51
|
Haase C, Gustafsson K, Mei S, Yeh SC, Richter D, Milosevic J, Turcotte R, Kharchenko PV, Sykes DB, Scadden DT, Lin CP. Image-seq: spatially resolved single-cell sequencing guided by in situ and in vivo imaging. Nat Methods 2022; 19:1622-1633. [PMID: 36424441 PMCID: PMC9718684 DOI: 10.1038/s41592-022-01673-2] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Accepted: 10/03/2022] [Indexed: 11/26/2022]
Abstract
Tissue function depends on cellular organization. While the properties of individual cells are increasingly being deciphered using powerful single-cell sequencing technologies, understanding their spatial organization and temporal evolution remains a major challenge. Here, we present Image-seq, a technology that provides single-cell transcriptional data on cells that are isolated from specific spatial locations under image guidance, thus preserving the spatial information of the target cells. It is compatible with in situ and in vivo imaging and can document the temporal and dynamic history of the cells being analyzed. Cell samples are isolated from intact tissue and processed with state-of-the-art library preparation protocols. The technique therefore combines spatial information with highly sensitive RNA sequencing readouts from individual, intact cells. We have used both high-throughput, droplet-based sequencing as well as SMARTseq-v4 library preparation to demonstrate its application to bone marrow and leukemia biology. We discovered that DPP4 is a highly upregulated gene during early progression of acute myeloid leukemia and that it marks a more proliferative subpopulation that is confined to specific bone marrow microenvironments. Furthermore, the ability of Image-seq to isolate viable, intact cells should make it compatible with a range of downstream single-cell analysis tools including multi-omics protocols.
Collapse
Affiliation(s)
- Christa Haase
- Wellman Center for Photomedicine, Massachusetts General Hospital, Boston, MA, USA.
- Center for Systems Biology, Massachusetts General Hospital, Boston, MA, USA.
| | - Karin Gustafsson
- Center for Regenerative Medicine, Massachusetts General Hospital, Boston, MA, USA
- Harvard Stem Cell Institute, Cambridge, MA, USA
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, USA
| | - Shenglin Mei
- Center for Regenerative Medicine, Massachusetts General Hospital, Boston, MA, USA
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA, USA
| | - Shu-Chi Yeh
- Wellman Center for Photomedicine, Massachusetts General Hospital, Boston, MA, USA
- Center for Systems Biology, Massachusetts General Hospital, Boston, MA, USA
- Department of Orthopaedics, Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, USA
| | - Dmitry Richter
- Wellman Center for Photomedicine, Massachusetts General Hospital, Boston, MA, USA
- Center for Systems Biology, Massachusetts General Hospital, Boston, MA, USA
| | - Jelena Milosevic
- Center for Regenerative Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Raphaël Turcotte
- Wellman Center for Photomedicine, Massachusetts General Hospital, Boston, MA, USA
- Center for Systems Biology, Massachusetts General Hospital, Boston, MA, USA
| | - Peter V Kharchenko
- Harvard Stem Cell Institute, Cambridge, MA, USA
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA, USA
- Altos Labs, San Diego, CA, USA
| | - David B Sykes
- Center for Regenerative Medicine, Massachusetts General Hospital, Boston, MA, USA
- Harvard Stem Cell Institute, Cambridge, MA, USA
| | - David T Scadden
- Center for Regenerative Medicine, Massachusetts General Hospital, Boston, MA, USA
- Harvard Stem Cell Institute, Cambridge, MA, USA
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, USA
| | - Charles P Lin
- Wellman Center for Photomedicine, Massachusetts General Hospital, Boston, MA, USA.
- Center for Systems Biology, Massachusetts General Hospital, Boston, MA, USA.
- Harvard Stem Cell Institute, Cambridge, MA, USA.
| |
Collapse
|
52
|
Hsieh WC, Budiarto BR, Wang YF, Lin CY, Gwo MC, So DK, Tzeng YS, Chen SY. Spatial multi-omics analyses of the tumor immune microenvironment. J Biomed Sci 2022; 29:96. [PMID: 36376874 PMCID: PMC9661775 DOI: 10.1186/s12929-022-00879-y] [Citation(s) in RCA: 51] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2022] [Accepted: 11/02/2022] [Indexed: 11/16/2022] Open
Abstract
In the past decade, single-cell technologies have revealed the heterogeneity of the tumor-immune microenvironment at the genomic, transcriptomic, and proteomic levels and have furthered our understanding of the mechanisms of tumor development. Single-cell technologies have also been used to identify potential biomarkers. However, spatial information about the tumor-immune microenvironment such as cell locations and cell-cell interactomes is lost in these approaches. Recently, spatial multi-omics technologies have been used to study transcriptomes, proteomes, and metabolomes of tumor-immune microenvironments in several types of cancer, and the data obtained from these methods has been combined with immunohistochemistry and multiparameter analysis to yield markers of cancer progression. Here, we review numerous cutting-edge spatial 'omics techniques, their application to study of the tumor-immune microenvironment, and remaining technical challenges.
Collapse
Affiliation(s)
- Wan-Chen Hsieh
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
- Genome and Systems Biology Degree Program, Academia Sinica and National Taiwan University, Taipei, Taiwan
| | - Bugi Ratno Budiarto
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
- Taiwan International Graduate Program in Molecular Medicine, National Yang Ming Chiao Tung University and Academia Sinica, Taipei, Taiwan
| | - Yi-Fu Wang
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Chih-Yu Lin
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Mao-Chun Gwo
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Dorothy Kazuno So
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
- Institute of Biotechnology, College of Bio-Resources and Agriculture, National Taiwan University, Taipei, Taiwan
| | - Yi-Shiuan Tzeng
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan.
| | - Shih-Yu Chen
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan.
- Genome and Systems Biology Degree Program, Academia Sinica and National Taiwan University, Taipei, Taiwan.
| |
Collapse
|
53
|
Kishi JY, Liu N, West ER, Sheng K, Jordanides JJ, Serrata M, Cepko CL, Saka SK, Yin P. Light-Seq: light-directed in situ barcoding of biomolecules in fixed cells and tissues for spatially indexed sequencing. Nat Methods 2022; 19:1393-1402. [PMID: 36216958 PMCID: PMC9636025 DOI: 10.1038/s41592-022-01604-1] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Accepted: 08/10/2022] [Indexed: 11/21/2022]
Abstract
We present Light-Seq, an approach for multiplexed spatial indexing of intact biological samples using light-directed DNA barcoding in fixed cells and tissues followed by ex situ sequencing. Light-Seq combines spatially targeted, rapid photocrosslinking of DNA barcodes onto complementary DNAs in situ with a one-step DNA stitching reaction to create pooled, spatially indexed sequencing libraries. This light-directed barcoding enables in situ selection of multiple cell populations in intact fixed tissue samples for full-transcriptome sequencing based on location, morphology or protein stains, without cellular dissociation. Applying Light-Seq to mouse retinal sections, we recovered thousands of differentially enriched transcripts from three cellular layers and discovered biomarkers for a very rare neuronal subtype, dopaminergic amacrine cells, from only four to eight individual cells per section. Light-Seq provides an accessible workflow to combine in situ imaging and protein staining with next generation sequencing of the same cells, leaving the sample intact for further analysis post-sequencing.
Collapse
Affiliation(s)
- Jocelyn Y Kishi
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, USA.
- Department of Systems Biology, Harvard Medical School, Boston, MA, USA.
| | - Ninning Liu
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, USA
| | - Emma R West
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA, USA
- Howard Hughes Medical Institute, Chevy Chase, MD, USA
| | - Kuanwei Sheng
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, USA
| | - Jack J Jordanides
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, USA
| | - Matthew Serrata
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, USA
| | - Constance L Cepko
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA, USA.
- Howard Hughes Medical Institute, Chevy Chase, MD, USA.
- Department of Ophthalmology, Harvard Medical School, Boston, MA, USA.
| | - Sinem K Saka
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, USA.
- Department of Systems Biology, Harvard Medical School, Boston, MA, USA.
- Genome Biology Unit, European Molecular Biology Laboratory (EMBL), Heidelberg, Germany.
| | - Peng Yin
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, USA.
- Department of Systems Biology, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
54
|
Yu Q, Jiang M, Wu L. Spatial transcriptomics technology in cancer research. Front Oncol 2022; 12:1019111. [PMID: 36313703 PMCID: PMC9606570 DOI: 10.3389/fonc.2022.1019111] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2022] [Accepted: 09/21/2022] [Indexed: 08/25/2023] Open
Abstract
In recent years, spatial transcriptomics (ST) technologies have developed rapidly and have been widely used in constructing spatial tissue atlases and characterizing spatiotemporal heterogeneity of cancers. Currently, ST has been used to profile spatial heterogeneity in multiple cancer types. Besides, ST is a benefit for identifying and comprehensively understanding special spatial areas such as tumor interface and tertiary lymphoid structures (TLSs), which exhibit unique tumor microenvironments (TMEs). Therefore, ST has also shown great potential to improve pathological diagnosis and identify novel prognostic factors in cancer. This review presents recent advances and prospects of applications on cancer research based on ST technologies as well as the challenges.
Collapse
Affiliation(s)
- Qichao Yu
- Beijing Genomics Institute (BGI)-Shenzhen, Shenzhen, China
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China
| | - Miaomiao Jiang
- Beijing Genomics Institute (BGI)-Shenzhen, Shenzhen, China
| | - Liang Wu
- Beijing Genomics Institute (BGI)-Shenzhen, Shenzhen, China
| |
Collapse
|
55
|
Kleino I, Frolovaitė P, Suomi T, Elo LL. Computational solutions for spatial transcriptomics. Comput Struct Biotechnol J 2022; 20:4870-4884. [PMID: 36147664 PMCID: PMC9464853 DOI: 10.1016/j.csbj.2022.08.043] [Citation(s) in RCA: 48] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Revised: 08/18/2022] [Accepted: 08/18/2022] [Indexed: 11/18/2022] Open
Abstract
Transcriptome level expression data connected to the spatial organization of the cells and molecules would allow a comprehensive understanding of how gene expression is connected to the structure and function in the biological systems. The spatial transcriptomics platforms may soon provide such information. However, the current platforms still lack spatial resolution, capture only a fraction of the transcriptome heterogeneity, or lack the throughput for large scale studies. The strengths and weaknesses in current ST platforms and computational solutions need to be taken into account when planning spatial transcriptomics studies. The basis of the computational ST analysis is the solutions developed for single-cell RNA-sequencing data, with advancements taking into account the spatial connectedness of the transcriptomes. The scRNA-seq tools are modified for spatial transcriptomics or new solutions like deep learning-based joint analysis of expression, spatial, and image data are developed to extract biological information in the spatially resolved transcriptomes. The computational ST analysis can reveal remarkable biological insights into spatial patterns of gene expression, cell signaling, and cell type variations in connection with cell type-specific signaling and organization in complex tissues. This review covers the topics that help choosing the platform and computational solutions for spatial transcriptomics research. We focus on the currently available ST methods and platforms and their strengths and limitations. Of the computational solutions, we provide an overview of the analysis steps and tools used in the ST data analysis. The compatibility with the data types and the tools provided by the current ST analysis frameworks are summarized.
Collapse
Key Words
- AOI, area of illumination
- BICCN, Brain Initiative Cell Census Network
- BOLORAMIS, barcoded oligonucleotides ligated on RNA amplified for multiplexed and parallel in situ analyses
- Baysor, Bayesian Segmentation of Spatial Transcriptomics Data
- BinSpect, Binary Spatial Extraction
- CCC, cell–cell communication
- CCI, cell–cell interactions
- CNV, copy-number variation
- Computational biology
- DSP, digital spatial profiling
- DbiT-Seq, Deterministic Barcoding in Tissue for spatial omics sequencing
- FA, factor analysis
- FFPE, formalin-fixed, paraffin-embedded
- FISH, fluorescence in situ hybridization
- FISSEQ, fluorescence in situ sequencing of RNA
- FOV, Field of view
- GRNs, gene regulation networks
- GSEA, gene set enrichment analysis
- GSVA, gene set variation analysis
- HDST, high definition spatial transcriptomics
- HMRF, hidden Markov random field
- ICG, interaction changed genes
- ISH, in situ hybridization
- ISS, in situ sequencing
- JSTA, Joint cell segmentation and cell type annotation
- KNN, k-nearest neighbor
- LCM, Laser Capture Microdissection
- LCM-seq, laser capture microdissection coupled with RNA sequencing
- LOH, loss of heterozygosity analysis
- MC, Molecular Cartography
- MERFISH, multiplexed error-robust FISH
- NMF (NNMF), Non-negative matrix factorization
- PCA, Principal Component Analysis
- PIXEL-seq, Polony (or DNA cluster)-indexed library-sequencing
- PL-lig, padlock ligation
- QC, quality control
- RNAseq, RNA sequencing
- ROI, region of interest
- SCENIC, Single-Cell rEgulatory Network Inference and Clustering
- SME, Spatial Morphological gene Expression normalization
- SPATA, SPAtial Transcriptomic Analysis
- ST Pipeline, Spatial Transcriptomics Pipeline
- ST, Spatial transcriptomics
- STARmap, spatially-resolved transcript amplicon readout mapping
- Single-cell analysis
- Spatial data analysis frameworks
- Spatial deconvolution
- Spatial transcriptomics
- TIVA, Transcriptome in Vivo Analysis
- TMA, tissue microarray
- TME, tumor micro environment
- UMAP, Uniform Manifold Approximation and Projection for Dimension Reduction
- UMI, unique molecular identifier
- ZipSeq, zipcoded sequencing.
- scRNA-seq, single-cell RNA sequencing
- scvi-tools, single-cell variational inference tools
- seqFISH, sequential fluorescence in situ hybridization
- sequ-smFISH, sequential single-molecule fluorescent in situ hybridization
- smFISH, single molecule FISH
- t-SNE, t-distributed stochastic neighbor embedding
Collapse
Affiliation(s)
- Iivari Kleino
- Turku Bioscience Centre, University of Turku and Åbo Akademi University Turku, Turku, Finland
| | - Paulina Frolovaitė
- Turku Bioscience Centre, University of Turku and Åbo Akademi University Turku, Turku, Finland
| | - Tomi Suomi
- Turku Bioscience Centre, University of Turku and Åbo Akademi University Turku, Turku, Finland
| | - Laura L. Elo
- Turku Bioscience Centre, University of Turku and Åbo Akademi University Turku, Turku, Finland
- Institute of Biomedicine, University of Turku, Turku, Finland
| |
Collapse
|
56
|
Hu Y, Zhang Y, Liu Y, Gao Y, San T, Li X, Song S, Yan B, Zhao Z. Advances in application of single-cell RNA sequencing in cardiovascular research. Front Cardiovasc Med 2022; 9:905151. [PMID: 35958408 PMCID: PMC9360414 DOI: 10.3389/fcvm.2022.905151] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Accepted: 07/05/2022] [Indexed: 11/13/2022] Open
Abstract
Single-cell RNA sequencing (scRNA-seq) provides high-resolution information on transcriptomic changes at the single-cell level, which is of great significance for distinguishing cell subtypes, identifying stem cell differentiation processes, and identifying targets for disease treatment. In recent years, emerging single-cell RNA sequencing technologies have been used to make breakthroughs regarding decoding developmental trajectories, phenotypic transitions, and cellular interactions in the cardiovascular system, providing new insights into cardiovascular disease. This paper reviews the technical processes of single-cell RNA sequencing and the latest progress based on single-cell RNA sequencing in the field of cardiovascular system research, compares single-cell RNA sequencing with other single-cell technologies, and summarizes the extended applications and advantages and disadvantages of single-cell RNA sequencing. Finally, the prospects for applying single-cell RNA sequencing in the field of cardiovascular research are discussed.
Collapse
Affiliation(s)
- Yue Hu
- Department of Cardiology, Jinan Central Hospital, Shandong University, Jinan, China
| | - Ying Zhang
- Department of Cardiology, Central Hospital Affiliated Shandong First Medical University, Jinan, China
| | - Yutong Liu
- Department of Cardiology, Jinan Central Hospital, Shandong University, Jinan, China
| | - Yan Gao
- Department of Research Center of Translational Medicine, Central Hospital Affiliated Shandong First Medical University, Jinan, China
| | - Tiantian San
- Department of Cardiology, Jinan Central Hospital, Shandong University, Jinan, China
| | - Xiaoying Li
- Department of Research Center of Translational Medicine, Central Hospital Affiliated Shandong First Medical University, Jinan, China
- Department of Emergency, Central Hospital Affiliated Shandong First Medical University, Jinan, China
| | - Sensen Song
- Department of Cardiology, Central Hospital Affiliated Shandong First Medical University, Jinan, China
| | - Binglong Yan
- Department of Cardiology, Central Hospital Affiliated Shandong First Medical University, Jinan, China
| | - Zhuo Zhao
- Department of Cardiology, Jinan Central Hospital, Shandong University, Jinan, China
- Department of Cardiology, Central Hospital Affiliated Shandong First Medical University, Jinan, China
- *Correspondence: Zhuo Zhao
| |
Collapse
|
57
|
Moffitt JR, Lundberg E, Heyn H. The emerging landscape of spatial profiling technologies. Nat Rev Genet 2022; 23:741-759. [PMID: 35859028 DOI: 10.1038/s41576-022-00515-3] [Citation(s) in RCA: 174] [Impact Index Per Article: 58.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/16/2022] [Indexed: 01/04/2023]
Abstract
Improved scale, multiplexing and resolution are establishing spatial nucleic acid and protein profiling methods as a major pillar for cellular atlas building of complex samples, from tissues to full organisms. Emerging methods yield omics measurements at resolutions covering the nano- to microscale, enabling the charting of cellular heterogeneity, complex tissue architectures and dynamic changes during development and disease. We present an overview of the developing landscape of in situ spatial genome, transcriptome and proteome technologies, exemplify their impact on cell biology and translational research, and discuss current challenges for their community-wide adoption. Among many transformative applications, we envision that spatial methods will map entire organs and enable next-generation pathology.
Collapse
Affiliation(s)
- Jeffrey R Moffitt
- Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, MA, USA.,Department of Microbiology, Harvard Medical School, Boston, MA, USA
| | - Emma Lundberg
- Science for Life Laboratory, School of Engineering Sciences in Chemistry, Biotechnology and Health, KTH - Royal Institute of Technology, Stockholm, Sweden.,Department of Bioengineering, Stanford University, Stanford, CA, USA.,Department of Pathology, Stanford University, Stanford, CA, USA.,Chan Zuckerberg Biohub, San Francisco, San Francisco, CA, USA
| | - Holger Heyn
- CNAG-CRG, Centre for Genomic Regulation (CRG), Barcelona Institute of Science and Technology (BIST), Barcelona, Spain. .,Universitat Pompeu Fabra (UPF), Barcelona, Spain.
| |
Collapse
|
58
|
Kersten K, Hu KH, Combes AJ, Samad B, Harwin T, Ray A, Rao AA, Cai E, Marchuk K, Artichoker J, Courau T, Shi Q, Belk J, Satpathy AT, Krummel MF. Spatiotemporal co-dependency between macrophages and exhausted CD8 + T cells in cancer. Cancer Cell 2022; 40:624-638.e9. [PMID: 35623342 PMCID: PMC9197962 DOI: 10.1016/j.ccell.2022.05.004] [Citation(s) in RCA: 154] [Impact Index Per Article: 51.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Revised: 02/08/2022] [Accepted: 05/04/2022] [Indexed: 12/11/2022]
Abstract
T cell exhaustion is a major impediment to antitumor immunity. However, it remains elusive how other immune cells in the tumor microenvironment (TME) contribute to this dysfunctional state. Here, we show that the biology of tumor-associated macrophages (TAMs) and exhausted T cells (Tex) in the TME is extensively linked. We demonstrate that in vivo depletion of TAMs reduces exhaustion programs in tumor-infiltrating CD8+ T cells and reinvigorates their effector potential. Reciprocally, transcriptional and epigenetic profiling reveals that Tex express factors that actively recruit monocytes to the TME and shape their differentiation. Using lattice light sheet microscopy, we show that TAM and CD8+ T cells engage in unique, long-lasting, antigen-specific synaptic interactions that fail to activate T cells but prime them for exhaustion, which is then accelerated in hypoxic conditions. Spatially resolved sequencing supports a spatiotemporal self-enforcing positive feedback circuit that is aligned to protect rather than destroy a tumor.
Collapse
Affiliation(s)
- Kelly Kersten
- Department of Pathology, University of California, San Francisco, San Francisco, CA 94143, USA; ImmunoX Initiative, University of California, San Francisco, San Francisco, CA 94143, USA; Parker Institute for Cancer Immunotherapy, San Francisco, CA 94129, USA
| | - Kenneth H Hu
- Department of Pathology, University of California, San Francisco, San Francisco, CA 94143, USA; ImmunoX Initiative, University of California, San Francisco, San Francisco, CA 94143, USA; Parker Institute for Cancer Immunotherapy, San Francisco, CA 94129, USA
| | - Alexis J Combes
- Department of Pathology, University of California, San Francisco, San Francisco, CA 94143, USA; ImmunoX Initiative, University of California, San Francisco, San Francisco, CA 94143, USA; UCSF CoLabs, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Bushra Samad
- Department of Pathology, University of California, San Francisco, San Francisco, CA 94143, USA; ImmunoX Initiative, University of California, San Francisco, San Francisco, CA 94143, USA; UCSF CoLabs, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Tory Harwin
- Department of Pathology, University of California, San Francisco, San Francisco, CA 94143, USA; ImmunoX Initiative, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Arja Ray
- Department of Pathology, University of California, San Francisco, San Francisco, CA 94143, USA; ImmunoX Initiative, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Arjun Arkal Rao
- Department of Pathology, University of California, San Francisco, San Francisco, CA 94143, USA; ImmunoX Initiative, University of California, San Francisco, San Francisco, CA 94143, USA; UCSF CoLabs, University of California, San Francisco, San Francisco, CA 94143, USA
| | - En Cai
- Department of Pathology, University of California, San Francisco, San Francisco, CA 94143, USA; ImmunoX Initiative, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Kyle Marchuk
- UCSF CoLabs, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Jordan Artichoker
- UCSF CoLabs, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Tristan Courau
- Department of Pathology, University of California, San Francisco, San Francisco, CA 94143, USA; ImmunoX Initiative, University of California, San Francisco, San Francisco, CA 94143, USA; UCSF CoLabs, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Quanming Shi
- Department of Personal Dynamic Regulomes, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Julia Belk
- Department of Computer Science, Stanford University, Stanford, CA 94305, USA
| | - Ansuman T Satpathy
- Department of Pathology, Stanford University, Stanford, CA 94305, USA; Gladstone-UCSF Institute of Genomic Immunology, San Francisco, CA 94158, USA; Parker Institute for Cancer Immunotherapy, San Francisco, CA 94129, USA
| | - Matthew F Krummel
- Department of Pathology, University of California, San Francisco, San Francisco, CA 94143, USA; ImmunoX Initiative, University of California, San Francisco, San Francisco, CA 94143, USA; UCSF CoLabs, University of California, San Francisco, San Francisco, CA 94143, USA; Parker Institute for Cancer Immunotherapy, San Francisco, CA 94129, USA.
| |
Collapse
|
59
|
Zheng B, Fang L. Spatially resolved transcriptomics provide a new method for cancer research. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2022; 41:179. [PMID: 35590346 PMCID: PMC9118771 DOI: 10.1186/s13046-022-02385-3] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/26/2022] [Accepted: 05/06/2022] [Indexed: 12/22/2022]
Abstract
A major feature of cancer is the heterogeneity, both intratumoral and intertumoral. Traditional single-cell techniques have given us a comprehensive understanding of the biological characteristics of individual tumor cells, but the lack of spatial context of the transcriptome has limited the study of cell-to-cell interaction patterns and hindered further exploration of tumor heterogeneity. In recent years, the advent of spatially resolved transcriptomics (SRT) technology has made possible the multidimensional analysis of the tumor microenvironment in the context of intact tissues. Different SRT methods are applicable to different working ranges due to different working principles. In this paper, we review the advantages and disadvantages of various current SRT methods and the overall idea of applying these techniques to oncology studies, hoping to help researchers find breakthroughs. Finally, we discussed the future direction of SRT technology, and deeper investigation into the complex mechanisms of tumor development from different perspectives through multi-omics fusion, paving the way for precisely targeted tumor therapy.
Collapse
Affiliation(s)
- Bowen Zheng
- Department of Breast and Thyroid Surgery, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, 200072, People's Republic of China
| | - Lin Fang
- Department of Breast and Thyroid Surgery, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, 200072, People's Republic of China.
| |
Collapse
|
60
|
Liu Z, Xie X, Huang Z, Lin F, Liu S, Chen Z, Qin S, Fan X, Chen PR. Spatially resolved cell tagging and surfaceome labeling via targeted photocatalytic decaging. Chem 2022. [DOI: 10.1016/j.chempr.2022.04.016] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
|
61
|
You R, Artichoker J, Ray A, Gonzalez Velozo H, Rock DA, Conner KP, Krummel MF. Visualizing Spatial and Stoichiometric Barriers to Bispecific T-cell Engager Efficacy. Cancer Immunol Res 2022; 10:698-712. [PMID: 35413104 DOI: 10.1158/2326-6066.cir-21-0594] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Revised: 01/09/2022] [Accepted: 04/04/2022] [Indexed: 11/16/2022]
Abstract
Bispecific T-cell engager (BiTE) molecules are biologic T cell-directing immunotherapies. Blinatumomab is approved for treatment of B-cell malignancies, but BiTE molecule development in solid tumors has been more challenging. Here, we employed intravital imaging to characterize exposure and pharmacodynamic response of an anti-muCD3/anti-huEGFRvIII mouse surrogate BiTE molecule in epidermal growth factor receptor variant III (EGFRvIII)-positive breast tumors implanted within immunocompetent mice. Our study revealed heterogeneous temporal and spatial dynamics of BiTE molecule extravasation into solid tumors, highlighting physical barriers to BiTE molecule function. We also discovered that high, homogeneous EGFRvIII expression on cancer cells was necessary for a BiTE molecule to efficiently clear tumors. Additionally, we found that resident tumor-infiltrating lymphocytes (TILs) were sufficient for optimal tumor killing only at high BiTE molecule dosage, whereas inclusion of peripheral T-cell recruitment was synergistic at moderate to low dosages. We report that deletion of stimulatory conventional type I DCs (cDC1) diminished BiTE molecule-induced T-cell activation and tumor clearance, suggesting that in situ antigen-presenting cell (APC) engagements modulate the extent of BiTE molecule efficacy. In summary, our work identified multiple requirements for optimal BiTE molecule efficacy in solid tumors, providing insights that could be harnessed for solid cancer immunotherapy development.
Collapse
Affiliation(s)
- Ran You
- Department of Pathology, University of California San Francisco, San Francisco, California
- ImmunoX Initiative, University of California San Francisco, San Francisco, California
| | - Jordan Artichoker
- ImmunoX Initiative, University of California San Francisco, San Francisco, California
- Biological Imaging Development CoLab, University of California San Francisco, San Francisco, California
| | - Arja Ray
- Department of Pathology, University of California San Francisco, San Francisco, California
- ImmunoX Initiative, University of California San Francisco, San Francisco, California
| | - Hugo Gonzalez Velozo
- Department of Anatomy, University of California San Francisco, San Francisco, California
| | - Dan A Rock
- Department of Pharmacokinetics and Drug Metabolism, Amgen, South San Francisco, California
| | - Kip P Conner
- Department of Pharmacokinetics and Drug Metabolism, Amgen, South San Francisco, California
| | - Matthew F Krummel
- Department of Pathology, University of California San Francisco, San Francisco, California
- ImmunoX Initiative, University of California San Francisco, San Francisco, California
- Biological Imaging Development CoLab, University of California San Francisco, San Francisco, California
| |
Collapse
|
62
|
Byrnes JR, Weeks AM, Shifrut E, Carnevale J, Kirkemo L, Ashworth A, Marson A, Wells JA. Hypoxia Is a Dominant Remodeler of the Effector T Cell Surface Proteome Relative to Activation and Regulatory T Cell Suppression. Mol Cell Proteomics 2022; 21:100217. [PMID: 35217172 PMCID: PMC9006863 DOI: 10.1016/j.mcpro.2022.100217] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Revised: 02/14/2022] [Accepted: 02/20/2022] [Indexed: 01/02/2023] Open
Abstract
Immunosuppressive factors in the tumor microenvironment (TME) impair T cell function and limit the antitumor immune response. T cell surface receptors and surface proteins that influence interactions and function in the TME are proven targets for cancer immunotherapy. However, how the entire surface proteome remodels in primary human T cells in response to specific suppressive factors in the TME remains to be broadly and systematically characterized. Here, using a reductionist cell culture approach with primary human T cells and stable isotopic labeling with amino acids in cell culture-based quantitative cell surface capture glycoproteomics, we examined how two immunosuppressive TME factors, regulatory T cells (Tregs) and hypoxia, globally affect the activated CD8+ surface proteome (surfaceome). Surprisingly, coculturing primary CD8+ T cells with Tregs only modestly affected the CD8+ surfaceome but did partially reverse activation-induced surfaceomic changes. In contrast, hypoxia drastically altered the CD8+ surfaceome in a manner consistent with both metabolic reprogramming and induction of an immunosuppressed state. The CD4+ T cell surfaceome similarly responded to hypoxia, revealing a common hypoxia-induced surface receptor program. Our surfaceomics findings suggest that hypoxic environments create a challenge for T cell activation. These studies provide global insight into how Tregs and hypoxia remodel the T cell surfaceome and we believe represent a valuable resource to inform future therapeutic efforts to enhance T cell function.
Collapse
Affiliation(s)
- James R Byrnes
- Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, California, USA
| | - Amy M Weeks
- Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, California, USA
| | - Eric Shifrut
- Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, California, USA; Gladstone Institutes, San Francisco, California, USA
| | - Julia Carnevale
- Department of Medicine, University of California, San Francisco, San Francisco, California, USA
| | - Lisa Kirkemo
- Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, California, USA
| | - Alan Ashworth
- Department of Medicine, University of California, San Francisco, San Francisco, California, USA; The Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, California, USA
| | - Alexander Marson
- Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, California, USA; Gladstone Institutes, San Francisco, California, USA; Department of Medicine, University of California, San Francisco, San Francisco, California, USA; The Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, California, USA; Innovative Genomics Institute, University of California, Berkeley, Berkeley, California, USA; Parker Institute for Cancer Immunotherapy, San Francisco, California, USA; Chan Zuckerberg Biohub, San Francisco, California, USA
| | - James A Wells
- Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, California, USA; Chan Zuckerberg Biohub, San Francisco, California, USA; Department of Cellular and Molecular Pharmacology, University of California, San Francisco, San Francisco, California, USA.
| |
Collapse
|
63
|
Liu H, Luo H, Xue Q, Qin S, Qiu S, Liu S, Lin J, Li JP, Chen PR. Antigen-Specific T Cell Detection via Photocatalytic Proximity Cell Labeling (PhoXCELL). J Am Chem Soc 2022; 144:5517-5526. [PMID: 35312320 DOI: 10.1021/jacs.2c00159] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Quantitative detection and characterization of antigen-specific T cells are crucial to our understanding of immune responses as well as the development of new immunotherapies. Herein, we report a spatiotemporally resolved method for the detection and quantification of cell-cell interactions via Photocatalytic proXimity CELl Labeling (PhoXCELL). The biocompatible photosensitizer dibromofluorescein (DBF) was leveraged and optimized as a nongenetic alternative of enzymatic approaches for efficient generation of singlet oxygen upon photoirradiation (520 nm) on the cell surface, which allowed the subsequent labeling of nearby oxidized proteins with primary aliphatic amine-based probes. We demonstrated that DBF-functionalized dendritic cells (DCs) could spatiotemporally label interacting T cells in immune synapses via rapid photoirradiation with quantitatively discriminated interaction strength, which revealed distinct gene signatures for T cells that strongly interact with antigen-pulsed DCs. Furthermore, we employed PhoXCELL to simultaneously detect tumor antigen-specific CD8+ as well as CD4+ T cells from tumor-infiltrating lymphocytes and draining lymph nodes in murine tumor models, enabling PhoXCELL as a powerful platform to identify antigen-specific T cells in T cell receptor (TCR)-relevant personal immunotherapy.
Collapse
Affiliation(s)
- Hongyu Liu
- Synthetic and Functional Biomolecules Center, Key Laboratory of Bioorganic Chemistry and Molecular Engineering of Ministry of Education, Beijing National Laboratory for Molecular Sciences, College of Chemistry and Molecular Engineering, Peking University, Beijing 100871, China.,State Key Laboratory of Coordination Chemistry, Chemistry and Biomedicine Innovation Center (ChemBIC), School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210023, China
| | - Huixin Luo
- Synthetic and Functional Biomolecules Center, Key Laboratory of Bioorganic Chemistry and Molecular Engineering of Ministry of Education, Beijing National Laboratory for Molecular Sciences, College of Chemistry and Molecular Engineering, Peking University, Beijing 100871, China.,State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Qi Xue
- State Key Laboratory of Coordination Chemistry, Chemistry and Biomedicine Innovation Center (ChemBIC), School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210023, China
| | - Shan Qin
- State Key Laboratory of Coordination Chemistry, Chemistry and Biomedicine Innovation Center (ChemBIC), School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210023, China
| | - Shuang Qiu
- State Key Laboratory of Coordination Chemistry, Chemistry and Biomedicine Innovation Center (ChemBIC), School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210023, China
| | - Shibo Liu
- Synthetic and Functional Biomolecules Center, Key Laboratory of Bioorganic Chemistry and Molecular Engineering of Ministry of Education, Beijing National Laboratory for Molecular Sciences, College of Chemistry and Molecular Engineering, Peking University, Beijing 100871, China
| | - Jian Lin
- Synthetic and Functional Biomolecules Center, Key Laboratory of Bioorganic Chemistry and Molecular Engineering of Ministry of Education, Beijing National Laboratory for Molecular Sciences, College of Chemistry and Molecular Engineering, Peking University, Beijing 100871, China
| | - Jie P Li
- State Key Laboratory of Coordination Chemistry, Chemistry and Biomedicine Innovation Center (ChemBIC), School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210023, China
| | - Peng R Chen
- Synthetic and Functional Biomolecules Center, Key Laboratory of Bioorganic Chemistry and Molecular Engineering of Ministry of Education, Beijing National Laboratory for Molecular Sciences, College of Chemistry and Molecular Engineering, Peking University, Beijing 100871, China.,Peking-Tsinghua Center for Life Sciences, Peking University, Beijing 100871, China
| |
Collapse
|
64
|
Abstract
The function of many biological systems, such as embryos, liver lobules, intestinal villi, and tumors, depends on the spatial organization of their cells. In the past decade, high-throughput technologies have been developed to quantify gene expression in space, and computational methods have been developed that leverage spatial gene expression data to identify genes with spatial patterns and to delineate neighborhoods within tissues. To comprehensively document spatial gene expression technologies and data-analysis methods, we present a curated review of literature on spatial transcriptomics dating back to 1987, along with a thorough analysis of trends in the field, such as usage of experimental techniques, species, tissues studied, and computational approaches used. Our Review places current methods in a historical context, and we derive insights about the field that can guide current research strategies. A companion supplement offers a more detailed look at the technologies and methods analyzed: https://pachterlab.github.io/LP_2021/ .
Collapse
|
65
|
Purohit V, Wagner A, Yosef N, Kuchroo VK. Systems-based approaches to study immunometabolism. Cell Mol Immunol 2022; 19:409-420. [PMID: 35121805 PMCID: PMC8891302 DOI: 10.1038/s41423-021-00783-9] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Accepted: 09/17/2021] [Indexed: 02/06/2023] Open
Abstract
Technical advances at the interface of biology and computation, such as single-cell RNA-sequencing (scRNA-seq), reveal new layers of complexity in cellular systems. An emerging area of investigation using the systems biology approach is the study of the metabolism of immune cells. The diverse spectra of immune cell phenotypes, sparsity of immune cell numbers in vivo, limitations in the number of metabolites identified, dynamic nature of cellular metabolism and metabolic fluxes, tissue specificity, and high dependence on the local milieu make investigations in immunometabolism challenging, especially at the single-cell level. In this review, we define the systemic nature of immunometabolism, summarize cell- and system-based approaches, and introduce mathematical modeling approaches for systems interrogation of metabolic changes in immune cells. We close the review by discussing the applications and shortcomings of metabolic modeling techniques. With systems-oriented studies of metabolism expected to become a mainstay of immunological research, an understanding of current approaches toward systems immunometabolism will help investigators make the best use of current resources and push the boundaries of the discipline.
Collapse
Affiliation(s)
- Vinee Purohit
- Evergrande Center for Immunologic Diseases and Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, 02141, USA
| | - Allon Wagner
- Department of Electrical Engineering and Computer Science, University of California, Berkeley, CA, 94720, USA
- Center for Computational Biology, University of California, Berkeley, CA, 94720, USA
| | - Nir Yosef
- Department of Electrical Engineering and Computer Science, University of California, Berkeley, CA, 94720, USA
- Center for Computational Biology, University of California, Berkeley, CA, 94720, USA
| | - Vijay K Kuchroo
- Evergrande Center for Immunologic Diseases and Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA.
- Broad Institute of MIT and Harvard, Cambridge, MA, 02141, USA.
| |
Collapse
|
66
|
Miao YR, Xia M, Luo M, Luo T, Yang M, Guo AY. ImmuCellAI-mouse: a tool for comprehensive prediction of mouse immune cell abundance and immune microenvironment depiction. Bioinformatics 2022; 38:785-791. [PMID: 34636837 DOI: 10.1093/bioinformatics/btab711] [Citation(s) in RCA: 61] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2021] [Revised: 09/22/2021] [Accepted: 10/08/2021] [Indexed: 02/03/2023] Open
Abstract
MOTIVATION Immune cells are important components of the immune system and are crucial for disease initiation, progression, prognosis and survival. Although several computational methods have been designed for predicting the abundance of immune cells, very few tools are applicable to mouse. Given that, mouse is the most widely used animal model in biomedical research, there is an urgent need to develop a precise algorithm for predicting mouse immune cells. RESULTS We developed a tool named Immune Cell Abundance Identifier for mouse (ImmuCellAI-mouse), for estimating the abundance of 36 immune cell (sub)types from gene expression data in a hierarchical strategy of three layers. Reference expression profiles and robust marker gene sets of immune cell types were curated. The abundance of cells in three layers was predicted separately by calculating the ssGSEA enrichment score of the expression deviation profile per cell type. Benchmark results showed high accuracy of ImmuCellAI-mouse in predicting most immune cell types, with correlation coefficients between predicted value and real cell proportion of most cell types being larger than 0.8. We applied ImmuCellAI-mouse to a mouse breast tumor dataset and revealed the dynamic change of immune cell infiltration during treatment, which is consistent with the findings of the original study but with more details. We also constructed an online server for ImmuCellAI-mouse, on which users can upload expression matrices for analysis. ImmuCellAI-mouse will be a useful tool for studying the immune microenvironment, cancer immunology and immunotherapy in mouse models, providing an indispensable supplement for human disease studies. AVAILABILITY AND IMPLEMENTATION Software is available at http://bioinfo.life.hust.edu.cn/ImmuCellAI-mouse/. SUPPLEMENTARY INFORMATION Supplementary data are available at Bioinformatics online.
Collapse
Affiliation(s)
- Ya-Ru Miao
- Center for Artificial Intelligence Biology, Hubei Bioinformatics and Molecular Imaging Key Laboratory, Department of Bioinformatics and Systems Biology, Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Mengxuan Xia
- Center for Artificial Intelligence Biology, Hubei Bioinformatics and Molecular Imaging Key Laboratory, Department of Bioinformatics and Systems Biology, Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Mei Luo
- Center for Artificial Intelligence Biology, Hubei Bioinformatics and Molecular Imaging Key Laboratory, Department of Bioinformatics and Systems Biology, Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Tao Luo
- Center for Artificial Intelligence Biology, Hubei Bioinformatics and Molecular Imaging Key Laboratory, Department of Bioinformatics and Systems Biology, Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Mei Yang
- Center for Artificial Intelligence Biology, Hubei Bioinformatics and Molecular Imaging Key Laboratory, Department of Bioinformatics and Systems Biology, Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
| | - An-Yuan Guo
- Center for Artificial Intelligence Biology, Hubei Bioinformatics and Molecular Imaging Key Laboratory, Department of Bioinformatics and Systems Biology, Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China.,Research Center of Clinical Medicine, Affiliated Hospital of Nantong University, Nantong 226001, China
| |
Collapse
|
67
|
Combes AJ, Samad B, Tsui J, Chew NW, Yan P, Reeder GC, Kushnoor D, Shen A, Davidson B, Barczak AJ, Adkisson M, Edwards A, Naser M, Barry KC, Courau T, Hammoudi T, Argüello RJ, Rao AA, Olshen AB, Cai C, Zhan J, Davis KC, Kelley RK, Chapman JS, Atreya CE, Patel A, Daud AI, Ha P, Diaz AA, Kratz JR, Collisson EA, Fragiadakis GK, Erle DJ, Boissonnas A, Asthana S, Chan V, Krummel MF, Fong L, Nelson A, Kumar R, Lee J, Burra A, Hsu J, Hackett C, Tolentino K, Sjarif J, Johnson P, Shao E, Abrau D, Lupin L, Shaw C, Collins Z, Lea T, Corvera C, Nakakura E, Carnevale J, Alvarado M, Loo K, Chen L, Chow M, Grandis J, Ryan W, El-Sayed I, Jablons D, Woodard G, Meng MW, Porten SP, Okada H, Tempero M, Ko A, Kirkwood K, Vandenberg S, Guevarra D, Oropeza E, Cyr C, Glenn P, Bolen J, Morton A, Eckalbar W. Discovering dominant tumor immune archetypes in a pan-cancer census. Cell 2022; 185:184-203.e19. [PMID: 34963056 PMCID: PMC8862608 DOI: 10.1016/j.cell.2021.12.004] [Citation(s) in RCA: 76] [Impact Index Per Article: 25.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2021] [Revised: 08/25/2021] [Accepted: 12/03/2021] [Indexed: 01/09/2023]
Abstract
Cancers display significant heterogeneity with respect to tissue of origin, driver mutations, and other features of the surrounding tissue. It is likely that individual tumors engage common patterns of the immune system-here "archetypes"-creating prototypical non-destructive tumor immune microenvironments (TMEs) and modulating tumor-targeting. To discover the dominant immune system archetypes, the University of California, San Francisco (UCSF) Immunoprofiler Initiative (IPI) processed 364 individual tumors across 12 cancer types using standardized protocols. Computational clustering of flow cytometry and transcriptomic data obtained from cell sub-compartments uncovered dominant patterns of immune composition across cancers. These archetypes were profound insofar as they also differentiated tumors based upon unique immune and tumor gene-expression patterns. They also partitioned well-established classifications of tumor biology. The IPI resource provides a template for understanding cancer immunity as a collection of dominant patterns of immune organization and provides a rational path forward to learn how to modulate these to improve therapy.
Collapse
Affiliation(s)
- Alexis J. Combes
- Department of Pathology, University of California San Francisco, San Francisco, CA 94143, USA,ImmunoX Initiative, University of California San Francisco, San Francisco, CA 94143, USA,UCSF Immunoprofiler Initiative, University of California San Francisco, San Francisco, CA 94143, USA,UCSF CoLabs, University of California San Francisco, San Francisco, CA 94143, USA,Correspondence: and
| | - Bushra Samad
- Department of Pathology, University of California San Francisco, San Francisco, CA 94143, USA,ImmunoX Initiative, University of California San Francisco, San Francisco, CA 94143, USA,UCSF Immunoprofiler Initiative, University of California San Francisco, San Francisco, CA 94143, USA,UCSF CoLabs, University of California San Francisco, San Francisco, CA 94143, USA
| | - Jessica Tsui
- Department of Pathology, University of California San Francisco, San Francisco, CA 94143, USA,ImmunoX Initiative, University of California San Francisco, San Francisco, CA 94143, USA,UCSF Immunoprofiler Initiative, University of California San Francisco, San Francisco, CA 94143, USA,UCSF CoLabs, University of California San Francisco, San Francisco, CA 94143, USA
| | - Nayvin W. Chew
- Department of Pathology, University of California San Francisco, San Francisco, CA 94143, USA,ImmunoX Initiative, University of California San Francisco, San Francisco, CA 94143, USA,UCSF Immunoprofiler Initiative, University of California San Francisco, San Francisco, CA 94143, USA,UCSF CoLabs, University of California San Francisco, San Francisco, CA 94143, USA
| | - Peter Yan
- Department of Pathology, University of California San Francisco, San Francisco, CA 94143, USA,ImmunoX Initiative, University of California San Francisco, San Francisco, CA 94143, USA,UCSF Immunoprofiler Initiative, University of California San Francisco, San Francisco, CA 94143, USA,UCSF CoLabs, University of California San Francisco, San Francisco, CA 94143, USA
| | - Gabriella C. Reeder
- Department of Pathology, University of California San Francisco, San Francisco, CA 94143, USA,ImmunoX Initiative, University of California San Francisco, San Francisco, CA 94143, USA,UCSF Immunoprofiler Initiative, University of California San Francisco, San Francisco, CA 94143, USA,UCSF CoLabs, University of California San Francisco, San Francisco, CA 94143, USA
| | - Divyashree Kushnoor
- Department of Pathology, University of California San Francisco, San Francisco, CA 94143, USA,ImmunoX Initiative, University of California San Francisco, San Francisco, CA 94143, USA,UCSF Immunoprofiler Initiative, University of California San Francisco, San Francisco, CA 94143, USA,UCSF CoLabs, University of California San Francisco, San Francisco, CA 94143, USA
| | - Alan Shen
- Department of Pathology, University of California San Francisco, San Francisco, CA 94143, USA,ImmunoX Initiative, University of California San Francisco, San Francisco, CA 94143, USA,UCSF Immunoprofiler Initiative, University of California San Francisco, San Francisco, CA 94143, USA,UCSF CoLabs, University of California San Francisco, San Francisco, CA 94143, USA
| | - Brittany Davidson
- ImmunoX Initiative, University of California San Francisco, San Francisco, CA 94143, USA,UCSF Immunoprofiler Initiative, University of California San Francisco, San Francisco, CA 94143, USA,UCSF CoLabs, University of California San Francisco, San Francisco, CA 94143, USA
| | - Andrea J. Barczak
- UCSF Immunoprofiler Initiative, University of California San Francisco, San Francisco, CA 94143, USA,UCSF CoLabs, University of California San Francisco, San Francisco, CA 94143, USA
| | - Michael Adkisson
- UCSF Immunoprofiler Initiative, University of California San Francisco, San Francisco, CA 94143, USA,UCSF CoLabs, University of California San Francisco, San Francisco, CA 94143, USA
| | - Austin Edwards
- Department of Pathology, University of California San Francisco, San Francisco, CA 94143, USA,ImmunoX Initiative, University of California San Francisco, San Francisco, CA 94143, USA,UCSF Immunoprofiler Initiative, University of California San Francisco, San Francisco, CA 94143, USA,UCSF CoLabs, University of California San Francisco, San Francisco, CA 94143, USA
| | - Mohammad Naser
- Department of Pathology, University of California San Francisco, San Francisco, CA 94143, USA,ImmunoX Initiative, University of California San Francisco, San Francisco, CA 94143, USA,UCSF Immunoprofiler Initiative, University of California San Francisco, San Francisco, CA 94143, USA,UCSF CoLabs, University of California San Francisco, San Francisco, CA 94143, USA
| | - Kevin C. Barry
- Translational Research Program, Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Tristan Courau
- Department of Pathology, University of California San Francisco, San Francisco, CA 94143, USA,ImmunoX Initiative, University of California San Francisco, San Francisco, CA 94143, USA,UCSF Immunoprofiler Initiative, University of California San Francisco, San Francisco, CA 94143, USA,UCSF CoLabs, University of California San Francisco, San Francisco, CA 94143, USA
| | - Taymour Hammoudi
- Department of Pathology, University of California San Francisco, San Francisco, CA 94143, USA,ImmunoX Initiative, University of California San Francisco, San Francisco, CA 94143, USA,UCSF Immunoprofiler Initiative, University of California San Francisco, San Francisco, CA 94143, USA,UCSF CoLabs, University of California San Francisco, San Francisco, CA 94143, USA
| | - Rafael J Argüello
- Aix Marseille University, CNRS, INSERM, CIML, Centre d’Immunologie de Marseille-Luminy, Marseille, FRANCE
| | - Arjun Arkal Rao
- Department of Pathology, University of California San Francisco, San Francisco, CA 94143, USA,ImmunoX Initiative, University of California San Francisco, San Francisco, CA 94143, USA,UCSF Immunoprofiler Initiative, University of California San Francisco, San Francisco, CA 94143, USA,UCSF CoLabs, University of California San Francisco, San Francisco, CA 94143, USA
| | - Adam B. Olshen
- UCSF Immunoprofiler Initiative, University of California San Francisco, San Francisco, CA 94143, USA,Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, CA 94143, USA,Department of Epidemiology and Biostatistics, University of California San Francisco, San Francisco, CA 94143, USA
| | | | - Cathy Cai
- Department of Pathology, University of California San Francisco, San Francisco, CA 94143, USA,ImmunoX Initiative, University of California San Francisco, San Francisco, CA 94143, USA,UCSF Immunoprofiler Initiative, University of California San Francisco, San Francisco, CA 94143, USA,UCSF CoLabs, University of California San Francisco, San Francisco, CA 94143, USA
| | - Jenny Zhan
- Department of Pathology, University of California San Francisco, San Francisco, CA 94143, USA,ImmunoX Initiative, University of California San Francisco, San Francisco, CA 94143, USA,UCSF Immunoprofiler Initiative, University of California San Francisco, San Francisco, CA 94143, USA,UCSF CoLabs, University of California San Francisco, San Francisco, CA 94143, USA
| | - Katelyn C. Davis
- Department of Pathology, University of California San Francisco, San Francisco, CA 94143, USA,ImmunoX Initiative, University of California San Francisco, San Francisco, CA 94143, USA,UCSF Immunoprofiler Initiative, University of California San Francisco, San Francisco, CA 94143, USA,UCSF CoLabs, University of California San Francisco, San Francisco, CA 94143, USA
| | - Robin K. Kelley
- Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, CA 94143, USA
| | - Jocelyn S. Chapman
- UCSF Immunoprofiler Initiative, University of California San Francisco, San Francisco, CA 94143, USA,Departments of Obstetrics, Gynecology, and Reproductive Sciences, Medicine, University of California San Francisco, San Francisco, CA 94143, USA
| | - Chloe E. Atreya
- UCSF Immunoprofiler Initiative, University of California San Francisco, San Francisco, CA 94143, USA,Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, CA 94143, USA,Department of Medicine, Division of Hematology and Oncology, University of California San Francisco, San Francisco, CA 94143, USA
| | - Amar Patel
- UCSF Immunoprofiler Initiative, University of California San Francisco, San Francisco, CA 94143, USA,Department of Medicine, Division of Hematology and Oncology, University of California San Francisco, San Francisco, CA 94143, USA
| | - Adil I. Daud
- Department of Medicine, Division of Hematology and Oncology, University of California San Francisco, San Francisco, CA 94143, USA,Department of Dermatology, University of California San Francisco, San Francisco, CA 94143, USA
| | - Patrick Ha
- Department of Otolaryngology, University of California San Francisco, San Francisco, CA 94143, USA
| | - Aaron A. Diaz
- Department of Neurological Surgery, University of California San Francisco, San Francisco, CA 94143, USA
| | - Johannes R. Kratz
- ImmunoX Initiative, University of California San Francisco, San Francisco, CA 94143, USA,Department of Surgery, University of California San Francisco, San Francisco, CA 94143, USA
| | - Eric A. Collisson
- UCSF Immunoprofiler Initiative, University of California San Francisco, San Francisco, CA 94143, USA,Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, CA 94143, USA,Department of Medicine, Division of Hematology and Oncology, University of California San Francisco, San Francisco, CA 94143, USA
| | - Gabriela K Fragiadakis
- ImmunoX Initiative, University of California San Francisco, San Francisco, CA 94143, USA,UCSF Immunoprofiler Initiative, University of California San Francisco, San Francisco, CA 94143, USA,UCSF CoLabs, University of California San Francisco, San Francisco, CA 94143, USA,Department of Medicine Division of Rheumatology, University of California San Francisco, San Francisco, CA 94143, USA
| | - David J. Erle
- ImmunoX Initiative, University of California San Francisco, San Francisco, CA 94143, USA,UCSF Immunoprofiler Initiative, University of California San Francisco, San Francisco, CA 94143, USA,UCSF CoLabs, University of California San Francisco, San Francisco, CA 94143, USA,Lung Biology Center, Department of Medicine and the Cardiovascular Research Institute, University of California San Francisco, San Francisco, CA 94143, USA
| | - Alexandre Boissonnas
- Sorbonne Université, INSERM, CNRS, Centre d’Immunologie et des Maladies Infectieuses - CIMI, Paris, France
| | - Saurabh Asthana
- UCSF Immunoprofiler Initiative, University of California San Francisco, San Francisco, CA 94143, USA,Department of Epidemiology and Biostatistics, University of California San Francisco, San Francisco, CA 94143, USA
| | - Vincent Chan
- ImmunoX Initiative, University of California San Francisco, San Francisco, CA 94143, USA,UCSF Immunoprofiler Initiative, University of California San Francisco, San Francisco, CA 94143, USA,Department of Microbiology and Immunology, University of California San Francisco, San Francisco, CA 94143, USA
| | - Matthew F. Krummel
- Department of Pathology, University of California San Francisco, San Francisco, CA 94143, USA,ImmunoX Initiative, University of California San Francisco, San Francisco, CA 94143, USA,UCSF Immunoprofiler Initiative, University of California San Francisco, San Francisco, CA 94143, USA,Correspondence: and
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
68
|
Chen WW, Liu W, Li Y, Wang J, Ren Y, Wang G, Chen C, Li H. Deciphering the Immune-Tumor Interplay During Early-Stage Lung Cancer Development via Single-Cell Technology. Front Oncol 2022; 11:716042. [PMID: 35047383 PMCID: PMC8761635 DOI: 10.3389/fonc.2021.716042] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Accepted: 11/08/2021] [Indexed: 12/19/2022] Open
Abstract
Lung cancer is the leading cause of cancer-related death worldwide. Cancer immunotherapy has shown great success in treating advanced-stage lung cancer but has yet been used to treat early-stage lung cancer, mostly due to lack of understanding of the tumor immune microenvironment in early-stage lung cancer. The immune system could both constrain and promote tumorigenesis in a process termed immune editing that can be divided into three phases, namely, elimination, equilibrium, and escape. Current understanding of the immune response toward tumor is mainly on the "escape" phase when the tumor is clinically detectable. The detailed mechanism by which tumor progenitor lesions was modulated by the immune system during early stage of lung cancer development remains elusive. The advent of single-cell sequencing technology enables tumor immunologists to address those fundamental questions. In this perspective, we will summarize our current understanding and big gaps about the immune response during early lung tumorigenesis. We will then present the state of the art of single-cell technology and then envision how single-cell technology could be used to address those questions. Advances in the understanding of the immune response and its dynamics during malignant transformation of pre-malignant lesion will shed light on how malignant cells interact with the immune system and evolve under immune selection. Such knowledge could then contribute to the development of precision and early intervention strategies toward lung malignancy.
Collapse
Affiliation(s)
- Wei-Wei Chen
- Department of Clinical Oncology, University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Wei Liu
- CAS Key Laboratory of Quantitative Engineering Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Yingze Li
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Jun Wang
- CAS Key Laboratory of Quantitative Engineering Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Yijiu Ren
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Guangsuo Wang
- Department of Thoracic Surgery, Shenzhen People’s Hospital, The Second Clinical Medical College, Jinan University, The First Affiliated Hospital, Southern University of Science and Technology, Shenzhen, China
| | - Chang Chen
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Hanjie Li
- CAS Key Laboratory of Quantitative Engineering Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| |
Collapse
|
69
|
Tang Q, Liu L, Guo Y, Zhang X, Zhang S, Jia Y, Du Y, Cheng B, Yang L, Huang Y, Chen X. Optical Cell Tagging for Spatially Resolved Single‐Cell RNA Sequencing. Angew Chem Int Ed Engl 2021. [DOI: 10.1002/ange.202113929] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Affiliation(s)
- Qi Tang
- College of Chemistry and Molecular Engineering Peking-Tsinghua Center for Life Sciences Beijing National Laboratory for Molecular Sciences Synthetic and Functional Biomolecules Center Key Laboratory of Bioorganic Chemistry and Molecular Engineering of Ministry of Education Peking University Beijing 100871 China
| | - Lu Liu
- Biomedical Pioneering Innovation Center (BIOPIC) School of Life Sciences College of Chemistry and Molecular Engineering Beijing Advanced Innovation Center for Genomics (ICG) Peking-Tsinghua Center for Life Science and Beijing National Laboratory for Molecular Sciences Peking University Beijing 100871 China
| | - Yilan Guo
- College of Chemistry and Molecular Engineering Peking-Tsinghua Center for Life Sciences Beijing National Laboratory for Molecular Sciences Synthetic and Functional Biomolecules Center Key Laboratory of Bioorganic Chemistry and Molecular Engineering of Ministry of Education Peking University Beijing 100871 China
| | - Xu Zhang
- College of Chemistry and Molecular Engineering Peking-Tsinghua Center for Life Sciences Beijing National Laboratory for Molecular Sciences Synthetic and Functional Biomolecules Center Key Laboratory of Bioorganic Chemistry and Molecular Engineering of Ministry of Education Peking University Beijing 100871 China
| | - Shaoran Zhang
- College of Chemistry and Molecular Engineering Peking-Tsinghua Center for Life Sciences Beijing National Laboratory for Molecular Sciences Synthetic and Functional Biomolecules Center Key Laboratory of Bioorganic Chemistry and Molecular Engineering of Ministry of Education Peking University Beijing 100871 China
| | - Yan Jia
- Renal Division Peking University First Hospital Beijing 100034 China
- Institute of Nephrology Key Laboratory of CKD Prevention and Treatment of Ministry of Education of China Peking University Beijing 100871 China
- Research Units of Diagnosis and Treatment of Immune-mediated Kidney Diseases Chinese Academy of Medical Sciences Beijing 100730 China
| | - Yifei Du
- College of Chemistry and Molecular Engineering Peking-Tsinghua Center for Life Sciences Beijing National Laboratory for Molecular Sciences Synthetic and Functional Biomolecules Center Key Laboratory of Bioorganic Chemistry and Molecular Engineering of Ministry of Education Peking University Beijing 100871 China
| | - Bo Cheng
- College of Chemistry and Molecular Engineering Peking-Tsinghua Center for Life Sciences Beijing National Laboratory for Molecular Sciences Synthetic and Functional Biomolecules Center Key Laboratory of Bioorganic Chemistry and Molecular Engineering of Ministry of Education Peking University Beijing 100871 China
| | - Li Yang
- Renal Division Peking University First Hospital Beijing 100034 China
- Institute of Nephrology Key Laboratory of CKD Prevention and Treatment of Ministry of Education of China Peking University Beijing 100871 China
- Research Units of Diagnosis and Treatment of Immune-mediated Kidney Diseases Chinese Academy of Medical Sciences Beijing 100730 China
| | - Yanyi Huang
- Biomedical Pioneering Innovation Center (BIOPIC) School of Life Sciences College of Chemistry and Molecular Engineering Beijing Advanced Innovation Center for Genomics (ICG) Peking-Tsinghua Center for Life Science and Beijing National Laboratory for Molecular Sciences Peking University Beijing 100871 China
| | - Xing Chen
- College of Chemistry and Molecular Engineering Peking-Tsinghua Center for Life Sciences Beijing National Laboratory for Molecular Sciences Synthetic and Functional Biomolecules Center Key Laboratory of Bioorganic Chemistry and Molecular Engineering of Ministry of Education Peking University Beijing 100871 China
| |
Collapse
|
70
|
Tang Q, Liu L, Guo Y, Zhang X, Zhang S, Jia Y, Du Y, Cheng B, Yang L, Huang Y, Chen X. Optical Cell Tagging for Spatially Resolved Single-Cell RNA Sequencing. Angew Chem Int Ed Engl 2021; 61:e202113929. [PMID: 34970821 DOI: 10.1002/anie.202113929] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Indexed: 01/13/2023]
Abstract
Single-cell RNA sequencing (scRNA-seq) has emerged as a powerful tool for profiling gene expression of distinct cell populations at the single-cell level. However, the information of the positions of cells within the multicellular samples is missing in scRNA-seq datasets. To overcome this limitation, we herein develop OpTAG (optical cell tagging) as a new chemical platform for attaching functional tags onto cell surfaces in a spatially resolved manner. With OpTAG, we establish OpTAG-seq, which enables spatially resolved scRNA-seq. We apply OpTAG-seq to investigate the spatially defined transcriptional program in migrating cancer cells and identified a list of genes that are potential regulators for cancer cell migration and invasion. OpTAG-seq provides a convenient method for mapping cellular heterogeneity with spatial information within multicellular biological systems.
Collapse
Affiliation(s)
- Qi Tang
- College of Chemistry and Molecular Engineering, Peking-Tsinghua Center for Life Sciences, Beijing National Laboratory for Molecular Sciences, Synthetic and Functional Biomolecules Center, Key Laboratory of Bioorganic Chemistry and Molecular Engineering of Ministry of Education, Peking University, Beijing, 100871, China
| | - Lu Liu
- Biomedical Pioneering Innovation Center (BIOPIC), School of Life Sciences, College of Chemistry and Molecular Engineering, Beijing Advanced Innovation Center for Genomics (ICG), Peking-Tsinghua Center for Life Science, and Beijing National Laboratory for Molecular Sciences, Peking University, Beijing, 100871, China
| | - Yilan Guo
- College of Chemistry and Molecular Engineering, Peking-Tsinghua Center for Life Sciences, Beijing National Laboratory for Molecular Sciences, Synthetic and Functional Biomolecules Center, Key Laboratory of Bioorganic Chemistry and Molecular Engineering of Ministry of Education, Peking University, Beijing, 100871, China
| | - Xu Zhang
- College of Chemistry and Molecular Engineering, Peking-Tsinghua Center for Life Sciences, Beijing National Laboratory for Molecular Sciences, Synthetic and Functional Biomolecules Center, Key Laboratory of Bioorganic Chemistry and Molecular Engineering of Ministry of Education, Peking University, Beijing, 100871, China
| | - Shaoran Zhang
- College of Chemistry and Molecular Engineering, Peking-Tsinghua Center for Life Sciences, Beijing National Laboratory for Molecular Sciences, Synthetic and Functional Biomolecules Center, Key Laboratory of Bioorganic Chemistry and Molecular Engineering of Ministry of Education, Peking University, Beijing, 100871, China
| | - Yan Jia
- Renal Division, Peking University First Hospital, Beijing, 100034, China.,Institute of Nephrology, Key Laboratory of CKD Prevention and Treatment of Ministry of Education of China, Peking University, Beijing, 100871, China.,Research Units of Diagnosis and Treatment of Immune-mediated Kidney Diseases, Chinese Academy of Medical Sciences, Beijing, 100730, China
| | - Yifei Du
- College of Chemistry and Molecular Engineering, Peking-Tsinghua Center for Life Sciences, Beijing National Laboratory for Molecular Sciences, Synthetic and Functional Biomolecules Center, Key Laboratory of Bioorganic Chemistry and Molecular Engineering of Ministry of Education, Peking University, Beijing, 100871, China
| | - Bo Cheng
- College of Chemistry and Molecular Engineering, Peking-Tsinghua Center for Life Sciences, Beijing National Laboratory for Molecular Sciences, Synthetic and Functional Biomolecules Center, Key Laboratory of Bioorganic Chemistry and Molecular Engineering of Ministry of Education, Peking University, Beijing, 100871, China
| | - Li Yang
- Renal Division, Peking University First Hospital, Beijing, 100034, China.,Institute of Nephrology, Key Laboratory of CKD Prevention and Treatment of Ministry of Education of China, Peking University, Beijing, 100871, China.,Research Units of Diagnosis and Treatment of Immune-mediated Kidney Diseases, Chinese Academy of Medical Sciences, Beijing, 100730, China
| | - Yanyi Huang
- Biomedical Pioneering Innovation Center (BIOPIC), School of Life Sciences, College of Chemistry and Molecular Engineering, Beijing Advanced Innovation Center for Genomics (ICG), Peking-Tsinghua Center for Life Science, and Beijing National Laboratory for Molecular Sciences, Peking University, Beijing, 100871, China
| | - Xing Chen
- College of Chemistry and Molecular Engineering, Peking-Tsinghua Center for Life Sciences, Beijing National Laboratory for Molecular Sciences, Synthetic and Functional Biomolecules Center, Key Laboratory of Bioorganic Chemistry and Molecular Engineering of Ministry of Education, Peking University, Beijing, 100871, China
| |
Collapse
|
71
|
From imaging a single cell to implementing precision medicine: an exciting new era. Emerg Top Life Sci 2021; 5:837-847. [PMID: 34889448 PMCID: PMC8786301 DOI: 10.1042/etls20210219] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 11/24/2021] [Accepted: 11/24/2021] [Indexed: 11/17/2022]
Abstract
In the age of high-throughput, single-cell biology, single-cell imaging has evolved not only in terms of technological advancements but also in its translational applications. The synchronous advancements of imaging and computational biology have produced opportunities of merging the two, providing the scientific community with tools towards observing, understanding, and predicting cellular and tissue phenotypes and behaviors. Furthermore, multiplexed single-cell imaging and machine learning algorithms now enable patient stratification and predictive diagnostics of clinical specimens. Here, we provide an overall summary of the advances in single-cell imaging, with a focus on high-throughput microscopy phenomics and multiplexed proteomic spatial imaging platforms. We also review various computational tools that have been developed in recent years for image processing and downstream applications used in biomedical sciences. Finally, we discuss how harnessing systems biology approaches and data integration across disciplines can further strengthen the exciting applications and future implementation of single-cell imaging on precision medicine.
Collapse
|
72
|
Lubitz GS, Brody JD. Not just neighbours: positive feedback between tumour-associated macrophages and exhausted T cells. Nat Rev Immunol 2021; 22:3. [PMID: 34789868 DOI: 10.1038/s41577-021-00660-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
- Gabrielle S Lubitz
- OxMS Preprint Journal Club, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| | - Joshua D Brody
- OxMS Preprint Journal Club, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| |
Collapse
|
73
|
Zhao E, Stone MR, Ren X, Guenthoer J, Smythe KS, Pulliam T, Williams SR, Uytingco CR, Taylor SEB, Nghiem P, Bielas JH, Gottardo R. Spatial transcriptomics at subspot resolution with BayesSpace. Nat Biotechnol 2021; 39:1375-1384. [PMID: 34083791 PMCID: PMC8763026 DOI: 10.1038/s41587-021-00935-2] [Citation(s) in RCA: 373] [Impact Index Per Article: 93.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Accepted: 04/26/2021] [Indexed: 11/09/2022]
Abstract
Recent spatial gene expression technologies enable comprehensive measurement of transcriptomic profiles while retaining spatial context. However, existing analysis methods do not address the limited resolution of the technology or use the spatial information efficiently. Here, we introduce BayesSpace, a fully Bayesian statistical method that uses the information from spatial neighborhoods for resolution enhancement of spatial transcriptomic data and for clustering analysis. We benchmark BayesSpace against current methods for spatial and non-spatial clustering and show that it improves identification of distinct intra-tissue transcriptional profiles from samples of the brain, melanoma, invasive ductal carcinoma and ovarian adenocarcinoma. Using immunohistochemistry and an in silico dataset constructed from scRNA-seq data, we show that BayesSpace resolves tissue structure that is not detectable at the original resolution and identifies transcriptional heterogeneity inaccessible to histological analysis. Our results illustrate BayesSpace's utility in facilitating the discovery of biological insights from spatial transcriptomic datasets.
Collapse
Affiliation(s)
- Edward Zhao
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
- Department of Biostatistics, University of Washington, Seattle, WA, USA
| | - Matthew R Stone
- Fred Hutch Innovation Laboratory, Immunotherapy Integrated Research Center, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Xing Ren
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Jamie Guenthoer
- Human Biology Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Kimberly S Smythe
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Thomas Pulliam
- Department of Medicine, Division of Dermatology, University of Washington, Seattle, WA, USA
| | | | | | | | - Paul Nghiem
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
- Department of Medicine, Division of Dermatology, University of Washington, Seattle, WA, USA
- Seattle Cancer Care Alliance, Seattle, WA, USA
| | - Jason H Bielas
- Fred Hutch Innovation Laboratory, Immunotherapy Integrated Research Center, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
- Translational Research Program, Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
- Department of Pathology, University of Washington, Seattle, WA, USA
| | - Raphael Gottardo
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA.
- Department of Biostatistics, University of Washington, Seattle, WA, USA.
| |
Collapse
|
74
|
Chen Y, Qian W, Lin L, Cai L, Yin K, Jiang S, Song J, Han RPS, Yang C. Mapping Gene Expression in the Spatial Dimension. SMALL METHODS 2021; 5:e2100722. [PMID: 34927963 DOI: 10.1002/smtd.202100722] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Revised: 08/25/2021] [Indexed: 06/14/2023]
Abstract
The main function and biological processes of tissues are determined by the combination of gene expression and spatial organization of their cells. RNA sequencing technologies have primarily interrogated gene expression without preserving the native spatial context of cells. However, the emergence of various spatially-resolved transcriptome analysis methods now makes it possible to map the gene expression to specific coordinates within tissues, enabling transcriptional heterogeneity between different regions, and for the localization of specific transcripts and novel spatial markers to be revealed. Hence, spatially-resolved transcriptome analysis technologies have broad utility in research into human disease and developmental biology. Here, recent advances in spatially-resolved transcriptome analysis methods are summarized, including experimental technologies and computational methods. Strengths, challenges, and potential applications of those methods are highlighted, and perspectives in this field are provided.
Collapse
Affiliation(s)
- Yingwen Chen
- The MOE Key Laboratory of Spectrochemical Analysis & Instrumentation, the Key Laboratory of Chemical Biology of Fujian Province, State Key Laboratory of Physical Chemistry of Solid Surfaces, Collaborative Innovation Center of Chemistry for Energy Materials, Department of Chemical Biology, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen, 361005, China
| | - Weizhou Qian
- The MOE Key Laboratory of Spectrochemical Analysis & Instrumentation, the Key Laboratory of Chemical Biology of Fujian Province, State Key Laboratory of Physical Chemistry of Solid Surfaces, Collaborative Innovation Center of Chemistry for Energy Materials, Department of Chemical Biology, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen, 361005, China
| | - Li Lin
- The MOE Key Laboratory of Spectrochemical Analysis & Instrumentation, the Key Laboratory of Chemical Biology of Fujian Province, State Key Laboratory of Physical Chemistry of Solid Surfaces, Collaborative Innovation Center of Chemistry for Energy Materials, Department of Chemical Biology, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen, 361005, China
| | - Linfeng Cai
- The MOE Key Laboratory of Spectrochemical Analysis & Instrumentation, the Key Laboratory of Chemical Biology of Fujian Province, State Key Laboratory of Physical Chemistry of Solid Surfaces, Collaborative Innovation Center of Chemistry for Energy Materials, Department of Chemical Biology, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen, 361005, China
| | - Kun Yin
- The MOE Key Laboratory of Spectrochemical Analysis & Instrumentation, the Key Laboratory of Chemical Biology of Fujian Province, State Key Laboratory of Physical Chemistry of Solid Surfaces, Collaborative Innovation Center of Chemistry for Energy Materials, Department of Chemical Biology, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen, 361005, China
| | - Shaowei Jiang
- The MOE Key Laboratory of Spectrochemical Analysis & Instrumentation, the Key Laboratory of Chemical Biology of Fujian Province, State Key Laboratory of Physical Chemistry of Solid Surfaces, Collaborative Innovation Center of Chemistry for Energy Materials, Department of Chemical Biology, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen, 361005, China
| | - Jia Song
- Institute of Molecular Medicine, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China
| | - Ray P S Han
- Jiangxi University of Traditional Chinese Medicine, Nanchang, Jiangxi, 33004, China
| | - Chaoyong Yang
- The MOE Key Laboratory of Spectrochemical Analysis & Instrumentation, the Key Laboratory of Chemical Biology of Fujian Province, State Key Laboratory of Physical Chemistry of Solid Surfaces, Collaborative Innovation Center of Chemistry for Energy Materials, Department of Chemical Biology, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen, 361005, China
- Institute of Molecular Medicine, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China
| |
Collapse
|
75
|
van der Leun AM, Hoekstra ME, Reinalda L, Scheele CLGJ, Toebes M, van de Graaff MJ, Chen LYY, Li H, Bercovich A, Lubling Y, David E, Thommen DS, Tanay A, van Rheenen J, Amit I, van Kasteren SI, Schumacher TN. Single-cell analysis of regions of interest (SCARI) using a photosensitive tag. Nat Chem Biol 2021; 17:1139-1147. [PMID: 34504322 PMCID: PMC7611907 DOI: 10.1038/s41589-021-00839-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Accepted: 06/24/2021] [Indexed: 11/25/2022]
Abstract
The functional activity and differentiation potential of cells are determined by their interactions with surrounding cells. Approaches that allow unbiased characterization of cell states while at the same time providing spatial information are of major value to assess this environmental influence. However, most current techniques are hampered by a tradeoff between spatial resolution and cell profiling depth. Here, we develop a photocage-based technology that allows isolation and in-depth analysis of live cells from regions of interest in complex ex vivo systems, including primary human tissues. The use of a highly sensitive 4-nitrophenyl(benzofuran) cage coupled to a set of nanobodies allows high-resolution photo-uncaging of different cell types in areas of interest. Single-cell RNA-sequencing of spatially defined CD8+ T cells is used to exemplify the feasibility of identifying location-dependent cell states. The technology described here provides a valuable tool for the analysis of spatially defined cells in diverse biological systems, including clinical samples.
Collapse
Affiliation(s)
- Anne M van der Leun
- Division of Molecular Oncology & Immunology, Oncode Institute, The Netherlands Cancer Institute, Amsterdam, Netherlands
| | - Mirjam E Hoekstra
- Division of Molecular Oncology & Immunology, Oncode Institute, The Netherlands Cancer Institute, Amsterdam, Netherlands
| | - Luuk Reinalda
- Department of Bio-Organic Synthesis, Leiden Institute of Chemistry, Leiden University, Leiden, Netherlands
| | - Colinda L G J Scheele
- Division of Molecular Pathology, Oncode Institute, The Netherlands Cancer Institute, Amsterdam, Netherlands
- VIB-KULeuven Center for Cancer Biology, Leuven, Belgium
| | - Mireille Toebes
- Division of Molecular Oncology & Immunology, Oncode Institute, The Netherlands Cancer Institute, Amsterdam, Netherlands
| | - Michel J van de Graaff
- Department of Bio-Organic Synthesis, Leiden Institute of Chemistry, Leiden University, Leiden, Netherlands
- SeraNovo, Leiden, Netherlands
| | - Linda Y Y Chen
- Division of Molecular Pathology, Oncode Institute, The Netherlands Cancer Institute, Amsterdam, Netherlands
| | - Hanjie Li
- Department of Immunology, Weizmann Institute of Science, Rehovot, Israel
- Shenzhen Institute of Synthetic Biology, Shenzhen, China
| | - Akhiad Bercovich
- Department of Computer Science and Applied Mathematics and Department of Biological Regulation, Weizmann Institute, Rehovot, Israel
| | - Yaniv Lubling
- Department of Computer Science and Applied Mathematics and Department of Biological Regulation, Weizmann Institute, Rehovot, Israel
- Cancer Research UK Cambridge Institute, Cambridge, UK
| | - Eyal David
- Department of Immunology, Weizmann Institute of Science, Rehovot, Israel
| | - Daniela S Thommen
- Division of Molecular Oncology & Immunology, The Netherlands Cancer Institute, Amsterdam, Netherlands
| | - Amos Tanay
- Department of Computer Science and Applied Mathematics and Department of Biological Regulation, Weizmann Institute, Rehovot, Israel
| | - Jacco van Rheenen
- Division of Molecular Pathology, Oncode Institute, The Netherlands Cancer Institute, Amsterdam, Netherlands
| | - Ido Amit
- Department of Immunology, Weizmann Institute of Science, Rehovot, Israel
| | - Sander I van Kasteren
- Department of Bio-Organic Synthesis, Leiden Institute of Chemistry, Leiden University, Leiden, Netherlands.
| | - Ton N Schumacher
- Division of Molecular Oncology & Immunology, Oncode Institute, The Netherlands Cancer Institute, Amsterdam, Netherlands.
- Department of Hematology, Leiden University Medical Center, Leiden, Netherlands.
| |
Collapse
|
76
|
Modavi C, Abate A. Change can be SCARI. Nat Chem Biol 2021; 17:1119-1120. [PMID: 34504323 DOI: 10.1038/s41589-021-00859-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
- Cyrus Modavi
- Department of Bioengineering and Therapeutic Sciences, University of California San Francisco, San Francisco, CA, USA
| | - Adam Abate
- Department of Bioengineering and Therapeutic Sciences, University of California San Francisco, San Francisco, CA, USA.
| |
Collapse
|
77
|
Im K, Combes AJ, Spitzer MH, Satpathy AT, Krummel MF. Archetypes of checkpoint-responsive immunity. Trends Immunol 2021; 42:960-974. [PMID: 34642094 PMCID: PMC8724347 DOI: 10.1016/j.it.2021.09.007] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Revised: 09/08/2021] [Accepted: 09/14/2021] [Indexed: 01/10/2023]
Abstract
Responsiveness to immune checkpoint blockade (ICB) therapy in cancer is currently predicted by disparate individual measures - with varying degrees of accuracy - including tumor mutation burden, tumor-infiltrating T cell densities, dendritic cell frequencies, and the expression of checkpoint ligands. We propose that many of these individual parameters are linked, forming two distinct 'reactive' immune archetypes - collections of cells and gene expression - in ICB-responsive patients. We hypothesize that these are 'seeds' of antitumor immunity and are supported by specific elements of the tumor microenvironment (TME) and by actions of the microbiome. Although removing 'immunosuppressive' factors in the TME is important, understanding and parsing reactive immunity is crucial for optimal prognosis and for engaging this biology with candidate therapies to increase tumor cure rates.
Collapse
Affiliation(s)
- Kwok Im
- Department of Pathology and ImmunoX Initiative, University of California at San Francisco, San Francisco, CA 94143, USA; UCSF CoLabs, University of California at San Francisco, San Francisco, CA 94143, USA
| | - Alexis J Combes
- Department of Pathology and ImmunoX Initiative, University of California at San Francisco, San Francisco, CA 94143, USA; UCSF CoLabs, University of California at San Francisco, San Francisco, CA 94143, USA
| | - Matthew H Spitzer
- Department of Otolaryngology, School of Medicine, University of California at San Francisco, San Franciso, CA 94143, USA
| | | | - Matthew F Krummel
- Department of Pathology and ImmunoX Initiative, University of California at San Francisco, San Francisco, CA 94143, USA.
| |
Collapse
|
78
|
Abstract
Spatial transcriptomics is a rapidly growing field that promises to comprehensively characterize tissue organization and architecture at the single-cell or subcellular resolution. Such information provides a solid foundation for mechanistic understanding of many biological processes in both health and disease that cannot be obtained by using traditional technologies. The development of computational methods plays important roles in extracting biological signals from raw data. Various approaches have been developed to overcome technology-specific limitations such as spatial resolution, gene coverage, sensitivity, and technical biases. Downstream analysis tools formulate spatial organization and cell-cell communications as quantifiable properties, and provide algorithms to derive such properties. Integrative pipelines further assemble multiple tools in one package, allowing biologists to conveniently analyze data from beginning to end. In this review, we summarize the state of the art of spatial transcriptomic data analysis methods and pipelines, and discuss how they operate on different technological platforms.
Collapse
Affiliation(s)
- Ruben Dries
- Department of Medicine, Boston University School of Medicine, Boston, Massachusetts 02118, USA
- Bioinformatics Graduate Program, Boston University, Boston, Massachusetts 02215, USA
- Section of Computational Biomedicine, Boston University School of Medicine, Boston, Massachusetts 02118, USA
| | - Jiaji Chen
- Department of Medicine, Boston University School of Medicine, Boston, Massachusetts 02118, USA
| | - Natalie Del Rossi
- Department of Genetics and Genomic Sciences, Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, New York 10029, USA
| | - Mohammed Muzamil Khan
- Department of Medicine, Boston University School of Medicine, Boston, Massachusetts 02118, USA
- Bioinformatics Graduate Program, Boston University, Boston, Massachusetts 02215, USA
- Section of Computational Biomedicine, Boston University School of Medicine, Boston, Massachusetts 02118, USA
| | - Adriana Sistig
- Department of Genetics and Genomic Sciences, Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, New York 10029, USA
| | - Guo-Cheng Yuan
- Department of Genetics and Genomic Sciences, Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, New York 10029, USA
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, New York 10029, USA
| |
Collapse
|
79
|
Longo SK, Guo MG, Ji AL, Khavari PA. Integrating single-cell and spatial transcriptomics to elucidate intercellular tissue dynamics. Nat Rev Genet 2021; 22:627-644. [PMID: 34145435 PMCID: PMC9888017 DOI: 10.1038/s41576-021-00370-8] [Citation(s) in RCA: 456] [Impact Index Per Article: 114.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/29/2021] [Indexed: 02/07/2023]
Abstract
Single-cell RNA sequencing (scRNA-seq) identifies cell subpopulations within tissue but does not capture their spatial distribution nor reveal local networks of intercellular communication acting in situ. A suite of recently developed techniques that localize RNA within tissue, including multiplexed in situ hybridization and in situ sequencing (here defined as high-plex RNA imaging) and spatial barcoding, can help address this issue. However, no method currently provides as complete a scope of the transcriptome as does scRNA-seq, underscoring the need for approaches to integrate single-cell and spatial data. Here, we review efforts to integrate scRNA-seq with spatial transcriptomics, including emerging integrative computational methods, and propose ways to effectively combine current methodologies.
Collapse
Affiliation(s)
- Sophia K. Longo
- Program in Epithelial Biology, Stanford University, Stanford, CA, USA,Stanford Cancer Institute, Stanford University, Stanford, CA, USA
| | - Margaret G. Guo
- Program in Epithelial Biology, Stanford University, Stanford, CA, USA,Stanford Cancer Institute, Stanford University, Stanford, CA, USA,Program in Biomedical Informatics, Stanford University, Stanford, CA, USA
| | - Andrew L. Ji
- Program in Epithelial Biology, Stanford University, Stanford, CA, USA,Stanford Cancer Institute, Stanford University, Stanford, CA, USA
| | - Paul A. Khavari
- Program in Epithelial Biology, Stanford University, Stanford, CA, USA,Stanford Cancer Institute, Stanford University, Stanford, CA, USA,Veterans Affairs Palo Alto Healthcare System, Palo Alto, CA, USA
| |
Collapse
|
80
|
Xu Y, Su GH, Ma D, Xiao Y, Shao ZM, Jiang YZ. Technological advances in cancer immunity: from immunogenomics to single-cell analysis and artificial intelligence. Signal Transduct Target Ther 2021; 6:312. [PMID: 34417437 PMCID: PMC8377461 DOI: 10.1038/s41392-021-00729-7] [Citation(s) in RCA: 62] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Revised: 07/06/2021] [Accepted: 07/18/2021] [Indexed: 02/07/2023] Open
Abstract
Immunotherapies play critical roles in cancer treatment. However, given that only a few patients respond to immune checkpoint blockades and other immunotherapeutic strategies, more novel technologies are needed to decipher the complicated interplay between tumor cells and the components of the tumor immune microenvironment (TIME). Tumor immunomics refers to the integrated study of the TIME using immunogenomics, immunoproteomics, immune-bioinformatics, and other multi-omics data reflecting the immune states of tumors, which has relied on the rapid development of next-generation sequencing. High-throughput genomic and transcriptomic data may be utilized for calculating the abundance of immune cells and predicting tumor antigens, referring to immunogenomics. However, as bulk sequencing represents the average characteristics of a heterogeneous cell population, it fails to distinguish distinct cell subtypes. Single-cell-based technologies enable better dissection of the TIME through precise immune cell subpopulation and spatial architecture investigations. In addition, radiomics and digital pathology-based deep learning models largely contribute to research on cancer immunity. These artificial intelligence technologies have performed well in predicting response to immunotherapy, with profound significance in cancer therapy. In this review, we briefly summarize conventional and state-of-the-art technologies in the field of immunogenomics, single-cell and artificial intelligence, and present prospects for future research.
Collapse
Affiliation(s)
- Ying Xu
- Key Laboratory of Breast Cancer in Shanghai, Department of Breast Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Guan-Hua Su
- Key Laboratory of Breast Cancer in Shanghai, Department of Breast Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Ding Ma
- Key Laboratory of Breast Cancer in Shanghai, Department of Breast Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Yi Xiao
- Key Laboratory of Breast Cancer in Shanghai, Department of Breast Surgery, Fudan University Shanghai Cancer Center, Shanghai, China.
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China.
| | - Zhi-Ming Shao
- Key Laboratory of Breast Cancer in Shanghai, Department of Breast Surgery, Fudan University Shanghai Cancer Center, Shanghai, China.
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China.
- Institutes of Biomedical Sciences, Fudan University, Shanghai, China.
| | - Yi-Zhou Jiang
- Key Laboratory of Breast Cancer in Shanghai, Department of Breast Surgery, Fudan University Shanghai Cancer Center, Shanghai, China.
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China.
| |
Collapse
|
81
|
Genshaft AS, Ziegler CGK, Tzouanas CN, Mead BE, Jaeger AM, Navia AW, King RP, Mana MD, Huang S, Mitsialis V, Snapper SB, Yilmaz ÖH, Jacks T, Van Humbeck JF, Shalek AK. Live cell tagging tracking and isolation for spatial transcriptomics using photoactivatable cell dyes. Nat Commun 2021; 12:4995. [PMID: 34404785 PMCID: PMC8371137 DOI: 10.1038/s41467-021-25279-y] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Accepted: 07/26/2021] [Indexed: 12/30/2022] Open
Abstract
A cell's phenotype and function are influenced by dynamic interactions with its microenvironment. To examine cellular spatiotemporal activity, we developed SPACECAT-Spatially PhotoActivatable Color Encoded Cell Address Tags-to annotate, track, and isolate cells while preserving viability. In SPACECAT, samples are stained with photocaged fluorescent molecules, and cells are labeled by uncaging those molecules with user-patterned near-UV light. SPACECAT offers single-cell precision and temporal stability across diverse cell and tissue types. Illustratively, we target crypt-like regions in patient-derived intestinal organoids to enrich for stem-like and actively mitotic cells, matching literature expectations. Moreover, we apply SPACECAT to ex vivo tissue sections from four healthy organs and an autochthonous lung tumor model. Lastly, we provide a computational framework to identify spatially-biased transcriptome patterns and enriched phenotypes. This minimally perturbative and broadly applicable method links cellular spatiotemporal and/or behavioral phenotypes with diverse downstream assays, enabling insights into the connections between tissue microenvironments and (dys)function.
Collapse
Affiliation(s)
- Alex S Genshaft
- Institute for Medical Engineering & Science, MIT, Cambridge, MA, USA
- Department of Chemistry, MIT, Cambridge, MA, USA
- Koch Institute for Integrative Cancer Research, MIT, Cambridge, MA, USA
- The Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Harvard-MIT Program in Health Sciences and Technology, Harvard Medical School, Cambridge, MA, USA
| | - Carly G K Ziegler
- Institute for Medical Engineering & Science, MIT, Cambridge, MA, USA
- Department of Chemistry, MIT, Cambridge, MA, USA
- Koch Institute for Integrative Cancer Research, MIT, Cambridge, MA, USA
- The Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Harvard-MIT Program in Health Sciences and Technology, Harvard Medical School, Cambridge, MA, USA
| | - Constantine N Tzouanas
- Institute for Medical Engineering & Science, MIT, Cambridge, MA, USA
- Department of Chemistry, MIT, Cambridge, MA, USA
- Koch Institute for Integrative Cancer Research, MIT, Cambridge, MA, USA
- The Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Harvard-MIT Program in Health Sciences and Technology, Harvard Medical School, Cambridge, MA, USA
| | - Benjamin E Mead
- Institute for Medical Engineering & Science, MIT, Cambridge, MA, USA
- Department of Chemistry, MIT, Cambridge, MA, USA
- Koch Institute for Integrative Cancer Research, MIT, Cambridge, MA, USA
- The Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Harvard-MIT Program in Health Sciences and Technology, Harvard Medical School, Cambridge, MA, USA
| | - Alex M Jaeger
- Koch Institute for Integrative Cancer Research, MIT, Cambridge, MA, USA
| | - Andrew W Navia
- Institute for Medical Engineering & Science, MIT, Cambridge, MA, USA
- Department of Chemistry, MIT, Cambridge, MA, USA
- Koch Institute for Integrative Cancer Research, MIT, Cambridge, MA, USA
- The Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Harvard-MIT Program in Health Sciences and Technology, Harvard Medical School, Cambridge, MA, USA
| | - Ryan P King
- Department of Chemistry, MIT, Cambridge, MA, USA
| | - Miyeko D Mana
- Koch Institute for Integrative Cancer Research, MIT, Cambridge, MA, USA
| | - Siyi Huang
- The Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, USA
- Department of Immunology & HMS Center for Immune Imaging, Harvard Medical School, Boston, MA, USA
| | - Vanessa Mitsialis
- Division of Gastroenterology, Hepatology, and Nutrition, Boston Children's Hospital, Boston, MA, USA
- Division of Gastroenterology, Brigham and Women's Hospital, Boston, MA, USA
| | - Scott B Snapper
- Division of Gastroenterology, Hepatology, and Nutrition, Boston Children's Hospital, Boston, MA, USA
- Division of Gastroenterology, Brigham and Women's Hospital, Boston, MA, USA
| | - Ömer H Yilmaz
- Koch Institute for Integrative Cancer Research, MIT, Cambridge, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Pathology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Tyler Jacks
- Koch Institute for Integrative Cancer Research, MIT, Cambridge, MA, USA
- Howard Hughes Medical Institute, Chevy Chase, MD, USA
| | | | - Alex K Shalek
- Institute for Medical Engineering & Science, MIT, Cambridge, MA, USA.
- Department of Chemistry, MIT, Cambridge, MA, USA.
- Koch Institute for Integrative Cancer Research, MIT, Cambridge, MA, USA.
- The Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, USA.
- Broad Institute of MIT and Harvard, Cambridge, MA, USA.
- Harvard-MIT Program in Health Sciences and Technology, Harvard Medical School, Cambridge, MA, USA.
| |
Collapse
|
82
|
Lê Cao KA, Abadi AJ, Davis-Marcisak EF, Hsu L, Arora A, Coullomb A, Deshpande A, Feng Y, Jeganathan P, Loth M, Meng C, Mu W, Pancaldi V, Sankaran K, Righelli D, Singh A, Sodicoff JS, Stein-O’Brien GL, Subramanian A, Welch JD, You Y, Argelaguet R, Carey VJ, Dries R, Greene CS, Holmes S, Love MI, Ritchie ME, Yuan GC, Culhane AC, Fertig E. Community-wide hackathons to identify central themes in single-cell multi-omics. Genome Biol 2021; 22:220. [PMID: 34353350 PMCID: PMC8340473 DOI: 10.1186/s13059-021-02433-9] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Affiliation(s)
- Kim-Anh Lê Cao
- Melbourne Integrative Genomics, School of Mathematics and Statistics, University of Melbourne, Melbourne, Australia
| | - Al J. Abadi
- Melbourne Integrative Genomics, School of Mathematics and Statistics, University of Melbourne, Melbourne, Australia
| | - Emily F. Davis-Marcisak
- McKusick-Nathans Institute of the Department of Genetic Medicine, Johns Hopkins School of Medicine, Baltimore, MD USA
| | - Lauren Hsu
- Data Science, Dana-Farber Cancer Institute, Boston, MA USA
- Department of Genetics, UNC, Chapel Hill, NC USA
| | - Arshi Arora
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY USA
| | - Alexis Coullomb
- Centre de Recherches en Cancérologie de Toulouse (INSERM), Université Paul Sabatier III, Toulouse, France
| | - Atul Deshpande
- Cancer Convergence Institute and Division of Quantitative Sciences, Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD USA
| | - Yuzhou Feng
- Melbourne Integrative Genomics, School of Mathematics and Statistics, University of Melbourne, Melbourne, Australia
| | | | - Melanie Loth
- Cancer Convergence Institute and Division of Quantitative Sciences, Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD USA
| | - Chen Meng
- Bavarian Center for Biomolecular Mass Spectrometry (BayBioMS), School of Life Sciences, Technical University of Munich, Munich, Germany
| | - Wancen Mu
- Department of Biostatistics, UNC, Chapel Hill, NC USA
| | - Vera Pancaldi
- Centre de Recherches en Cancérologie de Toulouse (INSERM), Université Paul Sabatier III, Toulouse, France
- Barcelona Supercomputing Center, Barcelona, Spain
| | - Kris Sankaran
- Department of Statistics, University of Wisconsin, Madison, WI USA
| | - Dario Righelli
- Department of Statistical Sciences, University of Padova, Padova, PD Italy
| | - Amrit Singh
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC Canada
- PROOF Centre of Excellence, Vancouver, BC Canada
| | - Joshua S. Sodicoff
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI USA
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI USA
| | - Genevieve L. Stein-O’Brien
- McKusick-Nathans Institute of the Department of Genetic Medicine, Johns Hopkins School of Medicine, Baltimore, MD USA
- Cancer Convergence Institute and Division of Quantitative Sciences, Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD USA
- Department of Neuroscience, Johns Hopkins University, Baltimore, MD USA
- Kavli Neuroscience Discovery Institute, Johns Hopkins University, Baltimore, MD USA
| | | | - Joshua D. Welch
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI USA
- Department of Computer Science and Engineering, University of Michigan, Ann Arbor, MI USA
| | - Yue You
- Epigenetics and Development Division, The Walter and Eliza Hall Institute of Medical Research, University of Melbourne, Melbourne, Australia
- Department of Medical Biology, University of Melbourne, Melbourne, Australia
| | | | - Vincent J. Carey
- Channing Division of Network Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA USA
| | - Ruben Dries
- Department of Hematology and Oncology, Boston Medical Center, Boston, MA USA
- Department of Computational Biomedicine, Boston University School of Medicine, Boston, MA USA
- Center for Regenerative Medicine (CReM), Boston University, Boston, MA USA
| | - Casey S. Greene
- Center for Health AI and Department of Biochemistry and Molecular Genetics, University of Colorado School of Medicine, Aurora, CO USA
| | - Susan Holmes
- Department of Statistics, Stanford University, Stanford, CA USA
| | - Michael I. Love
- Department of Biostatistics, UNC, Chapel Hill, NC USA
- Department of Genetics, UNC, Chapel Hill, NC USA
| | - Matthew E. Ritchie
- Epigenetics and Development Division, The Walter and Eliza Hall Institute of Medical Research, University of Melbourne, Melbourne, Australia
- School of Mathematics and Statistics, University of Melbourne, Melbourne, Australia
- Department of Medical Biology, University of Melbourne, Melbourne, Australia
| | - Guo-Cheng Yuan
- Department of Genetics and Genomic Sciences, Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, NY USA
| | - Aedin C. Culhane
- Data Science, Dana-Farber Cancer Institute, Boston, MA USA
- Biostatistics, Harvard TH Chan School of Public Health, Boston, MA USA
| | - Elana Fertig
- Cancer Convergence Institute and Division of Quantitative Sciences, Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD USA
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD USA
- Department of Applied Mathematics and Statistics, Johns Hopkins University Whiting School of Engineering, Baltimore, MD USA
| |
Collapse
|
83
|
Lewis SM, Asselin-Labat ML, Nguyen Q, Berthelet J, Tan X, Wimmer VC, Merino D, Rogers KL, Naik SH. Spatial omics and multiplexed imaging to explore cancer biology. Nat Methods 2021; 18:997-1012. [PMID: 34341583 DOI: 10.1038/s41592-021-01203-6] [Citation(s) in RCA: 271] [Impact Index Per Article: 67.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Accepted: 06/04/2021] [Indexed: 01/19/2023]
Abstract
Understanding intratumoral heterogeneity-the molecular variation among cells within a tumor-promises to address outstanding questions in cancer biology and improve the diagnosis and treatment of specific cancer subtypes. Single-cell analyses, especially RNA sequencing and other genomics modalities, have been transformative in revealing novel biomarkers and molecular regulators associated with tumor growth, metastasis and drug resistance. However, these approaches fail to provide a complete picture of tumor biology, as information on cellular location within the tumor microenvironment is lost. New technologies leveraging multiplexed fluorescence, DNA, RNA and isotope labeling enable the detection of tens to thousands of cancer subclones or molecular biomarkers within their native spatial context. The expeditious growth in these techniques, along with methods for multiomics data integration, promises to yield a more comprehensive understanding of cell-to-cell variation within and between individual tumors. Here we provide the current state and future perspectives on the spatial technologies expected to drive the next generation of research and diagnostic and therapeutic strategies for cancer.
Collapse
Affiliation(s)
- Sabrina M Lewis
- Advanced Technology and Biology Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia.,Immunology Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia.,Department of Medical Biology, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne, Melbourne, Victoria, Australia
| | - Marie-Liesse Asselin-Labat
- Department of Medical Biology, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne, Melbourne, Victoria, Australia.,Personalised Oncology Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
| | - Quan Nguyen
- Division of Genetics and Genomics, Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland, Australia
| | - Jean Berthelet
- Olivia Newton-John Cancer Research Institute, Heidelberg, Victoria, Australia.,School of Cancer Medicine, La Trobe University, Bundoora, Victoria, Australia
| | - Xiao Tan
- Division of Genetics and Genomics, Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland, Australia
| | - Verena C Wimmer
- Advanced Technology and Biology Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia.,Department of Medical Biology, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne, Melbourne, Victoria, Australia
| | - Delphine Merino
- Immunology Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia.,Department of Medical Biology, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne, Melbourne, Victoria, Australia.,Olivia Newton-John Cancer Research Institute, Heidelberg, Victoria, Australia.,School of Cancer Medicine, La Trobe University, Bundoora, Victoria, Australia
| | - Kelly L Rogers
- Advanced Technology and Biology Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia. .,Department of Medical Biology, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne, Melbourne, Victoria, Australia.
| | - Shalin H Naik
- Immunology Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia. .,Department of Medical Biology, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne, Melbourne, Victoria, Australia.
| |
Collapse
|
84
|
Fu T, Dai LJ, Wu SY, Xiao Y, Ma D, Jiang YZ, Shao ZM. Spatial architecture of the immune microenvironment orchestrates tumor immunity and therapeutic response. J Hematol Oncol 2021; 14:98. [PMID: 34172088 PMCID: PMC8234625 DOI: 10.1186/s13045-021-01103-4] [Citation(s) in RCA: 237] [Impact Index Per Article: 59.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2021] [Accepted: 06/03/2021] [Indexed: 02/08/2023] Open
Abstract
Tumors are not only aggregates of malignant cells but also well-organized complex ecosystems. The immunological components within tumors, termed the tumor immune microenvironment (TIME), have long been shown to be strongly related to tumor development, recurrence and metastasis. However, conventional studies that underestimate the potential value of the spatial architecture of the TIME are unable to completely elucidate its complexity. As innovative high-flux and high-dimensional technologies emerge, researchers can more feasibly and accurately detect and depict the spatial architecture of the TIME. These findings have improved our understanding of the complexity and role of the TIME in tumor biology. In this review, we first epitomized some representative emerging technologies in the study of the spatial architecture of the TIME and categorized the description methods used to characterize these structures. Then, we determined the functions of the spatial architecture of the TIME in tumor biology and the effects of the gradient of extracellular nonspecific chemicals (ENSCs) on the TIME. We also discussed the potential clinical value of our understanding of the spatial architectures of the TIME, as well as current limitations and future prospects in this novel field. This review will bring spatial architectures of the TIME, an emerging dimension of tumor ecosystem research, to the attention of more researchers and promote its application in tumor research and clinical practice.
Collapse
Affiliation(s)
- Tong Fu
- Department of Breast Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Key Laboratory of Breast Cancer in Shanghai, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Lei-Jie Dai
- Department of Breast Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Key Laboratory of Breast Cancer in Shanghai, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Song-Yang Wu
- Department of Breast Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Key Laboratory of Breast Cancer in Shanghai, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Yi Xiao
- Department of Breast Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Key Laboratory of Breast Cancer in Shanghai, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Ding Ma
- Department of Breast Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032, China.
- Key Laboratory of Breast Cancer in Shanghai, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China.
| | - Yi-Zhou Jiang
- Department of Breast Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032, China.
- Key Laboratory of Breast Cancer in Shanghai, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China.
| | - Zhi-Ming Shao
- Department of Breast Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032, China.
- Key Laboratory of Breast Cancer in Shanghai, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China.
| |
Collapse
|
85
|
Murphy KJ, Reed DA, Trpceski M, Herrmann D, Timpson P. Quantifying and visualising the nuances of cellular dynamics in vivo using intravital imaging. Curr Opin Cell Biol 2021; 72:41-53. [PMID: 34091131 DOI: 10.1016/j.ceb.2021.04.007] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Revised: 04/23/2021] [Accepted: 04/28/2021] [Indexed: 12/14/2022]
Abstract
Intravital imaging is a powerful technology used to quantify and track dynamic changes in live cells and tissues within an intact environment. The ability to watch cell biology in real-time 'as it happens' has provided novel insight into tissue homeostasis, as well as disease initiation, progression and response to treatment. In this minireview, we highlight recent advances in the field of intravital microscopy, touching upon advances in awake versus anaesthesia-based approaches, as well as the integration of biosensors into intravital imaging. We also discuss current challenges that, in our opinion, need to be overcome to further advance the field of intravital imaging at the single-cell, subcellular and molecular resolution to reveal nuances of cell behaviour that can be targeted in complex disease settings.
Collapse
Affiliation(s)
- Kendelle J Murphy
- Garvan Institute of Medical Research & The Kinghorn Cancer Centre, Cancer Theme, Sydney, NSW, 2010, Australia; St Vincent's Clinical School, Faculty of Medicine, UNSW Sydney, Sydney, NSW, 2010, Australia
| | - Daniel A Reed
- Garvan Institute of Medical Research & The Kinghorn Cancer Centre, Cancer Theme, Sydney, NSW, 2010, Australia; St Vincent's Clinical School, Faculty of Medicine, UNSW Sydney, Sydney, NSW, 2010, Australia
| | - Michael Trpceski
- Garvan Institute of Medical Research & The Kinghorn Cancer Centre, Cancer Theme, Sydney, NSW, 2010, Australia
| | - David Herrmann
- Garvan Institute of Medical Research & The Kinghorn Cancer Centre, Cancer Theme, Sydney, NSW, 2010, Australia; St Vincent's Clinical School, Faculty of Medicine, UNSW Sydney, Sydney, NSW, 2010, Australia.
| | - Paul Timpson
- Garvan Institute of Medical Research & The Kinghorn Cancer Centre, Cancer Theme, Sydney, NSW, 2010, Australia; St Vincent's Clinical School, Faculty of Medicine, UNSW Sydney, Sydney, NSW, 2010, Australia.
| |
Collapse
|
86
|
Nagle MP, Tam GS, Maltz E, Hemminger Z, Wollman R. Bridging scales: From cell biology to physiology using in situ single-cell technologies. Cell Syst 2021; 12:388-400. [PMID: 34015260 DOI: 10.1016/j.cels.2021.03.002] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Revised: 01/30/2021] [Accepted: 03/08/2021] [Indexed: 12/14/2022]
Abstract
Biological organization crosses multiple spatial scales: from molecular, cellular, to tissues and organs. The proliferation of molecular profiling technologies enables increasingly detailed cataloging of the components at each scale. However, the scarcity of spatial profiling has made it challenging to bridge across these scales. Emerging technologies based on highly multiplexed in situ profiling are paving the way to study the spatial organization of cells and tissues in greater detail. These new technologies provide the data needed to cross the scale from cell biology to physiology and identify the fundamental principles that govern tissue organization. Here, we provide an overview of these key technologies and discuss the current and future insights these powerful techniques enable.
Collapse
Affiliation(s)
- Maeve P Nagle
- Department of Chemistry and Biochemistry, University of California, Los Angeles, Los Angeles, CA 90095, USA; Institute for Quantitative and Computational Biosciences, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Gabriela S Tam
- Department of Chemistry and Biochemistry, University of California, Los Angeles, Los Angeles, CA 90095, USA; Institute for Quantitative and Computational Biosciences, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Evan Maltz
- Department of Chemistry and Biochemistry, University of California, Los Angeles, Los Angeles, CA 90095, USA; Institute for Quantitative and Computational Biosciences, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Zachary Hemminger
- Department of Chemistry and Biochemistry, University of California, Los Angeles, Los Angeles, CA 90095, USA; Institute for Quantitative and Computational Biosciences, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Roy Wollman
- Department of Chemistry and Biochemistry, University of California, Los Angeles, Los Angeles, CA 90095, USA; Institute for Quantitative and Computational Biosciences, University of California, Los Angeles, Los Angeles, CA 90095, USA; Department of Integrative Biology and Physiology, University of California, Los Angeles, Los Angeles, CA 90095, USA.
| |
Collapse
|
87
|
Schoenit A, Cavalcanti-Adam EA, Göpfrich K. Functionalization of Cellular Membranes with DNA Nanotechnology. Trends Biotechnol 2021; 39:1208-1220. [PMID: 33722382 DOI: 10.1016/j.tibtech.2021.02.002] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Revised: 01/29/2021] [Accepted: 02/01/2021] [Indexed: 02/07/2023]
Abstract
Due to its versatility and programmability, DNA nanotechnology has greatly expanded the experimental toolbox for biomedical research. Recent advances allow reliable and efficient functionalization of cellular plasma membranes with a variety of synthetic DNA constructs, ranging from single strands to complex 3D DNA origami. The scope for applications, which probe biophysical parameters or equip cells with novel functions, is rapidly increasing. These applications extend from programmed cellular connectivity and tissue engineering to molecular force measurements, controlled receptor-ligand interactions, membrane-anchored biosensors, and artificial transmembrane structures. Here, we give guidance on different strategies to functionalize cellular membranes with DNA nanotechnology and summarize current trends employing membrane-anchored DNA as a tool in biophysics, cell biology, and synthetic biology.
Collapse
Affiliation(s)
- Andreas Schoenit
- Biophysical Engineering Group, Max Planck Institute for Medical Research, Jahnstraße 29, D-69120 Heidelberg, Germany; Department of Cellular Biophysics, Max Planck Institute for Medical Research, Jahnstraße 29, D-69120 Heidelberg, Germany
| | - Elisabetta Ada Cavalcanti-Adam
- Department of Cellular Biophysics, Max Planck Institute for Medical Research, Jahnstraße 29, D-69120 Heidelberg, Germany.
| | - Kerstin Göpfrich
- Biophysical Engineering Group, Max Planck Institute for Medical Research, Jahnstraße 29, D-69120 Heidelberg, Germany; Department of Physics and Astronomy, Heidelberg University, D-69120 Heidelberg, Germany.
| |
Collapse
|
88
|
Maniatis S, Petrescu J, Phatnani H. Spatially resolved transcriptomics and its applications in cancer. Curr Opin Genet Dev 2021; 66:70-77. [PMID: 33434721 PMCID: PMC7969406 DOI: 10.1016/j.gde.2020.12.002] [Citation(s) in RCA: 53] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Revised: 11/25/2020] [Accepted: 12/08/2020] [Indexed: 02/06/2023]
Abstract
Spatially resolved transcriptomics (SRT) offers the promise of understanding cells and their modes of dysfunction in the context of intact tissues. Technologies for SRT have advanced rapidly with a large number being published in recent years. Diverse methods for SRT produce data at widely varying depth, throughput, accessibility and cost. Many published SRT methods have been demonstrated only in their labs of origin, while others have matured to the point of commercialization and widespread availability. Here we review technologies for SRT, and their application in studies of tumor heterogeneity.
Collapse
Affiliation(s)
- Silas Maniatis
- Center for Genomics of Neurodegenerative Disease, New York Genome Center, New York, NY, USA
| | - Joana Petrescu
- Center for Genomics of Neurodegenerative Disease, New York Genome Center, New York, NY, USA; Department of Neurology, Division of Neuromuscular Medicine, Columbia University, New York, NY, USA
| | - Hemali Phatnani
- Center for Genomics of Neurodegenerative Disease, New York Genome Center, New York, NY, USA; Department of Neurology, Division of Neuromuscular Medicine, Columbia University, New York, NY, USA.
| |
Collapse
|
89
|
Acosta J, Ssozi D, van Galen P. Single-Cell RNA Sequencing to Disentangle the Blood System. Arterioscler Thromb Vasc Biol 2021; 41:1012-1018. [PMID: 33441024 PMCID: PMC7901535 DOI: 10.1161/atvbaha.120.314654] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The blood system is often represented as a tree-like structure with stem cells that give rise to mature blood cell types through a series of demarcated steps. Although this representation has served as a model of hierarchical tissue organization for decades, single-cell technologies are shedding new light on the abundance of cell type intermediates and the molecular mechanisms that ensure balanced replenishment of differentiated cells. In this Brief Review, we exemplify new insights into blood cell differentiation generated by single-cell RNA sequencing, summarize considerations for the application of this technology, and highlight innovations that are leading the way to understand hematopoiesis at the resolution of single cells. Graphic Abstract: A graphic abstract is available for this article.
Collapse
Affiliation(s)
- Jean Acosta
- Division of Hematology, Brigham and Women's Hospital, Boston, MA. Department of Medicine, Harvard Medical School, Boston, MA. Broad Institute of MIT and Harvard, Cambridge, MA
| | - Daniel Ssozi
- Division of Hematology, Brigham and Women's Hospital, Boston, MA. Department of Medicine, Harvard Medical School, Boston, MA. Broad Institute of MIT and Harvard, Cambridge, MA
| | - Peter van Galen
- Division of Hematology, Brigham and Women's Hospital, Boston, MA. Department of Medicine, Harvard Medical School, Boston, MA. Broad Institute of MIT and Harvard, Cambridge, MA
| |
Collapse
|
90
|
Fiorentino J, Torres-Padilla ME, Scialdone A. Measuring and Modeling Single-Cell Heterogeneity and Fate Decision in Mouse Embryos. Annu Rev Genet 2020; 54:167-187. [PMID: 32867543 DOI: 10.1146/annurev-genet-021920-110200] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Cellular heterogeneity is a property of any living system; however, its relationship with cellular fate decision remains an open question. Recent technological advances have enabled valuable insights, especially in complex systems such as the mouse embryo. In this review, we discuss recent studies that characterize cellular heterogeneity at different levels during mouse development, from the two-cell stage up to gastrulation. In addition to key experimental findings, we review mathematical modeling approaches that help researchers interpret these findings. Disentangling the role of heterogeneity in cell fate decision will likely rely on the refined integration of experiments, large-scale omics data, and mathematical modeling, complemented by the use of synthetic embryos and gastruloids as promising in vitro models.
Collapse
Affiliation(s)
- Jonathan Fiorentino
- Institute of Epigenetics and Stem Cells (IES), Helmholtz Zentrum München, D-81377 München, Germany; .,Institute of Functional Epigenetics (IFE) and Institute of Computational Biology (ICB), Helmholtz Zentrum München, D-85764 Neuherberg, Germany
| | - Maria-Elena Torres-Padilla
- Institute of Epigenetics and Stem Cells (IES), Helmholtz Zentrum München, D-81377 München, Germany; .,Faculty of Biology, Ludwig-Maximilians Universität, D-82152 Planegg-Martinsried, Germany
| | - Antonio Scialdone
- Institute of Epigenetics and Stem Cells (IES), Helmholtz Zentrum München, D-81377 München, Germany; .,Institute of Functional Epigenetics (IFE) and Institute of Computational Biology (ICB), Helmholtz Zentrum München, D-85764 Neuherberg, Germany
| |
Collapse
|
91
|
A zipcode for transcriptomes. Nat Rev Genet 2020; 21:508-509. [DOI: 10.1038/s41576-020-0266-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
|