101
|
Thuilliez C, Moquin-Beaudry G, Khneisser P, Marques Da Costa ME, Karkar S, Boudhouche H, Drubay D, Audinot B, Geoerger B, Scoazec JY, Gaspar N, Marchais A. CellsFromSpace: a fast, accurate, and reference-free tool to deconvolve and annotate spatially distributed omics data. BIOINFORMATICS ADVANCES 2024; 4:vbae081. [PMID: 38915885 PMCID: PMC11194756 DOI: 10.1093/bioadv/vbae081] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Revised: 05/02/2024] [Accepted: 05/29/2024] [Indexed: 06/26/2024]
Abstract
Motivation Spatial transcriptomics enables the analysis of cell crosstalk in healthy and diseased organs by capturing the transcriptomic profiles of millions of cells within their spatial contexts. However, spatial transcriptomics approaches also raise new computational challenges for the multidimensional data analysis associated with spatial coordinates. Results In this context, we introduce a novel analytical framework called CellsFromSpace based on independent component analysis (ICA), which allows users to analyze various commercially available technologies without relying on a single-cell reference dataset. The ICA approach deployed in CellsFromSpace decomposes spatial transcriptomics data into interpretable components associated with distinct cell types or activities. ICA also enables noise or artifact reduction and subset analysis of cell types of interest through component selection. We demonstrate the flexibility and performance of CellsFromSpace using real-world samples to demonstrate ICA's ability to successfully identify spatially distributed cells as well as rare diffuse cells, and quantitatively deconvolute datasets from the Visium, Slide-seq, MERSCOPE, and CosMX technologies. Comparative analysis with a current alternative reference-free deconvolution tool also highlights CellsFromSpace's speed, scalability and accuracy in processing complex, even multisample datasets. CellsFromSpace also offers a user-friendly graphical interface enabling non-bioinformaticians to annotate and interpret components based on spatial distribution and contributor genes, and perform full downstream analysis. Availability and implementation CellsFromSpace (CFS) is distributed as an R package available from github at https://github.com/gustaveroussy/CFS along with tutorials, examples, and detailed documentation.
Collapse
Affiliation(s)
- Corentin Thuilliez
- INSERM U1015, Gustave Roussy Cancer Campus, Université Paris-Saclay, Villejuif F-94805, France
| | - Gaël Moquin-Beaudry
- INSERM U1015, Gustave Roussy Cancer Campus, Université Paris-Saclay, Villejuif F-94805, France
| | - Pierre Khneisser
- Department of Medical Biology and Pathology, Gustave Roussy Cancer Campus, Villejuif 94805, France
| | - Maria Eugenia Marques Da Costa
- INSERM U1015, Gustave Roussy Cancer Campus, Université Paris-Saclay, Villejuif F-94805, France
- Department of Pediatric and Adolescent Oncology, Gustave Roussy Cancer Campus, Université Paris-Saclay, Villejuif 94805, France
| | - Slim Karkar
- University Bordeaux, CNRS, IBGC, UMR, Bordeaux 33077, France
- Bordeaux Bioinformatic Center CBiB, University of Bordeaux, Bordeaux 33000, France
| | - Hanane Boudhouche
- INSERM U1015, Gustave Roussy Cancer Campus, Université Paris-Saclay, Villejuif F-94805, France
| | - Damien Drubay
- Office of Biostatistics and Epidemiology, Gustave Roussy, Université Paris-Saclay, Villejuif 94805, France
- Inserm, Université Paris-Saclay, CESP U1018, Oncostat, Labeled Ligue Contre le Cancer, Villejuif 94805, France
| | - Baptiste Audinot
- INSERM U1015, Gustave Roussy Cancer Campus, Université Paris-Saclay, Villejuif F-94805, France
| | - Birgit Geoerger
- INSERM U1015, Gustave Roussy Cancer Campus, Université Paris-Saclay, Villejuif F-94805, France
- Department of Pediatric and Adolescent Oncology, Gustave Roussy Cancer Campus, Université Paris-Saclay, Villejuif 94805, France
| | - Jean-Yves Scoazec
- Department of Medical Biology and Pathology, Gustave Roussy Cancer Campus, Villejuif 94805, France
| | - Nathalie Gaspar
- INSERM U1015, Gustave Roussy Cancer Campus, Université Paris-Saclay, Villejuif F-94805, France
- Department of Pediatric and Adolescent Oncology, Gustave Roussy Cancer Campus, Université Paris-Saclay, Villejuif 94805, France
| | - Antonin Marchais
- INSERM U1015, Gustave Roussy Cancer Campus, Université Paris-Saclay, Villejuif F-94805, France
- Department of Pediatric and Adolescent Oncology, Gustave Roussy Cancer Campus, Université Paris-Saclay, Villejuif 94805, France
| |
Collapse
|
102
|
Chu LX, Wang WJ, Gu XP, Wu P, Gao C, Zhang Q, Wu J, Jiang DW, Huang JQ, Ying XW, Shen JM, Jiang Y, Luo LH, Xu JP, Ying YB, Chen HM, Fang A, Feng ZY, An SH, Li XK, Wang ZG. Spatiotemporal multi-omics: exploring molecular landscapes in aging and regenerative medicine. Mil Med Res 2024; 11:31. [PMID: 38797843 PMCID: PMC11129507 DOI: 10.1186/s40779-024-00537-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Accepted: 05/07/2024] [Indexed: 05/29/2024] Open
Abstract
Aging and regeneration represent complex biological phenomena that have long captivated the scientific community. To fully comprehend these processes, it is essential to investigate molecular dynamics through a lens that encompasses both spatial and temporal dimensions. Conventional omics methodologies, such as genomics and transcriptomics, have been instrumental in identifying critical molecular facets of aging and regeneration. However, these methods are somewhat limited, constrained by their spatial resolution and their lack of capacity to dynamically represent tissue alterations. The advent of emerging spatiotemporal multi-omics approaches, encompassing transcriptomics, proteomics, metabolomics, and epigenomics, furnishes comprehensive insights into these intricate molecular dynamics. These sophisticated techniques facilitate accurate delineation of molecular patterns across an array of cells, tissues, and organs, thereby offering an in-depth understanding of the fundamental mechanisms at play. This review meticulously examines the significance of spatiotemporal multi-omics in the realms of aging and regeneration research. It underscores how these methodologies augment our comprehension of molecular dynamics, cellular interactions, and signaling pathways. Initially, the review delineates the foundational principles underpinning these methods, followed by an evaluation of their recent applications within the field. The review ultimately concludes by addressing the prevailing challenges and projecting future advancements in the field. Indubitably, spatiotemporal multi-omics are instrumental in deciphering the complexities inherent in aging and regeneration, thus charting a course toward potential therapeutic innovations.
Collapse
Affiliation(s)
- Liu-Xi Chu
- Affiliated Cixi Hospital, Wenzhou Medical University, Ningbo, 315300, Zhejiang, China
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China
- National Key Laboratory of Macromolecular Drug Development and Manufacturing, School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China
| | - Wen-Jia Wang
- State Key Laboratory of Bioelectronics, School of Biological Science & Medical Engineering, Southeast University, Nanjing, 210096, China
| | - Xin-Pei Gu
- School of Pharmaceutical Sciences, Guangdong Provincial Key Laboratory of New Drug Screening, Southern Medical University, Guangzhou, 510515, China
- Department of Human Anatomy, Shandong First Medical University & Shandong Academy of Medical Sciences, Taian, 271000, Shandong, China
| | - Ping Wu
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China
- National Key Laboratory of Macromolecular Drug Development and Manufacturing, School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China
| | - Chen Gao
- State Key Laboratory of Bioelectronics, School of Biological Science & Medical Engineering, Southeast University, Nanjing, 210096, China
| | - Quan Zhang
- Integrative Muscle Biology Laboratory, Division of Regenerative and Rehabilitative Sciences, University of Tennessee Health Science Center, Memphis, TN, 38163, United States
| | - Jia Wu
- Key Laboratory for Laboratory Medicine, Ministry of Education, Zhejiang Provincial Key Laboratory of Medical Genetics, School of Laboratory Medicine and Life Science, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China
| | - Da-Wei Jiang
- Affiliated Cixi Hospital, Wenzhou Medical University, Ningbo, 315300, Zhejiang, China
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China
- National Key Laboratory of Macromolecular Drug Development and Manufacturing, School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China
| | - Jun-Qing Huang
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China
- National Key Laboratory of Macromolecular Drug Development and Manufacturing, School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China
- Key Laboratory of Imaging Diagnosis and Minimally Invasive Intervention Research, Institute of Imaging Diagnosis and Minimally Invasive Intervention Research, the Fifth Affiliated Hospital of Wenzhou Medical University, Lishui Hospital of Zhejiang University, Lishui, 323000, Zhejiang, China
| | - Xin-Wang Ying
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China
- National Key Laboratory of Macromolecular Drug Development and Manufacturing, School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China
| | - Jia-Men Shen
- National Key Laboratory of Macromolecular Drug Development and Manufacturing, School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China
| | - Yi Jiang
- National Key Laboratory of Macromolecular Drug Development and Manufacturing, School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China
| | - Li-Hua Luo
- School and Hospital of Stomatology, Wenzhou Medical University, Wenzhou, 324025, Zhejiang, China
| | - Jun-Peng Xu
- Affiliated Cixi Hospital, Wenzhou Medical University, Ningbo, 315300, Zhejiang, China
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China
- National Key Laboratory of Macromolecular Drug Development and Manufacturing, School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China
| | - Yi-Bo Ying
- National Key Laboratory of Macromolecular Drug Development and Manufacturing, School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China
| | - Hao-Man Chen
- National Key Laboratory of Macromolecular Drug Development and Manufacturing, School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China
| | - Ao Fang
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China
- National Key Laboratory of Macromolecular Drug Development and Manufacturing, School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China
| | - Zun-Yong Feng
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China.
- National Key Laboratory of Macromolecular Drug Development and Manufacturing, School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China.
- Departments of Diagnostic Radiology, Surgery, Chemical and Biomolecular Engineering, and Biomedical Engineering, Yong Loo Lin School of Medicine and College of Design and Engineering, National University of Singapore, Singapore, 119074, Singapore.
- Clinical Imaging Research Centre, Centre for Translational Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117599, Singapore.
- Nanomedicine Translational Research Program, NUS Center for Nanomedicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117597, Singapore.
- Institute of Molecular and Cell Biology, Agency for Science, Technology, and Research (A*STAR), Singapore, 138673, Singapore.
| | - Shu-Hong An
- Department of Human Anatomy, Shandong First Medical University & Shandong Academy of Medical Sciences, Taian, 271000, Shandong, China.
| | - Xiao-Kun Li
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China.
- National Key Laboratory of Macromolecular Drug Development and Manufacturing, School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China.
| | - Zhou-Guang Wang
- Affiliated Cixi Hospital, Wenzhou Medical University, Ningbo, 315300, Zhejiang, China.
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China.
- National Key Laboratory of Macromolecular Drug Development and Manufacturing, School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China.
- Key Laboratory of Imaging Diagnosis and Minimally Invasive Intervention Research, Institute of Imaging Diagnosis and Minimally Invasive Intervention Research, the Fifth Affiliated Hospital of Wenzhou Medical University, Lishui Hospital of Zhejiang University, Lishui, 323000, Zhejiang, China.
| |
Collapse
|
103
|
Bitto V, Hönscheid P, Besso MJ, Sperling C, Kurth I, Baumann M, Brors B. Enhancing mass spectrometry imaging accessibility using convolutional autoencoders for deriving hypoxia-associated peptides from tumors. NPJ Syst Biol Appl 2024; 10:57. [PMID: 38802379 PMCID: PMC11130291 DOI: 10.1038/s41540-024-00385-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Accepted: 05/13/2024] [Indexed: 05/29/2024] Open
Abstract
Mass spectrometry imaging (MSI) allows to study cancer's intratumoral heterogeneity through spatially-resolved peptides, metabolites and lipids. Yet, in biomedical research MSI is rarely used for biomarker discovery. Besides its high dimensionality and multicollinearity, mass spectrometry (MS) technologies typically output mass-to-charge ratio values but not the biochemical compounds of interest. Our framework makes particularly low-abundant signals in MSI more accessible. We utilized convolutional autoencoders to aggregate features associated with tumor hypoxia, a parameter with significant spatial heterogeneity, in cancer xenograft models. We highlight that MSI captures these low-abundant signals and that autoencoders can preserve them in their latent space. The relevance of individual hyperparameters is demonstrated through ablation experiments, and the contribution from original features to latent features is unraveled. Complementing MSI with tandem MS from the same tumor model, multiple hypoxia-associated peptide candidates were derived. Compared to random forests alone, our autoencoder approach yielded more biologically relevant insights for biomarker discovery.
Collapse
Affiliation(s)
- Verena Bitto
- Division of Applied Bioinformatics, German Cancer Research Center (DKFZ), Heidelberg, Germany.
- Division of Radiooncology/Radiobiology, German Cancer Research Center (DKFZ), Heidelberg, Germany.
- HIDSS4Health - Helmholtz Information and Data Science School for Health, Karlsruhe/Heidelberg, Heidelberg, Germany.
- Faculty for Mathematics and Computer Science, Heidelberg University, Heidelberg, Germany.
| | - Pia Hönscheid
- National Center for Tumor Diseases (NCT), Partner Site Dresden, German Cancer Research Center (DKFZ), Heidelberg, Germany
- University Hospital Carl Gustav Carus (UKD), Technische Universität Dresden, Institute of Pathology, Dresden, Germany
- Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - María José Besso
- Division of Radiooncology/Radiobiology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Christian Sperling
- National Center for Tumor Diseases (NCT), Partner Site Dresden, German Cancer Research Center (DKFZ), Heidelberg, Germany
- University Hospital Carl Gustav Carus (UKD), Technische Universität Dresden, Institute of Pathology, Dresden, Germany
- Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Ina Kurth
- Division of Radiooncology/Radiobiology, German Cancer Research Center (DKFZ), Heidelberg, Germany
- OncoRay - National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Helmholtz-Zentrum Dresden - Rossendorf, Dresden, Germany
- German Cancer Consortium (DKTK), Core Center Heidelberg, Heidelberg, Germany
| | - Michael Baumann
- Division of Radiooncology/Radiobiology, German Cancer Research Center (DKFZ), Heidelberg, Germany
- OncoRay - National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Helmholtz-Zentrum Dresden - Rossendorf, Dresden, Germany
- German Cancer Consortium (DKTK), Core Center Heidelberg, Heidelberg, Germany
| | - Benedikt Brors
- Division of Applied Bioinformatics, German Cancer Research Center (DKFZ), Heidelberg, Germany
- German Cancer Consortium (DKTK), Core Center Heidelberg, Heidelberg, Germany
- National Center for Tumor Diseases (NCT), Heidelberg, Germany
- Medical Faculty Heidelberg and Faculty of Biosciences, Heidelberg University, Heidelberg, Germany
| |
Collapse
|
104
|
Moreno J, Gluud LL, Galsgaard ED, Hvid H, Mazzoni G, Das V. Identification of ligand and receptor interactions in CKD and MASH through the integration of single cell and spatial transcriptomics. PLoS One 2024; 19:e0302853. [PMID: 38768139 PMCID: PMC11104622 DOI: 10.1371/journal.pone.0302853] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Accepted: 04/10/2024] [Indexed: 05/22/2024] Open
Abstract
BACKGROUND Chronic Kidney Disease (CKD) and Metabolic dysfunction-associated steatohepatitis (MASH) are metabolic fibroinflammatory diseases. Combining single-cell (scRNAseq) and spatial transcriptomics (ST) could give unprecedented molecular disease understanding at single-cell resolution. A more comprehensive analysis of the cell-specific ligand-receptor (L-R) interactions could provide pivotal information about signaling pathways in CKD and MASH. To achieve this, we created an integrative analysis framework in CKD and MASH from two available human cohorts. RESULTS The analytical framework identified L-R pairs involved in cellular crosstalk in CKD and MASH. Interactions between cell types identified using scRNAseq data were validated by checking the spatial co-presence using the ST data and the co-expression of the communicating targets. Multiple L-R protein pairs identified are known key players in CKD and MASH, while others are novel potential targets previously observed only in animal models. CONCLUSION Our study highlights the importance of integrating different modalities of transcriptomic data for a better understanding of the molecular mechanisms. The combination of single-cell resolution from scRNAseq data, combined with tissue slide investigations and visualization of cell-cell interactions obtained through ST, paves the way for the identification of future potential therapeutic targets and developing effective therapies.
Collapse
Affiliation(s)
- Jaime Moreno
- Digital Science and Innovation, Computational Biology – AI & Digital Research, Novo Nordisk A/S, Maløv, Denmark
| | - Lise Lotte Gluud
- Gastro Unit, Copenhagen University Hospital Hvidovre, Hvidovre, Denmark
- Dept of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | | | - Henning Hvid
- Global Drug Discovery, Novo Nordisk A/S, Maløv, Denmark
| | - Gianluca Mazzoni
- Digital Science and Innovation, Computational Biology – AI & Digital Research, Novo Nordisk A/S, Maløv, Denmark
| | - Vivek Das
- Digital Science and Innovation, Computational Biology – AI & Digital Research, Novo Nordisk A/S, Maløv, Denmark
| |
Collapse
|
105
|
Zhu B, Gao S, Chen S, Yeung J, Bai Y, Huang AY, Yeo YY, Liao G, Mao S, Jiang ZG, Rodig SJ, Shalek AK, Nolan GP, Jiang S, Ma Z. Cross-domain information fusion for enhanced cell population delineation in single-cell spatial-omics data. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.12.593710. [PMID: 38798592 PMCID: PMC11118457 DOI: 10.1101/2024.05.12.593710] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/29/2024]
Abstract
Cell population delineation and identification is an essential step in single-cell and spatial-omics studies. Spatial-omics technologies can simultaneously measure information from three complementary domains related to this task: expression levels of a panel of molecular biomarkers at single-cell resolution, relative positions of cells, and images of tissue sections, but existing computational methods for performing this task on single-cell spatial-omics datasets often relinquish information from one or more domains. The additional reliance on the availability of "atlas" training or reference datasets limits cell type discovery to well-defined but limited cell population labels, thus posing major challenges for using these methods in practice. Successful integration of all three domains presents an opportunity for uncovering cell populations that are functionally stratified by their spatial contexts at cellular and tissue levels: the key motivation for employing spatial-omics technologies in the first place. In this work, we introduce Cell Spatio- and Neighborhood-informed Annotation and Patterning (CellSNAP), a self-supervised computational method that learns a representation vector for each cell in tissue samples measured by spatial-omics technologies at the single-cell or finer resolution. The learned representation vector fuses information about the corresponding cell across all three aforementioned domains. By applying CellSNAP to datasets spanning both spatial proteomic and spatial transcriptomic modalities, and across different tissue types and disease settings, we show that CellSNAP markedly enhances de novo discovery of biologically relevant cell populations at fine granularity, beyond current approaches, by fully integrating cells' molecular profiles with cellular neighborhood and tissue image information.
Collapse
Affiliation(s)
- Bokai Zhu
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Sheng Gao
- Department of Statistics and Data Science, The Wharton School, University of Pennsylvania, PA, United States
| | - Shuxiao Chen
- Department of Statistics and Data Science, The Wharton School, University of Pennsylvania, PA, United States
| | - Jason Yeung
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Yunhao Bai
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Amy Y Huang
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Massachusetts Institute of Technology, Cambridge, MA, USA
- Dana-Farber Cancer Institute, Boston, MA, USA
| | - Yao Yu Yeo
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Guanrui Liao
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
- Center of Hepato-Pancreato-Biliary Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, People's Republic of China
| | - Shulin Mao
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
- Division of Genetics and Genomics, Boston Children's Hospital, Harvard Medical School, Boston, MA, United States
- Program in Biological and Biomedical Sciences, Harvard Medical School, Boston, MA, USA
| | - Zhenghui G Jiang
- Division of Gastroenterology/Liver Center, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Scott J Rodig
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, United States
| | - Alex K Shalek
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA, USA
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Garry P Nolan
- Department of Pathology, Stanford University, Stanford, CA, United States
| | - Sizun Jiang
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Zongming Ma
- Department of Statistics and Data Science, Yale University, New Haven, CT, United States
| |
Collapse
|
106
|
Chen Y, Sun H, Luo Z, Mei Y, Xu Z, Tan J, Xie Y, Li M, Xia J, Yang B, Su B. Crosstalk between CD8 + T cells and mesenchymal stromal cells in intestine homeostasis and immunity. Adv Immunol 2024; 162:23-58. [PMID: 38866438 DOI: 10.1016/bs.ai.2024.02.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/14/2024]
Abstract
The intestine represents the most complex cellular network in the whole body. It is constantly faced with multiple types of immunostimulatory agents encompassing from food antigen, gut microbiome, metabolic waste products, and dead cell debris. Within the intestine, most T cells are found in three primary compartments: the organized gut-associated lymphoid tissue, the lamina propria, and the epithelium. The well-orchestrated epithelial-immune-microbial interaction is critically important for the precise immune response. The main role of intestinal mesenchymal stromal cells is to support a structural framework within the gut wall. However, recent evidence from stromal cell studies indicates that they also possess significant immunomodulatory functions, such as maintaining intestinal tolerance via the expression of PDL1/2 and MHC-II molecules, and promoting the development of CD103+ dendritic cells, and IgA+ plasma cells, thereby enhancing intestinal homeostasis. In this review, we will summarize the current understanding of CD8+ T cells and stromal cells alongside the intestinal tract and discuss the reciprocal interactions between T subsets and mesenchymal stromal cell populations. We will focus on how the tissue residency, migration, and function of CD8+ T cells could be potentially regulated by mesenchymal stromal cell populations and explore the molecular mediators, such as TGF-β, IL-33, and MHC-II molecules that might influence these processes. Finally, we discuss the potential pathophysiological impact of such interaction in intestine hemostasis as well as diseases of inflammation, infection, and malignancies.
Collapse
Affiliation(s)
- Yao Chen
- Shanghai Institute of Immunology, Department of Immunology and Microbiology, The Ministry of Education Key Laboratory of Cell Death and Differentiation, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Hongxiang Sun
- Shanghai Institute of Immunology, Department of Immunology and Microbiology, The Ministry of Education Key Laboratory of Cell Death and Differentiation, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhengnan Luo
- Shanghai Institute of Immunology, Department of Immunology and Microbiology, The Ministry of Education Key Laboratory of Cell Death and Differentiation, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yisong Mei
- Shanghai Institute of Immunology, Department of Immunology and Microbiology, The Ministry of Education Key Laboratory of Cell Death and Differentiation, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ziyang Xu
- Shanghai Institute of Immunology, Department of Immunology and Microbiology, The Ministry of Education Key Laboratory of Cell Death and Differentiation, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jianmei Tan
- Shanghai Institute of Immunology, Department of Immunology and Microbiology, The Ministry of Education Key Laboratory of Cell Death and Differentiation, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yiting Xie
- Shanghai Institute of Immunology, Department of Immunology and Microbiology, The Ministry of Education Key Laboratory of Cell Death and Differentiation, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Mengda Li
- Shanghai Institute of Immunology, Department of Immunology and Microbiology, The Ministry of Education Key Laboratory of Cell Death and Differentiation, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jiaqi Xia
- Shanghai Institute of Immunology, Department of Immunology and Microbiology, The Ministry of Education Key Laboratory of Cell Death and Differentiation, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Beichun Yang
- Shanghai Institute of Immunology, Department of Immunology and Microbiology, The Ministry of Education Key Laboratory of Cell Death and Differentiation, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Bing Su
- Shanghai Institute of Immunology, Department of Immunology and Microbiology, The Ministry of Education Key Laboratory of Cell Death and Differentiation, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Center for Immune-Related Diseases at Shanghai Institute of Immunology, Department of Gastroenterology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Shanghai Jiao Tong University School of Medicine-Yale Institute for Immune Metabolism, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Key Laboratory of Molecular Radiation Oncology of Hunan Province, Xiangya Hospital, Central South University, Changsha, China.
| |
Collapse
|
107
|
Li F, Xiang R, Liu Y, Hu G, Jiang Q, Jia T. Approaches and challenges in identifying, quantifying, and manipulating dynamic mitochondrial genome variations. Cell Signal 2024; 117:111123. [PMID: 38417637 DOI: 10.1016/j.cellsig.2024.111123] [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: 12/07/2023] [Revised: 02/14/2024] [Accepted: 02/25/2024] [Indexed: 03/01/2024]
Abstract
Mitochondria, the cellular powerhouses, possess their own unique genetic system, including replication, transcription, and translation. Studying these processes is crucial for comprehending mitochondrial disorders, energy production, and their related diseases. Over the past decades, various approaches have been applied in detecting and quantifying mitochondrial genome variations with also the purpose of manipulation of mitochondria or mitochondrial genome for therapeutics. Understanding the scope and limitations of above strategies is not only fundamental to the understanding of basic biology but also critical for exploring disease-related novel target(s), as well to develop innovative therapies. Here, this review provides an overview of different tools and techniques for accurate mitochondrial genome variations identification, quantification, and discuss novel strategies for the manipulation of mitochondria to develop innovative therapeutic interventions, through combining the insights gained from the study of mitochondrial genetics with ongoing single cell omics combined with advanced single molecular tools.
Collapse
Affiliation(s)
- Fei Li
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Run Xiang
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, China; Department of Thoracic Surgery, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu, China
| | - Yue Liu
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Guoliang Hu
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, China; Department of Thoracic Surgery, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu, China
| | - Quanbo Jiang
- Light, Nanomaterials, Nanotechnologies (L2n) Laboratory, CNRS EMR 7004, University of Technology of Troyes, 12 rue Marie Curie, 10004 Troyes, France
| | - Tao Jia
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, China; CNRS-UMR9187, INSERM U1196, PSL-Research University, 91405 Orsay, France; CNRS-UMR9187, INSERM U1196, Université Paris Saclay, 91405 Orsay, France.
| |
Collapse
|
108
|
Chen Y, Wu Y, Yan G, Zhang G. Tertiary lymphoid structures in cancer: maturation and induction. Front Immunol 2024; 15:1369626. [PMID: 38690273 PMCID: PMC11058640 DOI: 10.3389/fimmu.2024.1369626] [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: 01/17/2024] [Accepted: 04/05/2024] [Indexed: 05/02/2024] Open
Abstract
Tertiary lymphoid structure (TLS) is an ectopic lymphocyte aggregate formed in peripheral non-lymphoid tissues, including inflamed or cancerous tissue. Tumor-associated TLS serves as a prominent center of antigen presentation and adaptive immune activation within the periphery, which has exhibited positive prognostic value in various cancers. In recent years, the concept of maturity regarding TLS has been proposed and mature TLS, characterized by well-developed germinal centers, exhibits a more potent tumor-suppressive capacity with stronger significance. Meanwhile, more and more evidence showed that TLS can be induced by therapeutic interventions during cancer treatments. Thus, the evaluation of TLS maturity and the therapeutic interventions that induce its formation are critical issues in current TLS research. In this review, we aim to provide a comprehensive summary of the existing classifications for TLS maturity and therapeutic strategies capable of inducing its formation in tumors.
Collapse
Affiliation(s)
- Yulu Chen
- Department of Phototherapy, Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai, China
- Skin Cancer Center, Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai, China
- Institute of Photomedicine, School of Medicine, Tongji University, Shanghai, China
| | - Yuhao Wu
- Department of Phototherapy, Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai, China
- Skin Cancer Center, Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai, China
- Institute of Photomedicine, School of Medicine, Tongji University, Shanghai, China
| | - Guorong Yan
- Department of Phototherapy, Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai, China
- Skin Cancer Center, Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai, China
- Institute of Photomedicine, School of Medicine, Tongji University, Shanghai, China
| | - Guolong Zhang
- Department of Phototherapy, Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai, China
- Skin Cancer Center, Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai, China
- Institute of Photomedicine, School of Medicine, Tongji University, Shanghai, China
| |
Collapse
|
109
|
Wang L, Li M, Hwang TH. The 3D Revolution in Cancer Discovery. Cancer Discov 2024; 14:625-629. [PMID: 38571426 DOI: 10.1158/2159-8290.cd-23-1499] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/05/2024]
Abstract
SUMMARY The transition from 2D to 3D spatial profiling marks a revolutionary era in cancer research, offering unprecedented potential to enhance cancer diagnosis and treatment. This commentary outlines the experimental and computational advancements and challenges in 3D spatial molecular profiling, underscoring the innovation needed in imaging tools, software, artificial intelligence, and machine learning to overcome implementation hurdles and harness the full potential of 3D analysis in the field.
Collapse
Affiliation(s)
- Linghua Wang
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas
- UTHealth Houston Graduate School of Biomedical Sciences, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Mingyao Li
- Statistical Center for Single-Cell and Spatial Genomics, Department of Biostatistics, Epidemiology and Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Tae Hyun Hwang
- Department of Artificial Intelligence and Informatics, Mayo Clinic, Jacksonville, Florida
| |
Collapse
|
110
|
Shugar AL, Konger RL, Rohan CA, Travers JB, Kim YL. Mapping cutaneous field carcinogenesis of nonmelanoma skin cancer using mesoscopic imaging of pro-inflammation cues. Exp Dermatol 2024; 33:e15076. [PMID: 38610095 PMCID: PMC11034840 DOI: 10.1111/exd.15076] [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: 12/03/2023] [Revised: 03/24/2024] [Accepted: 03/30/2024] [Indexed: 04/14/2024]
Abstract
Nonmelanoma skin cancers remain the most widely diagnosed types of cancers globally. Thus, for optimal patient management, it has become imperative that we focus our efforts on the detection and monitoring of cutaneous field carcinogenesis. The concept of field cancerization (or field carcinogenesis), introduced by Slaughter in 1953 in the context of oral cancer, suggests that invasive cancer may emerge from a molecularly and genetically altered field affecting a substantial area of underlying tissue including the skin. A carcinogenic field alteration, present in precancerous tissue over a relatively large area, is not easily detected by routine visualization. Conventional dermoscopy and microscopy imaging are often limited in assessing the entire carcinogenic landscape. Recent efforts have suggested the use of noninvasive mesoscopic (between microscopic and macroscopic) optical imaging methods that can detect chronic inflammatory features to identify pre-cancerous and cancerous angiogenic changes in tissue microenvironments. This concise review covers major types of mesoscopic optical imaging modalities capable of assessing pro-inflammatory cues by quantifying blood haemoglobin parameters and hemodynamics. Importantly, these imaging modalities demonstrate the ability to detect angiogenesis and inflammation associated with actinically damaged skin. Representative experimental preclinical and human clinical studies using these imaging methods provide biological and clinical relevance to cutaneous field carcinogenesis in altered tissue microenvironments in the apparently normal epidermis and dermis. Overall, mesoscopic optical imaging modalities assessing chronic inflammatory hyperemia can enhance the understanding of cutaneous field carcinogenesis, offer a window of intervention and monitoring for actinic keratoses and nonmelanoma skin cancers and maximise currently available treatment options.
Collapse
Affiliation(s)
- Andrea L. Shugar
- Department of Pharmacology & Toxicology, Wright State University Boonshoft School of Medicine, Dayton, Ohio, USA
| | - Raymond L. Konger
- Department of Pathology and Laboratory Medicine, Indiana University School of Medicine, Indianapolis, Indiana, USA
- Department of Dermatology, Indiana University School of Medicine, Indianapolis, Indiana, USA
- Department of Pathology, Richard L. Roudebush Veterans Administration Hospital, Indianapolis, Indiana, USA
| | - Craig A. Rohan
- Department of Pharmacology & Toxicology, Wright State University Boonshoft School of Medicine, Dayton, Ohio, USA
- Department of Dermatology, Wright State University Boonshoft School of Medicine, Dayton, Ohio, USA
- Department of Medicine, Dayton Veterans Affairs Medical Center, Dayton, Ohio, USA
| | - Jeffrey B. Travers
- Department of Pharmacology & Toxicology, Wright State University Boonshoft School of Medicine, Dayton, Ohio, USA
- Department of Dermatology, Wright State University Boonshoft School of Medicine, Dayton, Ohio, USA
- Department of Medicine, Dayton Veterans Affairs Medical Center, Dayton, Ohio, USA
| | - Young L. Kim
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, Indiana, USA
- Purdue Institute for Cancer Research, Purdue University, West Lafayette, Indiana, USA
| |
Collapse
|
111
|
Arulraj T, Wang H, Ippolito A, Zhang S, Fertig EJ, Popel AS. Leveraging multi-omics data to empower quantitative systems pharmacology in immuno-oncology. Brief Bioinform 2024; 25:bbae131. [PMID: 38557676 PMCID: PMC10982948 DOI: 10.1093/bib/bbae131] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Revised: 02/20/2024] [Accepted: 03/08/2024] [Indexed: 04/04/2024] Open
Abstract
Understanding the intricate interactions of cancer cells with the tumor microenvironment (TME) is a pre-requisite for the optimization of immunotherapy. Mechanistic models such as quantitative systems pharmacology (QSP) provide insights into the TME dynamics and predict the efficacy of immunotherapy in virtual patient populations/digital twins but require vast amounts of multimodal data for parameterization. Large-scale datasets characterizing the TME are available due to recent advances in bioinformatics for multi-omics data. Here, we discuss the perspectives of leveraging omics-derived bioinformatics estimates to inform QSP models and circumvent the challenges of model calibration and validation in immuno-oncology.
Collapse
Affiliation(s)
- Theinmozhi Arulraj
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Hanwen Wang
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Alberto Ippolito
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Shuming Zhang
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Elana J Fertig
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Department of Oncology, and the Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Department of Applied Mathematics and Statistics, Johns Hopkins University, Baltimore, MD, USA
| | - Aleksander S Popel
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Department of Oncology, and the Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| |
Collapse
|
112
|
Hamacher C, Degen R, Franke M, Switacz VK, Fleck D, Katreddi RR, Hernandez-Clavijo A, Strauch M, Horio N, Hachgenei E, Spehr J, Liberles SD, Merhof D, Forni PE, Zimmer-Bensch G, Ben-Shaul Y, Spehr M. A revised conceptual framework for mouse vomeronasal pumping and stimulus sampling. Curr Biol 2024; 34:1206-1221.e6. [PMID: 38320553 PMCID: PMC10965388 DOI: 10.1016/j.cub.2024.01.036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2023] [Revised: 12/15/2023] [Accepted: 01/11/2024] [Indexed: 02/08/2024]
Abstract
The physiological performance of any sensory organ is determined by its anatomy and physical properties. Consequently, complex sensory structures with elaborate features have evolved to optimize stimulus detection. Understanding these structures and their physical nature forms the basis for mechanistic insights into sensory function. Despite its crucial role as a sensor for pheromones and other behaviorally instructive chemical cues, the vomeronasal organ (VNO) remains a poorly characterized mammalian sensory structure. Fundamental principles of its physico-mechanical function, including basic aspects of stimulus sampling, remain poorly explored. Here, we revisit the classical vasomotor pump hypothesis of vomeronasal stimulus uptake. Using advanced anatomical, histological, and physiological methods, we demonstrate that large parts of the lateral mouse VNO are composed of smooth muscle. Vomeronasal smooth muscle tissue comprises two subsets of fibers with distinct topography, structure, excitation-contraction coupling, and, ultimately, contractile properties. Specifically, contractions of a large population of noradrenaline-sensitive cells mediate both transverse and longitudinal lumen expansion, whereas cholinergic stimulation targets an adluminal group of smooth muscle fibers. The latter run parallel to the VNO's rostro-caudal axis and are ideally situated to mediate antagonistic longitudinal constriction of the lumen. This newly discovered arrangement implies a novel mode of function. Single-cell transcriptomics and pharmacological profiling reveal the receptor subtypes involved. Finally, 2D/3D tomography provides non-invasive insight into the intact VNO's anatomy and mechanics, enables measurement of luminal fluid volume, and allows an assessment of relative volume change upon noradrenergic stimulation. Together, we propose a revised conceptual framework for mouse vomeronasal pumping and, thus, stimulus sampling.
Collapse
Affiliation(s)
- Christoph Hamacher
- Department of Chemosensation, Institute for Biology II, RWTH Aachen University, 52074 Aachen, Germany
| | - Rudolf Degen
- Department of Chemosensation, Institute for Biology II, RWTH Aachen University, 52074 Aachen, Germany; Research Training Group 2416 MultiSenses - MultiScales, RWTH Aachen University, 52074 Aachen, Germany
| | - Melissa Franke
- Department of Chemosensation, Institute for Biology II, RWTH Aachen University, 52074 Aachen, Germany
| | - Victoria K Switacz
- Department of Chemosensation, Institute for Biology II, RWTH Aachen University, 52074 Aachen, Germany; Research Training Group 2416 MultiSenses - MultiScales, RWTH Aachen University, 52074 Aachen, Germany
| | - David Fleck
- Department of Chemosensation, Institute for Biology II, RWTH Aachen University, 52074 Aachen, Germany
| | - Raghu Ram Katreddi
- Department of Biological Sciences, The RNA Institute, University at Albany, Albany, NY 12222, USA
| | - Andres Hernandez-Clavijo
- Department of Chemosensation, Institute for Biology II, RWTH Aachen University, 52074 Aachen, Germany
| | - Martin Strauch
- Institute of Imaging and Computer Vision, RWTH Aachen University, 52074 Aachen, Germany
| | - Nao Horio
- Howard Hughes Medical Institute, Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA
| | - Enno Hachgenei
- Department of Production Metrology, Fraunhofer Institute for Production Technology, 52074 Aachen, Germany
| | - Jennifer Spehr
- Department of Chemosensation, Institute for Biology II, RWTH Aachen University, 52074 Aachen, Germany
| | - Stephen D Liberles
- Howard Hughes Medical Institute, Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA
| | - Dorit Merhof
- Research Training Group 2416 MultiSenses - MultiScales, RWTH Aachen University, 52074 Aachen, Germany; Institute of Imaging and Computer Vision, RWTH Aachen University, 52074 Aachen, Germany
| | - Paolo E Forni
- Department of Biological Sciences, The RNA Institute, University at Albany, Albany, NY 12222, USA
| | - Geraldine Zimmer-Bensch
- Research Training Group 2416 MultiSenses - MultiScales, RWTH Aachen University, 52074 Aachen, Germany; Department of Neuroepigenetics, Institute for Biology II, RWTH Aachen University, 52074 Aachen, Germany
| | - Yoram Ben-Shaul
- Department of Medical Neurobiology, Institute for Medical Research Israel Canada, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem 9112102, Israel
| | - Marc Spehr
- Department of Chemosensation, Institute for Biology II, RWTH Aachen University, 52074 Aachen, Germany; Research Training Group 2416 MultiSenses - MultiScales, RWTH Aachen University, 52074 Aachen, Germany.
| |
Collapse
|
113
|
Meroueh C, Warasnhe K, Tizhoosh HR, Shah VH, Ibrahim SH. Digital pathology and spatial omics in steatohepatitis: Clinical applications and discovery potentials. Hepatology 2024:01515467-990000000-00815. [PMID: 38517078 PMCID: PMC11669472 DOI: 10.1097/hep.0000000000000866] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/27/2024] [Accepted: 02/26/2024] [Indexed: 03/23/2024]
Abstract
Steatohepatitis with diverse etiologies is the most common histological manifestation in patients with liver disease. However, there are currently no specific histopathological features pathognomonic for metabolic dysfunction-associated steatotic liver disease, alcohol-associated liver disease, or metabolic dysfunction-associated steatotic liver disease with increased alcohol intake. Digitizing traditional pathology slides has created an emerging field of digital pathology, allowing for easier access, storage, sharing, and analysis of whole-slide images. Artificial intelligence (AI) algorithms have been developed for whole-slide images to enhance the accuracy and speed of the histological interpretation of steatohepatitis and are currently employed in biomarker development. Spatial biology is a novel field that enables investigators to map gene and protein expression within a specific region of interest on liver histological sections, examine disease heterogeneity within tissues, and understand the relationship between molecular changes and distinct tissue morphology. Here, we review the utility of digital pathology (using linear and nonlinear microscopy) augmented with AI analysis to improve the accuracy of histological interpretation. We will also discuss the spatial omics landscape with special emphasis on the strengths and limitations of established spatial transcriptomics and proteomics technologies and their application in steatohepatitis. We then highlight the power of multimodal integration of digital pathology augmented by machine learning (ML)algorithms with spatial biology. The review concludes with a discussion of the current gaps in knowledge, the limitations and premises of these tools and technologies, and the areas of future research.
Collapse
Affiliation(s)
- Chady Meroueh
- Department of Laboratory Medicine & Pathology, Mayo Clinic, Rochester, Minnesota, USA
| | - Khaled Warasnhe
- Division of Gastroenterology & Hepatology, Mayo Clinic, Rochester, Minnesota, USA
| | - H. R. Tizhoosh
- Department of Artificial Intelligence & Informatics, Mayo Clinic, Rochester, Minnesota, USA
| | - Vijay H. Shah
- Division of Gastroenterology & Hepatology, Mayo Clinic, Rochester, Minnesota, USA
| | - Samar H. Ibrahim
- Division of Gastroenterology & Hepatology, Mayo Clinic, Rochester, Minnesota, USA
- Division of Pediatric Gastroenterology & Hepatology, Mayo Clinic, Rochester, Minnesota, USA
| |
Collapse
|
114
|
Chen J, Ke R. Spatial analysis toolkits for RNA in situ sequencing. WILEY INTERDISCIPLINARY REVIEWS. RNA 2024; 15:e1842. [PMID: 38605484 DOI: 10.1002/wrna.1842] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Revised: 03/13/2024] [Accepted: 03/14/2024] [Indexed: 04/13/2024]
Abstract
Spatial transcriptomics (ST) is featured by high-throughput gene expression profiling within their native cell and tissue context, offering a means to investigate gene regulatory networks in tissue microenvironment. In situ sequencing (ISS) is an imaging-based ST technology that simultaneously detects hundreds to thousands of genes at subcellular resolution. As a highly reproducible and robust technique, ISS has been widely adapted and undergone a series of technical iterations. As the interest in ISS-based spatial transcriptomic analysis grows, scalable and integrated data analysis workflows are needed to facilitate the applications of ISS in different research fields. This review presents the state-of-the-art bioinformatic toolkits for ISS data analysis, which covers the upstream and downstream analysis workflows, including image analysis, cell segmentation, clustering, functional enrichment, detection of spatially variable genes and cell clusters, spatial cell-cell interactions, and trajectory inference. To assist the community in choosing the right tools for their research, the application of each tool and its compatibility with ISS data are reviewed in detailed. Finally, future perspectives and challenges concerning how to integrate heterogeneous tools into a user-friendly analysis pipeline are discussed. This article is categorized under: RNA Methods > RNA Analyses In Vitro and In Silico.
Collapse
Affiliation(s)
- Jiayu Chen
- School of Medicine, Huaqiao University, Xiamen, Fujian, China
| | - Rongqin Ke
- School of Medicine, Huaqiao University, Xiamen, Fujian, China
| |
Collapse
|
115
|
Zhang J, Zhang L, Gongol B, Hayes J, Borowsky A, Bailey-Serres J, Girke T. spatialHeatmap: visualizing spatial bulk and single-cell assays in anatomical images. NAR Genom Bioinform 2024; 6:lqae006. [PMID: 38312938 PMCID: PMC10836942 DOI: 10.1093/nargab/lqae006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2023] [Revised: 12/14/2023] [Accepted: 01/18/2024] [Indexed: 02/06/2024] Open
Abstract
Visualizing spatial assay data in anatomical images is vital for understanding biological processes in cell, tissue, and organ organizations. Technologies requiring this functionality include traditional one-at-a-time assays, and bulk and single-cell omics experiments, including RNA-seq and proteomics. The spatialHeatmap software provides a series of powerful new methods for these needs, and allows users to work with adequately formatted anatomical images from public collections or custom images. It colors the spatial features (e.g. tissues) annotated in the images according to the measured or predicted abundance levels of biomolecules (e.g. mRNAs) using a color key. This core functionality of the package is called a spatial heatmap plot. Single-cell data can be co-visualized in composite plots that combine spatial heatmaps with embedding plots of high-dimensional data. The resulting spatial context information is essential for gaining insights into the tissue-level organization of single-cell data, or vice versa. Additional core functionalities include the automated identification of biomolecules with spatially selective abundance patterns and clusters of biomolecules sharing similar abundance profiles. To appeal to both non-expert and computational users, spatialHeatmap provides a graphical and a command-line interface, respectively. It is distributed as a free, open-source Bioconductor package (https://bioconductor.org/packages/spatialHeatmap) that users can install on personal computers, shared servers, or cloud systems.
Collapse
Affiliation(s)
- Jianhai Zhang
- Institute for Integrative Genome Biology, Department of Botany and Plant Sciences, 1207F Genomics Building, University of California, Riverside, CA 92521, USA
| | - Le Zhang
- Institute for Integrative Genome Biology, Department of Botany and Plant Sciences, 1207F Genomics Building, University of California, Riverside, CA 92521, USA
| | - Brendan Gongol
- Institute for Integrative Genome Biology, Department of Botany and Plant Sciences, 1207F Genomics Building, University of California, Riverside, CA 92521, USA
| | - Jordan Hayes
- Institute for Integrative Genome Biology, Department of Botany and Plant Sciences, 1207F Genomics Building, University of California, Riverside, CA 92521, USA
| | - Alexander T Borowsky
- Center for Plant Cell Biology, Department of Botany and Plant Sciences, University of California, Riverside, Riverside, CA 92521, USA
| | - Julia Bailey-Serres
- Center for Plant Cell Biology, Department of Botany and Plant Sciences, University of California, Riverside, Riverside, CA 92521, USA
| | - Thomas Girke
- Institute for Integrative Genome Biology, Department of Botany and Plant Sciences, 1207F Genomics Building, University of California, Riverside, CA 92521, USA
| |
Collapse
|
116
|
Navikas V, Kowal J, Rodriguez D, Rivest F, Brajkovic S, Cassano M, Dupouy D. Semi-automated approaches for interrogating spatial heterogeneity of tissue samples. Sci Rep 2024; 14:5025. [PMID: 38424144 PMCID: PMC10904364 DOI: 10.1038/s41598-024-55387-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2023] [Accepted: 02/22/2024] [Indexed: 03/02/2024] Open
Abstract
Tissues are spatially orchestrated ecosystems composed of heterogeneous cell populations and non-cellular elements. Tissue components' interactions shape the biological processes that govern homeostasis and disease, thus comprehensive insights into tissues' composition are crucial for understanding their biology. Recently, advancements in the spatial biology field enabled the in-depth analyses of tissue architecture at single-cell resolution, while preserving the structural context. The increasing number of biomarkers analyzed, together with whole tissue imaging, generate datasets approaching several hundreds of gigabytes in size, which are rich sources of valuable knowledge but require investments in infrastructure and resources for extracting quantitative information. The analysis of multiplex whole-tissue images requires extensive training and experience in data analysis. Here, we showcase how a set of open-source tools can allow semi-automated image data extraction to study the spatial composition of tissues with a focus on tumor microenvironment (TME). With the use of Lunaphore COMET platform, we interrogated lung cancer specimens where we examined the expression of 20 biomarkers. Subsequently, the tissue composition was interrogated using an in-house optimized nuclei detection algorithm followed by a newly developed image artifact exclusion approach. Thereafter, the data was processed using several publicly available tools, highlighting the compatibility of COMET-derived data with currently available image analysis frameworks. In summary, we showcased an innovative semi-automated workflow that highlights the ease of adoption of multiplex imaging to explore TME composition at single-cell resolution using a simple slide in, data out approach. Our workflow is easily transferrable to various cohorts of specimens to provide a toolset for spatial cellular dissection of the tissue composition.
Collapse
Affiliation(s)
| | - Joanna Kowal
- Lunaphore Technologies SA, Tolochenaz, Switzerland
| | | | | | | | | | - Diego Dupouy
- Lunaphore Technologies SA, Tolochenaz, Switzerland.
| |
Collapse
|
117
|
Hsu JE, Ruiz L, Hwang Y, Guzman S, Cho CS, Cheng W, Si Y, Macpherson P, Schrank M, Jun G, Kang HM, Kim M, Brooks S, Lee JH. High-Resolution Spatial Transcriptomic Atlas of Mouse Soleus Muscle: Unveiling Single Cell and Subcellular Heterogeneity in Health and Denervation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.02.26.582103. [PMID: 38464282 PMCID: PMC10925160 DOI: 10.1101/2024.02.26.582103] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/12/2024]
Abstract
Skeletal muscle is essential for both movement and metabolic processes, characterized by a complex and ordered structure. Despite its importance, a detailed spatial map of gene expression within muscle tissue has been challenging to achieve due to the limitations of existing technologies, which struggle to provide high-resolution views. In this study, we leverage the Seq-Scope technique, an innovative method that allows for the observation of the entire transcriptome at an unprecedented submicron spatial resolution. By applying this technique to the mouse soleus muscle, we analyze and compare the gene expression profiles in both healthy conditions and following denervation, a process that mimics aspects of muscle aging. Our approach reveals detailed characteristics of muscle fibers, other cell types present within the muscle, and specific subcellular structures such as the postsynaptic nuclei at neuromuscular junctions, hybrid muscle fibers, and areas of localized expression of genes responsive to muscle injury, along with their histological context. The findings of this research significantly enhance our understanding of the diversity within the muscle cell transcriptome and its variation in response to denervation, a key factor in the decline of muscle function with age. This breakthrough in spatial transcriptomics not only deepens our knowledge of muscle biology but also sets the stage for the development of new therapeutic strategies aimed at mitigating the effects of aging on muscle health, thereby offering a more comprehensive insight into the mechanisms of muscle maintenance and degeneration in the context of aging and disease.
Collapse
Affiliation(s)
- Jer-En Hsu
- Department of Molecular & Integrative Physiology, University of Michigan, Ann Arbor, MI, USA
| | - Lloyd Ruiz
- Department of Molecular & Integrative Physiology, University of Michigan, Ann Arbor, MI, USA
| | - Yongha Hwang
- Department of Molecular & Integrative Physiology, University of Michigan, Ann Arbor, MI, USA
- Space Planning and Analysis, University of Michigan, Ann Arbor, MI, USA
| | - Steve Guzman
- Department of Molecular & Integrative Physiology, University of Michigan, Ann Arbor, MI, USA
| | - Chun-Seok Cho
- Department of Molecular & Integrative Physiology, University of Michigan, Ann Arbor, MI, USA
| | - Weiqiu Cheng
- Department of Biostatistics, University of Michigan, Ann Arbor, MI, USA
| | - Yichen Si
- Department of Biostatistics, University of Michigan, Ann Arbor, MI, USA
| | - Peter Macpherson
- Department of Molecular & Integrative Physiology, University of Michigan, Ann Arbor, MI, USA
| | - Mitchell Schrank
- Department of Molecular & Integrative Physiology, University of Michigan, Ann Arbor, MI, USA
| | - Goo Jun
- Human Genetics Center, University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Hyun-Min Kang
- Department of Biostatistics, University of Michigan, Ann Arbor, MI, USA
| | - Myungjin Kim
- Department of Molecular & Integrative Physiology, University of Michigan, Ann Arbor, MI, USA
| | - Susan Brooks
- Department of Molecular & Integrative Physiology, University of Michigan, Ann Arbor, MI, USA
| | - Jun Hee Lee
- Department of Molecular & Integrative Physiology, University of Michigan, Ann Arbor, MI, USA
| |
Collapse
|
118
|
Qiu Y, Wei K, Lin H, Liu Y, Lin C, Ke R. Combined amplification-based single-molecule fluorescence in situ hybridization with immunofluorescence for simultaneous in situ detection of RNAs and proteins. Biochem Biophys Res Commun 2024; 696:149508. [PMID: 38244312 DOI: 10.1016/j.bbrc.2024.149508] [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: 12/21/2023] [Accepted: 01/08/2024] [Indexed: 01/22/2024]
Abstract
We present a combined amplification-based single-molecule fluorescence in situ hybridization and immunofluorescence (asmFISH-IF) method for the detection of multiple RNAs and proteins simultaneously in cells and formaldehyde-fixed and paraffin-embedded tissue sections. We showed that performing asmFISH before immunofluorescence gives a better IF signal than the opposite. Our asmFISH-IF method could help study the interplay of RNA and protein, helping to understand their functions.
Collapse
Affiliation(s)
- Yinghui Qiu
- School of Medicine, Huaqiao University, Xiamen, Fujian, 361021, China; College of Materials Science and Engineering, Huaqiao University, Xiamen, Fujian, 361021, China
| | - Kaipeng Wei
- Department of Medical Laboratory Technology, Quanzhou Medical College, Quanzhou, Fujian, 362011, China
| | - Hui Lin
- Department of Pathology, The 910 Hospital, Quanzhou, 362000, Fujian, China
| | - Yanxiu Liu
- School of Medicine, Huaqiao University, Xiamen, Fujian, 361021, China
| | - Chen Lin
- School of Medicine, Huaqiao University, Xiamen, Fujian, 361021, China.
| | - Rongqin Ke
- School of Medicine, Huaqiao University, Xiamen, Fujian, 361021, China.
| |
Collapse
|
119
|
Helms HR, Oyama KA, Ware JP, Ibsen SD, Bertassoni LE. Multiplex Single-Cell Bioprinting for Engineering of Heterogeneous Tissue Constructs with Subcellular Spatial Resolution. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.02.01.578499. [PMID: 38352428 PMCID: PMC10862823 DOI: 10.1101/2024.02.01.578499] [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] [Indexed: 02/24/2024]
Abstract
Tissue development, function, and disease are largely driven by the spatial organization of individual cells and their cell-cell interactions. Precision engineered tissues with single-cell spatial resolution, therefore, have tremendous potential for next generation disease models, drug discovery, and regenerative therapeutics. Despite significant advancements in biofabrication approaches to improve feature resolution, strategies to fabricate tissues with the exact same organization of individual cells in their native cellular microenvironment have remained virtually non-existent to date. Here we report a method to spatially pattern single cells with up to eight cell phenotypes and subcellular spatial precision. As proof-of-concept we first demonstrate the ability to systematically assess the influence of cellular microenvironments on cell behavior by controllably altering the spatial arrangement of cell types in bioprinted precision cell-cell interaction arrays. We then demonstrate, for the first time, the ability to produce high-fidelity replicas of a patient's annotated cancer biopsy with subcellular resolution. The ability to replicate native cellular microenvironments marks a significant advancement for precision biofabricated in-vitro models, where heterogenous tissues can be engineered with single-cell spatial precision to advance our understanding of complex biological systems in a controlled and systematic manner.
Collapse
Affiliation(s)
- Haylie R Helms
- Knight Cancer Precision Biofabrication Hub, Knight Cancer Institute, Oregon Health and Science University, Portland, OR 97201, USA
- Department of Biomedical Engineering, School of Medicine, Oregon Health and Science University, Portland, OR 97201, USA
- Cancer Early Detection Advanced Research Center (CEDAR), Knight Cancer Institute, Oregon Health and Science University, Portland, OR 97201, USA
| | - Kody A Oyama
- Knight Cancer Precision Biofabrication Hub, Knight Cancer Institute, Oregon Health and Science University, Portland, OR 97201, USA
| | - Jason P Ware
- Department of Biomedical Engineering, School of Medicine, Oregon Health and Science University, Portland, OR 97201, USA
- Cancer Early Detection Advanced Research Center (CEDAR), Knight Cancer Institute, Oregon Health and Science University, Portland, OR 97201, USA
| | - Stuart D Ibsen
- Department of Biomedical Engineering, School of Medicine, Oregon Health and Science University, Portland, OR 97201, USA
- Cancer Early Detection Advanced Research Center (CEDAR), Knight Cancer Institute, Oregon Health and Science University, Portland, OR 97201, USA
| | - Luiz E Bertassoni
- Knight Cancer Precision Biofabrication Hub, Knight Cancer Institute, Oregon Health and Science University, Portland, OR 97201, USA
- Department of Biomedical Engineering, School of Medicine, Oregon Health and Science University, Portland, OR 97201, USA
- Cancer Early Detection Advanced Research Center (CEDAR), Knight Cancer Institute, Oregon Health and Science University, Portland, OR 97201, USA
- Division of Oncological Sciences, Knight Cancer Institute, Oregon Health and Science University, Portland, OR 97201, USA
- Division of Biomaterials and Biomechanics, Department of Oral Rehabilitation and Biosciences, School of Dentistry, Oregon Health and Science University, Portland, OR 97201, USA
- Center for Regenerative Medicine, School of Medicine, Oregon Health and Science University, Portland, OR 97201, USA
| |
Collapse
|
120
|
Persson PB, Bondke Persson A. Subcellular physiology. Acta Physiol (Oxf) 2024; 240:e14080. [PMID: 38205920 DOI: 10.1111/apha.14080] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Accepted: 12/21/2023] [Indexed: 01/12/2024]
Affiliation(s)
- Pontus B Persson
- Institute of Translational Physiology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | | |
Collapse
|
121
|
Beck M, Covino R, Hänelt I, Müller-McNicoll M. Understanding the cell: Future views of structural biology. Cell 2024; 187:545-562. [PMID: 38306981 DOI: 10.1016/j.cell.2023.12.017] [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: 10/04/2023] [Revised: 12/05/2023] [Accepted: 12/11/2023] [Indexed: 02/04/2024]
Abstract
Determining the structure and mechanisms of all individual functional modules of cells at high molecular detail has often been seen as equal to understanding how cells work. Recent technical advances have led to a flush of high-resolution structures of various macromolecular machines, but despite this wealth of detailed information, our understanding of cellular function remains incomplete. Here, we discuss present-day limitations of structural biology and highlight novel technologies that may enable us to analyze molecular functions directly inside cells. We predict that the progression toward structural cell biology will involve a shift toward conceptualizing a 4D virtual reality of cells using digital twins. These will capture cellular segments in a highly enriched molecular detail, include dynamic changes, and facilitate simulations of molecular processes, leading to novel and experimentally testable predictions. Transferring biological questions into algorithms that learn from the existing wealth of data and explore novel solutions may ultimately unveil how cells work.
Collapse
Affiliation(s)
- Martin Beck
- Max Planck Institute of Biophysics, Max-von-Laue-Straße 3, 60438 Frankfurt am Main, Germany; Goethe University Frankfurt, Frankfurt, Germany.
| | - Roberto Covino
- Frankfurt Institute for Advanced Studies, Ruth-Moufang-Straße 1, 60438 Frankfurt am Main, Germany.
| | - Inga Hänelt
- Goethe University Frankfurt, Frankfurt, Germany.
| | | |
Collapse
|
122
|
Zhao Y, Agyemang D, Liu Y, Mahoney M, Li S. Quantifying Interpretation Reproducibility in Vision Transformer Models with TAVAC. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.18.576252. [PMID: 38328179 PMCID: PMC10849480 DOI: 10.1101/2024.01.18.576252] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/09/2024]
Abstract
The use of deep learning algorithms to extract meaningful diagnostic features from biomedical images holds the promise to improve patient care given the expansion of digital pathology. Among these deep learning models, Vision Transformer (ViT) models have been demonstrated to capture long-range spatial relationships with more robust prediction power for image classification tasks than regular convolutional neural network (CNN) models, and also better model interpretability. Model interpretation is important for understanding and elucidating how a deep learning model makes predictions, especially for developing transparent models for digital pathology. However, like other deep learning algorithms, with limited annotated biomedical imaging datasets, ViT models are prone to poor performance due to overfitting, which can lead to false predictions due to random noise. Overfitting affects model interpretation when predictions are made out of random noise. To address this issue, we introduce a novel metric - Training Attention and Validation Attention Consistency (TAVAC) - for evaluating ViT model degree of overfitting on imaging datasets and quantifying the reproducibility of interpretation. Specifically, the model interpretation is performed by comparing the high-attention regions in the image between training and testing. We test the method on four publicly available image classification datasets and two independent breast cancer histological image datasets. All overfitted models exhibited significantly lower TAVAC scores than the good-fit models. The TAVAC score quantitatively measures the level of generalization of model interpretation on a fine-grained level for small groups of cells in each H&E image, which cannot be provided by traditional performance evaluation metrics like prediction accuracy. Furthermore, the application of TAVAC extends beyond medical diagnostic AI models; it enhances the monitoring of model interpretative reproducibility at pixel-resolution in basic research, to reveal critical spatial patterns and cellular structures essential to understanding biological processes and disease mechanisms. TAVAC sets a new standard for evaluating the performance of deep learning model interpretation and provides a method for determining the significance of high-attention regions detected from the attention map of the biomedical images.
Collapse
Affiliation(s)
- Yue Zhao
- The Jackson Laboratory for Genomic Medicine, Farmington, CT, USA
| | - Dylan Agyemang
- Department of Mathematics and Statistics, University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - Yang Liu
- The Jackson Laboratory for Genomic Medicine, Farmington, CT, USA
| | - Matt Mahoney
- The Jackson Laboratory for Mouse Genetics, Bar Harbor, ME, USA
| | - Sheng Li
- The Jackson Laboratory for Genomic Medicine, Farmington, CT, USA
- The Jackson Laboratory Cancer Center, Bar Harbor, ME, USA
- Department of Genetics and Genome Sciences, University of Connecticut School of Medicine, Farmington, CT, USA
- Department of Computer Science and Engineering, University of Connecticut School of Engineering, Storrs, CT
| |
Collapse
|
123
|
Cao L, Yang C, Hu L, Jiang W, Ren Y, Xia T, Xu M, Ji Y, Li M, Xu X, Li Y, Zhang Y, Fang S. Deciphering spatial domains from spatially resolved transcriptomics with Siamese graph autoencoder. Gigascience 2024; 13:giae003. [PMID: 38373745 PMCID: PMC10939418 DOI: 10.1093/gigascience/giae003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Revised: 12/26/2023] [Accepted: 01/10/2024] [Indexed: 02/21/2024] Open
Abstract
BACKGROUND Cell clustering is a pivotal aspect of spatial transcriptomics (ST) data analysis as it forms the foundation for subsequent data mining. Recent advances in spatial domain identification have leveraged graph neural network (GNN) approaches in conjunction with spatial transcriptomics data. However, such GNN-based methods suffer from representation collapse, wherein all spatial spots are projected onto a singular representation. Consequently, the discriminative capability of individual representation feature is limited, leading to suboptimal clustering performance. RESULTS To address this issue, we proposed SGAE, a novel framework for spatial domain identification, incorporating the power of the Siamese graph autoencoder. SGAE mitigates the information correlation at both sample and feature levels, thus improving the representation discrimination. We adapted this framework to ST analysis by constructing a graph based on both gene expression and spatial information. SGAE outperformed alternative methods by its effectiveness in capturing spatial patterns and generating high-quality clusters, as evaluated by the Adjusted Rand Index, Normalized Mutual Information, and Fowlkes-Mallows Index. Moreover, the clustering results derived from SGAE can be further utilized in the identification of 3-dimensional (3D) Drosophila embryonic structure with enhanced accuracy. CONCLUSIONS Benchmarking results from various ST datasets generated by diverse platforms demonstrate compelling evidence for the effectiveness of SGAE against other ST clustering methods. Specifically, SGAE exhibits potential for extension and application on multislice 3D reconstruction and tissue structure investigation. The source code and a collection of spatial clustering results can be accessed at https://github.com/STOmics/SGAE/.
Collapse
Affiliation(s)
- Lei Cao
- BGI Research, Beijing 102601, China
- BGI Research, Shenzhen 518083, China
| | - Chao Yang
- BGI Research, Beijing 102601, China
- BGI Research, Shenzhen 518083, China
| | - Luni Hu
- BGI Research, Beijing 102601, China
- BGI Research, Shenzhen 518083, China
| | - Wenjian Jiang
- BGI Research, Beijing 102601, China
- BGI Research, Shenzhen 518083, China
| | - Yating Ren
- School of Software, Beihang University, Beijing 100191, China
| | - Tianyi Xia
- BGI Research, Beijing 102601, China
- BGI Research, Shenzhen 518083, China
| | - Mengyang Xu
- BGI Research, Shenzhen 518083, China
- BGI Research, Qingdao 266555, China
| | | | - Mei Li
- BGI Research, Shenzhen 518083, China
| | - Xun Xu
- BGI Research, Wuhan 430074, China
| | - Yuxiang Li
- BGI Research, Shenzhen 518083, China
- BGI Research, Wuhan 430074, China
- Guangdong Bigdata Engineering Technology Research Center for Life Sciences, BGI Research, Shenzhen 518083, China
| | - Yong Zhang
- BGI Research, Shenzhen 518083, China
- BGI Research, Wuhan 430074, China
- Guangdong Bigdata Engineering Technology Research Center for Life Sciences, BGI Research, Shenzhen 518083, China
| | - Shuangsang Fang
- BGI Research, Beijing 102601, China
- BGI Research, Shenzhen 518083, China
| |
Collapse
|
124
|
Chen Y, Yang S, Yu K, Zhang J, Wu M, Zheng Y, Zhu Y, Dai J, Wang C, Zhu X, Dai Y, Sun Y, Wu T, Wang S. Spatial omics: An innovative frontier in aging research. Ageing Res Rev 2024; 93:102158. [PMID: 38056503 DOI: 10.1016/j.arr.2023.102158] [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: 08/28/2023] [Revised: 11/25/2023] [Accepted: 12/01/2023] [Indexed: 12/08/2023]
Abstract
Disentangling the impact of aging on health and disease has become critical as population aging progresses rapidly. Studying aging at the molecular level is complicated by the diverse aging profiles and dynamics. However, the examination of cellular states within aging tissues in situ is hampered by the lack of high-resolution spatial data. Emerging spatial omics technologies facilitate molecular and spatial analysis of tissues, providing direct access to precise information on various functional regions and serving as a favorable tool for unraveling the heterogeneity of aging. In this review, we summarize the recent advances in spatial omics application in multi-organ aging research, which has enhanced the understanding of aging mechanisms from multiple standpoints. We also discuss the main challenges in spatial omics research to date, the opportunities for further developing the technology, and the potential applications of spatial omics in aging and aging-related diseases.
Collapse
Affiliation(s)
- Ying Chen
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; National Clinical Research Center for Obstetrical and Gynecological Diseases, Wuhan, China; Ministry of Education, Key Laboratory of Cancer Invasion and Metastasis, Wuhan, China
| | - Shuhao Yang
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; National Clinical Research Center for Obstetrical and Gynecological Diseases, Wuhan, China; Ministry of Education, Key Laboratory of Cancer Invasion and Metastasis, Wuhan, China
| | - Kaixu Yu
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jinjin Zhang
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; National Clinical Research Center for Obstetrical and Gynecological Diseases, Wuhan, China; Ministry of Education, Key Laboratory of Cancer Invasion and Metastasis, Wuhan, China
| | - Meng Wu
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; National Clinical Research Center for Obstetrical and Gynecological Diseases, Wuhan, China; Ministry of Education, Key Laboratory of Cancer Invasion and Metastasis, Wuhan, China
| | - Yongqiang Zheng
- State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Centre, Sun Yat-sen University, Guangzhou, China
| | - Yun Zhu
- Department of Internal Medicine, Southern Illinois University School of Medicine, 801 N. Rutledge, P.O. Box 19628, Springfield, IL 62702, USA
| | - Jun Dai
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; National Clinical Research Center for Obstetrical and Gynecological Diseases, Wuhan, China; Ministry of Education, Key Laboratory of Cancer Invasion and Metastasis, Wuhan, China
| | - Chunyan Wang
- College of Science & Engineering Jinan University, Guangzhou, China
| | - Xiaoran Zhu
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; National Clinical Research Center for Obstetrical and Gynecological Diseases, Wuhan, China; Ministry of Education, Key Laboratory of Cancer Invasion and Metastasis, Wuhan, China
| | - Yun Dai
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; National Clinical Research Center for Obstetrical and Gynecological Diseases, Wuhan, China; Ministry of Education, Key Laboratory of Cancer Invasion and Metastasis, Wuhan, China
| | - Yunhong Sun
- Hubei Key Laboratory of Food Nutrition and Safety, MOE Key Lab of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Tong Wu
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; National Clinical Research Center for Obstetrical and Gynecological Diseases, Wuhan, China; Ministry of Education, Key Laboratory of Cancer Invasion and Metastasis, Wuhan, China.
| | - Shixuan Wang
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; National Clinical Research Center for Obstetrical and Gynecological Diseases, Wuhan, China; Ministry of Education, Key Laboratory of Cancer Invasion and Metastasis, Wuhan, China.
| |
Collapse
|
125
|
Rinaldi G, Loukas A, Sotillo J. Trematode Genomics and Proteomics. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2024; 1454:507-539. [PMID: 39008274 DOI: 10.1007/978-3-031-60121-7_13] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/16/2024]
Abstract
Trematode infections stand out as one of the frequently overlooked tropical diseases, despite their wide global prevalence and remarkable capacity to parasitize diverse host species and tissues. Furthermore, these parasites hold significant socio-economic, medical, veterinary and agricultural implications. Over the past decades, substantial strides have been taken to bridge the information gap concerning various "omic" tools, such as proteomics and genomics, in this field. In this edition of the book, we highlight recent progress in genomics and proteomics concerning trematodes with a particular focus on the advances made in the past 5 years. Additionally, we present insights into cutting-edge technologies employed in studying trematode biology and shed light on the available resources for exploring the molecular facets of this particular group of parasitic helminths.
Collapse
Affiliation(s)
- Gabriel Rinaldi
- Department of Life Sciences, Aberystwyth University, Aberystwyth, UK
| | - Alex Loukas
- Australian Institute of Tropical Health and Medicine, James Cook University, Cairns, QLD, Australia
| | - Javier Sotillo
- Laboratorio de Referencia e Investigación en Parasitología, Centro Nacional de Microbiología, Instituto de Salud Carlos III, Majadahonda, Spain.
| |
Collapse
|
126
|
Walsh LA, Quail DF. Decoding the tumor microenvironment with spatial technologies. Nat Immunol 2023; 24:1982-1993. [PMID: 38012408 DOI: 10.1038/s41590-023-01678-9] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2023] [Accepted: 10/10/2023] [Indexed: 11/29/2023]
Abstract
Visualization of the cellular heterogeneity and spatial architecture of the tumor microenvironment (TME) is becoming increasingly important to understand mechanisms of disease progression and therapeutic response. This is particularly relevant in the era of cancer immunotherapy, in which the contexture of immune cell positioning within the tumor landscape has been proven to affect efficacy. Although single-cell technologies have mostly replaced conventional approaches to analyze specific cellular subsets within tumors, those that integrate a spatial dimension are now on the rise. In this Review, we assess the strengths and limitations of emerging spatial technologies with a focus on their applications in tumor immunology, as well as forthcoming opportunities for artificial intelligence (AI) and the value of integrating multiomics datasets to achieve a holistic picture of the TME.
Collapse
Affiliation(s)
- Logan A Walsh
- Rosalind and Morris Goodman Cancer Institute, McGill University, Montreal, Quebec, Canada.
- Department of Human Genetics, Faculty of Medicine, McGill University, Montreal, Quebec, Canada.
| | - Daniela F Quail
- Rosalind and Morris Goodman Cancer Institute, McGill University, Montreal, Quebec, Canada.
- Department of Physiology, Faculty of Medicine, McGill University, Montreal, Quebec, Canada.
- Department of Medicine, Division of Experimental Medicine, McGill University, Montreal, Quebec, Canada.
| |
Collapse
|
127
|
Scipione CA, Hyduk SJ, Polenz CK, Cybulsky MI. Unveiling the Hidden Landscape of Arterial Diseases at Single-Cell Resolution. Can J Cardiol 2023; 39:1781-1794. [PMID: 37716639 DOI: 10.1016/j.cjca.2023.09.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Revised: 08/25/2023] [Accepted: 09/11/2023] [Indexed: 09/18/2023] Open
Abstract
High-resolution single-cell technologies have shed light on the pathogenesis of cardiovascular diseases by enabling the discovery of novel cellular and transcriptomic signatures associated with various conditions, and uncovering new contributions of inflammatory processes, immunity, metabolic stress, and risk factors. We review the information obtained from studies using single-cell technologies in tissues with atherosclerosis and aortic aneurysms. Insights are provided on the biology of endothelial, smooth muscle, and immune cells in the arterial intima and media. In addition to cellular diversity, numerous examples of plasticity and phenotype switching are highlighted and presented in the context of normal cell functions.
Collapse
Affiliation(s)
- Corey A Scipione
- Toronto General Hospital Research Institute, University Health Network, Toronto, Ontario, Canada; Departments of Laboratory Medicine and Pathobiology and Immunology, University of Toronto, Toronto, Ontario, Canada.
| | - Sharon J Hyduk
- Toronto General Hospital Research Institute, University Health Network, Toronto, Ontario, Canada
| | - Chanele K Polenz
- Toronto General Hospital Research Institute, University Health Network, Toronto, Ontario, Canada; Departments of Laboratory Medicine and Pathobiology and Immunology, University of Toronto, Toronto, Ontario, Canada
| | - Myron I Cybulsky
- Toronto General Hospital Research Institute, University Health Network, Toronto, Ontario, Canada; Departments of Laboratory Medicine and Pathobiology and Immunology, University of Toronto, Toronto, Ontario, Canada; Peter Munk Cardiac Centre, University Health Network, Toronto, Ontario, Canada.
| |
Collapse
|
128
|
Zhang S, Zhang YD, Shi DD, Wang Z. Therapeutic uses of oxytocin in stress-related neuropsychiatric disorders. Cell Biosci 2023; 13:216. [PMID: 38017588 PMCID: PMC10683256 DOI: 10.1186/s13578-023-01173-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Accepted: 11/18/2023] [Indexed: 11/30/2023] Open
Abstract
Oxytocin (OXT), produced and secreted in the paraventricular nucleus and supraoptic nucleus of magnocellular and parvocellular neurons. The diverse presence and activity of oxytocin suggests a potential for this neuropeptide in the pathogenesis and treatment of stress-related neuropsychiatric disorders (anxiety, depression and post-traumatic stress disorder (PTSD)). For a more comprehensive understanding of the mechanism of OXT's anti-stress action, the signaling cascade of OXT binding to targeting stress were summarized. Then the advance of OXT treatment in depression, anxiety, PTSD and the major projection region of OXT neuron were discussed. Further, the efficacy of endogenous and exogenous OXT in stress responses were highlighted in this review. To augment the level of OXT in stress-related neuropsychiatric disorders, current biological strategies were summarized to shed a light on the treatment of stress-induced psychiatric disorders. We also conclude some of the major puzzles in the therapeutic uses of OXT in stress-related neuropsychiatric disorders. Although some questions remain to be resolved, OXT has an enormous potential therapeutic use as a hormone that regulates stress responses.
Collapse
Affiliation(s)
- Sen Zhang
- Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, 600 Wan Ping Nan Road, Shanghai, 200030, China
- College of Physical Education and Health, East China Normal University, Shanghai, China
| | - Ying-Dan Zhang
- Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, 600 Wan Ping Nan Road, Shanghai, 200030, China
| | - Dong-Dong Shi
- Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, 600 Wan Ping Nan Road, Shanghai, 200030, China.
- Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Zhen Wang
- Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, 600 Wan Ping Nan Road, Shanghai, 200030, China.
- Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
- Institute of Psychological and Behavioral Science, Shanghai Jiao Tong University, Shanghai, China.
| |
Collapse
|
129
|
Velliou RI, Legaki AI, Nikolakopoulou P, Vlachogiannis NI, Chatzigeorgiou A. Liver endothelial cells in NAFLD and transition to NASH and HCC. Cell Mol Life Sci 2023; 80:314. [PMID: 37798474 PMCID: PMC11072568 DOI: 10.1007/s00018-023-04966-7] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Revised: 09/04/2023] [Accepted: 09/15/2023] [Indexed: 10/07/2023]
Abstract
Non-alcoholic fatty liver disease (NAFLD) is considered as the hepatic manifestation of metabolic syndrome, which is characterised by obesity, insulin resistance, hypercholesterolemia and hypertension. NAFLD is the most frequent liver disease worldwide and more than 10% of NAFLD patients progress to the inflammatory and fibrotic stage of non-alcoholic steatohepatitis (NASH), which can lead to end-stage liver disease including hepatocellular carcinoma (HCC), the most frequent primary malignant liver tumor. Liver sinusoidal endothelial cells (LSEC) are strategically positioned at the interface between blood and hepatic parenchyma. LSECs are highly specialized cells, characterised by the presence of transcellular pores, called fenestrae, and exhibit anti-inflammatory and anti-fibrotic characteristics under physiological conditions. However, during NAFLD development they undergo capillarisation and acquire a phenotype similar to vascular endothelial cells, actively promoting all pathophysiological aspects of NAFLD, including steatosis, inflammation, and fibrosis. LSEC dysfunction is critical for the progression to NASH and HCC while restoring LSEC homeostasis appears to be a promising approach to prevent NAFLD progression and its complications and even reverse tissue damage. In this review we present current information on the role of LSEC throughout the progressive phases of NAFLD, summarising in vitro and in vivo experimental evidence and data from human studies.
Collapse
Affiliation(s)
- Rallia-Iliana Velliou
- Department of Physiology, Medical School, National and Kapodistrian University of Athens, 75 Mikras Asias Str., 11527, Athens, Greece
| | - Aigli-Ioanna Legaki
- Department of Physiology, Medical School, National and Kapodistrian University of Athens, 75 Mikras Asias Str., 11527, Athens, Greece
| | - Polyxeni Nikolakopoulou
- Department of Physiology, Medical School, National and Kapodistrian University of Athens, 75 Mikras Asias Str., 11527, Athens, Greece
| | - Nikolaos I Vlachogiannis
- Department of Physiology, Medical School, National and Kapodistrian University of Athens, 75 Mikras Asias Str., 11527, Athens, Greece
| | - Antonios Chatzigeorgiou
- Department of Physiology, Medical School, National and Kapodistrian University of Athens, 75 Mikras Asias Str., 11527, Athens, Greece.
- Institute for Clinical Chemistry and Laboratory Medicine, University Hospital and Faculty of Medicine Carl Gustav Carus of TU Dresden, Fetscherstrasse 74, 01307, Dresden, Germany.
| |
Collapse
|
130
|
Ye Z, Lai Z, Zheng S, Chen Y. Spatial-Live: A lightweight and versatile tool for single cell spatial-omics data visualization. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.09.24.559173. [PMID: 37873441 PMCID: PMC10592799 DOI: 10.1101/2023.09.24.559173] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/25/2023]
Abstract
Single cell spatial-omics data visualization plays a pivotal role in unraveling the intricate spatial organization and heterogeneity of cellular systems. Although various software tools and packages have been developed for this purpose, challenges persist in terms of user-friendly accessibility, data integration, and interactivity. In this study, we introduce Spatial-Live, a lightweight and versatile viewer tool designed for flexible single-cell spatial-omics data visualization. Spatial-Live overcomes the fundamental limitations of two-dimensional (2D) orthographic modes by employing a layer-stacking strategy, enabling efficient rendering of diverse data types with interactive features, and enhancing visualization with richer information in a unified three-dimensional (3D) space.
Collapse
Affiliation(s)
- Zhenqing Ye
- Greehey Children’s Cancer Research Institute, University of Texas Health San Antonio, San Antonio, Texas, 78229, USA
- Department of Population Health Sciences, University of Texas Health San Antonio, San Antonio, Texas, 78229, USA
| | - Zhao Lai
- Greehey Children’s Cancer Research Institute, University of Texas Health San Antonio, San Antonio, Texas, 78229, USA
- Department of Molecular Medicine, University of Texas Health San Antonio, San Antonio, Texas, 78229, USA
| | - Siyuan Zheng
- Greehey Children’s Cancer Research Institute, University of Texas Health San Antonio, San Antonio, Texas, 78229, USA
- Department of Population Health Sciences, University of Texas Health San Antonio, San Antonio, Texas, 78229, USA
| | - Yidong Chen
- Greehey Children’s Cancer Research Institute, University of Texas Health San Antonio, San Antonio, Texas, 78229, USA
- Department of Population Health Sciences, University of Texas Health San Antonio, San Antonio, Texas, 78229, USA
| |
Collapse
|
131
|
Sans M, Chen Y, Thege FI, Dou R, Min J, Yip-Schneider M, Zhang J, Wu R, Irajizad E, Makino Y, Rajapakshe KI, Hurd MW, León-Letelier RA, Vykoukal J, Dennison JB, Do KA, Wolff RA, Guerrero PA, Kim MP, Schmidt CM, Maitra A, Hanash S, Fahrmann JF. Integrated spatial transcriptomics and lipidomics of precursor lesions of pancreatic cancer identifies enrichment of long chain sulfatide biosynthesis as an early metabolic alteration. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.08.14.553002. [PMID: 37645752 PMCID: PMC10462088 DOI: 10.1101/2023.08.14.553002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/31/2023]
Abstract
Background The development of diverse spatial profiling technologies has provided an unprecedented insight into molecular mechanisms driving cancer pathogenesis. Here, we conducted the first integrated cross-species assessment of spatial transcriptomics and spatial metabolomics alterations associated with progression of intraductal papillary mucinous neoplasms (IPMN), bona fide cystic precursors of pancreatic ductal adenocarcinoma (PDAC). Methods Matrix Assisted Laster Desorption/Ionization (MALDI) mass spectrometry (MS)-based spatial imaging and Visium spatial transcriptomics (ST) (10X Genomics) was performed on human resected IPMN tissues (N= 23) as well as pancreata from a mutant Kras;Gnas mouse model of IPMN. Findings were further compared with lipidomic analyses of cystic fluid from 89 patients with histologically confirmed IPMNs, as well as single-cell and bulk transcriptomic data of PDAC and normal tissues. Results MALDI-MS analyses of IPMN tissues revealed long-chain hydroxylated sulfatides, particularly the C24:0(OH) and C24:1(OH) species, to be selectively enriched in the IPMN and PDAC neoplastic epithelium. Integrated ST analyses confirmed that the cognate transcripts engaged in sulfatide biosynthesis, including UGT8, Gal3St1 , and FA2H , were co-localized with areas of sulfatide enrichment. Lipidomic analyses of cystic fluid identified several sulfatide species, including the C24:0(OH) and C24:1(OH) species, to be significantly elevated in patients with IPMN/PDAC compared to those with low-grade IPMN. Targeting of sulfatide metabolism via the selective galactosylceramide synthase inhibitor, UGT8-IN-1, resulted in ceramide-induced lethal mitophagy and subsequent cancer cell death in vitro , and attenuated tumor growth of mutant Kras;Gnas allografts. Transcript levels of UGT8 and FA2H were also selectively enriched in PDAC transcriptomic datasets compared to non-cancerous areas, and elevated tumoral UGT8 was prognostic for poor overall survival. Conclusion Enhanced sulfatide metabolism is an early metabolic alteration in cystic pre-cancerous lesions of the pancreas that persists through invasive neoplasia. Targeting sulfatide biosynthesis might represent an actionable vulnerability for cancer interception.
Collapse
|