatial transcriptomics method that uses molecular diffusion patterns to computationally reconstruct spatial data. To do so, we utilize a simple experimental protocol on two dimensional barcode arrays to establish an interaction network between barcodes via molecular diffusion. Sequencing these interactions generates a high dimensional matrix of interactions between different spatial barcodes. Then, we perform dimensionality reduction to regenerate a two-dimensional manifold, which represents the spatial locations of the barcode arrays. Surprisingly, we found that the UMAP algorithm, with minimal modifications can faithfully successfully reconstruct the arrays. We demonstrated that this method is compatible with capture array based spatial transcriptomics/genomics methods, Slide-seq and Slide-tags, with high fidelity. We systematically explore the fidelity of the reconstruction through comparisons with experimentally derived ground truth data, and demonstrate that reconstruction generates high quality spatial genomics data. We also scaled this technique to reconstruct high-resolution spatial information over areas up to 1.2 centimeters. This computational reconstruction method effectively converts spatial genomics measurements to molecular biology, enabling spatial transcriptomics with high accessibility, and scalability.
Collapse
Affiliation(s)
- Chenlei Hu
- Broad Institute of Harvard and MIT, Cambridge, MA, USA
- Department of Molecular and Cellular Biology, Harvard University, Cambridge, MA, USA
| | - Mehdi Borji
- Broad Institute of Harvard and MIT, Cambridge, MA, USA
| | | | - Vipin Kumar
- Broad Institute of Harvard and MIT, Cambridge, MA, USA
| | - Jackson A Weir
- Broad Institute of Harvard and MIT, Cambridge, MA, USA
- Biological and Biomedical Sciences Program, Harvard University, Cambridge, MA, USA
| | | | - Evan Z Macosko
- Broad Institute of Harvard and MIT, Cambridge, MA, USA
- Department of Psychiatry, Massachusetts General Hospital, Boston, MA, USA
| | - Fei Chen
- Broad Institute of Harvard and MIT, Cambridge, MA, USA
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, USA
| |
Collapse
|
10
|
Jena SG, Verma A, Engelhardt BE. Answering open questions in biology using spatial genomics and structured methods. BMC Bioinformatics 2024; 25:291. [PMID: 39232666 PMCID: PMC11375982 DOI: 10.1186/s12859-024-05912-5] [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: 10/10/2023] [Accepted: 08/22/2024] [Indexed: 09/06/2024] Open
Abstract
Genomics methods have uncovered patterns in a range of biological systems, but obscure important aspects of cell behavior: the shapes, relative locations, movement, and interactions of cells in space. Spatial technologies that collect genomic or epigenomic data while preserving spatial information have begun to overcome these limitations. These new data promise a deeper understanding of the factors that affect cellular behavior, and in particular the ability to directly test existing theories about cell state and variation in the context of morphology, location, motility, and signaling that could not be tested before. Rapid advancements in resolution, ease-of-use, and scale of spatial genomics technologies to address these questions also require an updated toolkit of statistical methods with which to interrogate these data. We present a framework to respond to this new avenue of research: four open biological questions that can now be answered using spatial genomics data paired with methods for analysis. We outline spatial data modalities for each open question that may yield specific insights, discuss how conflicting theories may be tested by comparing the data to conceptual models of biological behavior, and highlight statistical and machine learning-based tools that may prove particularly helpful to recover biological understanding.
Collapse
Affiliation(s)
- Siddhartha G Jena
- Department of Stem Cell and Regenerative Biology, Harvard, 7 Divinity Ave, Cambridge, MA, USA
| | - Archit Verma
- Gladstone Institutes, 1650 Owens Street, San Francisco, CA, 94158, USA
| | | |
Collapse
|
11
|
Johnston KG, Berackey BT, Tran KM, Gelber A, Yu Z, MacGregor GR, Mukamel EA, Tan Z, Green KN, Xu X. Single-cell spatial transcriptomics reveals distinct patterns of dysregulation in non-neuronal and neuronal cells induced by the Trem2 R47H Alzheimer's risk gene mutation. Mol Psychiatry 2024:10.1038/s41380-024-02651-0. [PMID: 39103533 DOI: 10.1038/s41380-024-02651-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Revised: 06/20/2024] [Accepted: 06/26/2024] [Indexed: 08/07/2024]
Abstract
The R47H missense mutation of the TREM2 gene is a known risk factor for development of Alzheimer's Disease. In this study, we analyze the impact of the Trem2R47H mutation on specific cell types in multiple cortical and subcortical brain regions in the context of wild-type and 5xFAD mouse background. We profile 19 mouse brain sections consisting of wild-type, Trem2R47H, 5xFAD and Trem2R47H; 5xFAD genotypes using MERFISH spatial transcriptomics, a technique that enables subcellular profiling of spatial gene expression. Spatial transcriptomics and neuropathology data are analyzed using our custom pipeline to identify plaque and Trem2R47H-induced transcriptomic dysregulation. We initially analyze cell type-specific transcriptomic alterations induced by plaque proximity. Next, we analyze spatial distributions of disease associated microglia and astrocytes, and how they vary between 5xFAD and Trem2R47H; 5xFAD mouse models. Finally, we analyze the impact of the Trem2R47H mutation on neuronal transcriptomes. The Trem2R47H mutation induces consistent upregulation of Bdnf and Ntrk2 across many cortical excitatory neuron types, independent of amyloid pathology. Spatial investigation of genotype enriched subclusters identified spatially localized neuronal subpopulations reduced in 5xFAD and Trem2R47H; 5xFAD mice. Overall, our MERFISH spatial transcriptomics analysis identifies glial and neuronal transcriptomic alterations induced independently by 5xFAD and Trem2R47H mutations, impacting inflammatory responses in microglia and astrocytes, and activity and BDNF signaling in neurons.
Collapse
Affiliation(s)
- Kevin G Johnston
- Department of Anatomy and Neurobiology, School of Medicine, University of California, Irvine, CA, 92697, USA
| | - Bereket T Berackey
- Department of Anatomy and Neurobiology, School of Medicine, University of California, Irvine, CA, 92697, USA
- Department of Biomedical Engineering, University of California, Irvine, CA, 92697, USA
| | - Kristine M Tran
- Department of Neurobiology and Behavior, Charlie Dunlop School of Biological Sciences, University of California, Irvine, CA, 92697, USA
| | - Alon Gelber
- Department of Cognitive Science, University of California, San Diego, CA, 92037, USA
| | - Zhaoxia Yu
- Department of Statistics, School of Computer and Information Science, University of California, Irvine, CA, 92697, USA
- Center for Neural Circuit Mapping, University of California, Irvine, CA, 92697, USA
| | - Grant R MacGregor
- Department of Developmental and Cell Biology, University of California, Irvine, CA, 92697, USA
- Institute for Memory Impairments and Neurological Disorders (UCI MIND), Irvine, USA
| | - Eran A Mukamel
- Department of Cognitive Science, University of California, San Diego, CA, 92037, USA
| | - Zhiqun Tan
- Department of Anatomy and Neurobiology, School of Medicine, University of California, Irvine, CA, 92697, USA
- Center for Neural Circuit Mapping, University of California, Irvine, CA, 92697, USA
- Institute for Memory Impairments and Neurological Disorders (UCI MIND), Irvine, USA
- Department of Molecular Biology and Biochemistry School of Biological Sciences, University of California, Irvine, CA, 92697, USA
| | - Kim N Green
- Department of Neurobiology and Behavior, Charlie Dunlop School of Biological Sciences, University of California, Irvine, CA, 92697, USA
- Institute for Memory Impairments and Neurological Disorders (UCI MIND), Irvine, USA
| | - Xiangmin Xu
- Department of Anatomy and Neurobiology, School of Medicine, University of California, Irvine, CA, 92697, USA.
- Department of Biomedical Engineering, University of California, Irvine, CA, 92697, USA.
- Center for Neural Circuit Mapping, University of California, Irvine, CA, 92697, USA.
- Institute for Memory Impairments and Neurological Disorders (UCI MIND), Irvine, USA.
| |
Collapse
|
12
|
Wang L, Xu W, Zhang S, Gundberg GC, Zheng CR, Wan Z, Mustafina K, Caliendo F, Sandt H, Kamm R, Weiss R. Sensing and guiding cell-state transitions by using genetically encoded endoribonuclease-mediated microRNA sensors. Nat Biomed Eng 2024:10.1038/s41551-024-01229-z. [PMID: 38982158 DOI: 10.1038/s41551-024-01229-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Accepted: 05/11/2024] [Indexed: 07/11/2024]
Abstract
Precisely sensing and guiding cell-state transitions via the conditional genetic activation of appropriate differentiation factors is challenging. Here we show that desired cell-state transitions can be guided via genetically encoded sensors, whereby endogenous cell-state-specific miRNAs regulate the translation of a constitutively transcribed endoribonuclease, which, in turn, controls the translation of a gene of interest. We used this approach to monitor several cell-state transitions, to enrich specific cell types and to automatically guide the multistep differentiation of human induced pluripotent stem cells towards a haematopoietic lineage via endothelial cells as an intermediate state. Such conditional activation of gene expression is durable and resistant to epigenetic silencing and could facilitate the monitoring of cell-state transitions in physiological and pathological conditions and eventually the 'rewiring' of cell-state transitions for applications in organoid-based disease modelling, cellular therapies and regenerative medicine.
Collapse
Affiliation(s)
- Lei Wang
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA.
- Synthetic Biology Center, Massachusetts Institute of Technology, Cambridge, MA, USA.
- Department of Bioengineering, Northeastern University, Boston, MA, USA.
- Department of Biology, Northeastern University, Boston, MA, USA.
| | - Wenlong Xu
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
- Synthetic Biology Center, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Shun Zhang
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, China
- State Key Laboratory of Organ Regeneration and Reconstruction, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Gregory C Gundberg
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
- Synthetic Biology Center, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Christine R Zheng
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Zhengpeng Wan
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Kamila Mustafina
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
- Synthetic Biology Center, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Fabio Caliendo
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
- Synthetic Biology Center, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Hayden Sandt
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
- Synthetic Biology Center, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Roger Kamm
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Ron Weiss
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA.
- Synthetic Biology Center, Massachusetts Institute of Technology, Cambridge, MA, USA.
- Department of Electrical Engineering and Computer Science, Massachusetts Institute of Technology, Cambridge, MA, USA.
| |
Collapse
|
13
|
Zou LS, Cable DM, Barrera-Lopez IA, Zhao T, Murray E, Aryee MJ, Chen F, Irizarry RA. Detection of allele-specific expression in spatial transcriptomics with spASE. Genome Biol 2024; 25:180. [PMID: 38978101 PMCID: PMC11229351 DOI: 10.1186/s13059-024-03317-4] [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: 11/02/2023] [Accepted: 06/20/2024] [Indexed: 07/10/2024] Open
Abstract
Spatial transcriptomics technologies permit the study of the spatial distribution of RNA at near-single-cell resolution genome-wide. However, the feasibility of studying spatial allele-specific expression (ASE) from these data remains uncharacterized. Here, we introduce spASE, a computational framework for detecting and estimating spatial ASE. To tackle the challenges presented by cell type mixtures and a low signal to noise ratio, we implement a hierarchical model involving additive mixtures of spatial smoothing splines. We apply our method to allele-resolved Visium and Slide-seq from the mouse cerebellum and hippocampus and report new insight into the landscape of spatial and cell type-specific ASE therein.
Collapse
Affiliation(s)
- Luli S Zou
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA, 02115, USA
- Department of Data Sciences, Dana-Farber Cancer Institute, Boston, MA, 02215, USA
- Broad Institute of Harvard and MIT, Cambridge, MA, 02142, USA
| | - Dylan M Cable
- Department of Data Sciences, Dana-Farber Cancer Institute, Boston, MA, 02215, USA
- Broad Institute of Harvard and MIT, Cambridge, MA, 02142, USA
- Department of Electrical Engineering and Computer Science, MIT, Cambridge, MA, 02139, USA
| | | | - Tongtong Zhao
- Broad Institute of Harvard and MIT, Cambridge, MA, 02142, USA
| | - Evan Murray
- Broad Institute of Harvard and MIT, Cambridge, MA, 02142, USA
| | - Martin J Aryee
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA, 02115, USA
- Department of Data Sciences, Dana-Farber Cancer Institute, Boston, MA, 02215, USA
- Broad Institute of Harvard and MIT, Cambridge, MA, 02142, USA
| | - Fei Chen
- Broad Institute of Harvard and MIT, Cambridge, MA, 02142, USA
| | - Rafael A Irizarry
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA, 02115, USA.
- Department of Data Sciences, Dana-Farber Cancer Institute, Boston, MA, 02215, USA.
| |
Collapse
|
14
|
Chen R, Nie P, Wang J, Wang GZ. Deciphering brain cellular and behavioral mechanisms: Insights from single-cell and spatial RNA sequencing. WILEY INTERDISCIPLINARY REVIEWS. RNA 2024; 15:e1865. [PMID: 38972934 DOI: 10.1002/wrna.1865] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Revised: 05/05/2024] [Accepted: 05/14/2024] [Indexed: 07/09/2024]
Abstract
The brain is a complex computing system composed of a multitude of interacting neurons. The computational outputs of this system determine the behavior and perception of every individual. Each brain cell expresses thousands of genes that dictate the cell's function and physiological properties. Therefore, deciphering the molecular expression of each cell is of great significance for understanding its characteristics and role in brain function. Additionally, the positional information of each cell can provide crucial insights into their involvement in local brain circuits. In this review, we briefly overview the principles of single-cell RNA sequencing and spatial transcriptomics, the potential issues and challenges in their data processing, and their applications in brain research. We further outline several promising directions in neuroscience that could be integrated with single-cell RNA sequencing, including neurodevelopment, the identification of novel brain microstructures, cognition and behavior, neuronal cell positioning, molecules and cells related to advanced brain functions, sleep-wake cycles/circadian rhythms, and computational modeling of brain function. We believe that the deep integration of these directions with single-cell and spatial RNA sequencing can contribute significantly to understanding the roles of individual cells or cell types in these specific functions, thereby making important contributions to addressing critical questions in those fields. This article is categorized under: RNA Evolution and Genomics > Computational Analyses of RNA RNA in Disease and Development > RNA in Development RNA in Disease and Development > RNA in Disease.
Collapse
Affiliation(s)
- Renrui Chen
- CAS Key Laboratory of Computational Biology, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Pengxing Nie
- CAS Key Laboratory of Computational Biology, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Jing Wang
- CAS Key Laboratory of Computational Biology, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Guang-Zhong Wang
- CAS Key Laboratory of Computational Biology, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| |
Collapse
|
15
|
Ye J, Gao X, Huang X, Huang S, Zeng D, Luo W, Zeng C, Lu C, Lu L, Huang H, Mo K, Huang J, Li S, Tang M, Wu T, Mai R, Luo M, Xie M, Wang S, Li Y, Lin Y, Liang R. Integrating Single-Cell and Spatial Transcriptomics to Uncover and Elucidate GP73-Mediated Pro-Angiogenic Regulatory Networks in Hepatocellular Carcinoma. RESEARCH (WASHINGTON, D.C.) 2024; 7:0387. [PMID: 38939041 PMCID: PMC11208919 DOI: 10.34133/research.0387] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Accepted: 04/21/2024] [Indexed: 06/29/2024]
Abstract
Hepatocellular carcinoma (HCC) was characterized as being hypervascular. In the present study, we generated a single-cell spatial transcriptomic landscape of the vasculogenic etiology of HCC and illustrated overexpressed Golgi phosphoprotein 73 (GP73) HCC cells exerting cellular communication with vascular endothelial cells with high pro-angiogenesis potential via multiple receptor-ligand interactions in the process of tumor vascular development. Specifically, we uncovered an interactive GP73-mediated regulatory network coordinated with c-Myc, lactate, Janus kinase 2/signal transducer and activator of transcription 3 (JAK2/STAT3) pathway, and endoplasmic reticulum stress (ERS) signals in HCC cells and elucidated its pro-angiogenic roles in vitro and in vivo. Mechanistically, we found that GP73, the pivotal hub gene, was activated by histone lactylation and c-Myc, which stimulated the phosphorylation of downstream STAT3 by directly binding STAT3 and simultaneously enhancing glucose-regulated protein 78 (GRP78)-induced ERS. STAT3 potentiates GP73-mediated pro-angiogenic functions. Clinically, serum GP73 levels were positively correlated with HCC response to anti-angiogenic regimens and were essential for a prognostic nomogram showing good predictive performance for determining 6-month and 1-year survival in patients with HCC treated with anti-angiogenic therapy. Taken together, the aforementioned data characterized the pro-angiogenic roles and mechanisms of a GP73-mediated network and proved that GP73 is a crucial tumor angiogenesis niche gene with favorable anti-angiogenic potential in the treatment of HCC.
Collapse
Affiliation(s)
- Jiazhou Ye
- Department of Hepatobiliary Surgery,
Guangxi Medical University Cancer Hospital, Nanning 530021, China
- Guangxi Liver Cancer Diagnosis and Treatment Project Technology Research Center, Nanning 530021, China
- Guangxi Key Laboratory of Basic and Translational Research for Colorectal Cancer, Nanning 530021, China
| | - Xing Gao
- Guangxi Liver Cancer Diagnosis and Treatment Project Technology Research Center, Nanning 530021, China
- Department of Digestive Oncology, Guangxi Medical University Cancer Hospital, Nanning 530021, China
| | - Xi Huang
- Guangxi Liver Cancer Diagnosis and Treatment Project Technology Research Center, Nanning 530021, China
- Department of Digestive Oncology, Guangxi Medical University Cancer Hospital, Nanning 530021, China
| | - Shilin Huang
- Guangxi Liver Cancer Diagnosis and Treatment Project Technology Research Center, Nanning 530021, China
- Department of Digestive Oncology, Guangxi Medical University Cancer Hospital, Nanning 530021, China
| | - Dandan Zeng
- Guangxi Liver Cancer Diagnosis and Treatment Project Technology Research Center, Nanning 530021, China
- Department of Digestive Oncology, Guangxi Medical University Cancer Hospital, Nanning 530021, China
| | - Wenfeng Luo
- Guangxi Liver Cancer Diagnosis and Treatment Project Technology Research Center, Nanning 530021, China
- Department of Digestive Oncology, Guangxi Medical University Cancer Hospital, Nanning 530021, China
| | - Can Zeng
- Department of Hepatobiliary Surgery,
Guangxi Medical University Cancer Hospital, Nanning 530021, China
- Guangxi Liver Cancer Diagnosis and Treatment Project Technology Research Center, Nanning 530021, China
| | - Cheng Lu
- Department of Hepatobiliary Surgery,
Guangxi Medical University Cancer Hospital, Nanning 530021, China
- Guangxi Liver Cancer Diagnosis and Treatment Project Technology Research Center, Nanning 530021, China
| | - Lu Lu
- Guangxi Liver Cancer Diagnosis and Treatment Project Technology Research Center, Nanning 530021, China
- Department of Digestive Oncology, Guangxi Medical University Cancer Hospital, Nanning 530021, China
| | - Hongyang Huang
- Department of Hepatobiliary Surgery,
Guangxi Medical University Cancer Hospital, Nanning 530021, China
- Guangxi Liver Cancer Diagnosis and Treatment Project Technology Research Center, Nanning 530021, China
| | - Kaixiang Mo
- Department of Hepatobiliary Surgery,
Guangxi Medical University Cancer Hospital, Nanning 530021, China
- Guangxi Liver Cancer Diagnosis and Treatment Project Technology Research Center, Nanning 530021, China
| | - Julu Huang
- Department of Hepatobiliary Surgery,
Guangxi Medical University Cancer Hospital, Nanning 530021, China
- Guangxi Liver Cancer Diagnosis and Treatment Project Technology Research Center, Nanning 530021, China
| | - Shizhou Li
- Department of Hepatobiliary Surgery,
Guangxi Medical University Cancer Hospital, Nanning 530021, China
- Guangxi Liver Cancer Diagnosis and Treatment Project Technology Research Center, Nanning 530021, China
| | - Minchao Tang
- Department of Hepatobiliary Surgery,
Guangxi Medical University Cancer Hospital, Nanning 530021, China
- Guangxi Liver Cancer Diagnosis and Treatment Project Technology Research Center, Nanning 530021, China
| | - Tianzhun Wu
- Guangxi Liver Cancer Diagnosis and Treatment Project Technology Research Center, Nanning 530021, China
- Department of Digestive Oncology, Guangxi Medical University Cancer Hospital, Nanning 530021, China
| | - Rongyun Mai
- Department of Hepatobiliary Surgery,
Guangxi Medical University Cancer Hospital, Nanning 530021, China
- Guangxi Liver Cancer Diagnosis and Treatment Project Technology Research Center, Nanning 530021, China
| | - Min Luo
- Guangxi Liver Cancer Diagnosis and Treatment Project Technology Research Center, Nanning 530021, China
- Department of Digestive Oncology, Guangxi Medical University Cancer Hospital, Nanning 530021, China
| | - Mingzhi Xie
- Guangxi Liver Cancer Diagnosis and Treatment Project Technology Research Center, Nanning 530021, China
- Department of Digestive Oncology, Guangxi Medical University Cancer Hospital, Nanning 530021, China
| | - Shan Wang
- Guangxi Liver Cancer Diagnosis and Treatment Project Technology Research Center, Nanning 530021, China
- Guangxi Key Laboratory of Basic and Translational Research for Colorectal Cancer, Nanning 530021, China
- Department of Research, Guangxi Medical University Cancer Hospital, Nanning 530021, China
| | - Yongqiang Li
- Guangxi Liver Cancer Diagnosis and Treatment Project Technology Research Center, Nanning 530021, China
- Department of Digestive Oncology, Guangxi Medical University Cancer Hospital, Nanning 530021, China
| | - Yan Lin
- Guangxi Liver Cancer Diagnosis and Treatment Project Technology Research Center, Nanning 530021, China
- Guangxi Key Laboratory of Basic and Translational Research for Colorectal Cancer, Nanning 530021, China
- Department of Digestive Oncology, Guangxi Medical University Cancer Hospital, Nanning 530021, China
| | - Rong Liang
- Guangxi Liver Cancer Diagnosis and Treatment Project Technology Research Center, Nanning 530021, China
- Guangxi Key Laboratory of Basic and Translational Research for Colorectal Cancer, Nanning 530021, China
- Department of Digestive Oncology, Guangxi Medical University Cancer Hospital, Nanning 530021, China
| |
Collapse
|
16
|
Chen H, Zuo H, Huang J, Liu J, Jiang L, Jiang C, Zhang S, Hu Q, Lai H, Yin B, Yang G, Mai G, Li B, Chi H. Unravelling infiltrating T-cell heterogeneity in kidney renal clear cell carcinoma: Integrative single-cell and spatial transcriptomic profiling. J Cell Mol Med 2024; 28:e18403. [PMID: 39031800 PMCID: PMC11190954 DOI: 10.1111/jcmm.18403] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Revised: 05/02/2024] [Accepted: 05/07/2024] [Indexed: 07/15/2024] Open
Abstract
Kidney renal clear cell carcinoma (KIRC) pathogenesis intricately involves immune system dynamics, particularly the role of T cells within the tumour microenvironment. Through a multifaceted approach encompassing single-cell RNA sequencing, spatial transcriptome analysis and bulk transcriptome profiling, we systematically explored the contribution of infiltrating T cells to KIRC heterogeneity. Employing high-density weighted gene co-expression network analysis (hdWGCNA), module scoring and machine learning, we identified a distinct signature of infiltrating T cell-associated genes (ITSGs). Spatial transcriptomic data were analysed using robust cell type decomposition (RCTD) to uncover spatial interactions. Further analyses included enrichment assessments, immune infiltration evaluations and drug susceptibility predictions. Experimental validation involved PCR experiments, CCK-8 assays, plate cloning assays, wound-healing assays and Transwell assays. Six subpopulations of infiltrating and proliferating T cells were identified in KIRC, with notable dynamics observed in mid- to late-stage disease progression. Spatial analysis revealed significant correlations between T cells and epithelial cells across varying distances within the tumour microenvironment. The ITSG-based prognostic model demonstrated robust predictive capabilities, implicating these genes in immune modulation and metabolic pathways and offering prognostic insights into drug sensitivity for 12 KIRC treatment agents. Experimental validation underscored the functional relevance of PPIB in KIRC cell proliferation, invasion and migration. Our study comprehensively characterizes infiltrating T-cell heterogeneity in KIRC using single-cell RNA sequencing and spatial transcriptome data. The stable prognostic model based on ITSGs unveils infiltrating T cells' prognostic potential, shedding light on the immune microenvironment and offering avenues for personalized treatment and immunotherapy.
Collapse
Affiliation(s)
- Haiqing Chen
- Department of General Surgery (Hepatopancreatobiliary Surgery), The Affiliated HospitalSouthwest Medical UniversityLuzhouChina
- School of Clinical Medicine, The Affiliated HospitalSouthwest Medical UniversityLuzhouChina
| | - Haoyuan Zuo
- Department of General Surgery (Hepatopancreatobiliary Surgery), The Affiliated HospitalSouthwest Medical UniversityLuzhouChina
- Department of General Surgery (Hepatopancreatobiliary Surgery)Deyang People's HospitalDeyangChina
| | - Jinbang Huang
- School of Clinical Medicine, The Affiliated HospitalSouthwest Medical UniversityLuzhouChina
| | - Jie Liu
- Department of General Surgery (Hepatopancreatobiliary Surgery), The Affiliated HospitalSouthwest Medical UniversityLuzhouChina
- Department of General SurgeryDazhou Central HospitalDazhouChina
| | - Lai Jiang
- School of Clinical Medicine, The Affiliated HospitalSouthwest Medical UniversityLuzhouChina
| | - Chenglu Jiang
- School of Clinical Medicine, The Affiliated HospitalSouthwest Medical UniversityLuzhouChina
| | - Shengke Zhang
- School of Clinical Medicine, The Affiliated HospitalSouthwest Medical UniversityLuzhouChina
| | - Qingwen Hu
- School of Clinical Medicine, The Affiliated HospitalSouthwest Medical UniversityLuzhouChina
| | - Haotian Lai
- School of Clinical Medicine, The Affiliated HospitalSouthwest Medical UniversityLuzhouChina
| | - Bangchao Yin
- Department of PathologySixth People's Hospital of YibinYibinChina
| | - Guanhu Yang
- Department of Specialty MedicineOhio UniversityAthensOhioUSA
| | - Gang Mai
- Department of General Surgery (Hepatopancreatobiliary Surgery), The Affiliated HospitalSouthwest Medical UniversityLuzhouChina
- Department of General Surgery (Hepatopancreatobiliary Surgery)Deyang People's HospitalDeyangChina
| | - Bo Li
- Department of General Surgery (Hepatopancreatobiliary Surgery), The Affiliated HospitalSouthwest Medical UniversityLuzhouChina
| | - Hao Chi
- School of Clinical Medicine, The Affiliated HospitalSouthwest Medical UniversityLuzhouChina
| |
Collapse
|
17
|
Zhang L, Xiong Z, Xiao M. A Review of the Application of Spatial Transcriptomics in Neuroscience. Interdiscip Sci 2024; 16:243-260. [PMID: 38374297 DOI: 10.1007/s12539-024-00603-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Revised: 12/29/2023] [Accepted: 01/02/2024] [Indexed: 02/21/2024]
Abstract
Since spatial transcriptomics can locate and distinguish the gene expression of functional genes in special regions and tissue, it is important for us to investigate the brain development, the development mechanism of brain diseases, and the relationship between brain structure and function in Neuroscience (or Brain science). While previous studies have introduced the crucial spatial transcriptomic techniques and data analysis methods, there are few studies to comprehensively overview the key methods, data resources, and technological applications of spatial transcriptomics in Neuroscience. For these reasons, we first investigate several common spatial transcriptomic data analysis approaches and data resources. Second, we introduce the applications of the spatial transcriptomic data analysis approaches in Neuroscience. Third, we summarize the integrating spatial transcriptomics with other technologies in Neuroscience. Finally, we discuss the challenges and future research directions of spatial transcriptomics in Neuroscience.
Collapse
Affiliation(s)
- Le Zhang
- College of Computer Science, Sichuan University, Chengdu, 610065, China
| | - Zhenqi Xiong
- College of Computer Science, Sichuan University, Chengdu, 610065, China
| | - Ming Xiao
- College of Computer Science, Sichuan University, Chengdu, 610065, China.
| |
Collapse
|
18
|
Ma Y, Shi W, Dong Y, Sun Y, Jin Q. Spatial Multi-Omics in Alzheimer's Disease: A Multi-Dimensional Approach to Understanding Pathology and Progression. Curr Issues Mol Biol 2024; 46:4968-4990. [PMID: 38785566 PMCID: PMC11119029 DOI: 10.3390/cimb46050298] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2024] [Revised: 05/14/2024] [Accepted: 05/15/2024] [Indexed: 05/25/2024] Open
Abstract
Alzheimer's Disease (AD) presents a complex neuropathological landscape characterized by hallmark amyloid plaques and neurofibrillary tangles, leading to progressive cognitive decline. Despite extensive research, the molecular intricacies contributing to AD pathogenesis are inadequately understood. While single-cell omics technology holds great promise for application in AD, particularly in deciphering the understanding of different cell types and analyzing rare cell types and transcriptomic expression changes, it is unable to provide spatial distribution information, which is crucial for understanding the pathological processes of AD. In contrast, spatial multi-omics research emerges as a promising and comprehensive approach to analyzing tissue cells, potentially better suited for addressing these issues in AD. This article focuses on the latest advancements in spatial multi-omics technology and compares various techniques. Additionally, we provide an overview of current spatial omics-based research results in AD. These technologies play a crucial role in facilitating new discoveries and advancing translational AD research in the future. Despite challenges such as balancing resolution, increasing throughput, and data analysis, the application of spatial multi-omics holds immense potential in revolutionizing our understanding of human disease processes and identifying new biomarkers and therapeutic targets, thereby potentially contributing to the advancement of AD research.
Collapse
Affiliation(s)
| | | | | | | | - Qiguan Jin
- College of Physical Education, Yangzhou University, Yangzhou 225127, China; (Y.M.); (W.S.); (Y.D.); (Y.S.)
| |
Collapse
|
19
|
Yu S, Li WV. spVC for the detection and interpretation of spatial gene expression variation. Genome Biol 2024; 25:103. [PMID: 38641849 PMCID: PMC11027374 DOI: 10.1186/s13059-024-03245-3] [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: 09/24/2023] [Accepted: 04/10/2024] [Indexed: 04/21/2024] Open
Abstract
Spatially resolved transcriptomics technologies have opened new avenues for understanding gene expression heterogeneity in spatial contexts. However, existing methods for identifying spatially variable genes often focus solely on statistical significance, limiting their ability to capture continuous expression patterns and integrate spot-level covariates. To address these challenges, we introduce spVC, a statistical method based on a generalized Poisson model. spVC seamlessly integrates constant and spatially varying effects of covariates, facilitating comprehensive exploration of gene expression variability and enhancing interpretability. Simulation and real data applications confirm spVC's accuracy in these tasks, highlighting its versatility in spatial transcriptomics analysis.
Collapse
Affiliation(s)
- Shan Yu
- Department of Statistics, Unversity of Virginia, Charlottesville, 22903, VA, USA.
| | - Wei Vivian Li
- Department of Statistics, University of California, Riverside, 92521, CA, USA.
| |
Collapse
|
20
|
Yuan Z, Zhao F, Lin S, Zhao Y, Yao J, Cui Y, Zhang XY, Zhao Y. Benchmarking spatial clustering methods with spatially resolved transcriptomics data. Nat Methods 2024; 21:712-722. [PMID: 38491270 DOI: 10.1038/s41592-024-02215-8] [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: 03/13/2023] [Accepted: 02/16/2024] [Indexed: 03/18/2024]
Abstract
Spatial clustering, which shares an analogy with single-cell clustering, has expanded the scope of tissue physiology studies from cell-centroid to structure-centroid with spatially resolved transcriptomics (SRT) data. Computational methods have undergone remarkable development in recent years, but a comprehensive benchmark study is still lacking. Here we present a benchmark study of 13 computational methods on 34 SRT data (7 datasets). The performance was evaluated on the basis of accuracy, spatial continuity, marker genes detection, scalability, and robustness. We found existing methods were complementary in terms of their performance and functionality, and we provide guidance for selecting appropriate methods for given scenarios. On testing additional 22 challenging datasets, we identified challenges in identifying noncontinuous spatial domains and limitations of existing methods, highlighting their inadequacies in handling recent large-scale tasks. Furthermore, with 145 simulated data, we examined the robustness of these methods against four different factors, and assessed the impact of pre- and postprocessing approaches. Our study offers a comprehensive evaluation of existing spatial clustering methods with SRT data, paving the way for future advancements in this rapidly evolving field.
Collapse
Affiliation(s)
- Zhiyuan Yuan
- Center for Medical Research and Innovation, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Fudan University, Shanghai, China.
- Institute of Science and Technology for Brain-Inspired Intelligence; MOE Key Laboratory of Computational Neuroscience and Brain-Inspired Intelligence; MOE Frontiers Center for Brain Science, Fudan University, Shanghai, China.
| | - Fangyuan Zhao
- Research Center for Ubiquitous Computing Systems, Institute of Computing Technology, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Senlin Lin
- Research Center for Ubiquitous Computing Systems, Institute of Computing Technology, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Yu Zhao
- Tencent AI Lab, Shenzhen, China
| | | | - Yan Cui
- Center for Medical Research and Innovation, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Fudan University, Shanghai, China
- Institute of Science and Technology for Brain-Inspired Intelligence; MOE Key Laboratory of Computational Neuroscience and Brain-Inspired Intelligence; MOE Frontiers Center for Brain Science, Fudan University, Shanghai, China
- Bioinformatics Center, Institute for Chemical Research, Kyoto University, Kyoto, Japan
| | - Xiao-Yong Zhang
- Center for Medical Research and Innovation, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Fudan University, Shanghai, China
| | - Yi Zhao
- Research Center for Ubiquitous Computing Systems, Institute of Computing Technology, Chinese Academy of Sciences, Beijing, China.
- University of Chinese Academy of Sciences, Beijing, China.
| |
Collapse
|
21
|
Lyu Y, Wu C, Sun W, Li Z. Regional analysis to delineate intrasample heterogeneity with RegionalST. Bioinformatics 2024; 40:btae186. [PMID: 38579257 PMCID: PMC11026142 DOI: 10.1093/bioinformatics/btae186] [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: 07/10/2023] [Revised: 03/06/2024] [Accepted: 04/03/2024] [Indexed: 04/07/2024] Open
Abstract
MOTIVATION Spatial transcriptomics has greatly contributed to our understanding of spatial and intra-sample heterogeneity, which could be crucial for deciphering the molecular basis of human diseases. Intra-tumor heterogeneity, e.g. may be associated with cancer treatment responses. However, the lack of computational tools for exploiting cross-regional information and the limited spatial resolution of current technologies present major obstacles to elucidating tissue heterogeneity. RESULTS To address these challenges, we introduce RegionalST, an efficient computational method that enables users to quantify cell type mixture and interactions, identify sub-regions of interest, and perform cross-region cell type-specific differential analysis for the first time. Our simulations and real data applications demonstrate that RegionalST is an efficient tool for visualizing and analyzing diverse spatial transcriptomics data, thereby enabling accurate and flexible exploration of tissue heterogeneity. Overall, RegionalST provides a one-stop destination for researchers seeking to delve deeper into the intricacies of spatial transcriptomics data. AVAILABILITY AND IMPLEMENTATION The implementation of our method is available as an open-source R/Bioconductor package with a user-friendly manual available at https://bioconductor.org/packages/release/bioc/html/RegionalST.html.
Collapse
Affiliation(s)
- Yue Lyu
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, United States
- Department of Biostatistics and Data Science, The University of Texas Health Science Center at Houston, Houston, TX 77030, United States
| | - Chong Wu
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, United States
| | - Wei Sun
- Biostatistics Program, Public Health Science Division, Fred Hutchinson Cancer Center, Seattle, WA 98109, United States
- Department of Biostatistics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27516, United States
- Department of Biostatistics, University of Washington, Seattle, WA 98195, United States
| | - Ziyi Li
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, United States
| |
Collapse
|
22
|
Guo X, Ning J, Chen Y, Liu G, Zhao L, Fan Y, Sun S. Recent advances in differential expression analysis for single-cell RNA-seq and spatially resolved transcriptomic studies. Brief Funct Genomics 2024; 23:95-109. [PMID: 37022699 DOI: 10.1093/bfgp/elad011] [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: 07/08/2022] [Revised: 12/09/2022] [Accepted: 03/10/2023] [Indexed: 04/07/2023] Open
Abstract
Differential expression (DE) analysis is a necessary step in the analysis of single-cell RNA sequencing (scRNA-seq) and spatially resolved transcriptomics (SRT) data. Unlike traditional bulk RNA-seq, DE analysis for scRNA-seq or SRT data has unique characteristics that may contribute to the difficulty of detecting DE genes. However, the plethora of DE tools that work with various assumptions makes it difficult to choose an appropriate one. Furthermore, a comprehensive review on detecting DE genes for scRNA-seq data or SRT data from multi-condition, multi-sample experimental designs is lacking. To bridge such a gap, here, we first focus on the challenges of DE detection, then highlight potential opportunities that facilitate further progress in scRNA-seq or SRT analysis, and finally provide insights and guidance in selecting appropriate DE tools or developing new computational DE methods.
Collapse
Affiliation(s)
- Xiya Guo
- School of Public Health, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
- Key Laboratory of Trace Elements and Endemic Diseases, Center for Single Cell Omics and Health, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Jin Ning
- School of Public Health, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
- Key Laboratory of Trace Elements and Endemic Diseases, Center for Single Cell Omics and Health, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Yuanze Chen
- School of Public Health, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
- Key Laboratory of Trace Elements and Endemic Diseases, Center for Single Cell Omics and Health, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Guoliang Liu
- School of Public Health, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
- Key Laboratory of Trace Elements and Endemic Diseases, Center for Single Cell Omics and Health, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Liyan Zhao
- School of Public Health, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
- Key Laboratory of Trace Elements and Endemic Diseases, Center for Single Cell Omics and Health, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Yue Fan
- School of Public Health, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
- Key Laboratory of Trace Elements and Endemic Diseases, Center for Single Cell Omics and Health, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Shiquan Sun
- School of Public Health, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
- Key Laboratory of Trace Elements and Endemic Diseases, Center for Single Cell Omics and Health, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| |
Collapse
|
23
|
Babu E, Sen S. Explore & actuate: the future of personalized medicine in oncology through emerging technologies. Curr Opin Oncol 2024; 36:93-101. [PMID: 38441149 DOI: 10.1097/cco.0000000000001016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/08/2024]
Abstract
PURPOSE OF REVIEW The future of medicine is aimed to equip the physician with tools to assess the individual health of the patient for the uniqueness of the disease that separates it from the rest. The integration of omics technologies into clinical practice, reviewed here, would open new avenues for addressing the spatial and temporal heterogeneity of cancer. The rising cancer burden patiently awaits the advent of such an approach to personalized medicine for routine clinical settings. RECENT FINDINGS To weigh the translational potential, multiple technologies were categorized based on the extractable information from the different types of samples used, to the various omic-levels of molecular information that each technology has been able to advance over the last 2 years. This review uses a multifaceted classification that helps to assess translational potential in a meaningful way toward clinical adaptation. SUMMARY The importance of distinguishing technologies based on the flow of information from exploration to actuation puts forth a framework that allows the clinicians to better adapt a chosen technology or use them in combination to enhance their goals toward personalized medicine.
Collapse
Affiliation(s)
- Erald Babu
- UM-DAE Centre for Excellence in Basic Sciences, School of Biological Sciences, University of Mumbai, Kalina Campus, Mumbai, Maharashtra, India
| | | |
Collapse
|
24
|
Garmire LX, Li Y, Huang Q, Xu C, Teichmann SA, Kaminski N, Pellegrini M, Nguyen Q, Teschendorff AE. Challenges and perspectives in computational deconvolution of genomics data. Nat Methods 2024; 21:391-400. [PMID: 38374264 DOI: 10.1038/s41592-023-02166-6] [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: 11/04/2022] [Accepted: 12/26/2023] [Indexed: 02/21/2024]
Abstract
Deciphering cell-type heterogeneity is crucial for systematically understanding tissue homeostasis and its dysregulation in diseases. Computational deconvolution is an efficient approach for estimating cell-type abundances from a variety of omics data. Despite substantial methodological progress in computational deconvolution in recent years, challenges are still outstanding. Here we enlist four important challenges related to computational deconvolution: the quality of the reference data, generation of ground truth data, limitations of computational methodologies, and benchmarking design and implementation. Finally, we make recommendations on reference data generation, new directions of computational methodologies, and strategies to promote rigorous benchmarking.
Collapse
Affiliation(s)
- Lana X Garmire
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI, USA.
| | - Yijun Li
- Department of Biostatistics, University of Michigan, Ann Arbor, MI, USA
| | - Qianhui Huang
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI, USA
| | - Chuan Xu
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, UK
| | | | - Naftali Kaminski
- Pulmonary, Critical Care & Sleep Medicine, Yale University School of Medicine, New Haven, CT, USA
| | - Matteo Pellegrini
- Molecular, Cell and Developmental Biology, University of California, Los Angeles, Los Angeles, CA, USA
| | - Quan Nguyen
- Institute for Molecular Bioscience, The University of Queensland and QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - Andrew E Teschendorff
- CAS Key Laboratory of Computational Biology, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
- UCL Cancer Institute, University College London, London, UK
| |
Collapse
|
25
|
Lin S, Zhao F, Wu Z, Yao J, Zhao Y, Yuan Z. Streamlining spatial omics data analysis with Pysodb. Nat Protoc 2024; 19:831-895. [PMID: 38135744 DOI: 10.1038/s41596-023-00925-5] [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: 04/04/2023] [Accepted: 10/02/2023] [Indexed: 12/24/2023]
Abstract
Advances in spatial omics technologies have improved the understanding of cellular organization in tissues, leading to the generation of complex and heterogeneous data and prompting the development of specialized tools for managing, loading and visualizing spatial omics data. The Spatial Omics Database (SODB) was established to offer a unified format for data storage and interactive visualization modules. Here we detail the use of Pysodb, a Python-based tool designed to enable the efficient exploration and loading of spatial datasets from SODB within a Python environment. We present seven case studies using Pysodb, detailing the interaction with various computational methods, ensuring reproducibility of experimental data and facilitating the integration of new data and alternative applications in SODB. The approach offers a reference for method developers by outlining label and metadata availability in representative spatial data that can be loaded by Pysodb. The tool is supplemented by a website ( https://protocols-pysodb.readthedocs.io/ ) with detailed information for benchmarking analysis, and allows method developers to focus on computational models by facilitating data processing. This protocol is designed for researchers with limited experience in computational biology. Depending on the dataset complexity, the protocol typically requires ~12 h to complete.
Collapse
Affiliation(s)
- Senlin Lin
- Institute of Computing Technology, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Fangyuan Zhao
- Institute of Computing Technology, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | | | | | - Yi Zhao
- Institute of Computing Technology, Chinese Academy of Sciences, Beijing, China.
- University of Chinese Academy of Sciences, Beijing, China.
| | - Zhiyuan Yuan
- Institute of Science and Technology for Brain-Inspired Intelligence, MOE Key Laboratory of Computational Neuroscience and Brain-Inspired Intelligence, MOE Frontiers Center for Brain Science, Fudan University, Shanghai, China.
- Center for Medical Research and Innovation, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Fudan University, Shanghai, China.
| |
Collapse
|
26
|
Mason K, Sathe A, Hess PR, Rong J, Wu CY, Furth E, Susztak K, Levinsohn J, Ji HP, Zhang N. Niche-DE: niche-differential gene expression analysis in spatial transcriptomics data identifies context-dependent cell-cell interactions. Genome Biol 2024; 25:14. [PMID: 38217002 PMCID: PMC10785550 DOI: 10.1186/s13059-023-03159-6] [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: 02/13/2023] [Accepted: 12/22/2023] [Indexed: 01/14/2024] Open
Abstract
Existing methods for analysis of spatial transcriptomic data focus on delineating the global gene expression variations of cell types across the tissue, rather than local gene expression changes driven by cell-cell interactions. We propose a new statistical procedure called niche-differential expression (niche-DE) analysis that identifies cell-type-specific niche-associated genes, which are differentially expressed within a specific cell type in the context of specific spatial niches. We further develop niche-LR, a method to reveal ligand-receptor signaling mechanisms that underlie niche-differential gene expression patterns. Niche-DE and niche-LR are applicable to low-resolution spot-based spatial transcriptomics data and data that is single-cell or subcellular in resolution.
Collapse
Affiliation(s)
- Kaishu Mason
- Department of Statistics and Data Science, The Wharton School, University of Pennsylvania, Philadelphia, USA
| | - Anuja Sathe
- Division of Oncology, Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Paul R Hess
- Department of Statistics and Data Science, The Wharton School, University of Pennsylvania, Philadelphia, USA
| | - Jiazhen Rong
- Genomics and Computational Biology Graduate Program, Perelman School of Medicine, University of Pennsylvania, Philadelphia, USA
| | - Chi-Yun Wu
- The Gladstone Institute, San Francisco, USA
| | - Emma Furth
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, USA
| | - Katalin Susztak
- Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Jonathan Levinsohn
- Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Hanlee P Ji
- Division of Oncology, Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Nancy Zhang
- Department of Statistics and Data Science, The Wharton School, University of Pennsylvania, Philadelphia, USA.
| |
Collapse
|
27
|
Rashidi A, Billingham LK, Zolp A, Chia TY, Silvers C, Katz JL, Park CH, Delay S, Boland L, Geng Y, Markwell SM, Dmello C, Arrieta VA, Zilinger K, Jacob IM, Lopez-Rosas A, Hou D, Castro B, Steffens AM, McCortney K, Walshon JP, Flowers MS, Lin H, Wang H, Zhao J, Sonabend A, Zhang P, Ahmed AU, Brat DJ, Heiland DH, Lee-Chang C, Lesniak MS, Chandel NS, Miska J. Myeloid cell-derived creatine in the hypoxic niche promotes glioblastoma growth. Cell Metab 2024; 36:62-77.e8. [PMID: 38134929 PMCID: PMC10842612 DOI: 10.1016/j.cmet.2023.11.013] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Revised: 05/08/2023] [Accepted: 11/27/2023] [Indexed: 12/24/2023]
Abstract
Glioblastoma (GBM) is a malignancy dominated by the infiltration of tumor-associated myeloid cells (TAMCs). Examination of TAMC metabolic phenotypes in mouse models and patients with GBM identified the de novo creatine metabolic pathway as a hallmark of TAMCs. Multi-omics analyses revealed that TAMCs surround the hypoxic peri-necrotic regions of GBM and express the creatine metabolic enzyme glycine amidinotransferase (GATM). Conversely, GBM cells located within these same regions are uniquely specific in expressing the creatine transporter (SLC6A8). We hypothesized that TAMCs provide creatine to tumors, promoting GBM progression. Isotopic tracing demonstrated that TAMC-secreted creatine is taken up by tumor cells. Creatine supplementation protected tumors from hypoxia-induced stress, which was abrogated with genetic ablation or pharmacologic inhibition of SLC6A8. Lastly, inhibition of creatine transport using the clinically relevant compound, RGX-202-01, blunted tumor growth and enhanced radiation therapy in vivo. This work highlights that myeloid-to-tumor transfer of creatine promotes tumor growth in the hypoxic niche.
Collapse
Affiliation(s)
- Aida Rashidi
- Department of Neurological Surgery, Lou and Jean Malnati Brain Tumor Institute, Feinberg School of Medicine, Northwestern University, 676 N St. Clair, Suite 2210, Chicago, IL 60611, USA
| | - Leah K Billingham
- Department of Neurological Surgery, Lou and Jean Malnati Brain Tumor Institute, Feinberg School of Medicine, Northwestern University, 676 N St. Clair, Suite 2210, Chicago, IL 60611, USA
| | - Andrew Zolp
- Department of Neurological Surgery, Lou and Jean Malnati Brain Tumor Institute, Feinberg School of Medicine, Northwestern University, 676 N St. Clair, Suite 2210, Chicago, IL 60611, USA
| | - Tzu-Yi Chia
- Department of Neurological Surgery, Lou and Jean Malnati Brain Tumor Institute, Feinberg School of Medicine, Northwestern University, 676 N St. Clair, Suite 2210, Chicago, IL 60611, USA
| | - Caylee Silvers
- Department of Neurological Surgery, Lou and Jean Malnati Brain Tumor Institute, Feinberg School of Medicine, Northwestern University, 676 N St. Clair, Suite 2210, Chicago, IL 60611, USA
| | - Joshua L Katz
- Department of Neurological Surgery, Lou and Jean Malnati Brain Tumor Institute, Feinberg School of Medicine, Northwestern University, 676 N St. Clair, Suite 2210, Chicago, IL 60611, USA
| | - Cheol H Park
- Department of Neurological Surgery, Lou and Jean Malnati Brain Tumor Institute, Feinberg School of Medicine, Northwestern University, 676 N St. Clair, Suite 2210, Chicago, IL 60611, USA
| | - Suzi Delay
- Department of Neurological Surgery, Lou and Jean Malnati Brain Tumor Institute, Feinberg School of Medicine, Northwestern University, 676 N St. Clair, Suite 2210, Chicago, IL 60611, USA
| | - Lauren Boland
- Stanley Manne Children's Research Institute, Ann & Robert H. Lurie Children's Hospital, Chicago, IL, USA
| | - Yuheng Geng
- Department of Neurological Surgery, Lou and Jean Malnati Brain Tumor Institute, Feinberg School of Medicine, Northwestern University, 676 N St. Clair, Suite 2210, Chicago, IL 60611, USA
| | - Steven M Markwell
- Department of Pathology, Feinberg School of Medicine, Northwestern University, 303 East Chicago Avenue, Chicago, IL 60611, USA
| | - Crismita Dmello
- Department of Neurological Surgery, Lou and Jean Malnati Brain Tumor Institute, Feinberg School of Medicine, Northwestern University, 676 N St. Clair, Suite 2210, Chicago, IL 60611, USA
| | - Victor A Arrieta
- Department of Neurological Surgery, Lou and Jean Malnati Brain Tumor Institute, Feinberg School of Medicine, Northwestern University, 676 N St. Clair, Suite 2210, Chicago, IL 60611, USA
| | - Kaylee Zilinger
- Department of Neurological Surgery, Lou and Jean Malnati Brain Tumor Institute, Feinberg School of Medicine, Northwestern University, 676 N St. Clair, Suite 2210, Chicago, IL 60611, USA
| | - Irene M Jacob
- Department of Neurological Surgery, Lou and Jean Malnati Brain Tumor Institute, Feinberg School of Medicine, Northwestern University, 676 N St. Clair, Suite 2210, Chicago, IL 60611, USA
| | - Aurora Lopez-Rosas
- Department of Neurological Surgery, Lou and Jean Malnati Brain Tumor Institute, Feinberg School of Medicine, Northwestern University, 676 N St. Clair, Suite 2210, Chicago, IL 60611, USA
| | - David Hou
- Department of Neurological Surgery, Lou and Jean Malnati Brain Tumor Institute, Feinberg School of Medicine, Northwestern University, 676 N St. Clair, Suite 2210, Chicago, IL 60611, USA
| | - Brandyn Castro
- Department of Neurological Surgery, Lou and Jean Malnati Brain Tumor Institute, Feinberg School of Medicine, Northwestern University, 676 N St. Clair, Suite 2210, Chicago, IL 60611, USA
| | - Alicia M Steffens
- Department of Neurological Surgery, Lou and Jean Malnati Brain Tumor Institute, Feinberg School of Medicine, Northwestern University, 676 N St. Clair, Suite 2210, Chicago, IL 60611, USA
| | - Kathleen McCortney
- Department of Neurological Surgery, Lou and Jean Malnati Brain Tumor Institute, Feinberg School of Medicine, Northwestern University, 676 N St. Clair, Suite 2210, Chicago, IL 60611, USA
| | - Jordain P Walshon
- Department of Neurological Surgery, Lou and Jean Malnati Brain Tumor Institute, Feinberg School of Medicine, Northwestern University, 676 N St. Clair, Suite 2210, Chicago, IL 60611, USA
| | - Mariah S Flowers
- Department of Neurological Surgery, Lou and Jean Malnati Brain Tumor Institute, Feinberg School of Medicine, Northwestern University, 676 N St. Clair, Suite 2210, Chicago, IL 60611, USA
| | - Hanchen Lin
- Department of Neurological Surgery, Lou and Jean Malnati Brain Tumor Institute, Feinberg School of Medicine, Northwestern University, 676 N St. Clair, Suite 2210, Chicago, IL 60611, USA
| | - Hanxiang Wang
- Department of Neurological Surgery, Lou and Jean Malnati Brain Tumor Institute, Feinberg School of Medicine, Northwestern University, 676 N St. Clair, Suite 2210, Chicago, IL 60611, USA
| | - Junfei Zhao
- Department of Systems Biology, Columbia University, New York, NY, USA
| | - Adam Sonabend
- Department of Neurological Surgery, Lou and Jean Malnati Brain Tumor Institute, Feinberg School of Medicine, Northwestern University, 676 N St. Clair, Suite 2210, Chicago, IL 60611, USA
| | - Peng Zhang
- Department of Neurological Surgery, Lou and Jean Malnati Brain Tumor Institute, Feinberg School of Medicine, Northwestern University, 676 N St. Clair, Suite 2210, Chicago, IL 60611, USA
| | - Atique U Ahmed
- Department of Neurological Surgery, Lou and Jean Malnati Brain Tumor Institute, Feinberg School of Medicine, Northwestern University, 676 N St. Clair, Suite 2210, Chicago, IL 60611, USA
| | - Daniel J Brat
- Department of Pathology, Feinberg School of Medicine, Northwestern University, 303 East Chicago Avenue, Chicago, IL 60611, USA
| | - Dieter H Heiland
- Department of Neurological Surgery, Lou and Jean Malnati Brain Tumor Institute, Feinberg School of Medicine, Northwestern University, 676 N St. Clair, Suite 2210, Chicago, IL 60611, USA; Microenvironment and Immunology Research Laboratory, Medical Center, University of Freiburg, 79106 Freiburg, Germany; Department of Neurosurgery, Medical Center, University of Freiburg, 79106 Freiburg, Germany. German Cancer Consortium (DKTK), partner site Freiburg, Freiburg, Germany
| | - Catalina Lee-Chang
- Department of Neurological Surgery, Lou and Jean Malnati Brain Tumor Institute, Feinberg School of Medicine, Northwestern University, 676 N St. Clair, Suite 2210, Chicago, IL 60611, USA
| | - Maciej S Lesniak
- Department of Neurological Surgery, Lou and Jean Malnati Brain Tumor Institute, Feinberg School of Medicine, Northwestern University, 676 N St. Clair, Suite 2210, Chicago, IL 60611, USA
| | - Navdeep S Chandel
- Department of Medicine, Feinberg School of Medicine, Northwestern University, 676 North St. Clair Street, Suite 2330, Chicago, IL 60611, USA
| | - Jason Miska
- Department of Neurological Surgery, Lou and Jean Malnati Brain Tumor Institute, Feinberg School of Medicine, Northwestern University, 676 N St. Clair, Suite 2210, Chicago, IL 60611, USA.
| |
Collapse
|
28
|
Zormpas E, Queen R, Comber A, Cockell SJ. Mapping the transcriptome: Realizing the full potential of spatial data analysis. Cell 2023; 186:5677-5689. [PMID: 38065099 DOI: 10.1016/j.cell.2023.11.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Revised: 09/04/2023] [Accepted: 11/02/2023] [Indexed: 12/24/2023]
Abstract
RNA sequencing in situ allows for whole-transcriptome characterization at high resolution, while retaining spatial information. These data present an analytical challenge for bioinformatics-how to leverage spatial information effectively? Properties of data with a spatial dimension require special handling, which necessitate a different set of statistical and inferential considerations when compared to non-spatial data. The geographical sciences primarily use spatial data and have developed methods to analye them. Here we discuss the challenges associated with spatial analysis and examine how we can take advantage of practice from the geographical sciences to realize the full potential of spatial information in transcriptomic datasets.
Collapse
Affiliation(s)
- Eleftherios Zormpas
- Biosciences Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne NE2 4HH, UK
| | - Rachel Queen
- Biosciences Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne NE2 4HH, UK; Bioinformatics Support Unit, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne NE2 4HH, UK
| | - Alexis Comber
- School of Geography and Leeds Institute for Data Analytics, University of Leeds, Leeds LS2 9NL, UK
| | - Simon J Cockell
- Biosciences Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne NE2 4HH, UK; School of Biomedical, Nutritional and Sport Sciences, Faculty of Medical Sciences, Newcastle upon Tyne NE2 4HH, UK.
| |
Collapse
|
29
|
Chen H, Lee YJ, Ovando JA, Rosas L, Rojas M, Mora AL, Bar-Joseph Z, Lugo-Martinez J. scResolve: Recovering single cell expression profiles from multi-cellular spatial transcriptomics. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.12.18.572269. [PMID: 38187629 PMCID: PMC10769299 DOI: 10.1101/2023.12.18.572269] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/09/2024]
Abstract
Many popular spatial transcriptomics techniques lack single-cell resolution. Instead, these methods measure the collective gene expression for each location from a mixture of cells, potentially containing multiple cell types. Here, we developed scResolve, a method for recovering single-cell expression profiles from spatial transcriptomics measurements at multi-cellular resolution. scResolve accurately restores expression profiles of individual cells at their locations, which is unattainable from cell type deconvolution. Applications of scResolve on human breast cancer data and human lung disease data demonstrate that scResolve enables cell type-specific differential gene expression analysis between different tissue contexts and accurate identification of rare cell populations. The spatially resolved cellular-level expression profiles obtained through scResolve facilitate more flexible and precise spatial analysis that complements raw multi-cellular level analysis.
Collapse
Affiliation(s)
- Hao Chen
- Ray and Stephanie Lane Computational Biology Department, School of Computer Science, Carnegie Mellon University, Pittsburgh, PA 15213, USA
| | - Young Je Lee
- Ray and Stephanie Lane Computational Biology Department, School of Computer Science, Carnegie Mellon University, Pittsburgh, PA 15213, USA
| | - Jose A. Ovando
- Dorothy M. Davis Heart and Lung Research Institute, Division of Pulmonary, Critical Care and Sleep Medicine, Department of Internal Medicine, Ohio State University, Columbus, OH 43210, USA
| | - Lorena Rosas
- Dorothy M. Davis Heart and Lung Research Institute, Division of Pulmonary, Critical Care and Sleep Medicine, Department of Internal Medicine, Ohio State University, Columbus, OH 43210, USA
| | - Mauricio Rojas
- Dorothy M. Davis Heart and Lung Research Institute, Division of Pulmonary, Critical Care and Sleep Medicine, Department of Internal Medicine, Ohio State University, Columbus, OH 43210, USA
| | - Ana L. Mora
- Dorothy M. Davis Heart and Lung Research Institute, Division of Pulmonary, Critical Care and Sleep Medicine, Department of Internal Medicine, Ohio State University, Columbus, OH 43210, USA
| | - Ziv Bar-Joseph
- Ray and Stephanie Lane Computational Biology Department, School of Computer Science, Carnegie Mellon University, Pittsburgh, PA 15213, USA
- Machine Learning Department, School of Computer Science, Carnegie Mellon University, Pittsburgh, PA 15213, USA
| | - Jose Lugo-Martinez
- Ray and Stephanie Lane Computational Biology Department, School of Computer Science, Carnegie Mellon University, Pittsburgh, PA 15213, USA
| |
Collapse
|
30
|
Jiang X, Dong L, Wang S, Wen Z, Chen M, Xu L, Xiao G, Li Q. Reconstructing Spatial Transcriptomics at the Single-cell Resolution with BayesDeep. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.12.07.570715. [PMID: 38106214 PMCID: PMC10723442 DOI: 10.1101/2023.12.07.570715] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2023]
Abstract
Spatially resolved transcriptomics (SRT) techniques have revolutionized the characterization of molecular profiles while preserving spatial and morphological context. However, most next-generation sequencing-based SRT techniques are limited to measuring gene expression in a confined array of spots, capturing only a fraction of the spatial domain. Typically, these spots encompass gene expression from a few to hundreds of cells, underscoring a critical need for more detailed, single-cell resolution SRT data to enhance our understanding of biological functions within the tissue context. Addressing this challenge, we introduce BayesDeep, a novel Bayesian hierarchical model that leverages cellular morphological data from histology images, commonly paired with SRT data, to reconstruct SRT data at the single-cell resolution. BayesDeep effectively model count data from SRT studies via a negative binomial regression model. This model incorporates explanatory variables such as cell types and nuclei-shape information for each cell extracted from the paired histology image. A feature selection scheme is integrated to examine the association between the morphological and molecular profiles, thereby improving the model robustness. We applied BayesDeep to two real SRT datasets, successfully demonstrating its capability to reconstruct SRT data at the single-cell resolution. This advancement not only yields new biological insights but also significantly enhances various downstream analyses, such as pseudotime and cell-cell communication.
Collapse
Affiliation(s)
- Xi Jiang
- Quantitative Biomedical Research Center, Peter O’Donnell Jr. School of Public Health, The University of Texas Southwestern Medical Center, Dallas, Texas, U.S.A
- Department of Statistics and Data Science, Southern Methodist University, Dallas, Texas, U.S.A
| | - Lei Dong
- Quantitative Biomedical Research Center, Peter O’Donnell Jr. School of Public Health, The University of Texas Southwestern Medical Center, Dallas, Texas, U.S.A
| | - Shidan Wang
- Quantitative Biomedical Research Center, Peter O’Donnell Jr. School of Public Health, The University of Texas Southwestern Medical Center, Dallas, Texas, U.S.A
| | - Zhuoyu Wen
- Quantitative Biomedical Research Center, Peter O’Donnell Jr. School of Public Health, The University of Texas Southwestern Medical Center, Dallas, Texas, U.S.A
| | - Mingyi Chen
- Department of Pathology, The University of Texas Southwestern Medical Center, Dallas, Texas, U.S.A
| | - Lin Xu
- Quantitative Biomedical Research Center, Peter O’Donnell Jr. School of Public Health, The University of Texas Southwestern Medical Center, Dallas, Texas, U.S.A
- Department of Pediatrics, Division of Hematology/Oncology, University of Texas Southwestern Medical Center, Dallas, Texas, U.S.A
| | - Guanghua Xiao
- Quantitative Biomedical Research Center, Peter O’Donnell Jr. School of Public Health, The University of Texas Southwestern Medical Center, Dallas, Texas, U.S.A
| | - Qiwei Li
- Department of Mathematical Sciences, The University of Texas at Dallas, Richardson, Texas, U.S.A
| |
Collapse
|
31
|
Johnston K, Berackey BB, Tran KM, Gelber A, Yu Z, MacGregor G, Mukamel EA, Tan Z, Green K, Xu X. Single cell spatial transcriptomics reveals distinct patterns of dysregulation in non-neuronal and neuronal cells induced by the Trem2R47H Alzheimer's risk gene mutation. RESEARCH SQUARE 2023:rs.3.rs-3656139. [PMID: 38106071 PMCID: PMC10723554 DOI: 10.21203/rs.3.rs-3656139/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2023]
Abstract
INTRODUCTION The R47H missense mutation of the TREM2 gene is a strong risk factor for development of Alzheimer's Disease. We investigate cell-type-specific spatial transcriptomic changes induced by the Trem2R47H mutation to determine the impacts of this mutation on transcriptional dysregulation. METHODS We profiled 15 mouse brain sections consisting of wild-type, Trem2R47H, 5xFAD and Trem2R47H; 5xFAD genotypes using MERFISH spatial transcriptomics. Single-cell spatial transcriptomics and neuropathology data were analyzed using our custom pipeline to identify plaque and Trem2R47H induced transcriptomic dysregulation. RESULTS The Trem2R47H mutation induced consistent upregulation of Bdnf and Ntrk2 across many cortical excitatory neuron types, independent of amyloid pathology. Spatial investigation of genotype enriched subclusters identified spatially localized neuronal subpopulations reduced in 5xFAD and Trem2R47H; 5xFAD mice. CONCLUSION Spatial transcriptomics analysis identifies glial and neuronal transcriptomic alterations induced independently by 5xFAD and Trem2R47H mutations, impacting inflammatory responses in microglia and astrocytes, and activity and BDNF signaling in neurons.
Collapse
|
32
|
Toninelli M, Rossetti G, Pagani M. Charting the tumor microenvironment with spatial profiling technologies. Trends Cancer 2023; 9:1085-1096. [PMID: 37673713 DOI: 10.1016/j.trecan.2023.08.004] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Revised: 08/04/2023] [Accepted: 08/10/2023] [Indexed: 09/08/2023]
Abstract
In recent years technologies that can achieve readouts at cellular resolution such as single-cell RNA sequencing (scRNA-seq) have provided a comprehensive characterization of the cellular proportions and phenotypes that populate the tumor microenvironment (TME). However, because of the sample dissociation steps required by these protocols, they fail to capture information related to the intricate spatial context in which cells operate as well as their dense networks of interactions. Spatial profiling technologies have recently emerged as a valuable way to investigate the physical organization of cells crowding the TME in intact tissues. In this review we first discuss how spatial profiling technologies have propelled TME characterization, and then explore their potential to improve both diagnosis and prognosis for cancer patients in the clinic.
Collapse
Affiliation(s)
- Mattia Toninelli
- IFOM ETS, The AIRC Institute of Molecular Oncology, Milan, Italy
| | - Grazisa Rossetti
- IFOM ETS, The AIRC Institute of Molecular Oncology, Milan, Italy
| | - Massimiliano Pagani
- IFOM ETS, The AIRC Institute of Molecular Oncology, Milan, Italy; Department of Medical Biotechnology and Translational Medicine (BIOMETRA), Università degli Studi, Milan, Italy.
| |
Collapse
|
33
|
Wan X, Xiao J, Tam SST, Cai M, Sugimura R, Wang Y, Wan X, Lin Z, Wu AR, Yang C. Integrating spatial and single-cell transcriptomics data using deep generative models with SpatialScope. Nat Commun 2023; 14:7848. [PMID: 38030617 PMCID: PMC10687049 DOI: 10.1038/s41467-023-43629-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Accepted: 11/13/2023] [Indexed: 12/01/2023] Open
Abstract
The rapid emergence of spatial transcriptomics (ST) technologies is revolutionizing our understanding of tissue spatial architecture and biology. Although current ST methods, whether based on next-generation sequencing (seq-based approaches) or fluorescence in situ hybridization (image-based approaches), offer valuable insights, they face limitations either in cellular resolution or transcriptome-wide profiling. To address these limitations, we present SpatialScope, a unified approach integrating scRNA-seq reference data and ST data using deep generative models. With innovation in model and algorithm designs, SpatialScope not only enhances seq-based ST data to achieve single-cell resolution, but also accurately infers transcriptome-wide expression levels for image-based ST data. We demonstrate SpatialScope's utility through simulation studies and real data analysis from both seq-based and image-based ST approaches. SpatialScope provides spatial characterization of tissue structures at transcriptome-wide single-cell resolution, facilitating downstream analysis, including detecting cellular communication through ligand-receptor interactions, localizing cellular subtypes, and identifying spatially differentially expressed genes.
Collapse
Affiliation(s)
- Xiaomeng Wan
- Department of Mathematics, The Hong Kong University of Science and Technology, Hong Kong SAR, China
| | - Jiashun Xiao
- Shenzhen Research Institute of Big Data, Shenzhen, 518172, China
| | - Sindy Sing Ting Tam
- Division of Life Science, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong SAR, China
| | - Mingxuan Cai
- Department of Biostatistics, City University of Hong Kong, Hong Kong SAR, China
| | - Ryohichi Sugimura
- Li Ka Shing Faculty of Medicine, School of Biomedical Sciences, University of Hong Kong, Hong Kong SAR, China
| | - Yang Wang
- Department of Mathematics, The Hong Kong University of Science and Technology, Hong Kong SAR, China
- Guangdong-Hong Kong-Macao Joint Laboratory for Data-Driven Fluid Mechanics and Engineering Applications, The Hong Kong University of Science and Technology, Hong Kong SAR, China
- Big Data Bio-Intelligence Lab, The Hong Kong University of Science and Technology, Hong Kong SAR, China
| | - Xiang Wan
- Shenzhen Research Institute of Big Data, Shenzhen, 518172, China
| | - Zhixiang Lin
- Department of Statistics, The Chinese University of Hong Kong, Hong Kong SAR, China.
| | - Angela Ruohao Wu
- Division of Life Science, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong SAR, China.
- Department of Chemical and Biological Engineering, The Hong Kong University of Science and Technology, Hong Kong SAR, China.
- Center for Aging Science, The Hong Kong University of Science and Technology, Hong Kong SAR, China.
- State Key Laboratory of Molecular Neuroscience, The Hong Kong University of Science and Technology, Hong Kong SAR, China.
| | - Can Yang
- Department of Mathematics, The Hong Kong University of Science and Technology, Hong Kong SAR, China.
- Guangdong-Hong Kong-Macao Joint Laboratory for Data-Driven Fluid Mechanics and Engineering Applications, The Hong Kong University of Science and Technology, Hong Kong SAR, China.
- Big Data Bio-Intelligence Lab, The Hong Kong University of Science and Technology, Hong Kong SAR, China.
| |
Collapse
|
34
|
Zhang C, Dong K, Aihara K, Chen L, Zhang S. STAMarker: determining spatial domain-specific variable genes with saliency maps in deep learning. Nucleic Acids Res 2023; 51:e103. [PMID: 37811885 PMCID: PMC10639070 DOI: 10.1093/nar/gkad801] [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: 12/08/2022] [Revised: 08/26/2023] [Accepted: 09/19/2023] [Indexed: 10/10/2023] Open
Abstract
Spatial transcriptomics characterizes gene expression profiles while retaining the information of the spatial context, providing an unprecedented opportunity to understand cellular systems. One of the essential tasks in such data analysis is to determine spatially variable genes (SVGs), which demonstrate spatial expression patterns. Existing methods only consider genes individually and fail to model the inter-dependence of genes. To this end, we present an analytic tool STAMarker for robustly determining spatial domain-specific SVGs with saliency maps in deep learning. STAMarker is a three-stage ensemble framework consisting of graph-attention autoencoders, multilayer perceptron (MLP) classifiers, and saliency map computation by the backpropagated gradient. We illustrate the effectiveness of STAMarker and compare it with serveral commonly used competing methods on various spatial transcriptomic data generated by different platforms. STAMarker considers all genes at once and is more robust when the dataset is very sparse. STAMarker could identify spatial domain-specific SVGs for characterizing spatial domains and enable in-depth analysis of the region of interest in the tissue section.
Collapse
Affiliation(s)
- Chihao Zhang
- NCMIS, CEMS, RCSDS, Academy of Mathematics and Systems Science, Chinese Academy of Sciences, Beijing 100190, China
- School of Mathematical Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Kangning Dong
- NCMIS, CEMS, RCSDS, Academy of Mathematics and Systems Science, Chinese Academy of Sciences, Beijing 100190, China
- School of Mathematical Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Kazuyuki Aihara
- International Research Center for Neurointelligence, The University of Tokyo Institutes for Advanced Study, The University of Tokyo, Tokyo 113-0033, Japan
| | - Luonan Chen
- Key Laboratory of Systems Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai 200031, China
- Key Laboratory of Systems Health Science of Zhejiang Province, School of Life Science, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou 310024, China
- School of Life Science and Technology, Shanghai Tech University, Shanghai 201210, China
- Guangdong Institute of Intelligence Science and Technology, Hengqin, Zhuhai, Guangdong 519031, China
| | - Shihua Zhang
- NCMIS, CEMS, RCSDS, Academy of Mathematics and Systems Science, Chinese Academy of Sciences, Beijing 100190, China
- School of Mathematical Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
- Key Laboratory of Systems Health Science of Zhejiang Province, School of Life Science, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou 310024, China
| |
Collapse
|
35
|
Kim H, Kumar A, Lövkvist C, Palma AM, Martin P, Kim J, Bhoopathi P, Trevino J, Fisher P, Madan E, Gogna R, Won KJ. CellNeighborEX: deciphering neighbor-dependent gene expression from spatial transcriptomics data. Mol Syst Biol 2023; 19:e11670. [PMID: 37815040 PMCID: PMC10632736 DOI: 10.15252/msb.202311670] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Revised: 09/14/2023] [Accepted: 09/18/2023] [Indexed: 10/11/2023] Open
Abstract
Cells have evolved their communication methods to sense their microenvironments and send biological signals. In addition to communication using ligands and receptors, cells use diverse channels including gap junctions to communicate with their immediate neighbors. Current approaches, however, cannot effectively capture the influence of various microenvironments. Here, we propose a novel approach to investigate cell neighbor-dependent gene expression (CellNeighborEX) in spatial transcriptomics (ST) data. To categorize cells based on their microenvironment, CellNeighborEX uses direct cell location or the mixture of transcriptome from multiple cells depending on ST technologies. For each cell type, CellNeighborEX identifies diverse gene sets associated with partnering cell types, providing further insight. We found that cells express different genes depending on their neighboring cell types in various tissues including mouse embryos, brain, and liver cancer. Those genes are associated with critical biological processes such as development or metastases. We further validated that gene expression is induced by neighboring partners via spatial visualization. The neighbor-dependent gene expression suggests new potential genes involved in cell-cell interactions beyond what ligand-receptor co-expression can discover.
Collapse
Affiliation(s)
- Hyobin Kim
- Department of Computational BiomedicineCedars‐Sinai Medical CenterHollywoodCAUSA
- Biotech Research and Innovation Centre (BRIC)University of CopenhagenCopenhagenDenmark
| | - Amit Kumar
- Massey Cancer CenterVirginia Commonwealth UniversityRichmondVAUSA
- School of Medicine, Institute of Molecular MedicineVirginia Commonwealth UniversityRichmondVAUSA
- Department of Human and Molecular Genetics, School of MedicineVirginia Commonwealth UniversityRichmondVAUSA
| | - Cecilia Lövkvist
- Novo Nordisk Foundation Center for Stem Cell Medicine, reNEWUniversity of CopenhagenCopenhagenDenmark
| | - António M Palma
- Massey Cancer CenterVirginia Commonwealth UniversityRichmondVAUSA
- School of Medicine, Institute of Molecular MedicineVirginia Commonwealth UniversityRichmondVAUSA
- Instituto Superior TecnicoUniversidade de LisboaLisboaPortugal
| | - Patrick Martin
- Department of Computational BiomedicineCedars‐Sinai Medical CenterHollywoodCAUSA
- Biotech Research and Innovation Centre (BRIC)University of CopenhagenCopenhagenDenmark
| | - Junil Kim
- School of Systems Biomedical ScienceSoongsil UniversitySeoulKorea
| | - Praveen Bhoopathi
- Massey Cancer CenterVirginia Commonwealth UniversityRichmondVAUSA
- School of Medicine, Institute of Molecular MedicineVirginia Commonwealth UniversityRichmondVAUSA
- Department of Human and Molecular Genetics, School of MedicineVirginia Commonwealth UniversityRichmondVAUSA
| | - Jose Trevino
- Massey Cancer CenterVirginia Commonwealth UniversityRichmondVAUSA
- Department of Surgery, School of MedicineVirginia Commonwealth UniversityRichmondVAUSA
| | - Paul Fisher
- Massey Cancer CenterVirginia Commonwealth UniversityRichmondVAUSA
- School of Medicine, Institute of Molecular MedicineVirginia Commonwealth UniversityRichmondVAUSA
- Department of Human and Molecular Genetics, School of MedicineVirginia Commonwealth UniversityRichmondVAUSA
| | - Esha Madan
- Massey Cancer CenterVirginia Commonwealth UniversityRichmondVAUSA
- School of Medicine, Institute of Molecular MedicineVirginia Commonwealth UniversityRichmondVAUSA
- Department of Human and Molecular Genetics, School of MedicineVirginia Commonwealth UniversityRichmondVAUSA
- Department of Surgery, School of MedicineVirginia Commonwealth UniversityRichmondVAUSA
| | - Rajan Gogna
- Massey Cancer CenterVirginia Commonwealth UniversityRichmondVAUSA
- School of Medicine, Institute of Molecular MedicineVirginia Commonwealth UniversityRichmondVAUSA
- Department of Human and Molecular Genetics, School of MedicineVirginia Commonwealth UniversityRichmondVAUSA
- Department of Surgery, School of MedicineVirginia Commonwealth UniversityRichmondVAUSA
| | - Kyoung Jae Won
- Department of Computational BiomedicineCedars‐Sinai Medical CenterHollywoodCAUSA
- Biotech Research and Innovation Centre (BRIC)University of CopenhagenCopenhagenDenmark
| |
Collapse
|
36
|
Mangoli A, Wu S, Liu HQ, Aksu M, Jain V, Foreman BE, Regal JA, Weidenhammer LB, Stewart CE, Guerra Garcia ME, Hocke E, Abramson K, Williams NT, Luo L, Deland K, Attardi L, Abe K, Hashizume R, Ashley DM, Becher OJ, Kirsch DG, Gregory SG, Reitman ZJ. Ataxia-telangiectasia mutated ( Atm ) disruption sensitizes spatially-directed H3.3K27M/TP53 diffuse midline gliomas to radiation therapy. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.10.18.562892. [PMID: 37904990 PMCID: PMC10614905 DOI: 10.1101/2023.10.18.562892] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/02/2023]
Abstract
Diffuse midline gliomas (DMGs) are lethal brain tumors characterized by p53-inactivating mutations and oncohistone H3.3K27M mutations that rewire the cellular response to genotoxic stress, which presents therapeutic opportunities. We used RCAS/tv-a retroviruses and Cre recombinase to inactivate p53 and induce K27M in the native H3f3a allele in a lineage- and spatially-directed manner, yielding primary mouse DMGs. Genetic or pharmacologic disruption of the DNA damage response kinase Ataxia-telangiectasia mutated (ATM) enhanced the efficacy of focal brain irradiation, extending mouse survival. This finding suggests that targeting ATM will enhance the efficacy of radiation therapy for p53-mutant DMG but not p53-wildtype DMG. We used spatial in situ transcriptomics and an allelic series of primary murine DMG models with different p53 mutations to identify transactivation-independent p53 activity as a key mediator of such radiosensitivity. These studies deeply profile a genetically faithful and versatile model of a lethal brain tumor to identify resistance mechanisms for a therapeutic strategy currently in clinical trials.
Collapse
|
37
|
Carbonetto P, Luo K, Sarkar A, Hung A, Tayeb K, Pott S, Stephens M. GoM DE: interpreting structure in sequence count data with differential expression analysis allowing for grades of membership. Genome Biol 2023; 24:236. [PMID: 37858253 PMCID: PMC10588049 DOI: 10.1186/s13059-023-03067-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Accepted: 09/20/2023] [Indexed: 10/21/2023] Open
Abstract
Parts-based representations, such as non-negative matrix factorization and topic modeling, have been used to identify structure from single-cell sequencing data sets, in particular structure that is not as well captured by clustering or other dimensionality reduction methods. However, interpreting the individual parts remains a challenge. To address this challenge, we extend methods for differential expression analysis by allowing cells to have partial membership to multiple groups. We call this grade of membership differential expression (GoM DE). We illustrate the benefits of GoM DE for annotating topics identified in several single-cell RNA-seq and ATAC-seq data sets.
Collapse
Affiliation(s)
- Peter Carbonetto
- Department of Human Genetics, University of Chicago, Chicago, IL, USA
- Research Computing Center, University of Chicago, Chicago, IL, USA
| | - Kaixuan Luo
- Department of Human Genetics, University of Chicago, Chicago, IL, USA
| | - Abhishek Sarkar
- Department of Human Genetics, University of Chicago, Chicago, IL, USA
- Vesalius Therapeutics, Cambridge, MA, USA
| | - Anthony Hung
- Department of Human Genetics, University of Chicago, Chicago, IL, USA
- Section of Genetic Medicine, University of Chicago, Chicago, IL, USA
| | - Karl Tayeb
- Department of Human Genetics, University of Chicago, Chicago, IL, USA
- Committee on Genetics, Genomics and Systems Biology, University of Chicago, Chicago, IL, USA
| | - Sebastian Pott
- Department of Human Genetics, University of Chicago, Chicago, IL, USA
- Section of Genetic Medicine, University of Chicago, Chicago, IL, USA
| | - Matthew Stephens
- Department of Human Genetics, University of Chicago, Chicago, IL, USA.
- Department of Statistics, University of Chicago, Chicago, IL, USA.
| |
Collapse
|
38
|
Chitra U, Arnold BJ, Sarkar H, Ma C, Lopez-Darwin S, Sanno K, Raphael BJ. Mapping the topography of spatial gene expression with interpretable deep learning. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.10.10.561757. [PMID: 37873258 PMCID: PMC10592770 DOI: 10.1101/2023.10.10.561757] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/25/2023]
Abstract
Spatially resolved transcriptomics technologies provide high-throughput measurements of gene expression in a tissue slice, but the sparsity of this data complicates the analysis of spatial gene expression patterns such as gene expression gradients. We address these issues by deriving a topographic map of a tissue slice-analogous to a map of elevation in a landscape-using a novel quantity called the isodepth. Contours of constant isodepth enclose spatial domains with distinct cell type composition, while gradients of the isodepth indicate spatial directions of maximum change in gene expression. We develop GASTON, an unsupervised and interpretable deep learning algorithm that simultaneously learns the isodepth, spatial gene expression gradients, and piecewise linear functions of the isodepth that model both continuous gradients and discontinuous spatial variation in the expression of individual genes. We validate GASTON by showing that it accurately identifies spatial domains and marker genes across several biological systems. In SRT data from the brain, GASTON reveals gradients of neuronal differentiation and firing, and in SRT data from a tumor sample, GASTON infers gradients of metabolic activity and epithelial-mesenchymal transition (EMT)-related gene expression in the tumor microenvironment.
Collapse
Affiliation(s)
- Uthsav Chitra
- Department of Computer Science, Princeton University, Princeton, NJ, USA
| | - Brian J. Arnold
- Department of Computer Science, Princeton University, Princeton, NJ, USA
- Center for Statistics and Machine Learning, Princeton University, Princeton, NJ, USA
| | - Hirak Sarkar
- Department of Computer Science, Princeton University, Princeton, NJ, USA
- Ludwig Cancer Institute, Princeton Branch, Princeton University, Princeton, NJ, USA
| | - Cong Ma
- Department of Computer Science, Princeton University, Princeton, NJ, USA
| | | | - Kohei Sanno
- Department of Computer Science, Princeton University, Princeton, NJ, USA
- Center for Statistics and Machine Learning, Princeton University, Princeton, NJ, USA
| | | |
Collapse
|
39
|
Zhou R, Yang G, Zhang Y, Wang Y. Spatial transcriptomics in development and disease. MOLECULAR BIOMEDICINE 2023; 4:32. [PMID: 37806992 PMCID: PMC10560656 DOI: 10.1186/s43556-023-00144-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Accepted: 08/29/2023] [Indexed: 10/10/2023] Open
Abstract
The proper functioning of diverse biological systems depends on the spatial organization of their cells, a critical factor for biological processes like shaping intricate tissue functions and precisely determining cell fate. Nonetheless, conventional bulk or single-cell RNA sequencing methods were incapable of simultaneously capturing both gene expression profiles and the spatial locations of cells. Hence, a multitude of spatially resolved technologies have emerged, offering a novel dimension for investigating regional gene expression, spatial domains, and interactions between cells. Spatial transcriptomics (ST) is a method that maps gene expression in tissue while preserving spatial information. It can reveal cellular heterogeneity, spatial organization and functional interactions in complex biological systems. ST can also complement and integrate with other omics methods to provide a more comprehensive and holistic view of biological systems at multiple levels of resolution. Since the advent of ST, new methods offering higher throughput and resolution have become available, holding significant potential to expedite fresh insights into comprehending biological complexity. Consequently, a rapid increase in associated research has occurred, using these technologies to unravel the spatial complexity during developmental processes or disease conditions. In this review, we summarize the recent advancement of ST in historical, technical, and application contexts. We compare different types of ST methods based on their principles and workflows, and present the bioinformatics tools for analyzing and integrating ST data with other modalities. We also highlight the applications of ST in various domains of biomedical research, especially development and diseases. Finally, we discuss the current limitations and challenges in the field, and propose the future directions of ST.
Collapse
Affiliation(s)
- Ran Zhou
- Department of Neurosurgery, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Gaoxia Yang
- Department of Neurosurgery, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, 610041, China
- National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Yan Zhang
- National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China.
| | - Yuan Wang
- Department of Neurosurgery, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, 610041, China.
| |
Collapse
|
40
|
Voigt AP, Mullin NK, Navratil EM, Flamme-Wiese MJ, Lin LC, Scheetz TE, Han IC, Stone EM, Tucker BA, Mullins RF. Gene Expression Within a Human Choroidal Neovascular Membrane Using Spatial Transcriptomics. Invest Ophthalmol Vis Sci 2023; 64:40. [PMID: 37878301 PMCID: PMC10615143 DOI: 10.1167/iovs.64.13.40] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Accepted: 10/07/2023] [Indexed: 10/26/2023] Open
Abstract
Purpose Macular neovascularization is a relatively common and potentially visually devastating complication of age-related macular degeneration. In macular neovascularization, pathologic angiogenesis can originate from either the choroid or the retina, but we have limited understanding of how different cell types become dysregulated in this dynamic process. Methods To study how gene expression is altered in focal areas of pathology, we performed spatial RNA sequencing on a human donor eye with macular neovascularization as well as a healthy control donor. We performed differential expression to identify genes enriched within the area of macular neovascularization and used deconvolution algorithms to predict the originating cell type of these dysregulated genes. Results Within the area of neovascularization, endothelial cells demonstrated increased expression of genes related to Rho family GTPase signaling and integrin signaling. Likewise, VEGF and TGFB1 were identified as potential upstream regulators that could drive the observed gene expression changes produced by endothelial and retinal pigment epithelium cells in the macular neovascularization donor. These spatial gene expression profiles were compared to previous single-cell gene expression experiments in human age-related macular degeneration as well as a model of laser-induced neovascularization in mice. As a secondary aim, we investigated regional gene expression patterns within the macular neural retina and between the macular and peripheral choroid. Conclusions Overall, this study spatially analyzes gene expression across the retina, retinal pigment epithelium, and choroid in health and describes a set of candidate molecules that become dysregulated in macular neovascularization.
Collapse
Affiliation(s)
- Andrew P. Voigt
- Department of Ophthalmology and Visual Sciences, the University of Iowa Carver College of Medicine, Iowa City, Iowa, United States
- Institute for Vision Research, the University of Iowa, Iowa City, Iowa, United States
| | - Nathaniel K. Mullin
- Department of Ophthalmology and Visual Sciences, the University of Iowa Carver College of Medicine, Iowa City, Iowa, United States
- Institute for Vision Research, the University of Iowa, Iowa City, Iowa, United States
| | - Emma M. Navratil
- Department of Ophthalmology and Visual Sciences, the University of Iowa Carver College of Medicine, Iowa City, Iowa, United States
- Institute for Vision Research, the University of Iowa, Iowa City, Iowa, United States
| | - Miles J. Flamme-Wiese
- Department of Ophthalmology and Visual Sciences, the University of Iowa Carver College of Medicine, Iowa City, Iowa, United States
- Institute for Vision Research, the University of Iowa, Iowa City, Iowa, United States
| | - Li-Chun Lin
- University of Iowa Neuroscience Institute, the University of Iowa Carver College of Medicine, Iowa City, Iowa, United States
| | - Todd E. Scheetz
- Department of Ophthalmology and Visual Sciences, the University of Iowa Carver College of Medicine, Iowa City, Iowa, United States
- Institute for Vision Research, the University of Iowa, Iowa City, Iowa, United States
| | - Ian C. Han
- Department of Ophthalmology and Visual Sciences, the University of Iowa Carver College of Medicine, Iowa City, Iowa, United States
- Institute for Vision Research, the University of Iowa, Iowa City, Iowa, United States
| | - Edwin M. Stone
- Department of Ophthalmology and Visual Sciences, the University of Iowa Carver College of Medicine, Iowa City, Iowa, United States
- Institute for Vision Research, the University of Iowa, Iowa City, Iowa, United States
| | - Budd A. Tucker
- Department of Ophthalmology and Visual Sciences, the University of Iowa Carver College of Medicine, Iowa City, Iowa, United States
- Institute for Vision Research, the University of Iowa, Iowa City, Iowa, United States
| | - Robert F. Mullins
- Department of Ophthalmology and Visual Sciences, the University of Iowa Carver College of Medicine, Iowa City, Iowa, United States
- Institute for Vision Research, the University of Iowa, Iowa City, Iowa, United States
| |
Collapse
|
41
|
Zhou X, Dong K, Zhang S. Integrating spatial transcriptomics data across different conditions, technologies and developmental stages. NATURE COMPUTATIONAL SCIENCE 2023; 3:894-906. [PMID: 38177758 DOI: 10.1038/s43588-023-00528-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Accepted: 09/05/2023] [Indexed: 01/06/2024]
Abstract
With the rapid generation of spatial transcriptomics (ST) data, integrative analysis of multiple ST datasets from different conditions, technologies and developmental stages is becoming increasingly important. Here we present a graph attention neural network called STAligner for integrating and aligning ST datasets, enabling spatially aware data integration, simultaneous spatial domain identification and downstream comparative analysis. We apply STAligner to ST datasets of the human cortex slices from different samples, the mouse olfactory bulb slices generated by two profiling technologies, the mouse hippocampus tissue slices under normal and Alzheimer's disease conditions, and the spatiotemporal atlases of mouse organogenesis. STAligner efficiently captures the shared tissue structures across different slices, the disease-related substructures and the dynamical changes during mouse embryonic development. In addition, the shared spatial domain and nearest-neighbor pairs identified by STAligner can be further considered as corresponding pairs to guide the three-dimensional reconstruction of consecutive slices, achieving more accurate local structure-guided registration than the existing method.
Collapse
Affiliation(s)
- Xiang Zhou
- NCMIS, CEMS, RCSDS, Academy of Mathematics and Systems Science, Chinese Academy of Sciences, Beijing, China
| | - Kangning Dong
- NCMIS, CEMS, RCSDS, Academy of Mathematics and Systems Science, Chinese Academy of Sciences, Beijing, China
| | - Shihua Zhang
- NCMIS, CEMS, RCSDS, Academy of Mathematics and Systems Science, Chinese Academy of Sciences, Beijing, China.
- School of Mathematical Sciences, University of Chinese Academy of Sciences, Beijing, China.
- Key Laboratory of Systems Health Science of Zhejiang Province, School of Life Science, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, China.
| |
Collapse
|
42
|
Cuomo ASE, Nathan A, Raychaudhuri S, MacArthur DG, Powell JE. Single-cell genomics meets human genetics. Nat Rev Genet 2023; 24:535-549. [PMID: 37085594 PMCID: PMC10784789 DOI: 10.1038/s41576-023-00599-5] [Citation(s) in RCA: 24] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/29/2023] [Indexed: 04/23/2023]
Abstract
Single-cell genomic technologies are revealing the cellular composition, identities and states in tissues at unprecedented resolution. They have now scaled to the point that it is possible to query samples at the population level, across thousands of individuals. Combining single-cell information with genotype data at this scale provides opportunities to link genetic variation to the cellular processes underpinning key aspects of human biology and disease. This strategy has potential implications for disease diagnosis, risk prediction and development of therapeutic solutions. But, effectively integrating large-scale single-cell genomic data, genetic variation and additional phenotypic data will require advances in data generation and analysis methods. As single-cell genetics begins to emerge as a field in its own right, we review its current state and the challenges and opportunities ahead.
Collapse
Affiliation(s)
- Anna S E Cuomo
- Garvan Institute of Medical Research, Darlinghurst, Sydney, New South Wales, Australia.
- Centre for Population Genomics, Garvan Institute of Medical Research, Sydney, New South Wales, Australia.
- Centre for Population Genomics, Murdoch Children's Research Institute, Melbourne, Victoria, Australia.
| | - Aparna Nathan
- Center for Data Sciences, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
- Divisions of Rheumatology and Genetics, Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA, USA
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Soumya Raychaudhuri
- Center for Data Sciences, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
- Divisions of Rheumatology and Genetics, Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA, USA
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Daniel G MacArthur
- Centre for Population Genomics, Garvan Institute of Medical Research, Sydney, New South Wales, Australia
- Centre for Population Genomics, Murdoch Children's Research Institute, Melbourne, Victoria, Australia
| | - Joseph E Powell
- Garvan Institute of Medical Research, Darlinghurst, Sydney, New South Wales, Australia.
- UNSW Cellular Genomics Futures Institute, University of New South Wales, Sydney, New South Wales, Australia.
| |
Collapse
|
43
|
Chen H, Li D, Bar-Joseph Z. SCS: cell segmentation for high-resolution spatial transcriptomics. Nat Methods 2023; 20:1237-1243. [PMID: 37429992 DOI: 10.1038/s41592-023-01939-3] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Accepted: 06/02/2023] [Indexed: 07/12/2023]
Abstract
Spatial transcriptomics promises to greatly improve our understanding of tissue organization and cell-cell interactions. While most current platforms for spatial transcriptomics only offer multi-cellular resolution, with 10-15 cells per spot, recent technologies provide a much denser spot placement leading to subcellular resolution. A key challenge for these newer methods is cell segmentation and the assignment of spots to cells. Traditional image-based segmentation methods are limited and do not make full use of the information profiled by spatial transcriptomics. Here we present subcellular spatial transcriptomics cell segmentation (SCS), which combines imaging data with sequencing data to improve cell segmentation accuracy. SCS assigns spots to cells by adaptively learning the position of each spot relative to the center of its cell using a transformer neural network. SCS was tested on two new subcellular spatial transcriptomics technologies and outperformed traditional image-based segmentation methods. SCS achieved better accuracy, identified more cells and provided more realistic cell size estimation. Subcellular analysis of RNAs using SCS spot assignments provides information on RNA localization and further supports the segmentation results.
Collapse
Affiliation(s)
- Hao Chen
- Computational Biology Department, School of Computer Science, Carnegie Mellon University, Pittsburgh, PA, USA
| | - Dongshunyi Li
- Computational Biology Department, School of Computer Science, Carnegie Mellon University, Pittsburgh, PA, USA
| | - Ziv Bar-Joseph
- Computational Biology Department, School of Computer Science, Carnegie Mellon University, Pittsburgh, PA, USA.
- Machine Learning Department, School of Computer Science, Carnegie Mellon University, Pittsburgh, PA, USA.
| |
Collapse
|
44
|
Merotto L, Zopoglou M, Zackl C, Finotello F. Next-generation deconvolution of transcriptomic data to investigate the tumor microenvironment. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2023; 382:103-143. [PMID: 38225101 DOI: 10.1016/bs.ircmb.2023.05.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/17/2024]
Abstract
Methods for in silico deconvolution of bulk transcriptomics can characterize the cellular composition of the tumor microenvironment, quantifying the abundance of cell types associated with patients' prognosis and response to therapy. While first-generation deconvolution methods rely on precomputed, transcriptional signatures of a handful of cell types, second-generation methods can be trained with single-cell data to disentangle more fine-grained cell phenotypes and states. These novel approaches can also be applied to spatial transcriptomic data to reveal the spatial organization of tumors. In this review, we describe state-of-the-art deconvolution methods (first-generation, second-generation, and spatial) which can be used to investigate the tumor microenvironment, discussing their strengths and limitations. We conclude with an outlook on the challenges that need to be overcome to unlock the full potential of next-generation deconvolution for oncology and the life sciences.
Collapse
Affiliation(s)
- Lorenzo Merotto
- Universität Innsbruck, Department of Molecular Biology, Digital Science Center (DiSC), Innsbruck, Austria
| | - Maria Zopoglou
- Universität Innsbruck, Department of Molecular Biology, Digital Science Center (DiSC), Innsbruck, Austria
| | - Constantin Zackl
- Universität Innsbruck, Department of Molecular Biology, Digital Science Center (DiSC), Innsbruck, Austria
| | - Francesca Finotello
- Universität Innsbruck, Department of Molecular Biology, Digital Science Center (DiSC), Innsbruck, Austria.
| |
Collapse
|
45
|
Voigt AP, Mullin NK, Navratil EM, Flamme-Wiese MJ, Lin LC, Scheetz TE, Han IC, Stone EM, Tucker BA, Mullins RF. GENE EXPRESSION WITHIN A HUMAN CHOROIDAL NEOVASCULAR MEMBRANE USING SPATIAL TRANSCRIPTOMICS. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.06.16.544770. [PMID: 37398429 PMCID: PMC10312719 DOI: 10.1101/2023.06.16.544770] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/04/2023]
Abstract
Macular neovascularization is a relatively common and potentially visually devastating complication of age-related macular degeneration. In macular neovascularization, pathologic angiogenesis can originate from either the choroid or the retina, but we have limited understanding of how different cell types become dysregulated in this dynamic process. In this study, we performed spatial RNA sequencing on a human donor eye with macular neovascularization as well as a healthy control donor. We identified genes enriched within the area of macular neovascularization and used deconvolution algorithms to predict the originating cell type of these dysregulated genes. Within the area of neovascularization, endothelial cells were predicted to increase expression of genes related to Rho family GTPase signaling and integrin signaling. Likewise, VEGF and TGFB1 were identified as potential upstream regulators that could drive the observed gene expression changes produced by endothelial and retinal pigment epithelium cells in the macular neovascularization donor. These spatial gene expression profiles were compared to previous single-cell gene expression experiments in human age-related macular degeneration as well as a model of laser-induced neovascularization in mice. As a secondary aim, we also investigated spatial gene expression patterns within the macular neural retina and between the macular and peripheral choroid. We recapitulated previously described regional-specific gene expression patterns across both tissues. Overall, this study spatially analyzes gene expression across the retina, retinal pigment epithelium, and choroid in health and describes a set of candidate molecules that become dysregulated in macular neovascularization.
Collapse
Affiliation(s)
- Andrew P. Voigt
- Department of Ophthalmology and Visual Sciences, the University of Iowa Carver College of Medicine, Iowa City, IA, 52242
- Institute for Vision Research, the University of Iowa, Iowa City, IA, 52242
| | - Nathaniel K. Mullin
- Department of Ophthalmology and Visual Sciences, the University of Iowa Carver College of Medicine, Iowa City, IA, 52242
- Institute for Vision Research, the University of Iowa, Iowa City, IA, 52242
| | - Emma M. Navratil
- Department of Ophthalmology and Visual Sciences, the University of Iowa Carver College of Medicine, Iowa City, IA, 52242
- Institute for Vision Research, the University of Iowa, Iowa City, IA, 52242
| | - Miles J. Flamme-Wiese
- Department of Ophthalmology and Visual Sciences, the University of Iowa Carver College of Medicine, Iowa City, IA, 52242
- Institute for Vision Research, the University of Iowa, Iowa City, IA, 52242
| | - Li-Chun Lin
- University of Iowa Neuroscience Institute, the University of Iowa Carver College of Medicine, Iowa City, IA, 52242
| | - Todd E. Scheetz
- Department of Ophthalmology and Visual Sciences, the University of Iowa Carver College of Medicine, Iowa City, IA, 52242
- Institute for Vision Research, the University of Iowa, Iowa City, IA, 52242
| | - Ian C. Han
- Department of Ophthalmology and Visual Sciences, the University of Iowa Carver College of Medicine, Iowa City, IA, 52242
- Institute for Vision Research, the University of Iowa, Iowa City, IA, 52242
| | - Edwin M. Stone
- Department of Ophthalmology and Visual Sciences, the University of Iowa Carver College of Medicine, Iowa City, IA, 52242
- Institute for Vision Research, the University of Iowa, Iowa City, IA, 52242
| | - Budd A. Tucker
- Department of Ophthalmology and Visual Sciences, the University of Iowa Carver College of Medicine, Iowa City, IA, 52242
- Institute for Vision Research, the University of Iowa, Iowa City, IA, 52242
| | - Robert F. Mullins
- Department of Ophthalmology and Visual Sciences, the University of Iowa Carver College of Medicine, Iowa City, IA, 52242
- Institute for Vision Research, the University of Iowa, Iowa City, IA, 52242
| |
Collapse
|
46
|
Chen H, Li D, Bar-Joseph Z. SCS: cell segmentation for high-resolution spatial transcriptomics. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.01.11.523658. [PMID: 37398213 PMCID: PMC10312435 DOI: 10.1101/2023.01.11.523658] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/04/2023]
Abstract
Spatial transcriptomics promises to greatly improve our understanding of tissue organization and cell-cell interactions. While most current platforms for spatial transcriptomics only offer multi-cellular resolution, with 10-15 cells per spot, recent technologies provide a much denser spot placement leading to sub-cellular resolution. A key challenge for these newer methods is cell segmentation and the assignment of spots to cells. Traditional image-based segmentation methods are limited and do not make full use of the information profiled by spatial transcrip-tomics. Here we present SCS, which combines imaging data with sequencing data to improve cell segmentation accuracy. SCS assigns spots to cells by adaptively learning the position of each spot relative to the center of its cell using a transformer neural network. SCS was tested on two new sub-cellular spatial transcriptomics technologies and outperformed traditional image-based segmentation methods. SCS achieved better accuracy, identified more cells, and provided more realistic cell size estimation. Sub-cellular analysis of RNAs using SCS spots assignments provides information on RNA localization and further supports the segmentation results.
Collapse
|
47
|
Bhargava R. Digital Histopathology by Infrared Spectroscopic Imaging. ANNUAL REVIEW OF ANALYTICAL CHEMISTRY (PALO ALTO, CALIF.) 2023; 16:205-230. [PMID: 37068745 PMCID: PMC10408309 DOI: 10.1146/annurev-anchem-101422-090956] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Infrared (IR) spectroscopic imaging records spatially resolved molecular vibrational spectra, enabling a comprehensive measurement of the chemical makeup and heterogeneity of biological tissues. Combining this novel contrast mechanism in microscopy with the use of artificial intelligence can transform the practice of histopathology, which currently relies largely on human examination of morphologic patterns within stained tissue. First, this review summarizes IR imaging instrumentation especially suited to histopathology, analyses of its performance, and major trends. Second, an overview of data processing methods and application of machine learning is given, with an emphasis on the emerging use of deep learning. Third, a discussion on workflows in pathology is provided, with four categories proposed based on the complexity of methods and the analytical performance needed. Last, a set of guidelines, termed experimental and analytical specifications for spectroscopic imaging in histopathology, are proposed to help standardize the diversity of approaches in this emerging area.
Collapse
Affiliation(s)
- Rohit Bhargava
- Department of Bioengineering; Department of Electrical and Computer Engineering; Department of Mechanical Science and Engineering; Department of Chemical and Biomolecular Engineering; Department of Chemistry; Cancer Center at Illinois; and Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Urbana, Illinois, USA;
| |
Collapse
|
48
|
Lee AJ, Cahill R, Abbasi-Asl R. Machine Learning for Uncovering Biological Insights in Spatial Transcriptomics Data. ARXIV 2023:arXiv:2303.16725v1. [PMID: 37033464 PMCID: PMC10081350] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 04/11/2023]
Abstract
Development and homeostasis in multicellular systems both require exquisite control over spatial molecular pattern formation and maintenance. Advances in spatially-resolved and high-throughput molecular imaging methods such as multiplexed immunofluorescence and spatial transcriptomics (ST) provide exciting new opportunities to augment our fundamental understanding of these processes in health and disease. The large and complex datasets resulting from these techniques, particularly ST, have led to rapid development of innovative machine learning (ML) tools primarily based on deep learning techniques. These ML tools are now increasingly featured in integrated experimental and computational workflows to disentangle signals from noise in complex biological systems. However, it can be difficult to understand and balance the different implicit assumptions and methodologies of a rapidly expanding toolbox of analytical tools in ST. To address this, we summarize major ST analysis goals that ML can help address and current analysis trends. We also describe four major data science concepts and related heuristics that can help guide practitioners in their choices of the right tools for the right biological questions.
Collapse
|
49
|
Xing Y, Zan C, Liu L. Recent advances in understanding neuronal diversity and neural circuit complexity across different brain regions using single-cell sequencing. Front Neural Circuits 2023; 17:1007755. [PMID: 37063385 PMCID: PMC10097998 DOI: 10.3389/fncir.2023.1007755] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2022] [Accepted: 02/16/2023] [Indexed: 04/18/2023] Open
Abstract
Neural circuits are characterized as interconnecting neuron networks connected by synapses. Some kinds of gene expression and/or functional changes of neurons and synaptic connections may result in aberrant neural circuits, which has been recognized as one crucial pathological mechanism for the onset of many neurological diseases. Gradual advances in single-cell sequencing approaches with strong technological advantages, as exemplified by high throughput and increased resolution for live cells, have enabled it to assist us in understanding neuronal diversity across diverse brain regions and further transformed our knowledge of cellular building blocks of neural circuits through revealing numerous molecular signatures. Currently published transcriptomic studies have elucidated various neuronal subpopulations as well as their distribution across prefrontal cortex, hippocampus, hypothalamus, and dorsal root ganglion, etc. Better characterization of brain region-specific circuits may shed light on new pathological mechanisms involved and assist in selecting potential targets for the prevention and treatment of specific neurological disorders based on their established roles. Given diverse neuronal populations across different brain regions, we aim to give a brief sketch of current progress in understanding neuronal diversity and neural circuit complexity according to their locations. With the special focus on the application of single-cell sequencing, we thereby summarize relevant region-specific findings. Considering the importance of spatial context and connectivity in neural circuits, we also discuss a few published results obtained by spatial transcriptomics. Taken together, these single-cell sequencing data may lay a mechanistic basis for functional identification of brain circuit components, which links their molecular signatures to anatomical regions, connectivity, morphology, and physiology. Furthermore, the comprehensive characterization of neuron subtypes, their distributions, and connectivity patterns via single-cell sequencing is critical for understanding neural circuit properties and how they generate region-dependent interactions in different context.
Collapse
Affiliation(s)
- Yu Xing
- Department of Neurology, Beidahuang Industry Group General Hospital, Harbin, China
| | - Chunfang Zan
- Institute for Stroke and Dementia Research (ISD), LMU Klinikum, Ludwig-Maximilian-University (LMU), Munich, Germany
| | - Lu Liu
- Munich Medical Research School (MMRS), LMU Klinikum, Ludwig-Maximilian-University (LMU), Munich, Germany
- *Correspondence: Lu Liu, ,
| |
Collapse
|
50
|
Liu S, Iorgulescu JB, Li S, Borji M, Barrera-Lopez IA, Shanmugam V, Lyu H, Morriss JW, Garcia ZN, Murray E, Reardon DA, Yoon CH, Braun DA, Livak KJ, Wu CJ, Chen F. Spatial maps of T cell receptors and transcriptomes reveal distinct immune niches and interactions in the adaptive immune response. Immunity 2022; 55:1940-1952.e5. [PMID: 36223726 PMCID: PMC9745674 DOI: 10.1016/j.immuni.2022.09.002] [Citation(s) in RCA: 52] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Revised: 06/21/2022] [Accepted: 08/31/2022] [Indexed: 12/15/2022]
Abstract
T cells mediate antigen-specific immune responses to disease through the specificity and diversity of their clonotypic T cell receptors (TCRs). Determining the spatial distributions of T cell clonotypes in tissues is essential to understanding T cell behavior, but spatial sequencing methods remain unable to profile the TCR repertoire. Here, we developed Slide-TCR-seq, a 10-μm-resolution method, to sequence whole transcriptomes and TCRs within intact tissues. We confirmed the ability of Slide-TCR-seq to map the characteristic locations of T cells and their receptors in mouse spleen. In human lymphoid germinal centers, we identified spatially distinct TCR repertoires. Profiling T cells in renal cell carcinoma and melanoma specimens revealed heterogeneous immune responses: T cell states and infiltration differed intra- and inter-clonally, and adjacent tumor and immune cells exhibited distinct gene expression. Altogether, our method yields insights into the spatial relationships between clonality, neighboring cell types, and gene expression that drive T cell responses.
Collapse
Affiliation(s)
- Sophia Liu
- Biophysics Program, Harvard University, Boston, MA 02115, USA; Harvard-MIT Division of Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - J Bryan Iorgulescu
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA; Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Shuqiang Li
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA; Translational Immunogenomics Lab, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - Mehdi Borji
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | | | - Vignesh Shanmugam
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Haoxiang Lyu
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA; Translational Immunogenomics Lab, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - Julia W Morriss
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Zoe N Garcia
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Evan Murray
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - David A Reardon
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA; Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Charles H Yoon
- Department of Surgical Oncology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - David A Braun
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA; Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA; Yale Center of Cellular and Molecular Oncology, Yale School of Medicine, New Haven, CT 06511, USA
| | - Kenneth J Livak
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA; Translational Immunogenomics Lab, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - Catherine J Wu
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA; Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA; Division of Stem Cell Transplantation and Cellular Therapies, Dana-Farber Cancer Institute, Boston, MA 02215, USA.
| | - Fei Chen
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA 02138, USA.
| |
Collapse
|