1
|
Kulasinghe A, Berrell N, Donovan ML, Nilges BS. Spatial-Omics Methods and Applications. Methods Mol Biol 2025; 2880:101-146. [PMID: 39900756 DOI: 10.1007/978-1-0716-4276-4_5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2025]
Abstract
Traditional tissue profiling approaches have evolved from bulk studies to single-cell analysis over the last decade; however, the spatial context in tissues and microenvironments has always been lost. Over the last 5 years, spatial technologies have emerged that enabled researchers to investigate tissues in situ for proteins and transcripts without losing anatomy and histology. The field of spatial-omics enables highly multiplexed analysis of biomolecules like RNAs and proteins in their native spatial context-and has matured from initial proof-of-concept studies to a thriving field with widespread applications from basic research to translational and clinical studies. While there has been wide adoption of spatial technologies, there remain challenges with the standardization of methodologies, sample compatibility, throughput, resolution, and ease of use. In this chapter, we discuss the current state of the field and highlight technological advances and limitations.
Collapse
Affiliation(s)
- Arutha Kulasinghe
- Frazer Institute, Faculty of Medicine, University of Queensland, Brisbane, QLD, Australia
- Queensland Spatial Biology Centre, Wesley Research Institute, The Wesley Hospital, Auchenflower, QLD, Australia
| | - Naomi Berrell
- Queensland Spatial Biology Centre, Wesley Research Institute, The Wesley Hospital, Auchenflower, QLD, Australia
| | - Meg L Donovan
- Queensland Spatial Biology Centre, Wesley Research Institute, The Wesley Hospital, Auchenflower, QLD, Australia
| | | |
Collapse
|
2
|
Cipurko D, Ueda T, Mei L, Chevrier N. Repurposing large-format microarrays for scalable spatial transcriptomics. Nat Methods 2025; 22:145-155. [PMID: 39562752 DOI: 10.1038/s41592-024-02501-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Accepted: 10/08/2024] [Indexed: 11/21/2024]
Abstract
Spatiomolecular analyses are key to study tissue functions and malfunctions. However, we lack profiling tools for spatial transcriptomics that are easy to adopt, low cost and scalable in terms of sample size and number. Here, we describe a method, Array-seq, to repurpose classical oligonucleotide microarrays for spatial transcriptomics profiling. We generate Array-seq slides from microarrays carrying custom-design probes that contain common sequences flanking unique barcodes at known coordinates. Then we perform a simple, two-step reaction that produces mRNA capture probes across all spots on the microarray. We demonstrate that Array-seq yields spatial transcriptomes with high detection sensitivity and localization specificity using histological sections from mouse tissues as test systems. Moreover, we show that the large surface area of Array-seq slides yields spatial transcriptomes (i) at high throughput by profiling multi-organ sections, (ii) in three dimensions by processing serial sections from one sample, and (iii) across whole human organs. Thus, by combining classical DNA microarrays and next-generation sequencing, we have created a simple and flexible platform for spatiomolecular studies of small-to-large specimens at scale.
Collapse
Affiliation(s)
- Denis Cipurko
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL, USA
- Medical Scientist Training Program, University of Chicago, Chicago, IL, USA
| | - Tatsuki Ueda
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL, USA
| | - Linghan Mei
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL, USA
| | - Nicolas Chevrier
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL, USA.
| |
Collapse
|
3
|
An J, Lu Y, Chen Y, Chen Y, Zhou Z, Chen J, Peng C, Huang R, Peng F. Spatial transcriptomics in breast cancer: providing insight into tumor heterogeneity and promoting individualized therapy. Front Immunol 2024; 15:1499301. [PMID: 39749323 PMCID: PMC11693744 DOI: 10.3389/fimmu.2024.1499301] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2024] [Accepted: 12/05/2024] [Indexed: 01/04/2025] Open
Abstract
A comprehensive understanding of tumor heterogeneity, tumor microenvironment and the mechanisms of drug resistance is fundamental to advancing breast cancer research. While single-cell RNA sequencing has resolved the issue of "temporal dynamic expression" of genes at the single-cell level, the lack of spatial information still prevents us from gaining a comprehensive understanding of breast cancer. The introduction and application of spatial transcriptomics addresses this limitation. As the annual technical method of 2020, spatial transcriptomics preserves the spatial location of tissues and resolves RNA-seq data to help localize and differentiate the active expression of functional genes within a specific tissue region, enabling the study of spatial location attributes of gene locations and cellular tissue environments. In the context of breast cancer, spatial transcriptomics can assist in the identification of novel breast cancer subtypes and spatially discriminative features that show promise for individualized precise treatment. This article summarized the key technical approaches, recent advances in spatial transcriptomics and its applications in breast cancer, and discusses the limitations of current spatial transcriptomics methods and the prospects for future development, with a view to advancing the application of this technology in clinical practice.
Collapse
Affiliation(s)
- Junsha An
- West China School of Pharmacy, Sichuan University, Chengdu, China
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- Cardiovascular Department, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yajie Lu
- West China School of Pharmacy, Sichuan University, Chengdu, China
| | - Yuxi Chen
- West China School of Pharmacy, Sichuan University, Chengdu, China
| | - Yuling Chen
- West China School of Pharmacy, Sichuan University, Chengdu, China
| | - Zhaokai Zhou
- Department of Clinical Medicine, Zhengzhou University, Zhengzhou, China
| | - Jianping Chen
- School of Chinese Medicine, The University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Cheng Peng
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Ruizhen Huang
- Cardiovascular Department, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Fu Peng
- West China School of Pharmacy, Sichuan University, Chengdu, China
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, Sichuan University, Chengdu, China
| |
Collapse
|
4
|
Jia D, Cui M, Divsalar A, Khattab TA, Al-Qahtani SD, Cheung E, Ding X. Derivative Technologies of Expansion Microscopy and Applications in Biomedicine. Chembiochem 2024:e202400795. [PMID: 39681518 DOI: 10.1002/cbic.202400795] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2024] [Revised: 11/20/2024] [Accepted: 12/16/2024] [Indexed: 12/18/2024]
Abstract
Expansion microscopy (ExM) is an innovative super-resolution imaging technique that utilizes physical expansion to magnify biological samples, facilitating the visualization of cellular structures that are challenging to observe using traditional optical microscopes. The fundamental principle of ExM revolves around employing a specialized hydrogel to uniformly expand biological samples, thereby achieving super-resolution imaging under conventional optical imaging conditions. This technology finds application not only in various biological samples such as cells and tissue sections, but also enables super-resolution imaging of large biological molecules including proteins, nucleic acids, and metabolite molecules. In recent years, numerous researchers have delved into ExM, resulting in the continuous development of a range of derivative technologies that optimize experimental protocols and broaden practical application fields. This article presents a comprehensive review of these derivative technologies, highlighting the utilization of ExM for anchoring nucleic acids, proteins, and other biological molecules, as well as its applications in biomedicine. Furthermore, this review offers insights into the future development prospects of ExM technology.
Collapse
Affiliation(s)
- Dongling Jia
- School of Pharmacy, Shanghai University of Medicine & Health Sciences, Shanghai, 201318, China
| | - Minhui Cui
- School of Pharmacy, Shanghai University of Medicine & Health Sciences, Shanghai, 201318, China
| | - Adeleh Divsalar
- Department of Cell & Molecular Sciences, Faculty of Biological Sciences, Kharazmi University, Tehran, 15719-14911, Iran
| | - Tawfik A Khattab
- Dyeing, Printing and Auxiliaries Department, Textile Research and Technology Institute National Research Centre, Cairo, 12622, Egypt
| | - Salhah D Al-Qahtani
- Department of Chemistry, College of Science, Princess Nourah bint Abdulrahman University, P.O. Box 84428, Riyadh, 11671, Saudi Arabia
| | - Edwin Cheung
- Cancer Centre, Centre for Precision Medicine Research and Training, Faculty of Health Science, University of Macau Taipa, 999078, Macau, SAR
| | - Xianting Ding
- Institute for Personalized Medicine, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, 200030, China
| |
Collapse
|
5
|
Cao J, Zheng Z, Sun D, Chen X, Cheng R, Lv T, An Y, Zheng J, Song J, Wu L, Yang C. Decoder-seq enhances mRNA capture efficiency in spatial RNA sequencing. Nat Biotechnol 2024; 42:1735-1746. [PMID: 38228777 DOI: 10.1038/s41587-023-02086-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Accepted: 12/04/2023] [Indexed: 01/18/2024]
Abstract
Spatial transcriptomics technologies with high resolution often lack high sensitivity in mRNA detection. Here we report a dendrimeric DNA coordinate barcoding design for spatial RNA sequencing (Decoder-seq), which offers both high sensitivity and high resolution. Decoder-seq combines dendrimeric nanosubstrates with microfluidic coordinate barcoding to generate spatial arrays with a DNA density approximately ten times higher than previously reported methods while maintaining flexibility in resolution. We show that the high RNA capture efficiency of Decoder-seq improved the detection of lowly expressed olfactory receptor (Olfr) genes in mouse olfactory bulbs and contributed to the discovery of a unique layer enrichment pattern for two Olfr genes. The near-cellular resolution provided by Decoder-seq has enabled the construction of a spatial single-cell atlas of the mouse hippocampus, revealing dendrite-enriched mRNAs in neurons. When applying Decoder-seq to human renal cell carcinomas, we dissected the heterogeneous tumor microenvironment across different cancer subtypes and identified spatial gradient-expressed genes related to epithelial-mesenchymal transition with the potential to predict tumor prognosis and progression.
Collapse
Affiliation(s)
- Jiao Cao
- Institute of Molecular Medicine, Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine, Department of Urology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhong Zheng
- Institute of Molecular Medicine, Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine, Department of Urology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Di Sun
- Institute of Molecular Medicine, Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine, Department of Urology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xin Chen
- Institute of Molecular Medicine, Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine, Department of Urology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Rui Cheng
- Institute of Molecular Medicine, Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine, Department of Urology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Tianpeng Lv
- Institute of Molecular Medicine, Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine, Department of Urology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yu An
- Institute of Molecular Medicine, Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine, Department of Urology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Junhua Zheng
- Institute of Molecular Medicine, Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine, Department of Urology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Jia Song
- Institute of Molecular Medicine, Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine, Department of Urology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Lingling Wu
- Institute of Molecular Medicine, Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine, Department of Urology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Chaoyong Yang
- Institute of Molecular Medicine, Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine, Department of Urology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
- The MOE Key Laboratory of Spectrochemical Analysis & Instrumentation, State Key Laboratory of Physical Chemical of Solid Surfaces, Department of Chemical Biology, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen, China.
| |
Collapse
|
6
|
Wang S, Shin TW, Yoder HB, McMillan RB, Su H, Liu Y, Zhang C, Leung KS, Yin P, Kiessling LL, Boyden ES. Single-shot 20-fold expansion microscopy. Nat Methods 2024; 21:2128-2134. [PMID: 39394503 PMCID: PMC11541206 DOI: 10.1038/s41592-024-02454-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2023] [Accepted: 09/09/2024] [Indexed: 10/13/2024]
Abstract
Expansion microscopy (ExM) is in increasingly widespread use throughout biology because its isotropic physical magnification enables nanoimaging on conventional microscopes. To date, ExM methods either expand specimens to a limited range (~4-10× linearly) or achieve larger expansion factors through iterating the expansion process a second time (~15-20× linearly). Here, we present an ExM protocol that achieves ~20× expansion (yielding <20-nm resolution on a conventional microscope) in a single expansion step, achieving the performance of iterative expansion with the simplicity of a single-shot protocol. This protocol, which we call 20ExM, supports postexpansion staining for brain tissue, which can facilitate biomolecular labeling. 20ExM may find utility in many areas of biological investigation requiring high-resolution imaging.
Collapse
Affiliation(s)
- Shiwei Wang
- McGovern Institute for Brain Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Tay Won Shin
- McGovern Institute for Brain Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Media Arts and Sciences, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Harley B Yoder
- McGovern Institute for Brain Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Electrical Engineering and Computer Science, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Ryan B McMillan
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, USA
- Department of Systems Biology, Harvard Medical School, Boston, MA, USA
- Biophysics PhD Program, Harvard University, Cambridge, MA, USA
| | - Hanquan Su
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, USA
- Department of Systems Biology, Harvard Medical School, Boston, MA, USA
| | - Yixi Liu
- McGovern Institute for Brain Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Electrical Engineering and Computer Science, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Chi Zhang
- McGovern Institute for Brain Research, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Kylie S Leung
- McGovern Institute for Brain Research, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Peng Yin
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, USA
- Department of Systems Biology, Harvard Medical School, Boston, MA, USA
| | - Laura L Kiessling
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA, USA.
- Broad Institute of MIT and Harvard, Cambridge, MA, USA.
- Koch Institute, Massachusetts Institute of Technology, Cambridge, MA, USA.
| | - Edward S Boyden
- McGovern Institute for Brain Research, Massachusetts Institute of Technology, Cambridge, MA, USA.
- Media Arts and Sciences, Massachusetts Institute of Technology, Cambridge, MA, USA.
- Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA, USA.
- Koch Institute, Massachusetts Institute of Technology, Cambridge, MA, USA.
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA.
- Center for Neurobiological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA.
- K. Lisa Yang Center for Bionics, Massachusetts Institute of Technology, Cambridge, MA, USA.
- Howard Hughes Medical Institute, Cambridge, MA, USA.
| |
Collapse
|
7
|
Abstract
The ability to localize hundreds of macromolecules to discrete locations, structures and cell types in a tissue is a powerful approach to understand the cellular and spatial organization of an organ. Spatially resolved transcriptomic technologies enable mapping of transcripts at single-cell or near single-cell resolution in a multiplex manner. The rapid development of spatial transcriptomic technologies has accelerated the pace of discovery in several fields, including nephrology. Its application to preclinical models and human samples has provided spatial information about new cell types discovered by single-cell sequencing and new insights into the cell-cell interactions within neighbourhoods, and has improved our understanding of the changes that occur in response to injury. Integration of spatial transcriptomic technologies with other omics methods, such as proteomics and spatial epigenetics, will further facilitate the generation of comprehensive molecular atlases, and provide insights into the dynamic relationships of molecular components in homeostasis and disease. This Review provides an overview of current and emerging spatial transcriptomic methods, their applications and remaining challenges for the field.
Collapse
Affiliation(s)
- Sanjay Jain
- Division of Nephrology, Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA.
| | - Michael T Eadon
- Division of Nephrology, Department of Medicine, Indiana University School of Medicine, Indianapolis, IN, USA.
| |
Collapse
|
8
|
Mahmutovic Persson I, Bozovic G, Westergren-Thorsson G, Rolandsson Enes S. Spatial lung imaging in clinical and translational settings. Breathe (Sheff) 2024; 20:230224. [PMID: 39360023 PMCID: PMC11444490 DOI: 10.1183/20734735.0224-2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Accepted: 07/05/2024] [Indexed: 10/04/2024] Open
Abstract
For many severe lung diseases, non-invasive biomarkers from imaging could improve early detection of lung injury or disease onset, establish a diagnosis, or help follow-up disease progression and treatment strategies. Imaging of the thorax and lung is challenging due to its size, respiration movement, transferred cardiac pulsation, vast density range and gravitation sensitivity. However, there is extensive ongoing research in this fast-evolving field. Recent improvements in spatial imaging have allowed us to study the three-dimensional structure of the lung, providing both spatial architecture and transcriptomic information at single-cell resolution. This fast progression, however, comes with several challenges, including significant image file storage and network capacity issues, increased costs, data processing and analysis, the role of artificial intelligence and machine learning, and mechanisms to combine several modalities. In this review, we provide an overview of advances and current issues in the field of spatial lung imaging.
Collapse
Affiliation(s)
- Irma Mahmutovic Persson
- Lund University BioImaging Centre (LBIC), Faculty of Medicine, Lund University, Lund, Sweden
- Respiratory Immunopharmacology, Experimental Medical Science, Faculty of Medicine, Lund University, Lund, Sweden
| | - Gracijela Bozovic
- Department of Clinical Sciences, Radiology, Lund University, Lund, Sweden
- Department of Medical Imaging and Clinical Physiology, Skåne University Hospital, Lund, Sweden
| | - Gunilla Westergren-Thorsson
- Lund University BioImaging Centre (LBIC), Faculty of Medicine, Lund University, Lund, Sweden
- Lung Biology, Experimental Medical Science, Faculty of Medicine, Lund University, Lund, Sweden
| | - Sara Rolandsson Enes
- Lung Biology, Experimental Medical Science, Faculty of Medicine, Lund University, Lund, Sweden
| |
Collapse
|
9
|
Huang X, Zhao X, Li Y, Feng Y, Zhang G, Wang Q, Xu C. Combining Bulk and Single Cell RNA-Sequencing Data to Identify Hub Genes of Fibroblasts in Dilated Cardiomyopathy. J Inflamm Res 2024; 17:5375-5388. [PMID: 39161677 PMCID: PMC11330748 DOI: 10.2147/jir.s470860] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Accepted: 08/02/2024] [Indexed: 08/21/2024] Open
Abstract
Background Dilated cardiomyopathy (DCM) is the second leading cause of heart failure, with intricate pathophysiological underpinnings. In order to shed fresh light on the mechanistic research of DCM, we combined bulk RNA-seq and single-cell RNA-seq (scRNA-seq) data to examine significant cells and genes implicated in the disease. Methods This analysis employed publicly accessible bulk RNA-seq and scRNA-seq DCM datasets. The scRNA-seq data underwent normalization, principal component, and t-distribution stochastic neighbor embedding analysis. Cell-to-cell communication networks and activity analysis were conducted using CellChat. Utilizing enrichment analysis, the marker genes' role in the active cells was evaluated. After screening by limma software and weighted gene co-expression network analysis, the differentially expressed genes (DEGs) served as hub genes. Furthermore, these hub genes were subjected to immunological studies, transcription factor expression, and gene set enrichment. Lastly, the expression of the four hub genes and their connection to DCM were verified using the rat models. Results Fibroblasts and monocytes were chosen as hub cells from among the eight identified cell clusters; their marker genes intersected with DEGs to yield six hub genes. In addition, the six hub genes and the essential module genes intersected to yield four essential genes (ASPN, SFRP4, LUM, and FRZB) that were connected to the Wnt signaling pathway and highly expressed in fibroblast. The four hub DEGs had an expression pattern in the DCM rat model experiment results that was in line with the findings of the bioinformatics study. Additionally, there was a strong correlation between decreased cardiac function and the up-regulation of ASPN, SFRP4, LUM, and FRZB. Conclusion Ultimately, bulk RNA-seq and scRNA-seq data identified fibroblasts and monocytes as the main cell types implicated in DCM. The highly expressed genes ASPN, FRZB, LUM, and SFRP4 in fibroblasts may aid in the mechanistic investigation of DCM.
Collapse
Affiliation(s)
- Xiaoyan Huang
- Shaanxi Provincial Key Laboratory of Infection and Immune Diseases, Shaanxi Provincial People’s Hospital, Xi’an, People’s Republic of China
- Shaanxi Engineering Research Center of Cell Immunology, Shaanxi Provincial People’s Hospital, Xi’an, People’s Republic of China
| | - Xiangrong Zhao
- Shaanxi Provincial Key Laboratory of Infection and Immune Diseases, Shaanxi Provincial People’s Hospital, Xi’an, People’s Republic of China
- Shaanxi Engineering Research Center of Cell Immunology, Shaanxi Provincial People’s Hospital, Xi’an, People’s Republic of China
| | - Yaping Li
- Shaanxi Provincial Key Laboratory of Infection and Immune Diseases, Shaanxi Provincial People’s Hospital, Xi’an, People’s Republic of China
- Shaanxi Engineering Research Center of Cell Immunology, Shaanxi Provincial People’s Hospital, Xi’an, People’s Republic of China
| | - Yangmeng Feng
- Shaanxi Provincial Key Laboratory of Infection and Immune Diseases, Shaanxi Provincial People’s Hospital, Xi’an, People’s Republic of China
- Shaanxi Engineering Research Center of Cell Immunology, Shaanxi Provincial People’s Hospital, Xi’an, People’s Republic of China
| | - Guoan Zhang
- Department of Cardiovascular Surgery, Shaanxi Provincial People’s Hospital, Xi’an, People’s Republic of China
| | - Qiyu Wang
- Department of Graduate School, Yan’an University, Yan’an, People’s Republic of China
| | - Cuixiang Xu
- Shaanxi Provincial Key Laboratory of Infection and Immune Diseases, Shaanxi Provincial People’s Hospital, Xi’an, People’s Republic of China
- Shaanxi Engineering Research Center of Cell Immunology, Shaanxi Provincial People’s Hospital, Xi’an, People’s Republic of China
| |
Collapse
|
10
|
Fortner A, Bucur O. Multiplexed spatial transcriptomics methods and the application of expansion microscopy. Front Cell Dev Biol 2024; 12:1378875. [PMID: 39105173 PMCID: PMC11298486 DOI: 10.3389/fcell.2024.1378875] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Accepted: 06/10/2024] [Indexed: 08/07/2024] Open
Abstract
While spatial transcriptomics has undeniably revolutionized our ability to study cellular organization, it has driven the development of a great number of innovative transcriptomics methods, which can be classified into in situ sequencing (ISS) methods, in situ hybridization (ISH) techniques, and next-generation sequencing (NGS)-based sequencing with region capture. These technologies not only refine our understanding of cellular processes, but also open up new possibilities for breakthroughs in various research domains. One challenge of spatial transcriptomics experiments is the limitation of RNA detection due to optical crowding of RNA in the cells. Expansion microscopy (ExM), characterized by the controlled enlargement of biological specimens, offers a means to achieve super-resolution imaging, overcoming the diffraction limit inherent in conventional microscopy and enabling precise visualization of RNA in spatial transcriptomics methods. In this review, we elaborate on ISS, ISH and NGS-based spatial transcriptomic protocols and on how performance of these techniques can be extended by the combination of these protocols with ExM. Moving beyond the techniques and procedures, we highlight the broader implications of transcriptomics in biology and medicine. These include valuable insight into the spatial organization of gene expression in cells within tissues, aid in the identification and the distinction of cell types and subpopulations and understanding of molecular mechanisms and intercellular changes driving disease development.
Collapse
Affiliation(s)
- Andra Fortner
- Medical School, Ruprecht-Karls-Universität Heidelberg, Heidelberg, Germany
- Victor Babes National Institute of Pathology, Bucharest, Romania
| | - Octavian Bucur
- Victor Babes National Institute of Pathology, Bucharest, Romania
- Genomics Research and Development Institute, Bucharest, Romania
| |
Collapse
|
11
|
Valihrach L, Zucha D, Abaffy P, Kubista M. A practical guide to spatial transcriptomics. Mol Aspects Med 2024; 97:101276. [PMID: 38776574 DOI: 10.1016/j.mam.2024.101276] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Revised: 04/30/2024] [Accepted: 05/10/2024] [Indexed: 05/25/2024]
Abstract
Spatial transcriptomics is revolutionizing modern biology, offering researchers an unprecedented ability to unravel intricate gene expression patterns within tissues. From pioneering techniques to newly commercialized platforms, the field of spatial transcriptomics has evolved rapidly, ushering in a new era of understanding across various disciplines, from developmental biology to disease research. This dynamic expansion is reflected in the rapidly growing number of technologies and data analysis techniques developed and introduced. However, the expanding landscape presents a considerable challenge for researchers, especially newcomers to the field, as staying informed about these advancements becomes increasingly complex. To address this challenge, we have prepared an updated review with a particular focus on technologies that have reached commercialization and are, therefore, accessible to a broad spectrum of potential new users. In this review, we present the fundamental principles of spatial transcriptomic methods, discuss the challenges in data analysis, provide insights into experimental considerations, offer information about available resources for spatial transcriptomics, and conclude with a guide for method selection and a forward-looking perspective. Our aim is to serve as a guiding resource for both experienced users and newcomers navigating the complex realm of spatial transcriptomics in this era of rapid development. We intend to equip researchers with the necessary knowledge to make informed decisions and contribute to the cutting-edge research that spatial transcriptomics offers.
Collapse
Affiliation(s)
- Lukas Valihrach
- Laboratory of Gene Expression, Institute of Biotechnology of the Czech Academy of Sciences, Vestec, Czech Republic; Department of Cellular Neurophysiology, Institute of Experimental Medicine of the Czech Academy of Sciences, Prague, Czech Republic.
| | - Daniel Zucha
- Laboratory of Gene Expression, Institute of Biotechnology of the Czech Academy of Sciences, Vestec, Czech Republic; Department of Informatics and Chemistry, Faculty of Chemical Technology, University of Chemistry and Technology, Prague, Czech Republic
| | - Pavel Abaffy
- Laboratory of Gene Expression, Institute of Biotechnology of the Czech Academy of Sciences, Vestec, Czech Republic
| | - Mikael Kubista
- Laboratory of Gene Expression, Institute of Biotechnology of the Czech Academy of Sciences, Vestec, Czech Republic.
| |
Collapse
|
12
|
Shi W, Zhang J, Huang S, Fan Q, Cao J, Zeng J, Wu L, Yang C. Next-Generation Sequencing-Based Spatial Transcriptomics: A Perspective from Barcoding Chemistry. JACS AU 2024; 4:1723-1743. [PMID: 38818076 PMCID: PMC11134576 DOI: 10.1021/jacsau.4c00118] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Revised: 03/22/2024] [Accepted: 03/26/2024] [Indexed: 06/01/2024]
Abstract
Gene expression profiling of tissue cells with spatial context is in high demand to reveal cell types, locations, and intercellular or molecular interactions for physiological and pathological studies. With rapid advances in barcoding chemistry and sequencing chemistry, spatially resolved transcriptome (SRT) techniques have emerged to quantify spatial gene expression in tissue samples by correlating transcripts with their spatial locations using diverse strategies. These techniques provide both physical tissue structure and molecular characteristics and are poised to revolutionize many fields, such as developmental biology, neuroscience, oncology, and histopathology. In this context, this Perspective focuses on next-generation sequencing-based SRT methods, particularly highlighting spatial barcoding chemistry. It delves into optically manipulated spatial indexing methods and DNA array-barcoded spatial indexing methods by exploring current advances, challenges, and future development directions in this nascent field.
Collapse
Affiliation(s)
- Weixiong Shi
- Institute
of Molecular Medicine, Shanghai Key Laboratory for Nucleic Acid Chemistry
and Nanomedicine, Renji Hospital, Shanghai
Jiao Tong University School of Medicine, Shanghai 200127, China
- The
MOE Key Laboratory of Spectrochemical Analysis & Instrumentation,
Discipline of Intelligent Instrument and Equipment, Department of
Chemical Biology, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen 361005, China
| | - Jing Zhang
- State
Key Laboratory of Cellular Stress Biology, School of Life Sciences,
Faculty of Medicine and Life Sciences, Xiamen
University, Xiamen 361102, China
| | - Shanqing Huang
- The
MOE Key Laboratory of Spectrochemical Analysis & Instrumentation,
Discipline of Intelligent Instrument and Equipment, Department of
Chemical Biology, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen 361005, China
| | - Qian Fan
- Institute
of Molecular Medicine, Shanghai Key Laboratory for Nucleic Acid Chemistry
and Nanomedicine, Renji Hospital, Shanghai
Jiao Tong University School of Medicine, Shanghai 200127, China
| | - Jiao Cao
- Institute
of Molecular Medicine, Shanghai Key Laboratory for Nucleic Acid Chemistry
and Nanomedicine, Renji Hospital, Shanghai
Jiao Tong University School of Medicine, Shanghai 200127, China
| | - Jun Zeng
- Institute
of Molecular Medicine, Shanghai Key Laboratory for Nucleic Acid Chemistry
and Nanomedicine, Renji Hospital, Shanghai
Jiao Tong University School of Medicine, Shanghai 200127, China
| | - Lingling Wu
- Institute
of Molecular Medicine, Shanghai Key Laboratory for Nucleic Acid Chemistry
and Nanomedicine, Renji Hospital, Shanghai
Jiao Tong University School of Medicine, Shanghai 200127, China
| | - Chaoyong Yang
- Institute
of Molecular Medicine, Shanghai Key Laboratory for Nucleic Acid Chemistry
and Nanomedicine, Renji Hospital, Shanghai
Jiao Tong University School of Medicine, Shanghai 200127, China
- The
MOE Key Laboratory of Spectrochemical Analysis & Instrumentation,
Discipline of Intelligent Instrument and Equipment, Department of
Chemical Biology, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen 361005, China
- State
Key Laboratory of Cellular Stress Biology, School of Life Sciences,
Faculty of Medicine and Life Sciences, Xiamen
University, Xiamen 361102, China
| |
Collapse
|
13
|
Cao Y, Zhao X, Tang S, Jiang Q, Li S, Li S, Chen S. scButterfly: a versatile single-cell cross-modality translation method via dual-aligned variational autoencoders. Nat Commun 2024; 15:2973. [PMID: 38582890 PMCID: PMC10998864 DOI: 10.1038/s41467-024-47418-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2023] [Accepted: 03/28/2024] [Indexed: 04/08/2024] Open
Abstract
Recent advancements for simultaneously profiling multi-omics modalities within individual cells have enabled the interrogation of cellular heterogeneity and molecular hierarchy. However, technical limitations lead to highly noisy multi-modal data and substantial costs. Although computational methods have been proposed to translate single-cell data across modalities, broad applications of the methods still remain impeded by formidable challenges. Here, we propose scButterfly, a versatile single-cell cross-modality translation method based on dual-aligned variational autoencoders and data augmentation schemes. With comprehensive experiments on multiple datasets, we provide compelling evidence of scButterfly's superiority over baseline methods in preserving cellular heterogeneity while translating datasets of various contexts and in revealing cell type-specific biological insights. Besides, we demonstrate the extensive applications of scButterfly for integrative multi-omics analysis of single-modality data, data enhancement of poor-quality single-cell multi-omics, and automatic cell type annotation of scATAC-seq data. Moreover, scButterfly can be generalized to unpaired data training, perturbation-response analysis, and consecutive translation.
Collapse
Affiliation(s)
- Yichuan Cao
- School of Mathematical Sciences and LPMC, Nankai University, Tianjin, 300071, China
| | - Xiamiao Zhao
- School of Mathematical Sciences and LPMC, Nankai University, Tianjin, 300071, China
| | - Songming Tang
- School of Mathematical Sciences and LPMC, Nankai University, Tianjin, 300071, China
| | - Qun Jiang
- MOE Key Laboratory of Bioinformatics and Bioinformatics Division of BNRIST, Department of Automation, Tsinghua University, 100084, Beijing, China
| | - Sijie Li
- School of Mathematical Sciences and LPMC, Nankai University, Tianjin, 300071, China
| | - Siyu Li
- School of Statistics and Data Science, Nankai University, Tianjin, 300071, China
| | - Shengquan Chen
- School of Mathematical Sciences and LPMC, Nankai University, Tianjin, 300071, China.
| |
Collapse
|
14
|
Kiessling P, Kuppe C. Spatial multi-omics: novel tools to study the complexity of cardiovascular diseases. Genome Med 2024; 16:14. [PMID: 38238823 PMCID: PMC10795303 DOI: 10.1186/s13073-024-01282-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2023] [Accepted: 01/02/2024] [Indexed: 01/22/2024] Open
Abstract
Spatial multi-omic studies have emerged as a promising approach to comprehensively analyze cells in tissues, enabling the joint analysis of multiple data modalities like transcriptome, epigenome, proteome, and metabolome in parallel or even the same tissue section. This review focuses on the recent advancements in spatial multi-omics technologies, including novel data modalities and computational approaches. We discuss the advancements in low-resolution and high-resolution spatial multi-omics methods which can resolve up to 10,000 of individual molecules at subcellular level. By applying and integrating these techniques, researchers have recently gained valuable insights into the molecular circuits and mechanisms which govern cell biology along the cardiovascular disease spectrum. We provide an overview of current data analysis approaches, with a focus on data integration of multi-omic datasets, highlighting strengths and weaknesses of various computational pipelines. These tools play a crucial role in analyzing and interpreting spatial multi-omics datasets, facilitating the discovery of new findings, and enhancing translational cardiovascular research. Despite nontrivial challenges, such as the need for standardization of experimental setups, data analysis, and improved computational tools, the application of spatial multi-omics holds tremendous potential in revolutionizing our understanding of human disease processes and the identification of novel biomarkers and therapeutic targets. Exciting opportunities lie ahead for the spatial multi-omics field and will likely contribute to the advancement of personalized medicine for cardiovascular diseases.
Collapse
Affiliation(s)
- Paul Kiessling
- Department of Nephrology, Rheumatology, and Clinical Immunology, University Hospital RWTH Aachen, Aachen, Germany
| | - Christoph Kuppe
- Department of Nephrology, Rheumatology, and Clinical Immunology, University Hospital RWTH Aachen, Aachen, Germany.
| |
Collapse
|