1
|
Zorab JM, Li H, Awasthi R, Schinasi A, Cho Y, O'Loughlin T, Wu X. Serotonin and neurotensin inputs in the vCA1 dictate opposing social valence. Nature 2025:10.1038/s41586-025-08809-2. [PMID: 40307550 DOI: 10.1038/s41586-025-08809-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Accepted: 02/19/2025] [Indexed: 05/02/2025]
Abstract
The ability to evaluate valence of a social agent based on social experience is essential for an animal's survival in its social group1. Although hippocampal circuits have been implicated in distinguishing novel and familiar conspecifics2-7, it remains unclear how social valence is constructed on the basis of social history and what mechanisms underlie the heightened valence versatility in dynamic relationships. Here we demonstrate that the ventral (v)CA1 integrates serotonin (5-HT) inputs from the dorsal raphe and neurotensin inputs from the paraventricular nucleus of the thalamus (PVT) to determine positive or negative valence of conspecific representations. Specifically, during an appetitive social interaction 5-HT is released into the vCA1 and disinhibits pyramidal neurons through 5-HT1B receptors, whereas neurotensin is released during an aversive social interaction and potentiates vCA1 neurons directly through NTR1s. Optogenetic silencing of dorsal raphe 5-HT and PVT neurotensin inputs into the vCA1 impairs positive and negative social valence, respectively, and excitation flexibly switches valence assignment. These results show how aversive and rewarding social experiences are linked to conspecific identity through converging dorsal raphe 5-HT and PVT neurotensin signals in the vCA1 that instruct opposing valence, and represent a synaptic switch for flexible social valence computation.
Collapse
Affiliation(s)
- Julia M Zorab
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Huanhuan Li
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Richa Awasthi
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Anna Schinasi
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Yoonjeong Cho
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Thomas O'Loughlin
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Xiaoting Wu
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| |
Collapse
|
2
|
Li L, Guo Y, Jing W, Tang X, Zeng J, Hou Z, Song Y, He A, Li H, Zhu L, Lu Y, Li X. Cell-Type Specific Circuits in the Mammillary Body for Place and Object Recognition Memory. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2409397. [PMID: 39928529 PMCID: PMC11967786 DOI: 10.1002/advs.202409397] [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: 08/08/2024] [Revised: 01/16/2025] [Indexed: 02/12/2025]
Abstract
Mammillary body (MB) is traditionally viewed as a structural node of an anatomic circuit for emotion and memory. However, little is known about its molecular and cellular organizations. Here, a discovery that MB contains four subtypes of neurons that occupy different spatial subregions is reported. Of these, two subtypes of neurons are tagged by parvalbumin (PV) and dopamine receptor-D2 (Drd2) markers. PV neurons are spontaneously active, whereas Drd2 neurons are inactive at rest and generate rebound bursts. These two distinct electrophysiological properties are encoded by Kcnn4 and Cacna1h. PV and Drd2 neurons generate two distinct cell-type specific circuits by receiving inputs from two discrete subiculum neuronal classes. Gain- and loss-of-function studies on these cortical-subcortical circuits demonstrate their differential roles for place and object recognition memory. This finding provides a comprehensive molecular and structural atlas of MB neurons at single-cell resolution and reveals that MB contains molecularly, structurally, and functionally dissociable streams within its serial architecture.
Collapse
Affiliation(s)
- Lanfang Li
- Wuhan Center of Brain ScienceHuazhong University of Science and TechnologyWuhan430030China
- Innovation Center of Brain Medical SciencesMinistry of Education of the People's Republic of ChinaWuhan430030China
- Department of PathophysiologySchool of Basic Medicine and Tongji Medical CollegeHuazhong University of Science and TechnologyWuhan430030China
| | - Yiqing Guo
- Wuhan Center of Brain ScienceHuazhong University of Science and TechnologyWuhan430030China
- Innovation Center of Brain Medical SciencesMinistry of Education of the People's Republic of ChinaWuhan430030China
- Department of PathophysiologySchool of Basic Medicine and Tongji Medical CollegeHuazhong University of Science and TechnologyWuhan430030China
| | - Wei Jing
- Wuhan Center of Brain ScienceHuazhong University of Science and TechnologyWuhan430030China
- Innovation Center of Brain Medical SciencesMinistry of Education of the People's Republic of ChinaWuhan430030China
- Department of AnatomySchool of Basic Medicine and Tongji Medical CollegeHuazhong University of Science and TechnologyWuhan430030China
| | - Xiaomei Tang
- Wuhan Center of Brain ScienceHuazhong University of Science and TechnologyWuhan430030China
- Innovation Center of Brain Medical SciencesMinistry of Education of the People's Republic of ChinaWuhan430030China
- Department of PathophysiologySchool of Basic Medicine and Tongji Medical CollegeHuazhong University of Science and TechnologyWuhan430030China
| | - Jinyu Zeng
- Wuhan Center of Brain ScienceHuazhong University of Science and TechnologyWuhan430030China
- Innovation Center of Brain Medical SciencesMinistry of Education of the People's Republic of ChinaWuhan430030China
- Department of PathophysiologySchool of Basic Medicine and Tongji Medical CollegeHuazhong University of Science and TechnologyWuhan430030China
| | - Zhenye Hou
- Wuhan Center of Brain ScienceHuazhong University of Science and TechnologyWuhan430030China
- Innovation Center of Brain Medical SciencesMinistry of Education of the People's Republic of ChinaWuhan430030China
- Department of PathophysiologySchool of Basic Medicine and Tongji Medical CollegeHuazhong University of Science and TechnologyWuhan430030China
| | - Yige Song
- Wuhan Center of Brain ScienceHuazhong University of Science and TechnologyWuhan430030China
- Innovation Center of Brain Medical SciencesMinistry of Education of the People's Republic of ChinaWuhan430030China
- Department of PathophysiologySchool of Basic Medicine and Tongji Medical CollegeHuazhong University of Science and TechnologyWuhan430030China
| | - Aodi He
- Wuhan Center of Brain ScienceHuazhong University of Science and TechnologyWuhan430030China
- Innovation Center of Brain Medical SciencesMinistry of Education of the People's Republic of ChinaWuhan430030China
- Department of AnatomySchool of Basic Medicine and Tongji Medical CollegeHuazhong University of Science and TechnologyWuhan430030China
| | - Hao Li
- Wuhan Center of Brain ScienceHuazhong University of Science and TechnologyWuhan430030China
- Innovation Center of Brain Medical SciencesMinistry of Education of the People's Republic of ChinaWuhan430030China
- Department of PathophysiologySchool of Basic Medicine and Tongji Medical CollegeHuazhong University of Science and TechnologyWuhan430030China
| | - Ling‐Qiang Zhu
- Wuhan Center of Brain ScienceHuazhong University of Science and TechnologyWuhan430030China
- Innovation Center of Brain Medical SciencesMinistry of Education of the People's Republic of ChinaWuhan430030China
- Department of PathophysiologySchool of Basic Medicine and Tongji Medical CollegeHuazhong University of Science and TechnologyWuhan430030China
| | - Youming Lu
- Wuhan Center of Brain ScienceHuazhong University of Science and TechnologyWuhan430030China
- Innovation Center of Brain Medical SciencesMinistry of Education of the People's Republic of ChinaWuhan430030China
- Department of PhysiologySchool of Basic Medicine and Tongji Medical CollegeHuazhong University of Science and TechnologyWuhan4030030China
| | - Xinyan Li
- Wuhan Center of Brain ScienceHuazhong University of Science and TechnologyWuhan430030China
- Innovation Center of Brain Medical SciencesMinistry of Education of the People's Republic of ChinaWuhan430030China
- Department of AnatomySchool of Basic Medicine and Tongji Medical CollegeHuazhong University of Science and TechnologyWuhan430030China
| |
Collapse
|
3
|
Zhao PA, Li R, Adewunmi T, Garber J, Gustafson C, Kim J, Malone J, Savage A, Skene P, Li XJ. SPARROW reveals microenvironment-zone-specific cell states in healthy and diseased tissues. Cell Syst 2025; 16:101235. [PMID: 40112778 DOI: 10.1016/j.cels.2025.101235] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2024] [Revised: 10/23/2024] [Accepted: 02/19/2025] [Indexed: 03/22/2025]
Abstract
Spatially resolved transcriptomics technologies have advanced our understanding of cellular characteristics within tissue contexts. However, current analytical tools often treat cell-type inference and cellular neighborhood identification as separate and hard clustering processes, limiting comparability across scales and samples. SPARROW addresses these challenges by jointly learning latent embeddings and soft clusterings of cell types and cellular organization. It outperformed state-of-the-art methods in cell-type inference and microenvironment zone delineation and uncovered zone-specific cell states in human and mouse tissues that competing methods missed. By integrating spatially resolved transcriptomics and single-cell RNA sequencing (scRNA-seq) data in a shared latent space, SPARROW achieves single-cell spatial resolution and whole-transcriptome coverage, enabling the discovery of both established and unknown microenvironment zone-specific ligand-receptor interactions in the human tonsil. Overall, SPARROW is a computational framework that provides a comprehensive characterization of tissue features across scales, samples, and conditions.
Collapse
Affiliation(s)
- Peiyao A Zhao
- Allen Institute for Immunology, Seattle, WA 98109, USA.
| | - Ruoxin Li
- Allen Institute for Immunology, Seattle, WA 98109, USA
| | - Temi Adewunmi
- Allen Institute for Immunology, Seattle, WA 98109, USA
| | | | | | - June Kim
- Allen Institute for Immunology, Seattle, WA 98109, USA
| | | | - Adam Savage
- Allen Institute for Immunology, Seattle, WA 98109, USA
| | - Peter Skene
- Allen Institute for Immunology, Seattle, WA 98109, USA
| | - Xiao-Jun Li
- Allen Institute for Immunology, Seattle, WA 98109, USA.
| |
Collapse
|
4
|
Israel U, Marks M, Dilip R, Li Q, Yu C, Laubscher E, Iqbal A, Pradhan E, Ates A, Abt M, Brown C, Pao E, Li S, Pearson-Goulart A, Perona P, Gkioxari G, Barnowski R, Yue Y, Van Valen D. CellSAM: A Foundation Model for Cell Segmentation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2023.11.17.567630. [PMID: 38045277 PMCID: PMC10690226 DOI: 10.1101/2023.11.17.567630] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/05/2023]
Abstract
Cells are a fundamental unit of biological organization, and identifying them in imaging data - cell segmentation - is a critical task for various cellular imaging experiments. While deep learning methods have led to substantial progress on this problem, most models are specialist models that work well for specific domains but cannot be applied across domains or scale well with large amounts of data. In this work, we present CellSAM, a universal model for cell segmentation that generalizes across diverse cellular imaging data. CellSAM builds on top of the Segment Anything Model (SAM) by developing a prompt engineering approach for mask generation. We train an object detector, CellFinder, to automatically detect cells and prompt SAM to generate segmentations. We show that this approach allows a single model to achieve human-level performance for segmenting images of mammalian cells, yeast, and bacteria collected across various imaging modalities. We show that CellSAM has strong zero-shot performance and can be improved with a few examples via few-shot learning. Additionally, we demonstrate how CellSAM can be applied across diverse bioimage analysis workflows. A deployed version of CellSAM is available at https://cellsam.deepcell.org/.
Collapse
Affiliation(s)
- Uriah Israel
- Division of Biology and Biological Engineering, Caltech
- Division of Computing and Mathematical Science, Caltech
| | - Markus Marks
- Division of Engineering and Applied Science, Caltech
- Division of Computing and Mathematical Science, Caltech
| | - Rohit Dilip
- Division of Computing and Mathematical Science, Caltech
| | - Qilin Li
- Division of Engineering and Applied Science, Caltech
| | - Changhua Yu
- Division of Biology and Biological Engineering, Caltech
| | | | - Ahamed Iqbal
- Division of Biology and Biological Engineering, Caltech
| | - Elora Pradhan
- Division of Biology and Biological Engineering, Caltech
| | - Ada Ates
- Division of Biology and Biological Engineering, Caltech
| | - Martin Abt
- Division of Biology and Biological Engineering, Caltech
| | - Caitlin Brown
- Division of Biology and Biological Engineering, Caltech
| | - Edward Pao
- Division of Biology and Biological Engineering, Caltech
| | - Shenyi Li
- Division of Biology and Biological Engineering, Caltech
| | | | - Pietro Perona
- Division of Engineering and Applied Science, Caltech
- Division of Computing and Mathematical Science, Caltech
| | | | | | - Yisong Yue
- Division of Computing and Mathematical Science, Caltech
| | - David Van Valen
- Division of Biology and Biological Engineering, Caltech
- Howard Hughes Medical Institute
| |
Collapse
|
5
|
Semba T, Ishimoto T. Spatial analysis by current multiplexed imaging technologies for the molecular characterisation of cancer tissues. Br J Cancer 2024; 131:1737-1747. [PMID: 39438630 PMCID: PMC11589153 DOI: 10.1038/s41416-024-02882-6] [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: 06/29/2024] [Revised: 10/09/2024] [Accepted: 10/11/2024] [Indexed: 10/25/2024] Open
Abstract
Tumours are composed of tumour cells and the surrounding tumour microenvironment (TME), and the molecular characterisation of the various elements of the TME and their interactions is essential for elucidating the mechanisms of tumour progression and developing better therapeutic strategies. Multiplex imaging is a technique that can quantify the expression of multiple protein markers on the same tissue section while maintaining spatial positioning, and this method has been rapidly developed in cancer research in recent years. Many multiplex imaging technologies and spatial analysis methods are emerging, and the elucidation of their principles and features is essential. In this review, we provide an overview of the latest multiplex imaging techniques by type of imaging and staining method and an introduction to image analysis methods, primarily focusing on spatial cellular properties, providing deeper insight into tumour organisation and spatial molecular biology in the TME.
Collapse
Affiliation(s)
- Takashi Semba
- Division of Carcinogenesis, The Cancer Institute, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Takatsugu Ishimoto
- Division of Carcinogenesis, The Cancer Institute, Japanese Foundation for Cancer Research, Tokyo, Japan.
| |
Collapse
|
6
|
Peroni E, Calistri E, Amato R, Gottardi M, Rosato A. Spatial-transcriptomic profiling: a new lens for understanding myelofibrosis pathophysiology. Cell Commun Signal 2024; 22:510. [PMID: 39434124 PMCID: PMC11492555 DOI: 10.1186/s12964-024-01877-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Accepted: 10/05/2024] [Indexed: 10/23/2024] Open
Abstract
Myelofibrosis (MF) is a complex myeloproliferative neoplasm characterized by abnormal hematopoietic stem cell proliferation and subsequent bone marrow (BM) fibrosis. First documented in the late 19th century, MF has since been extensively studied to unravel its pathophysiology, clinical phenotypes, and therapeutic interventions. MF can be classified into primary and secondary forms, both driven by mutations in genes such as JAK2, CALR, and MPL, which activate the JAK-STAT signaling pathway. These driver mutations are frequently accompanied by additional non-driver mutations in genes like TET2, SRSF2, and TP53, contributing to disease complexity. The BM microenvironment, consisting of stromal cells, extracellular matrix, and cytokines such as TGF-β and TNF-α, plays a critical role in fibrosis and aberrant hematopoiesis. Clinically, MF manifests with symptoms ranging from anemia, splenomegaly, and fatigue to severe complications such as leukemic transformation. Splenomegaly, caused by extramedullary hematopoiesis, leads to abdominal discomfort and early satiety. Current therapeutic strategies include JAK inhibitors like Ruxolitinib, which target the JAK-STAT pathway, alongside supportive treatments such as blood transfusions, erythropoiesis-stimulating agents and developing combinatorial approaches. Allogeneic hematopoietic stem cell transplantation remains the only curative option, though it is limited to younger, high-risk patients. Recently approved JAK inhibitors, including Fedratinib, Pacritinib, and Momelotinib, have expanded the therapeutic landscape. Spatially Resolved Transcriptomics (SRT) has revolutionized the study of gene expression within the spatial context of tissues, providing unprecedented insights into cellular heterogeneity, spatial gene regulation, and microenvironmental interactions, including stromal-hematopoietic dynamics. SRT enables high-resolution mapping of gene expression in the BM and spleen, revealing molecular signatures, spatial heterogeneity, and pathological niches that drive disease progression. These technologies elucidate the role of the spleen in MF, highlighting its transformation into a site of abnormal hematopoietic activity, fibrotic changes, and immune cell infiltration, functioning as a "tumor surrogate." By profiling diverse cell populations and molecular alterations within the BM and spleen, SRT facilitates a deeper understanding of MF pathophysiology, helping identify novel therapeutic targets and biomarkers. Ultimately, integrating spatial transcriptomics into MF research promises to enhance diagnostic precision and therapeutic innovation, addressing the multifaceted challenges of this disease.
Collapse
Affiliation(s)
- Edoardo Peroni
- Immunology and Molecular Oncology Unit, Veneto Institute of Oncology, IOV-IRCCS, Padova, 35128, Italy.
| | - Elisabetta Calistri
- Onco-Hematology, Department of Oncology, Veneto Institute of Oncology, IOV-IRCCS, Padua, 31033, Italy
| | - Rosario Amato
- Medical Genetics Unit, Mater Domini University Hospital, Catanzaro, 88100, Italy
- Immuno-Genetics Lab, Department of Health Science, Medical School, University "Magna Graecia" of Catanzaro, Catanzaro, 88100, Italy
| | - Michele Gottardi
- Onco-Hematology, Department of Oncology, Veneto Institute of Oncology, IOV-IRCCS, Padua, 31033, Italy
| | - Antonio Rosato
- Immunology and Molecular Oncology Unit, Veneto Institute of Oncology, IOV-IRCCS, Padova, 35128, Italy
- Department of Surgery Oncology and Gastroenterology, University of Padova, Padova, 35122, Italy
| |
Collapse
|
7
|
Kim H, Kim KE, Madan E, Martin P, Gogna R, Rhee HW, Won KJ. Unveiling contact-mediated cellular crosstalk. Trends Genet 2024; 40:868-879. [PMID: 38906738 DOI: 10.1016/j.tig.2024.05.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Revised: 05/28/2024] [Accepted: 05/29/2024] [Indexed: 06/23/2024]
Abstract
Cell-cell interactions orchestrate complex functions in multicellular organisms, forming a regulatory network for diverse biological processes. Their disruption leads to disease states. Recent advancements - including single-cell sequencing and spatial transcriptomics, coupled with powerful bioengineering and molecular tools - have revolutionized our understanding of how cells respond to each other. Notably, spatial transcriptomics allows us to analyze gene expression changes based on cell proximity, offering a unique window into the impact of cell-cell contact. Additionally, computational approaches are being developed to decipher how cell contact governs the symphony of cellular responses. This review explores these cutting-edge approaches, providing valuable insights into deciphering the intricate cellular changes influenced by cell-cell communication.
Collapse
Affiliation(s)
- Hyobin Kim
- Department of Computational Biomedicine, Cedars-Sinai Medical Center, West, Hollywood, CA, USA
| | - Kwang-Eun Kim
- Department of Convergence Medicine, Yonsei University Wonju College of Medicine, Wonju, South Korea; Department of Chemistry, Seoul National University, Seoul, South Korea
| | - Esha Madan
- Massey Cancer Center, Virginia Commonwealth University, Richmond, VA, USA; School of Medicine, Institute of Molecular Medicine, Virginia Commonwealth University, Richmond, VA, USA; Department of Human and Molecular Genetics, School of Medicine, Virginia Commonwealth University, Richmond, VA, USA
| | - Patrick Martin
- Department of Computational Biomedicine, Cedars-Sinai Medical Center, West, Hollywood, CA, USA
| | - Rajan Gogna
- Massey Cancer Center, Virginia Commonwealth University, Richmond, VA, USA; School of Medicine, Institute of Molecular Medicine, Virginia Commonwealth University, Richmond, VA, USA; Department of Human and Molecular Genetics, School of Medicine, Virginia Commonwealth University, Richmond, VA, USA
| | - Hyun-Woo Rhee
- Department of Chemistry, Seoul National University, Seoul, South Korea.
| | - Kyoung-Jae Won
- Department of Computational Biomedicine, Cedars-Sinai Medical Center, West, Hollywood, CA, USA.
| |
Collapse
|
8
|
Polonsky M, Gerhardt LMS, Yun J, Koppitch K, Colón KL, Amrhein H, Wold B, Zheng S, Yuan GC, Thomson M, Cai L, McMahon AP. Spatial transcriptomics defines injury specific microenvironments and cellular interactions in kidney regeneration and disease. Nat Commun 2024; 15:7010. [PMID: 39237549 PMCID: PMC11377535 DOI: 10.1038/s41467-024-51186-z] [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: 01/02/2024] [Accepted: 08/01/2024] [Indexed: 09/07/2024] Open
Abstract
Kidney injury disrupts the intricate renal architecture and triggers limited regeneration, together with injury-invoked inflammation and fibrosis. Deciphering the molecular pathways and cellular interactions driving these processes is challenging due to the complex tissue structure. Here, we apply single cell spatial transcriptomics to examine ischemia-reperfusion injury in the mouse kidney. Spatial transcriptomics reveals injury-specific and spatially-dependent gene expression patterns in distinct cellular microenvironments within the kidney and predicts Clcf1-Crfl1 in a molecular interplay between persistently injured proximal tubule cells and their neighboring fibroblasts. Immune cell types play a critical role in organ repair. Spatial analysis identifies cellular microenvironments resembling early tertiary lymphoid structures and associated molecular pathways. Collectively, this study supports a focus on molecular interactions in cellular microenvironments to enhance understanding of injury, repair and disease.
Collapse
Affiliation(s)
- Michal Polonsky
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA
| | - Louisa M S Gerhardt
- Department of Stem Cell Biology and Regenerative Medicine, Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research, Keck School of Medicine of the University of Southern California, Los Angeles, California, USA
- Fifth Department of Medicine, University Medical Centre Mannheim, University of Heidelberg, Mannheim, Germany
| | - Jina Yun
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA
| | - Kari Koppitch
- Department of Stem Cell Biology and Regenerative Medicine, Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research, Keck School of Medicine of the University of Southern California, Los Angeles, California, USA
| | - Katsuya Lex Colón
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA
| | - Henry Amrhein
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA
| | - Barbara Wold
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA
| | - Shiwei Zheng
- Department of Genetics and Genomic Sciences and Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Guo-Cheng Yuan
- Department of Genetics and Genomic Sciences and Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Matt Thomson
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA
| | - Long Cai
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA.
| | - Andrew P McMahon
- Department of Stem Cell Biology and Regenerative Medicine, Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research, Keck School of Medicine of the University of Southern California, Los Angeles, California, USA.
| |
Collapse
|
9
|
Liu X, Peng T, Xu M, Lin S, Hu B, Chu T, Liu B, Xu Y, Ding W, Li L, Cao C, Wu P. Spatial multi-omics: deciphering technological landscape of integration of multi-omics and its applications. J Hematol Oncol 2024; 17:72. [PMID: 39182134 PMCID: PMC11344930 DOI: 10.1186/s13045-024-01596-9] [Citation(s) in RCA: 16] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Accepted: 08/09/2024] [Indexed: 08/27/2024] Open
Abstract
The emergence of spatial multi-omics has helped address the limitations of single-cell sequencing, which often leads to the loss of spatial context among cell populations. Integrated analysis of the genome, transcriptome, proteome, metabolome, and epigenome has enhanced our understanding of cell biology and the molecular basis of human diseases. Moreover, this approach offers profound insights into the interactions between intracellular and intercellular molecular mechanisms involved in the development, physiology, and pathogenesis of human diseases. In this comprehensive review, we examine current advancements in multi-omics technologies, focusing on their evolution and refinement over the past decade, including improvements in throughput and resolution, modality integration, and accuracy. We also discuss the pivotal contributions of spatial multi-omics in revealing spatial heterogeneity, constructing detailed spatial atlases, deciphering spatial crosstalk in tumor immunology, and advancing translational research and cancer therapy through precise spatial mapping.
Collapse
Affiliation(s)
- Xiaojie Liu
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- National Clinical Research Center for Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Cancer Biology Research Center (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Ting Peng
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- National Clinical Research Center for Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Cancer Biology Research Center (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Miaochun Xu
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- National Clinical Research Center for Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Cancer Biology Research Center (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Shitong Lin
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- National Clinical Research Center for Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Cancer Biology Research Center (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Bai Hu
- Department of Gynecologic Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- National Clinical Research Center for Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Cancer Biology Research Center (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Tian Chu
- Department of Gynecologic Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- National Clinical Research Center for Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Cancer Biology Research Center (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Binghan Liu
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- National Clinical Research Center for Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Cancer Biology Research Center (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Yashi Xu
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- National Clinical Research Center for Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Cancer Biology Research Center (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Wencheng Ding
- Department of Gynecologic Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- National Clinical Research Center for Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Cancer Biology Research Center (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Li Li
- Department of Gynecologic Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- National Clinical Research Center for Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Cancer Biology Research Center (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Canhui Cao
- Department of Gynecologic Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China.
- National Clinical Research Center for Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China.
- Cancer Biology Research Center (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China.
| | - Peng Wu
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China.
- National Clinical Research Center for Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China.
- Cancer Biology Research Center (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China.
| |
Collapse
|
10
|
Dezem FS, Arjumand W, DuBose H, Morosini NS, Plummer J. Spatially Resolved Single-Cell Omics: Methods, Challenges, and Future Perspectives. Annu Rev Biomed Data Sci 2024; 7:131-153. [PMID: 38768396 DOI: 10.1146/annurev-biodatasci-102523-103640] [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: 05/22/2024]
Abstract
Overlaying omics data onto spatial biological dimensions has been a promising technology to provide high-resolution insights into the interactome and cellular heterogeneity relative to the organization of the molecular microenvironment of tissue samples in normal and disease states. Spatial omics can be categorized into three major modalities: (a) next-generation sequencing-based assays, (b) imaging-based spatially resolved transcriptomics approaches including in situ hybridization/in situ sequencing, and (c) imaging-based spatial proteomics. These modalities allow assessment of transcripts and proteins at a cellular level, generating large and computationally challenging datasets. The lack of standardized computational pipelines to analyze and integrate these nonuniform structured data has made it necessary to apply artificial intelligence and machine learning strategies to best visualize and translate their complexity. In this review, we summarize the currently available techniques and computational strategies, highlight their advantages and limitations, and discuss their future prospects in the scientific field.
Collapse
Affiliation(s)
- Felipe Segato Dezem
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
- Center for Spatial Omics, St. Jude Children's Research Hospital, Memphis, Tennessee, USA;
| | - Wani Arjumand
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
- Center for Spatial Omics, St. Jude Children's Research Hospital, Memphis, Tennessee, USA;
| | - Hannah DuBose
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
- Center for Spatial Omics, St. Jude Children's Research Hospital, Memphis, Tennessee, USA;
| | - Natalia Silva Morosini
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
- Center for Spatial Omics, St. Jude Children's Research Hospital, Memphis, Tennessee, USA;
| | - Jasmine Plummer
- Department of Cellular and Molecular Biology and Comprehensive Cancer Center, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
- Center for Spatial Omics, St. Jude Children's Research Hospital, Memphis, Tennessee, USA;
| |
Collapse
|
11
|
Jackson KC, Booeshaghi AS, Gálvez-Merchán Á, Moses L, Chari T, Kim A, Pachter L. Identification of spatial homogeneous regions in tissues with concordex. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.06.28.546949. [PMID: 39071320 PMCID: PMC11275758 DOI: 10.1101/2023.06.28.546949] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/30/2024]
Abstract
Spatial homogeneous regions (SHRs) in tissues are domains that are homogeneous with respect to cell type composition. We present a method for identifying SHRs using spatial transcriptomics data, and demonstrate that it is efficient and effective at finding SHRs for a wide variety of tissue types. The method is implemented in a tool called concordex, which relies on analysis of k-nearest-neighbor (kNN) graphs. The concordex tool is also useful for analysis of non-spatial transcriptomics data, and can elucidate the extent of concordance between partitions of cells derived from clustering algorithms, and transcriptomic similarity as represented in kNN graphs.
Collapse
Affiliation(s)
- Kayla C Jackson
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA
- Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - A Sina Booeshaghi
- Department of Bioengineering, University of California, Berkeley, CA, USA
| | | | - Lambda Moses
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA
| | - Tara Chari
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA
| | | | - Lior Pachter
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA
- Department of Computing and Mathematical Sciences, California Institute of Technology, Pasadena, CA, USA
| |
Collapse
|
12
|
Liao L, Martin PCN, Kim H, Panahandeh S, Won KJ. Data enhancement in the age of spatial biology. Adv Cancer Res 2024; 163:39-70. [PMID: 39271267 DOI: 10.1016/bs.acr.2024.06.008] [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: 09/15/2024]
Abstract
Unveiling the intricate interplay of cells in their native environment lies at the heart of understanding fundamental biological processes and unraveling disease mechanisms, particularly in complex diseases like cancer. Spatial transcriptomics (ST) offers a revolutionary lens into the spatial organization of gene expression within tissues, empowering researchers to study both cell heterogeneity and microenvironments in health and disease. However, current ST technologies often face limitations in either resolution or the number of genes profiled simultaneously. Integrating ST data with complementary sources, such as single-cell transcriptomics and detailed tissue staining images, presents a powerful solution to overcome these limitations. This review delves into the computational approaches driving the integration of spatial transcriptomics with other data types. By illuminating the key challenges and outlining the current algorithmic solutions, we aim to highlight the immense potential of these methods to revolutionize our understanding of cancer biology.
Collapse
Affiliation(s)
- Linbu Liao
- Biotech Research and Innovation Centre (BRIC), University of Copenhagen, Denmark; Samuel Oschin Cancer Center, Cedars-Sinai Medical Center, Los Angeles, CA, United States
| | - Patrick C N Martin
- Department of Computational Biomedicine, Cedars-Sinai Medical Center, Los Angeles, CA, United States
| | - Hyobin Kim
- Department of Computational Biomedicine, Cedars-Sinai Medical Center, Los Angeles, CA, United States
| | - Sanaz Panahandeh
- Department of Computational Biomedicine, Cedars-Sinai Medical Center, Los Angeles, CA, United States
| | - Kyoung Jae Won
- Department of Computational Biomedicine, Cedars-Sinai Medical Center, Los Angeles, CA, United States.
| |
Collapse
|
13
|
Ma Y, Shi W, Dong Y, Sun Y, Jin Q. Spatial Multi-Omics in Alzheimer's Disease: A Multi-Dimensional Approach to Understanding Pathology and Progression. Curr Issues Mol Biol 2024; 46:4968-4990. [PMID: 38785566 PMCID: PMC11119029 DOI: 10.3390/cimb46050298] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2024] [Revised: 05/14/2024] [Accepted: 05/15/2024] [Indexed: 05/25/2024] Open
Abstract
Alzheimer's Disease (AD) presents a complex neuropathological landscape characterized by hallmark amyloid plaques and neurofibrillary tangles, leading to progressive cognitive decline. Despite extensive research, the molecular intricacies contributing to AD pathogenesis are inadequately understood. While single-cell omics technology holds great promise for application in AD, particularly in deciphering the understanding of different cell types and analyzing rare cell types and transcriptomic expression changes, it is unable to provide spatial distribution information, which is crucial for understanding the pathological processes of AD. In contrast, spatial multi-omics research emerges as a promising and comprehensive approach to analyzing tissue cells, potentially better suited for addressing these issues in AD. This article focuses on the latest advancements in spatial multi-omics technology and compares various techniques. Additionally, we provide an overview of current spatial omics-based research results in AD. These technologies play a crucial role in facilitating new discoveries and advancing translational AD research in the future. Despite challenges such as balancing resolution, increasing throughput, and data analysis, the application of spatial multi-omics holds immense potential in revolutionizing our understanding of human disease processes and identifying new biomarkers and therapeutic targets, thereby potentially contributing to the advancement of AD research.
Collapse
Affiliation(s)
| | | | | | | | - Qiguan Jin
- College of Physical Education, Yangzhou University, Yangzhou 225127, China; (Y.M.); (W.S.); (Y.D.); (Y.S.)
| |
Collapse
|
14
|
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
|
15
|
Hao M, Luo E, Chen Y, Wu Y, Li C, Chen S, Gao H, Bian H, Gu J, Wei L, Zhang X. STEM enables mapping of single-cell and spatial transcriptomics data with transfer learning. Commun Biol 2024; 7:56. [PMID: 38184694 PMCID: PMC10771471 DOI: 10.1038/s42003-023-05640-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Accepted: 11/27/2023] [Indexed: 01/08/2024] Open
Abstract
Profiling spatial variations of cellular composition and transcriptomic characteristics is important for understanding the physiology and pathology of tissues. Spatial transcriptomics (ST) data depict spatial gene expression but the currently dominating high-throughput technology is yet not at single-cell resolution. Single-cell RNA-sequencing (SC) data provide high-throughput transcriptomic information at the single-cell level but lack spatial information. Integrating these two types of data would be ideal for revealing transcriptomic landscapes at single-cell resolution. We develop the method STEM (SpaTially aware EMbedding) for this purpose. It uses deep transfer learning to encode both ST and SC data into a unified spatially aware embedding space, and then uses the embeddings to infer SC-ST mapping and predict pseudo-spatial adjacency between cells in SC data. Semi-simulation and real data experiments verify that the embeddings preserved spatial information and eliminated technical biases between SC and ST data. We apply STEM to human squamous cell carcinoma and hepatic lobule datasets to uncover the localization of rare cell types and reveal cell-type-specific gene expression variation along a spatial axis. STEM is powerful for mapping SC and ST data to build single-cell level spatial transcriptomic landscapes, and can provide mechanistic insights into the spatial heterogeneity and microenvironments of tissues.
Collapse
Affiliation(s)
- Minsheng Hao
- MOE Key Laboratory of Bioinformatics and Bioinformatics Division, BNRIST, Department of Automation, Tsinghua University, Beijing, 100084, China
| | - Erpai Luo
- MOE Key Laboratory of Bioinformatics and Bioinformatics Division, BNRIST, Department of Automation, Tsinghua University, Beijing, 100084, China
| | - Yixin Chen
- MOE Key Laboratory of Bioinformatics and Bioinformatics Division, BNRIST, Department of Automation, Tsinghua University, Beijing, 100084, China
| | - Yanhong Wu
- MOE Key Laboratory of Bioinformatics and Bioinformatics Division, BNRIST, Department of Automation, Tsinghua University, Beijing, 100084, China
| | - Chen Li
- MOE Key Laboratory of Bioinformatics and Bioinformatics Division, BNRIST, Department of Automation, Tsinghua University, Beijing, 100084, China
| | - Sijie Chen
- MOE Key Laboratory of Bioinformatics and Bioinformatics Division, BNRIST, Department of Automation, Tsinghua University, Beijing, 100084, China
| | - Haoxiang Gao
- MOE Key Laboratory of Bioinformatics and Bioinformatics Division, BNRIST, Department of Automation, Tsinghua University, Beijing, 100084, China
| | - Haiyang Bian
- MOE Key Laboratory of Bioinformatics and Bioinformatics Division, BNRIST, Department of Automation, Tsinghua University, Beijing, 100084, China
| | - Jin Gu
- MOE Key Laboratory of Bioinformatics and Bioinformatics Division, BNRIST, Department of Automation, Tsinghua University, Beijing, 100084, China
| | - Lei Wei
- MOE Key Laboratory of Bioinformatics and Bioinformatics Division, BNRIST, Department of Automation, Tsinghua University, Beijing, 100084, China.
| | - Xuegong Zhang
- MOE Key Laboratory of Bioinformatics and Bioinformatics Division, BNRIST, Department of Automation, Tsinghua University, Beijing, 100084, China.
- School of Life Sciences and School of Medicine, Center for Synthetic and Systems Biology, Tsinghua University, Beijing, 100084, China.
| |
Collapse
|
16
|
Polonsky M, Gerhardt LMS, Yun J, Koppitch K, Colón KL, Amrhein H, Zheng S, Yuan GC, Thomson M, Cai L, McMahon AP. Spatial transcriptomics defines injury-specific microenvironments in the adult mouse kidney and novel cellular interactions in regeneration and disease. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.11.22.568217. [PMID: 38045285 PMCID: PMC10690238 DOI: 10.1101/2023.11.22.568217] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/05/2023]
Abstract
Kidney injury disrupts the intricate renal architecture and triggers limited regeneration, and injury-invoked inflammation and fibrosis. Deciphering molecular pathways and cellular interactions driving these processes is challenging due to the complex renal architecture. Here, we applied single cell spatial transcriptomics to examine ischemia-reperfusion injury in the mouse kidney. Spatial transcriptomics revealed injury-specific and spatially-dependent gene expression patterns in distinct cellular microenvironments within the kidney and predicted Clcf1-Crfl1 in a molecular interplay between persistently injured proximal tubule cells and neighboring fibroblasts. Immune cell types play a critical role in organ repair. Spatial analysis revealed cellular microenvironments resembling early tertiary lymphoid structures and identified associated molecular pathways. Collectively, this study supports a focus on molecular interactions in cellular microenvironments to enhance understanding of injury, repair and disease.
Collapse
Affiliation(s)
- Michal Polonsky
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA
| | - Louisa M. S. Gerhardt
- Department of Stem Cell Biology and Regenerative Medicine, Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research, Keck School of Medicine of the University of Southern California, Los Angeles, California
| | - Jina Yun
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA
| | - Kari Koppitch
- Department of Stem Cell Biology and Regenerative Medicine, Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research, Keck School of Medicine of the University of Southern California, Los Angeles, California
| | - Katsuya Lex Colón
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA
| | - Henry Amrhein
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA
| | - Shiwei Zheng
- Department of Genetics and Genomic Sciences and Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Guo-Cheng Yuan
- Department of Genetics and Genomic Sciences and Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Matt Thomson
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA
| | - Long Cai
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA
| | - Andrew P. McMahon
- Department of Stem Cell Biology and Regenerative Medicine, Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research, Keck School of Medicine of the University of Southern California, Los Angeles, California
| |
Collapse
|
17
|
Paas-Oliveros E, Hernández-Lemus E, de Anda-Jáuregui G. Computational single cell oncology: state of the art. Front Genet 2023; 14:1256991. [PMID: 38028624 PMCID: PMC10663273 DOI: 10.3389/fgene.2023.1256991] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Accepted: 10/24/2023] [Indexed: 12/01/2023] Open
Abstract
Single cell computational analysis has emerged as a powerful tool in the field of oncology, enabling researchers to decipher the complex cellular heterogeneity that characterizes cancer. By leveraging computational algorithms and bioinformatics approaches, this methodology provides insights into the underlying genetic, epigenetic and transcriptomic variations among individual cancer cells. In this paper, we present a comprehensive overview of single cell computational analysis in oncology, discussing the key computational techniques employed for data processing, analysis, and interpretation. We explore the challenges associated with single cell data, including data quality control, normalization, dimensionality reduction, clustering, and trajectory inference. Furthermore, we highlight the applications of single cell computational analysis, including the identification of novel cell states, the characterization of tumor subtypes, the discovery of biomarkers, and the prediction of therapy response. Finally, we address the future directions and potential advancements in the field, including the development of machine learning and deep learning approaches for single cell analysis. Overall, this paper aims to provide a roadmap for researchers interested in leveraging computational methods to unlock the full potential of single cell analysis in understanding cancer biology with the goal of advancing precision oncology. For this purpose, we also include a notebook that instructs on how to apply the recommended tools in the Preprocessing and Quality Control section.
Collapse
Affiliation(s)
- Ernesto Paas-Oliveros
- Computational Genomics Division, National Institute of Genomic Medicine, Mexico City, Mexico
| | - Enrique Hernández-Lemus
- Computational Genomics Division, National Institute of Genomic Medicine, Mexico City, Mexico
- Center for Complexity Sciences, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Guillermo de Anda-Jáuregui
- Computational Genomics Division, National Institute of Genomic Medicine, Mexico City, Mexico
- Center for Complexity Sciences, Universidad Nacional Autónoma de México, Mexico City, Mexico
- Investigadores por Mexico, Conahcyt, Mexico City, Mexico
| |
Collapse
|
18
|
Liang G, Yin H, Ding F. Technical Advances and Applications of Spatial Transcriptomics. GEN BIOTECHNOLOGY 2023; 2:384-398. [PMID: 39544230 PMCID: PMC11562938 DOI: 10.1089/genbio.2023.0032] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/17/2024]
Abstract
Transcriptomics is one of the largest areas of research in biological sciences. Aside from RNA expression levels, the significance of RNA spatial context has also been unveiled in the recent decade, playing a critical role in diverse biological processes, from subcellular kinetic regulation to cell communication, from tissue architecture to tumor microenvironment, and more. To systematically unravel the positional patterns of RNA molecules across subcellular, cellular, and tissue levels, spatial transcriptomics techniques have emerged and rapidly became an irreplaceable tool set. Herein, we review and compare current spatial transcriptomics techniques on their methods, advantages, and limitations, as well as applications across a wide range of biological investigations. This review serves as a comprehensive guide to spatial transcriptomics for researchers interested in adopting this powerful suite of technologies.
Collapse
Affiliation(s)
- Guohao Liang
- Department of Biomedical Engineering, University of California, Irvine, Irvine, California, USA
| | - Hong Yin
- Department of Biomedical Engineering, University of California, Irvine, Irvine, California, USA
| | - Fangyuan Ding
- Center for Synthetic Biology, Center for Complex Biological Systems, Department of Developmental and Cell Biology, and Department of Pharmaceutical Sciences, University of California, Irvine, California, USA
| |
Collapse
|
19
|
Jung N, Kim TK. Spatial transcriptomics in neuroscience. Exp Mol Med 2023; 55:2105-2115. [PMID: 37779145 PMCID: PMC10618223 DOI: 10.1038/s12276-023-01093-y] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 07/07/2023] [Accepted: 07/09/2023] [Indexed: 10/03/2023] Open
Abstract
The brain is one of the most complex living tissue types and is composed of an exceptional diversity of cell types displaying unique functional connectivity. Single-cell RNA sequencing (scRNA-seq) can be used to efficiently map the molecular identities of the various cell types in the brain by providing the transcriptomic profiles of individual cells isolated from the tissue. However, the lack of spatial context in scRNA-seq prevents a comprehensive understanding of how different configurations of cell types give rise to specific functions in individual brain regions and how each distinct cell is connected to form a functional unit. To understand how the various cell types contribute to specific brain functions, it is crucial to correlate the identities of individual cells obtained through scRNA-seq with their spatial information in intact tissue. Spatial transcriptomics (ST) can resolve the complex spatial organization of cell types in the brain and their connectivity. Various ST tools developed during the past decade based on imaging and sequencing technology have permitted the creation of functional atlases of the brain and have pulled the properties of neural circuits into ever-sharper focus. In this review, we present a summary of several ST tools and their applications in neuroscience and discuss the unprecedented insights these tools have made possible.
Collapse
Affiliation(s)
- Namyoung Jung
- Department of Life Sciences, Pohang University of Science and Technology (POSTECH), Pohang, Gyeongbuk, 37673, Republic of Korea
| | - Tae-Kyung Kim
- Department of Life Sciences, Pohang University of Science and Technology (POSTECH), Pohang, Gyeongbuk, 37673, Republic of Korea.
- Institute for Convergence Research and Education in Advanced Technology, Yonsei University, Seoul, 03722, Republic of Korea.
| |
Collapse
|
20
|
Seal S, Bitler BG, Ghosh D. SMASH: Scalable Method for Analyzing Spatial Heterogeneity of genes in spatial transcriptomics data. PLoS Genet 2023; 19:e1010983. [PMID: 37862362 PMCID: PMC10619839 DOI: 10.1371/journal.pgen.1010983] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Revised: 11/01/2023] [Accepted: 09/19/2023] [Indexed: 10/22/2023] Open
Abstract
In high-throughput spatial transcriptomics (ST) studies, it is of great interest to identify the genes whose level of expression in a tissue covaries with the spatial location of cells/spots. Such genes, also known as spatially variable genes (SVGs), can be crucial to the biological understanding of both structural and functional characteristics of complex tissues. Existing methods for detecting SVGs either suffer from huge computational demand or significantly lack statistical power. We propose a non-parametric method termed SMASH that achieves a balance between the above two problems. We compare SMASH with other existing methods in varying simulation scenarios demonstrating its superior statistical power and robustness. We apply the method to four ST datasets from different platforms uncovering interesting biological insights.
Collapse
Affiliation(s)
- Souvik Seal
- Department of Public Health Sciences, School of Medicine, Medical University of South Carolina, Charleston, South Carolina, United States of America
| | - Benjamin G. Bitler
- Department of Obstetrics and Gynecology, School of Medicine, University of Colorado Denver Anschutz Medical Campus, Aurora, Colorado, United States of America
| | - Debashis Ghosh
- Department of Biostatistics and Informatics, Colorado School of Public Health, University of Colorado Denver Anschutz Medical Campus, Aurora, Colorado, United States of America
| |
Collapse
|
21
|
Halawani R, Buchert M, Chen YPP. Deep learning exploration of single-cell and spatially resolved cancer transcriptomics to unravel tumour heterogeneity. Comput Biol Med 2023; 164:107274. [PMID: 37506451 DOI: 10.1016/j.compbiomed.2023.107274] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2023] [Revised: 07/03/2023] [Accepted: 07/16/2023] [Indexed: 07/30/2023]
Abstract
Tumour heterogeneity is one of the critical confounding aspects in decoding tumour growth. Malignant cells display variations in their gene transcription profiles and mutation spectra even when originating from a single progenitor cell. Single-cell and spatial transcriptomics sequencing have recently emerged as key technologies for unravelling tumour heterogeneity. Single-cell sequencing promotes individual cell-type identification through transcriptome-wide gene expression measurements of each cell. Spatial transcriptomics facilitates identification of cell-cell interactions and the structural organization of heterogeneous cells within a tumour tissue through associating spatial RNA abundance of cells at distinct spots in the tissue section. However, extracting features and analyzing single-cell and spatial transcriptomics data poses challenges. Single-cell transcriptome data is extremely noisy and its sparse nature and dropouts can lead to misinterpretation of gene expression and the misclassification of cell types. Deep learning predictive power can overcome data challenges, provide high-resolution analysis and enhance precision oncology applications that involve early cancer prognosis, diagnosis, patient survival estimation and anti-cancer therapy planning. In this paper, we provide a background to and review of the recent progress of deep learning frameworks to investigate tumour heterogeneity using both single-cell and spatial transcriptomics data types.
Collapse
Affiliation(s)
- Raid Halawani
- Department of Computer Science and Information Technology, La Trobe University, Melbourne, Australia
| | - Michael Buchert
- School of Cancer Medicine, La Trobe University, Melbourne, Victoria, Australia; Olivia Newton-John Cancer Research Institute, Melbourne, Victoria, Australia
| | - Yi-Ping Phoebe Chen
- Department of Computer Science and Information Technology, La Trobe University, Melbourne, Australia.
| |
Collapse
|
22
|
Jackson KC, Pachter L. A standard for sharing spatial transcriptomics data. CELL GENOMICS 2023; 3:100374. [PMID: 37601972 PMCID: PMC10435375 DOI: 10.1016/j.xgen.2023.100374] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/22/2023]
Abstract
Spatial transcriptomic technologies have the potential to reveal critical relationships between the function of genes and cells and their spatial organization. Here, we provide a sharing model for spatial transcriptomics data with the aim of establishing a set of primary data and metadata needed to reproduce analyses and facilitate computational methods development.
Collapse
Affiliation(s)
- Kayla C. Jackson
- Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA
| | - Lior Pachter
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA
- Department of Computing and Mathematical Sciences, California Institute of Technology, Pasadena, CA, USA
| |
Collapse
|
23
|
Vandereyken K, Sifrim A, Thienpont B, Voet T. Methods and applications for single-cell and spatial multi-omics. Nat Rev Genet 2023; 24:494-515. [PMID: 36864178 PMCID: PMC9979144 DOI: 10.1038/s41576-023-00580-2] [Citation(s) in RCA: 461] [Impact Index Per Article: 230.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/20/2023] [Indexed: 03/04/2023]
Abstract
The joint analysis of the genome, epigenome, transcriptome, proteome and/or metabolome from single cells is transforming our understanding of cell biology in health and disease. In less than a decade, the field has seen tremendous technological revolutions that enable crucial new insights into the interplay between intracellular and intercellular molecular mechanisms that govern development, physiology and pathogenesis. In this Review, we highlight advances in the fast-developing field of single-cell and spatial multi-omics technologies (also known as multimodal omics approaches), and the computational strategies needed to integrate information across these molecular layers. We demonstrate their impact on fundamental cell biology and translational research, discuss current challenges and provide an outlook to the future.
Collapse
Affiliation(s)
- Katy Vandereyken
- KU Leuven Institute for Single Cell Omics (LISCO), University of Leuven, KU Leuven, Leuven, Belgium
- Department of Human Genetics, University of Leuven, KU Leuven, Leuven, Belgium
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, USA
| | - Alejandro Sifrim
- KU Leuven Institute for Single Cell Omics (LISCO), University of Leuven, KU Leuven, Leuven, Belgium
- Department of Human Genetics, University of Leuven, KU Leuven, Leuven, Belgium
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, USA
| | - Bernard Thienpont
- KU Leuven Institute for Single Cell Omics (LISCO), University of Leuven, KU Leuven, Leuven, Belgium
- Department of Human Genetics, University of Leuven, KU Leuven, Leuven, Belgium
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, USA
| | - Thierry Voet
- KU Leuven Institute for Single Cell Omics (LISCO), University of Leuven, KU Leuven, Leuven, Belgium.
- Department of Human Genetics, University of Leuven, KU Leuven, Leuven, Belgium.
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, USA.
| |
Collapse
|
24
|
Zhang X, Lu P, Shen X. Morphologies and potential roles of telocytes in nervous tissue. CLINICAL AND TRANSLATIONAL DISCOVERY 2023; 3. [DOI: 10.1002/ctd2.186] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Accepted: 03/16/2023] [Indexed: 01/03/2025]
Abstract
AbstractStructurally similar cells have been found and termed telocytes (TCs) since the first characterisation of interstitial Cajal‐like cells in 1911. TCs are a novel and peculiar interstitial cell type with a small cellular body, markedly long cell processes named telopodes and a wide distribution in numerous tissues throughout the body. Besides specific morphological characteristics and immunohistochemical profiles, TCs build three‐dimensional mixed networks through homocellular (connection to each other) and/or heterocellular contacts (connection with other cell types), interaction with extracellular matrix and their vicinity to nerve endings, and thus might play, as part of an integrated system, roles in maintaining organ/tissue function. In this mini‐review, we summarise physical properties, general characteristics and distribution of TCs in diverse organs and tissues, focusing on their potential functions in nervous tissue and current challenges in investigating TCs as a distinct cell type.
Collapse
Affiliation(s)
- Xu Zhang
- Institute for Reproductive Health Shanghai Institute for Biomedical and Pharmaceutical Technologies Shanghai China
| | - Ping Lu
- Institute for Reproductive Health Shanghai Institute for Biomedical and Pharmaceutical Technologies Shanghai China
| | - Xiaorong Shen
- Institute for Reproductive Health Shanghai Institute for Biomedical and Pharmaceutical Technologies Shanghai China
| |
Collapse
|
25
|
Seal S, Bitler BG, Ghosh D. SMASH: Scalable Method for Analyzing Spatial Heterogeneity of genes in spatial transcriptomics data. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.03.23.533980. [PMID: 36993287 PMCID: PMC10055313 DOI: 10.1101/2023.03.23.533980] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/30/2023]
Abstract
In high-throughput spatial transcriptomics (ST) studies, it is of great interest to identify the genes whose level of expression in a tissue covaries with the spatial location of cells/spots. Such genes, also known as spatially variable genes (SVGs), can be crucial to the biological understanding of both structural and functional characteristics of complex tissues. Existing methods for detecting SVGs either suffer from huge computational demand or significantly lack statistical power. We propose a non-parametric method termed SMASH that achieves a balance between the above two problems. We compare SMASH with other existing methods in varying simulation scenarios demonstrating its superior statistical power and robustness. We apply the method to four ST datasets from different platforms revealing interesting biological insights.
Collapse
Affiliation(s)
- Souvik Seal
- Department of Public Health Sciences, School of Medicine, Medical University of South Carolina, Charleston, USA
| | - Benjamin G. Bitler
- Department of Obstetrics and Gynecology, School of Medicine, University of Colorado Denver - Anschutz Medical Campus, Aurora, USA
| | - Debashis Ghosh
- Department of Biostatistics and Informatics, Colorado School of Public Health, University of Colorado Denver - Anschutz Medical Campus, Aurora, USA
| |
Collapse
|
26
|
Bagley JR, Denes LT, McCarthy JJ, Wang ET, Murach KA. The myonuclear domain in adult skeletal muscle fibres: past, present and future. J Physiol 2023; 601:723-741. [PMID: 36629254 PMCID: PMC9931674 DOI: 10.1113/jp283658] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Accepted: 01/06/2023] [Indexed: 01/12/2023] Open
Abstract
Most cells in the body are mononuclear whereas skeletal muscle fibres are uniquely multinuclear. The nuclei of muscle fibres (myonuclei) are usually situated peripherally which complicates the equitable distribution of gene products. Myonuclear abundance can also change under conditions such as hypertrophy and atrophy. Specialised zones in muscle fibres have different functions and thus distinct synthetic demands from myonuclei. The complex structure and regulatory requirements of multinuclear muscle cells understandably led to the hypothesis that myonuclei govern defined 'domains' to maintain homeostasis and facilitate adaptation. The purpose of this review is to provide historical context for the myonuclear domain and evaluate its veracity with respect to mRNA and protein distribution resulting from myonuclear transcription. We synthesise insights from past and current in vitro and in vivo genetically modified models for studying the myonuclear domain under dynamic conditions. We also cover the most contemporary knowledge on mRNA and protein transport in muscle cells. Insights from emerging technologies such as single myonuclear RNA-sequencing further inform our discussion of the myonuclear domain. We broadly conclude: (1) the myonuclear domain can be flexible during muscle fibre growth and atrophy, (2) the mechanisms and role of myonuclear loss and motility deserve further consideration, (3) mRNA in muscle is actively transported via microtubules and locally restricted, but proteins may travel far from a myonucleus of origin and (4) myonuclear transcriptional specialisation extends beyond the classic neuromuscular and myotendinous populations. A deeper understanding of the myonuclear domain in muscle may promote effective therapies for ageing and disease.
Collapse
Affiliation(s)
- James R. Bagley
- Muscle Physiology Laboratory, Department of Kinesiology, San Francisco State University, San Francisco, California
| | | | - John J. McCarthy
- The Center for Muscle Biology, University of Kentucky, Lexington, Kentucky
- Department of Physiology, College of Medicine, University of Kentucky
| | - Eric T. Wang
- Department of Molecular Genetics and Microbiology, Center for NeuroGenetics, University of Florida, Gainesville, Florida
- Myology Institute, University of Florida
- Genetics Institute, University of Florida
| | - Kevin A. Murach
- Exercise Science Research Center, Department of Health, Human Performance, and Recreation, University of Arkansas, Fayetteville, Arkansas
- Cell and Molecular Biology Graduate Program, University of Arkansas
| |
Collapse
|
27
|
Milanese JS, Marcotte R, Costain WJ, Kablar B, Drouin S. Roles of Skeletal Muscle in Development: A Bioinformatics and Systems Biology Overview. ADVANCES IN ANATOMY, EMBRYOLOGY, AND CELL BIOLOGY 2023; 236:21-55. [PMID: 37955770 DOI: 10.1007/978-3-031-38215-4_2] [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: 11/14/2023]
Abstract
The ability to assess various cellular events consequent to perturbations, such as genetic mutations, disease states and therapies, has been recently revolutionized by technological advances in multiple "omics" fields. The resulting deluge of information has enabled and necessitated the development of tools required to both process and interpret the data. While of tremendous value to basic researchers, the amount and complexity of the data has made it extremely difficult to manually draw inference and identify factors key to the study objectives. The challenges of data reduction and interpretation are being met by the development of increasingly complex tools that integrate disparate knowledge bases and synthesize coherent models based on current biological understanding. This chapter presents an example of how genomics data can be integrated with biological network analyses to gain further insight into the developmental consequences of genetic perturbations. State of the art methods for conducting similar studies are discussed along with modern methods used to analyze and interpret the data.
Collapse
Affiliation(s)
| | - Richard Marcotte
- Human Health Therapeutics, National Research Council of Canada , Montreal, QC, Canada
| | - Willard J Costain
- Human Health Therapeutics, National Research Council of Canada, Ottawa, ON, Canada
| | - Boris Kablar
- Department of Medical Neuroscience, Anatomy and Pathology, Faculty of Medicine, Dalhousie University, Halifax, NS, Canada
| | - Simon Drouin
- Human Health Therapeutics, National Research Council of Canada , Montreal, QC, Canada.
| |
Collapse
|
28
|
Xing Y, Zan C, Liu L. Recent advances in understanding neuronal diversity and neural circuit complexity across different brain regions using single-cell sequencing. Front Neural Circuits 2023; 17:1007755. [PMID: 37063385 PMCID: PMC10097998 DOI: 10.3389/fncir.2023.1007755] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2022] [Accepted: 02/16/2023] [Indexed: 04/18/2023] Open
Abstract
Neural circuits are characterized as interconnecting neuron networks connected by synapses. Some kinds of gene expression and/or functional changes of neurons and synaptic connections may result in aberrant neural circuits, which has been recognized as one crucial pathological mechanism for the onset of many neurological diseases. Gradual advances in single-cell sequencing approaches with strong technological advantages, as exemplified by high throughput and increased resolution for live cells, have enabled it to assist us in understanding neuronal diversity across diverse brain regions and further transformed our knowledge of cellular building blocks of neural circuits through revealing numerous molecular signatures. Currently published transcriptomic studies have elucidated various neuronal subpopulations as well as their distribution across prefrontal cortex, hippocampus, hypothalamus, and dorsal root ganglion, etc. Better characterization of brain region-specific circuits may shed light on new pathological mechanisms involved and assist in selecting potential targets for the prevention and treatment of specific neurological disorders based on their established roles. Given diverse neuronal populations across different brain regions, we aim to give a brief sketch of current progress in understanding neuronal diversity and neural circuit complexity according to their locations. With the special focus on the application of single-cell sequencing, we thereby summarize relevant region-specific findings. Considering the importance of spatial context and connectivity in neural circuits, we also discuss a few published results obtained by spatial transcriptomics. Taken together, these single-cell sequencing data may lay a mechanistic basis for functional identification of brain circuit components, which links their molecular signatures to anatomical regions, connectivity, morphology, and physiology. Furthermore, the comprehensive characterization of neuron subtypes, their distributions, and connectivity patterns via single-cell sequencing is critical for understanding neural circuit properties and how they generate region-dependent interactions in different context.
Collapse
Affiliation(s)
- Yu Xing
- Department of Neurology, Beidahuang Industry Group General Hospital, Harbin, China
| | - Chunfang Zan
- Institute for Stroke and Dementia Research (ISD), LMU Klinikum, Ludwig-Maximilian-University (LMU), Munich, Germany
| | - Lu Liu
- Munich Medical Research School (MMRS), LMU Klinikum, Ludwig-Maximilian-University (LMU), Munich, Germany
- *Correspondence: Lu Liu, ,
| |
Collapse
|
29
|
Ya D, Zhang Y, Cui Q, Jiang Y, Yang J, Tian N, Xiang W, Lin X, Li Q, Liao R. Application of spatial transcriptome technologies to neurological diseases. Front Cell Dev Biol 2023; 11:1142923. [PMID: 36936681 PMCID: PMC10020196 DOI: 10.3389/fcell.2023.1142923] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Accepted: 02/23/2023] [Indexed: 03/06/2023] Open
Abstract
Spatial transcriptome technology acquires gene expression profiles while retaining spatial location information, it displays the gene expression properties of cells in situ. Through the investigation of cell heterogeneity, microenvironment, function, and cellular interactions, spatial transcriptome technology can deeply explore the pathogenic mechanisms of cell-type-specific responses and spatial localization in neurological diseases. The present article overviews spatial transcriptome technologies based on microdissection, in situ hybridization, in situ sequencing, in situ capture, and live cell labeling. Each technology is described along with its methods, detection throughput, spatial resolution, benefits, and drawbacks. Furthermore, their applications in neurodegenerative disease, neuropsychiatric illness, stroke and epilepsy are outlined. This information can be used to understand disease mechanisms, pick therapeutic targets, and establish biomarkers.
Collapse
Affiliation(s)
- Dongshan Ya
- Laboratory of Neuroscience, Affiliated Hospital of Guilin Medical University, Guilin Medical University, Guilin, China
- Department of Neurology, Affiliated Hospital of Guilin Medical University, Guilin Medical University, Guilin, China
| | - Yingmei Zhang
- Laboratory of Neuroscience, Affiliated Hospital of Guilin Medical University, Guilin Medical University, Guilin, China
- Department of Neurology, Affiliated Hospital of Guilin Medical University, Guilin Medical University, Guilin, China
| | - Qi Cui
- Laboratory of Neuroscience, Affiliated Hospital of Guilin Medical University, Guilin Medical University, Guilin, China
- Department of Neurology, Affiliated Hospital of Guilin Medical University, Guilin Medical University, Guilin, China
| | - Yanlin Jiang
- Department of Pharmacology, Affiliated Hospital of Guilin Medical University, Guilin Medical University, Guilin, China
| | - Jiaxin Yang
- Laboratory of Neuroscience, Affiliated Hospital of Guilin Medical University, Guilin Medical University, Guilin, China
- Department of Neurology, Affiliated Hospital of Guilin Medical University, Guilin Medical University, Guilin, China
| | - Ning Tian
- Laboratory of Neuroscience, Affiliated Hospital of Guilin Medical University, Guilin Medical University, Guilin, China
- Guangxi Clinical Research Center for Neurological Diseases, Affiliated Hospital of Guilin Medical University, Guilin Medical University, Guilin, China
| | - Wenjing Xiang
- Department of Neurology ward 2, Guilin People’s Hospital, Guilin, China
| | - Xiaohui Lin
- Department of Geriatrics, Affiliated Hospital of Guilin Medical University, Guilin Medical University, Guilin, China
| | - Qinghua Li
- Laboratory of Neuroscience, Affiliated Hospital of Guilin Medical University, Guilin Medical University, Guilin, China
- Department of Neurology, Affiliated Hospital of Guilin Medical University, Guilin Medical University, Guilin, China
- Guangxi Clinical Research Center for Neurological Diseases, Affiliated Hospital of Guilin Medical University, Guilin Medical University, Guilin, China
| | - Rujia Liao
- Laboratory of Neuroscience, Affiliated Hospital of Guilin Medical University, Guilin Medical University, Guilin, China
- Department of Neurology, Affiliated Hospital of Guilin Medical University, Guilin Medical University, Guilin, China
- Guangxi Clinical Research Center for Neurological Diseases, Affiliated Hospital of Guilin Medical University, Guilin Medical University, Guilin, China
- *Correspondence: Rujia Liao,
| |
Collapse
|
30
|
Lund JB, Lindberg EL, Maatz H, Pottbaecker F, Hübner N, Lippert C. AntiSplodge: a neural-network-based RNA-profile deconvolution pipeline designed for spatial transcriptomics. NAR Genom Bioinform 2022; 4:lqac073. [PMID: 36225530 PMCID: PMC9549785 DOI: 10.1093/nargab/lqac073] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2021] [Revised: 08/30/2022] [Accepted: 09/30/2022] [Indexed: 11/14/2022] Open
Abstract
With the current surge of spatial transcriptomics (ST) studies, researchers are exploring the deep interactive cell-play directly in tissues, in situ. However, with the current technologies, measurements consist of mRNA transcript profiles of mixed origin. Recently, applications have been proposed to tackle the deconvolution process, to gain knowledge about which cell types (SC) are found within. This is usually done by incorporating metrics from single-cell (SC) RNA, from similar tissues. Yet, most existing tools are cumbersome, and we found them hard to integrate and properly utilize. Therefore, we present AntiSplodge, a simple feed-forward neural-network-based pipeline designed to effective deconvolute ST profiles by utilizing synthetic ST profiles derived from real-life SC datasets. AntiSplodge is designed to be easy, fast and intuitive while still being lightweight. To demonstrate AntiSplodge, we deconvolute the human heart and verify correctness across time points. We further deconvolute the mouse brain, where spot patterns correctly follow that of the underlying tissue. In particular, for the hippocampus from where the cells originate. Furthermore, AntiSplodge demonstrates top of the line performance when compared to current state-of-the-art tools. Software availability: https://github.com/HealthML/AntiSplodge/.
Collapse
Affiliation(s)
- Jesper B Lund
- Digital Health & Machine Learning Research Group, Hasso Plattner Institut for Digital Engineering, Potsdam, Germany
| | - Eric L Lindberg
- Cardiovascular and Metabolic Sciences, Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
| | - Henrike Maatz
- Cardiovascular and Metabolic Sciences, Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, Berlin, Germany
| | - Fabian Pottbaecker
- Digital Health & Machine Learning Research Group, Hasso Plattner Institut for Digital Engineering, Potsdam, Germany
| | - Norbert Hübner
- Cardiovascular and Metabolic Sciences, Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, Berlin, Germany
- Charite, Universitätsmedizin Berlin, Berlin, Germany
| | - Christoph Lippert
- Digital Health & Machine Learning Research Group, Hasso Plattner Institut for Digital Engineering, Potsdam, Germany
- Hasso Plattner Institute for Digital Health at Mount Sinai, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| |
Collapse
|
31
|
Furia L, Pelicci S, Perillo F, Bolognesi MM, Pelicci PG, Facciotti F, Cattoretti G, Faretta M. Automated multimodal fluorescence microscopy for hyperplex spatial-proteomics: Coupling microfluidic-based immunofluorescence to high resolution, high sensitivity, three-dimensional analysis of histological slides. Front Oncol 2022; 12:960734. [PMCID: PMC9606676 DOI: 10.3389/fonc.2022.960734] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2022] [Accepted: 09/27/2022] [Indexed: 11/13/2022] Open
Abstract
In situ multiplexing analysis and in situ transcriptomics are now providing revolutionary tools to achieve the comprehension of the molecular basis of cancer and to progress towards personalized medicine to fight the disease. The complexity of these tasks requires a continuous interplay among different technologies during all the phases of the experimental procedures. New tools are thus needed and their characterization in terms of performances and limits is mandatory to reach the best resolution and sensitivity. We propose here a new experimental pipeline to obtain an optimized costs-to-benefits ratio thanks to the alternate employment of automated and manual procedures during all the phases of a multiplexing experiment from sample preparation to image collection and analysis. A comparison between ultra-fast and automated immunofluorescence staining and standard staining protocols has been carried out to compare the performances in terms of antigen saturation, background, signal-to-noise ratio and total duration. We then developed specific computational tools to collect data by automated analysis-driven fluorescence microscopy. Computer assisted selection of targeted areas with variable magnification and resolution allows employing confocal microscopy for a 3D high resolution analysis. Spatial resolution and sensitivity were thus maximized in a framework where the amount of stored data and the total requested time for the procedure were optimized and reduced with respect to a standard experimental approach.
Collapse
Affiliation(s)
- Laura Furia
- Department of Experimental Oncology, European Institute of Oncology IRCCS, Milan, Italy
| | - Simone Pelicci
- Department of Experimental Oncology, European Institute of Oncology IRCCS, Milan, Italy
| | - Federica Perillo
- Department of Experimental Oncology, European Institute of Oncology IRCCS, Milan, Italy
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
| | | | - Pier Giuseppe Pelicci
- Department of Experimental Oncology, European Institute of Oncology IRCCS, Milan, Italy
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
| | - Federica Facciotti
- Department of Experimental Oncology, European Institute of Oncology IRCCS, Milan, Italy
- Department of Biotechnology and Biosciences, University of Milan-Bicocca, Milan, Italy
| | - Giorgio Cattoretti
- Department of Medicine and Surgery, Università di Milano-Bicocca, Monza, Italy
| | - Mario Faretta
- Department of Experimental Oncology, European Institute of Oncology IRCCS, Milan, Italy
- *Correspondence: Mario Faretta,
| |
Collapse
|
32
|
Kleino I, Frolovaitė P, Suomi T, Elo LL. Computational solutions for spatial transcriptomics. Comput Struct Biotechnol J 2022; 20:4870-4884. [PMID: 36147664 PMCID: PMC9464853 DOI: 10.1016/j.csbj.2022.08.043] [Citation(s) in RCA: 56] [Impact Index Per Article: 18.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Revised: 08/18/2022] [Accepted: 08/18/2022] [Indexed: 11/18/2022] Open
Abstract
Transcriptome level expression data connected to the spatial organization of the cells and molecules would allow a comprehensive understanding of how gene expression is connected to the structure and function in the biological systems. The spatial transcriptomics platforms may soon provide such information. However, the current platforms still lack spatial resolution, capture only a fraction of the transcriptome heterogeneity, or lack the throughput for large scale studies. The strengths and weaknesses in current ST platforms and computational solutions need to be taken into account when planning spatial transcriptomics studies. The basis of the computational ST analysis is the solutions developed for single-cell RNA-sequencing data, with advancements taking into account the spatial connectedness of the transcriptomes. The scRNA-seq tools are modified for spatial transcriptomics or new solutions like deep learning-based joint analysis of expression, spatial, and image data are developed to extract biological information in the spatially resolved transcriptomes. The computational ST analysis can reveal remarkable biological insights into spatial patterns of gene expression, cell signaling, and cell type variations in connection with cell type-specific signaling and organization in complex tissues. This review covers the topics that help choosing the platform and computational solutions for spatial transcriptomics research. We focus on the currently available ST methods and platforms and their strengths and limitations. Of the computational solutions, we provide an overview of the analysis steps and tools used in the ST data analysis. The compatibility with the data types and the tools provided by the current ST analysis frameworks are summarized.
Collapse
Key Words
- AOI, area of illumination
- BICCN, Brain Initiative Cell Census Network
- BOLORAMIS, barcoded oligonucleotides ligated on RNA amplified for multiplexed and parallel in situ analyses
- Baysor, Bayesian Segmentation of Spatial Transcriptomics Data
- BinSpect, Binary Spatial Extraction
- CCC, cell–cell communication
- CCI, cell–cell interactions
- CNV, copy-number variation
- Computational biology
- DSP, digital spatial profiling
- DbiT-Seq, Deterministic Barcoding in Tissue for spatial omics sequencing
- FA, factor analysis
- FFPE, formalin-fixed, paraffin-embedded
- FISH, fluorescence in situ hybridization
- FISSEQ, fluorescence in situ sequencing of RNA
- FOV, Field of view
- GRNs, gene regulation networks
- GSEA, gene set enrichment analysis
- GSVA, gene set variation analysis
- HDST, high definition spatial transcriptomics
- HMRF, hidden Markov random field
- ICG, interaction changed genes
- ISH, in situ hybridization
- ISS, in situ sequencing
- JSTA, Joint cell segmentation and cell type annotation
- KNN, k-nearest neighbor
- LCM, Laser Capture Microdissection
- LCM-seq, laser capture microdissection coupled with RNA sequencing
- LOH, loss of heterozygosity analysis
- MC, Molecular Cartography
- MERFISH, multiplexed error-robust FISH
- NMF (NNMF), Non-negative matrix factorization
- PCA, Principal Component Analysis
- PIXEL-seq, Polony (or DNA cluster)-indexed library-sequencing
- PL-lig, padlock ligation
- QC, quality control
- RNAseq, RNA sequencing
- ROI, region of interest
- SCENIC, Single-Cell rEgulatory Network Inference and Clustering
- SME, Spatial Morphological gene Expression normalization
- SPATA, SPAtial Transcriptomic Analysis
- ST Pipeline, Spatial Transcriptomics Pipeline
- ST, Spatial transcriptomics
- STARmap, spatially-resolved transcript amplicon readout mapping
- Single-cell analysis
- Spatial data analysis frameworks
- Spatial deconvolution
- Spatial transcriptomics
- TIVA, Transcriptome in Vivo Analysis
- TMA, tissue microarray
- TME, tumor micro environment
- UMAP, Uniform Manifold Approximation and Projection for Dimension Reduction
- UMI, unique molecular identifier
- ZipSeq, zipcoded sequencing.
- scRNA-seq, single-cell RNA sequencing
- scvi-tools, single-cell variational inference tools
- seqFISH, sequential fluorescence in situ hybridization
- sequ-smFISH, sequential single-molecule fluorescent in situ hybridization
- smFISH, single molecule FISH
- t-SNE, t-distributed stochastic neighbor embedding
Collapse
Affiliation(s)
- Iivari Kleino
- Turku Bioscience Centre, University of Turku and Åbo Akademi University Turku, Turku, Finland
| | - Paulina Frolovaitė
- Turku Bioscience Centre, University of Turku and Åbo Akademi University Turku, Turku, Finland
| | - Tomi Suomi
- Turku Bioscience Centre, University of Turku and Åbo Akademi University Turku, Turku, Finland
| | - Laura L. Elo
- Turku Bioscience Centre, University of Turku and Åbo Akademi University Turku, Turku, Finland
- Institute of Biomedicine, University of Turku, Turku, Finland
| |
Collapse
|
33
|
Zhang C, Chen R, Zhang Y. Accurate inference of genome-wide spatial expression with iSpatial. SCIENCE ADVANCES 2022; 8:eabq0990. [PMID: 36026447 PMCID: PMC9417177 DOI: 10.1126/sciadv.abq0990] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Accepted: 07/13/2022] [Indexed: 06/15/2023]
Abstract
Spatially resolved transcriptomic analyses can reveal molecular insights underlying tissue structure and context-dependent cell-cell or cell-environment interaction. Because of the current technical limitation, obtaining genome-wide spatial transcriptome at single-cell resolution is challenging. Here, we developed a new algorithm named iSpatial to derive the spatial pattern of the entire transcriptome by integrating spatial transcriptomic and single-cell RNA-seq datasets. Compared to other existing methods, iSpatial has higher accuracy in predicting gene expression and spatial distribution. Furthermore, it reduces false-positive and false-negative signals in the original datasets. By testing iSpatial with multiple spatial transcriptomic datasets, we demonstrate its wide applicability to datasets from different tissues and by different techniques. Thus, we provide a computational approach to reveal spatial organization of the entire transcriptome at single-cell resolution. With numerous high-quality datasets available in the public domain, iSpatial provides a unique way to understand the structure and function of complex tissues and disease processes.
Collapse
Affiliation(s)
- Chao Zhang
- Howard Hughes Medical Institute, Boston Children’s Hospital, Boston, MA 02115, USA
- Program in Cellular and Molecular Medicine, Boston Children’s Hospital, Boston, MA 02115, USA
- Division of Hematology/Oncology, Department of Pediatrics, Boston Children’s Hospital, Boston, MA 02115, USA
| | - Renchao Chen
- Howard Hughes Medical Institute, Boston Children’s Hospital, Boston, MA 02115, USA
- Program in Cellular and Molecular Medicine, Boston Children’s Hospital, Boston, MA 02115, USA
- Division of Hematology/Oncology, Department of Pediatrics, Boston Children’s Hospital, Boston, MA 02115, USA
| | - Yi Zhang
- Howard Hughes Medical Institute, Boston Children’s Hospital, Boston, MA 02115, USA
- Program in Cellular and Molecular Medicine, Boston Children’s Hospital, Boston, MA 02115, USA
- Division of Hematology/Oncology, Department of Pediatrics, Boston Children’s Hospital, Boston, MA 02115, USA
- Department of Genetics, Harvard Medical School, Boston, MA 02115, USA
- Harvard Stem Cell Institute, WAB-149G, 200 Longwood Avenue, Boston, MA 02115, USA
| |
Collapse
|
34
|
Shan X, Chen J, Dong K, Zhou W, Zhang S. Deciphering the Spatial Modular Patterns of Tissues by Integrating Spatial and Single-Cell Transcriptomic Data. J Comput Biol 2022; 29:650-663. [PMID: 35727094 DOI: 10.1089/cmb.2021.0617] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Single-cell RNA sequencing (scRNA-seq) provides a powerful tool to analyze the expression level of tissues at a cellular resolution. However, it could not capture the spatial organization of cells in a tissue. The spatially resolved transcriptomics technologies (ST) have been developed to address this issue. However, the emerging STs are still inefficient at single-cell resolution and/or fail to capture the sufficient reads. To this end, we adopted a partial least squares-based method (spatial modular patterns [SpaMOD]) to simultaneously integrate the two data modalities, as well as the networks related to cells and spots, to identify the cell-spot comodules for deciphering the SpaMOD of tissues. We applied SpaMOD to three paired scRNA-seq and ST datasets, derived from the mouse brain, granuloma, and pancreatic ductal adenocarcinoma, respectively. The identified cell-spot comodules provide detailed biological insights into the spatial relationships between cell populations and their spatial locations in the tissue.
Collapse
Affiliation(s)
- Xu Shan
- Department of Software Engineering, Yunnan University, Kunming, China
| | - Jinyu Chen
- College of Statistics and Data Science, Faculty of Science, Beijing University of Technology, Beijing, China
| | - Kangning Dong
- NCMIS, CEMS, RCSDS, Academy of Mathematics and Systems Science, Chinese Academy of Sciences, Beijing, China
| | - Wei Zhou
- Department of Software Engineering, Yunnan University, Kunming, China
| | - Shihua Zhang
- NCMIS, CEMS, RCSDS, Academy of Mathematics and Systems Science, Chinese Academy of Sciences, Beijing, China.,School of Mathematical Sciences, University of Chinese Academy of Sciences, Beijing, China.,Center for Excellence in Animal Evolution and Genetics, Chinese Academy of Sciences, Kunming, China.,Key Laboratory of Systems Biology, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Hangzhou, China
| |
Collapse
|
35
|
Kashyap D, Pal D, Sharma R, Garg VK, Goel N, Koundal D, Zaguia A, Koundal S, Belay A. Global Increase in Breast Cancer Incidence: Risk Factors and Preventive Measures. BIOMED RESEARCH INTERNATIONAL 2022; 2022:9605439. [PMID: 35480139 PMCID: PMC9038417 DOI: 10.1155/2022/9605439] [Citation(s) in RCA: 252] [Impact Index Per Article: 84.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Revised: 02/25/2022] [Accepted: 03/21/2022] [Indexed: 02/07/2023]
Abstract
Breast cancer is a global cause for concern owing to its high incidence around the world. The alarming increase in breast cancer cases emphasizes the management of disease at multiple levels. The management should start from the beginning that includes stringent cancer screening or cancer registry to effective diagnostic and treatment strategies. Breast cancer is highly heterogeneous at morphology as well as molecular levels and needs different therapeutic regimens based on the molecular subtype. Breast cancer patients with respective subtype have different clinical outcome prognoses. Breast cancer heterogeneity emphasizes the advanced molecular testing that will help on-time diagnosis and improved survival. Emerging fields such as liquid biopsy and artificial intelligence would help to under the complexity of breast cancer disease and decide the therapeutic regimen that helps in breast cancer management. In this review, we have discussed various risk factors and advanced technology available for breast cancer diagnosis to combat the worst breast cancer status and areas that need to be focused for the better management of breast cancer.
Collapse
Affiliation(s)
- Dharambir Kashyap
- Department of Histopathology, Postgraduate Institute of Medical Education and Research, Chandigarh 160012, India
| | - Deeksha Pal
- Department of Translational and Regenerative Medicine, Postgraduate Institute of Medical Education and Research, Chandigarh 160012, India
| | - Riya Sharma
- Department of Pulmonary Medicine, Postgraduate Institute of Medical Education and Research, Chandigarh 160012, India
| | - Vivek Kumar Garg
- Department of Medical Laboratory Technology, University Institute of Applied Health Sciences, Chandigarh University (Gharuan), Mohali 140313, India
| | - Neelam Goel
- Department of Information Technology, University Institute of Engineering & Technology, Panjab University, Chandigarh 160014, India
| | - Deepika Koundal
- Department of Systemics, School of Computer Science, University of Petroleum & Energy Studies, Dehradun, India
| | - Atef Zaguia
- Department of computer science, College of Computers and Information Technology, Taif University, P.O. BOX 11099, Taif 21944, Saudi Arabia
| | - Shubham Koundal
- Department of Medical Laboratory Technology, University Institute of Applied Health Sciences, Chandigarh University (Gharuan), Mohali 140313, India
| | - Assaye Belay
- Department of Statistics, Mizan-Tepi University, Ethiopia
| |
Collapse
|
36
|
Christopher JA, Geladaki A, Dawson CS, Vennard OL, Lilley KS. Subcellular Transcriptomics and Proteomics: A Comparative Methods Review. Mol Cell Proteomics 2022; 21:100186. [PMID: 34922010 PMCID: PMC8864473 DOI: 10.1016/j.mcpro.2021.100186] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Revised: 11/16/2021] [Accepted: 12/13/2021] [Indexed: 12/23/2022] Open
Abstract
The internal environment of cells is molecularly crowded, which requires spatial organization via subcellular compartmentalization. These compartments harbor specific conditions for molecules to perform their biological functions, such as coordination of the cell cycle, cell survival, and growth. This compartmentalization is also not static, with molecules trafficking between these subcellular neighborhoods to carry out their functions. For example, some biomolecules are multifunctional, requiring an environment with differing conditions or interacting partners, and others traffic to export such molecules. Aberrant localization of proteins or RNA species has been linked to many pathological conditions, such as neurological, cancer, and pulmonary diseases. Differential expression studies in transcriptomics and proteomics are relatively common, but the majority have overlooked the importance of subcellular information. In addition, subcellular transcriptomics and proteomics data do not always colocate because of the biochemical processes that occur during and after translation, highlighting the complementary nature of these fields. In this review, we discuss and directly compare the current methods in spatial proteomics and transcriptomics, which include sequencing- and imaging-based strategies, to give the reader an overview of the current tools available. We also discuss current limitations of these strategies as well as future developments in the field of spatial -omics.
Collapse
Affiliation(s)
- Josie A Christopher
- Department of Biochemistry, Cambridge Centre for Proteomics, University of Cambridge, Cambridge, UK; Milner Therapeutics Institute, Jeffrey Cheah Biomedical Centre, Cambridge, UK
| | - Aikaterini Geladaki
- Department of Biochemistry, Cambridge Centre for Proteomics, University of Cambridge, Cambridge, UK; Department of Genetics, University of Cambridge, Cambridge, UK
| | - Charlotte S Dawson
- Department of Biochemistry, Cambridge Centre for Proteomics, University of Cambridge, Cambridge, UK; Milner Therapeutics Institute, Jeffrey Cheah Biomedical Centre, Cambridge, UK
| | - Owen L Vennard
- Department of Biochemistry, Cambridge Centre for Proteomics, University of Cambridge, Cambridge, UK; Milner Therapeutics Institute, Jeffrey Cheah Biomedical Centre, Cambridge, UK
| | - Kathryn S Lilley
- Department of Biochemistry, Cambridge Centre for Proteomics, University of Cambridge, Cambridge, UK; Milner Therapeutics Institute, Jeffrey Cheah Biomedical Centre, Cambridge, UK.
| |
Collapse
|
37
|
Wang Y, Eddison M, Fleishman G, Weigert M, Xu S, Wang T, Rokicki K, Goina C, Henry FE, Lemire AL, Schmidt U, Yang H, Svoboda K, Myers EW, Saalfeld S, Korff W, Sternson SM, Tillberg PW. EASI-FISH for thick tissue defines lateral hypothalamus spatio-molecular organization. Cell 2021; 184:6361-6377.e24. [PMID: 34875226 DOI: 10.1016/j.cell.2021.11.024] [Citation(s) in RCA: 93] [Impact Index Per Article: 23.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2021] [Revised: 08/22/2021] [Accepted: 11/12/2021] [Indexed: 11/17/2022]
Abstract
Determining the spatial organization and morphological characteristics of molecularly defined cell types is a major bottleneck for characterizing the architecture underpinning brain function. We developed Expansion-Assisted Iterative Fluorescence In Situ Hybridization (EASI-FISH) to survey gene expression in brain tissue, as well as a turnkey computational pipeline to rapidly process large EASI-FISH image datasets. EASI-FISH was optimized for thick brain sections (300 μm) to facilitate reconstruction of spatio-molecular domains that generalize across brains. Using the EASI-FISH pipeline, we investigated the spatial distribution of dozens of molecularly defined cell types in the lateral hypothalamic area (LHA), a brain region with poorly defined anatomical organization. Mapping cell types in the LHA revealed nine spatially and molecularly defined subregions. EASI-FISH also facilitates iterative reanalysis of scRNA-seq datasets to determine marker-genes that further dissociated spatial and morphological heterogeneity. The EASI-FISH pipeline democratizes mapping molecularly defined cell types, enabling discoveries about brain organization.
Collapse
Affiliation(s)
- Yuhan Wang
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA 20147, USA
| | - Mark Eddison
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA 20147, USA
| | - Greg Fleishman
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA 20147, USA
| | - Martin Weigert
- Max Planck Institute of Molecular Cell Biology and Genetics, 01307 Dresden, Germany; Institute of Bioengineering, School of Life Sciences, EPFL, 1015 Lausanne, Switzerland
| | - Shengjin Xu
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA 20147, USA
| | - Tim Wang
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA 20147, USA
| | - Konrad Rokicki
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA 20147, USA
| | - Cristian Goina
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA 20147, USA
| | - Fredrick E Henry
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA 20147, USA
| | - Andrew L Lemire
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA 20147, USA
| | - Uwe Schmidt
- Max Planck Institute of Molecular Cell Biology and Genetics, 01307 Dresden, Germany
| | - Hui Yang
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA 20147, USA; Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, New York 10461, USA
| | - Karel Svoboda
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA 20147, USA
| | - Eugene W Myers
- Max Planck Institute of Molecular Cell Biology and Genetics, 01307 Dresden, Germany
| | - Stephan Saalfeld
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA 20147, USA
| | - Wyatt Korff
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA 20147, USA
| | - Scott M Sternson
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA 20147, USA; Department of Neurosciences, Howard Hughes Medical Institute, University of California, San Diego, La Jolla, CA 92093, USA.
| | - Paul W Tillberg
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA 20147, USA.
| |
Collapse
|
38
|
Missarova A, Jain J, Butler A, Ghazanfar S, Stuart T, Brusko M, Wasserfall C, Nick H, Brusko T, Atkinson M, Satija R, Marioni JC. geneBasis: an iterative approach for unsupervised selection of targeted gene panels from scRNA-seq. Genome Biol 2021; 22:333. [PMID: 34872616 PMCID: PMC8650258 DOI: 10.1186/s13059-021-02548-z] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Accepted: 11/19/2021] [Indexed: 12/13/2022] Open
Abstract
scRNA-seq datasets are increasingly used to identify gene panels that can be probed using alternative technologies, such as spatial transcriptomics, where choosing the best subset of genes is vital. Existing methods are limited by a reliance on pre-existing cell type labels or by difficulties in identifying markers of rare cells. We introduce an iterative approach, geneBasis, for selecting an optimal gene panel, where each newly added gene captures the maximum distance between the true manifold and the manifold constructed using the currently selected gene panel. Our approach outperforms existing strategies and can resolve cell types and subtle cell state differences.
Collapse
Affiliation(s)
- Alsu Missarova
- European Molecular Biology Laboratory, European Bioinformatics Institute, Wellcome Genome Campus, Cambridge, UK
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, UK
| | | | - Andrew Butler
- New York Genome Center, New York, USA
- Center for Genomics and Systems Biology, NYU, New York, USA
| | - Shila Ghazanfar
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, UK
| | - Tim Stuart
- New York Genome Center, New York, USA
- Center for Genomics and Systems Biology, NYU, New York, USA
| | - Maigan Brusko
- Department of Pathology, Immunology, and Laboratory Medicine, College of Medicine, University of Florida, Jacksonville, USA
| | - Clive Wasserfall
- Department of Pathology, Immunology, and Laboratory Medicine, College of Medicine, University of Florida, Jacksonville, USA
| | - Harry Nick
- Department of Neuroscience, College of Medicine, University of Florida, Jacksonville, USA
| | - Todd Brusko
- Department of Pathology, Immunology, and Laboratory Medicine, College of Medicine, University of Florida, Jacksonville, USA
| | - Mark Atkinson
- Department of Pathology, Immunology, and Laboratory Medicine, College of Medicine, University of Florida, Jacksonville, USA
| | - Rahul Satija
- New York Genome Center, New York, USA.
- Center for Genomics and Systems Biology, NYU, New York, USA.
| | - John C Marioni
- European Molecular Biology Laboratory, European Bioinformatics Institute, Wellcome Genome Campus, Cambridge, UK.
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, UK.
- Wellcome Sanger Institute, Wellcome Genome Campus, Cambridge, UK.
| |
Collapse
|
39
|
Ahmed R, Augustine R, Valera E, Ganguli A, Mesaeli N, Ahmad IS, Bashir R, Hasan A. Spatial mapping of cancer tissues by OMICS technologies. Biochim Biophys Acta Rev Cancer 2021; 1877:188663. [PMID: 34861353 DOI: 10.1016/j.bbcan.2021.188663] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2021] [Revised: 11/15/2021] [Accepted: 11/26/2021] [Indexed: 12/14/2022]
Abstract
Spatial mapping of heterogeneity in gene expression in cancer tissues can improve our understanding of cancers and help in the rapid detection of cancers with high accuracy and reliability. Significant advancements have been made in recent years in OMICS technologies, which possess the strong potential to be applied in the spatial mapping of biopsy tissue samples and their molecular profiling to a single-cell level. The clinical application of OMICS technologies in spatial profiling of cancer tissues is also advancing. The current review presents recent advancements and prospects of applying OMICS technologies to the spatial mapping of various analytes in cancer tissues. We benchmark the current state of the art in the field to advance existing OMICS technologies for high throughput spatial profiling. The factors taken into consideration include spatial resolution, types of biomolecules, number of different biomolecules that can be detected from the same assay, labeled versus label-free approaches, and approximate time required for each assay. Further advancements are still needed for the widespread application of OMICs technologies in performing fast and high throughput spatial mapping of cancer tissues as well as their effective use in research and clinical applications.
Collapse
Affiliation(s)
- Rashid Ahmed
- Department of Mechanical and Industrial Engineering, College of Engineering, Qatar University, Doha 2713, Qatar; Biomedical Research Center (BRC), Qatar University, Doha 2713, Qatar; Nick Holonyak Jr. Micro and Nanotechnology Laboratory, University of Illinois at Urbana Champaign, IL, USA
| | - Robin Augustine
- Department of Mechanical and Industrial Engineering, College of Engineering, Qatar University, Doha 2713, Qatar; Biomedical Research Center (BRC), Qatar University, Doha 2713, Qatar
| | - Enrique Valera
- Nick Holonyak Jr. Micro and Nanotechnology Laboratory, University of Illinois at Urbana Champaign, IL, USA; Department of Bioengineering, University of Illinois at Urbana Champaign, IL, USA
| | - Anurup Ganguli
- Nick Holonyak Jr. Micro and Nanotechnology Laboratory, University of Illinois at Urbana Champaign, IL, USA; Department of Bioengineering, University of Illinois at Urbana Champaign, IL, USA
| | - Nasrin Mesaeli
- Department of Biochemistry, Weill Cornell Medicine in Qatar, Qatar Foundation, Doha, Qatar
| | - Irfan S Ahmad
- Nick Holonyak Jr. Micro and Nanotechnology Laboratory, University of Illinois at Urbana Champaign, IL, USA
| | - Rashid Bashir
- Nick Holonyak Jr. Micro and Nanotechnology Laboratory, University of Illinois at Urbana Champaign, IL, USA; Department of Bioengineering, University of Illinois at Urbana Champaign, IL, USA; Carle Illinois College of Medicine, University of Illinois at Urbana-Champaign, Champaign, IL, USA.
| | - Anwarul Hasan
- Department of Mechanical and Industrial Engineering, College of Engineering, Qatar University, Doha 2713, Qatar; Biomedical Research Center (BRC), Qatar University, Doha 2713, Qatar.
| |
Collapse
|
40
|
Endo M, Maruoka H, Okabe S. Advanced Technologies for Local Neural Circuits in the Cerebral Cortex. Front Neuroanat 2021; 15:757499. [PMID: 34803616 PMCID: PMC8595196 DOI: 10.3389/fnana.2021.757499] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Accepted: 10/13/2021] [Indexed: 11/13/2022] Open
Abstract
The neural network in the brain can be viewed as an integrated system assembled from a large number of local neural circuits specialized for particular brain functions. Activities of neurons in local neural circuits are thought to be organized both spatially and temporally under the rules optimized for their roles in information processing. It is well perceived that different areas of the mammalian neocortex have specific cognitive functions and distinct computational properties. However, the organizational principles of the local neural circuits in different cortical regions have not yet been clarified. Therefore, new research principles and related neuro-technologies that enable efficient and precise recording of large-scale neuronal activities and synaptic connections are necessary. Innovative technologies for structural analysis, including tissue clearing and expansion microscopy, have enabled super resolution imaging of the neural circuits containing thousands of neurons at a single synapse resolution. The imaging resolution and volume achieved by new technologies are beyond the limits of conventional light or electron microscopic methods. Progress in genome editing and related technologies has made it possible to label and manipulate specific cell types and discriminate activities of multiple cell types. These technologies will provide a breakthrough for multiscale analysis of the structure and function of local neural circuits. This review summarizes the basic concepts and practical applications of the emerging technologies and new insight into local neural circuits obtained by these technologies.
Collapse
Affiliation(s)
| | | | - Shigeo Okabe
- Department of Cellular Neurobiology, Graduate School of Medicine and Faculty of Medicine, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
41
|
Asada K, Takasawa K, Machino H, Takahashi S, Shinkai N, Bolatkan A, Kobayashi K, Komatsu M, Kaneko S, Okamoto K, Hamamoto R. Single-Cell Analysis Using Machine Learning Techniques and Its Application to Medical Research. Biomedicines 2021; 9:1513. [PMID: 34829742 PMCID: PMC8614827 DOI: 10.3390/biomedicines9111513] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2021] [Revised: 10/06/2021] [Accepted: 10/19/2021] [Indexed: 01/14/2023] Open
Abstract
In recent years, the diversity of cancer cells in tumor tissues as a result of intratumor heterogeneity has attracted attention. In particular, the development of single-cell analysis technology has made a significant contribution to the field; technologies that are centered on single-cell RNA sequencing (scRNA-seq) have been reported to analyze cancer constituent cells, identify cell groups responsible for therapeutic resistance, and analyze gene signatures of resistant cell groups. However, although single-cell analysis is a powerful tool, various issues have been reported, including batch effects and transcriptional noise due to gene expression variation and mRNA degradation. To overcome these issues, machine learning techniques are currently being introduced for single-cell analysis, and promising results are being reported. In addition, machine learning has also been used in various ways for single-cell analysis, such as single-cell assay of transposase accessible chromatin sequencing (ATAC-seq), chromatin immunoprecipitation sequencing (ChIP-seq) analysis, and multi-omics analysis; thus, it contributes to a deeper understanding of the characteristics of human diseases, especially cancer, and supports clinical applications. In this review, we present a comprehensive introduction to the implementation of machine learning techniques in medical research for single-cell analysis, and discuss their usefulness and future potential.
Collapse
Affiliation(s)
- Ken Asada
- Cancer Translational Research Team, RIKEN Center for Advanced Intelligence Project, 1-4-1 Nihonbashi, Chuo-ku, Tokyo 103-0027, Japan; (K.T.); (H.M.); (S.T.); (N.S.); (A.B.); (M.K.)
| | - Ken Takasawa
- Cancer Translational Research Team, RIKEN Center for Advanced Intelligence Project, 1-4-1 Nihonbashi, Chuo-ku, Tokyo 103-0027, Japan; (K.T.); (H.M.); (S.T.); (N.S.); (A.B.); (M.K.)
| | - Hidenori Machino
- Cancer Translational Research Team, RIKEN Center for Advanced Intelligence Project, 1-4-1 Nihonbashi, Chuo-ku, Tokyo 103-0027, Japan; (K.T.); (H.M.); (S.T.); (N.S.); (A.B.); (M.K.)
| | - Satoshi Takahashi
- Cancer Translational Research Team, RIKEN Center for Advanced Intelligence Project, 1-4-1 Nihonbashi, Chuo-ku, Tokyo 103-0027, Japan; (K.T.); (H.M.); (S.T.); (N.S.); (A.B.); (M.K.)
| | - Norio Shinkai
- Cancer Translational Research Team, RIKEN Center for Advanced Intelligence Project, 1-4-1 Nihonbashi, Chuo-ku, Tokyo 103-0027, Japan; (K.T.); (H.M.); (S.T.); (N.S.); (A.B.); (M.K.)
- Department of NCC Cancer Science, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-ku, Tokyo 113-8510, Japan
| | - Amina Bolatkan
- Cancer Translational Research Team, RIKEN Center for Advanced Intelligence Project, 1-4-1 Nihonbashi, Chuo-ku, Tokyo 103-0027, Japan; (K.T.); (H.M.); (S.T.); (N.S.); (A.B.); (M.K.)
- Division of Medical AI Research and Development, National Cancer Center Research Institute, 5-1-1 Tsukiji, Chuo-ku, Tokyo 104-0045, Japan; (K.K.); (S.K.)
| | - Kazuma Kobayashi
- Division of Medical AI Research and Development, National Cancer Center Research Institute, 5-1-1 Tsukiji, Chuo-ku, Tokyo 104-0045, Japan; (K.K.); (S.K.)
| | - Masaaki Komatsu
- Cancer Translational Research Team, RIKEN Center for Advanced Intelligence Project, 1-4-1 Nihonbashi, Chuo-ku, Tokyo 103-0027, Japan; (K.T.); (H.M.); (S.T.); (N.S.); (A.B.); (M.K.)
| | - Syuzo Kaneko
- Division of Medical AI Research and Development, National Cancer Center Research Institute, 5-1-1 Tsukiji, Chuo-ku, Tokyo 104-0045, Japan; (K.K.); (S.K.)
| | - Koji Okamoto
- Division of Cancer Differentiation, National Cancer Center Research Institute, 5-1-1 Tsukiji, Chuo-ku, Tokyo 104-0045, Japan;
| | - Ryuji Hamamoto
- Department of NCC Cancer Science, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-ku, Tokyo 113-8510, Japan
- Division of Medical AI Research and Development, National Cancer Center Research Institute, 5-1-1 Tsukiji, Chuo-ku, Tokyo 104-0045, Japan; (K.K.); (S.K.)
| |
Collapse
|
42
|
Eichenberger BT, Zhan Y, Rempfler M, Giorgetti L, Chao JA. deepBlink: threshold-independent detection and localization of diffraction-limited spots. Nucleic Acids Res 2021; 49:7292-7297. [PMID: 34197605 PMCID: PMC8287908 DOI: 10.1093/nar/gkab546] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Revised: 05/12/2021] [Accepted: 06/23/2021] [Indexed: 11/13/2022] Open
Abstract
Detection of diffraction-limited spots in single-molecule microscopy images is traditionally performed with mathematical operators designed for idealized spots. This process requires manual tuning of parameters that is time-consuming and not always reliable. We have developed deepBlink, a neural network-based method to detect and localize spots automatically. We demonstrate that deepBlink outperforms other state-of-the-art methods across six publicly available datasets containing synthetic and experimental data.
Collapse
Affiliation(s)
- Bastian Th Eichenberger
- Friedrich Miescher Institute for Biomedical Research, 4058 Basel, Switzerland.,University of Basel, 4003 Basel, Switzerland
| | - YinXiu Zhan
- Friedrich Miescher Institute for Biomedical Research, 4058 Basel, Switzerland
| | - Markus Rempfler
- Friedrich Miescher Institute for Biomedical Research, 4058 Basel, Switzerland
| | - Luca Giorgetti
- Friedrich Miescher Institute for Biomedical Research, 4058 Basel, Switzerland
| | - Jeffrey A Chao
- Friedrich Miescher Institute for Biomedical Research, 4058 Basel, Switzerland
| |
Collapse
|
43
|
Dai G, Feinberg AW, Wan LQ. Recent Advances in Cellular and Molecular Bioengineering for Building and Translation of Biological Systems. Cell Mol Bioeng 2021; 14:293-308. [PMID: 34055096 PMCID: PMC8147909 DOI: 10.1007/s12195-021-00676-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Accepted: 05/07/2021] [Indexed: 11/24/2022] Open
Abstract
In January of 2020, the Biomedical Engineering Society (BMES)- Cellular and Molecular Bioengineering (CMBE) conference was held in Puerto Rico and themed “Vision 2020: Emerging Technologies to Elucidate the Rule of Life.” The annual BME-CMBE conference gathered worldwide leaders and discussed successes and challenges in engineering biological systems and their translation. The goal of this report is to present the research frontiers in this field and provide perspectives on successful engineering and translation towards the clinic. We hope that this report serves as a constructive guide in shaping the future of research and translation of engineered biological systems.
Collapse
Affiliation(s)
- Guohao Dai
- Department of Bioengineering, Northeastern University, 805 Columbus Ave, ISEC 224, Boston, MA 02115 USA
| | - Adam W Feinberg
- Departments of Biomedical Engineering & Materials Science & Engineering, Carnegie Mellon University, 5000 Forbes Avenue, Pittsburgh, PA 15213 USA
| | - Leo Q Wan
- Departments of Biomedical Engineering & Biological Sciences, Rensselaer Polytechnic Institute, Biotech 2147, 110 8th Street, Troy, NY 12180 USA
| |
Collapse
|
44
|
Assigning transcriptomic class in the trigeminal ganglion using multiplex in situ hybridization and machine learning. Pain 2021; 161:2212-2224. [PMID: 32379225 DOI: 10.1097/j.pain.0000000000001911] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2020] [Accepted: 04/10/2020] [Indexed: 11/25/2022]
Abstract
ABSTRACT Single cell sequencing has provided unprecedented information about the transcriptomic diversity of somatosensory systems. Here, we describe a simple and versatile in situ hybridization (ISH)-based approach for mapping this information back to the tissue. We illustrate the power of this approach by demonstrating that ISH localization with just 8 probes is sufficient to distinguish all major classes of neurons in sections of the trigeminal ganglion. To further simplify the approach and make transcriptomic class assignment and cell segmentation automatic, we developed a machine learning approach for analyzing images from multiprobe ISH experiments. We demonstrate the power of in situ class assignment by examining the expression patterns of voltage-gated sodium channels that play roles in distinct somatosensory processes and pain. Specifically, this analysis resolves intrinsic problems with single cell sequencing related to the sparseness of data leading to ambiguity about gene expression patterns. We also used the multiplex in situ approach to study the projection fields of the different neuronal classes. Our results demonstrate that the surface of the eye and meninges are targeted by broad arrays of neural classes despite their very different sensory properties but exhibit idiotypic patterns of innervation at a quantitative level. Very surprisingly, itch-related neurons extensively innervated the meninges, indicating that these transcriptomic cell classes are not simply labeled lines for triggering itch. Together, these results substantiate the importance of a sensory neuron's peripheral and central connections as well as its transcriptomic class in determining its role in sensation.
Collapse
|
45
|
Nagle MP, Tam GS, Maltz E, Hemminger Z, Wollman R. Bridging scales: From cell biology to physiology using in situ single-cell technologies. Cell Syst 2021; 12:388-400. [PMID: 34015260 DOI: 10.1016/j.cels.2021.03.002] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Revised: 01/30/2021] [Accepted: 03/08/2021] [Indexed: 12/14/2022]
Abstract
Biological organization crosses multiple spatial scales: from molecular, cellular, to tissues and organs. The proliferation of molecular profiling technologies enables increasingly detailed cataloging of the components at each scale. However, the scarcity of spatial profiling has made it challenging to bridge across these scales. Emerging technologies based on highly multiplexed in situ profiling are paving the way to study the spatial organization of cells and tissues in greater detail. These new technologies provide the data needed to cross the scale from cell biology to physiology and identify the fundamental principles that govern tissue organization. Here, we provide an overview of these key technologies and discuss the current and future insights these powerful techniques enable.
Collapse
Affiliation(s)
- Maeve P Nagle
- Department of Chemistry and Biochemistry, University of California, Los Angeles, Los Angeles, CA 90095, USA; Institute for Quantitative and Computational Biosciences, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Gabriela S Tam
- Department of Chemistry and Biochemistry, University of California, Los Angeles, Los Angeles, CA 90095, USA; Institute for Quantitative and Computational Biosciences, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Evan Maltz
- Department of Chemistry and Biochemistry, University of California, Los Angeles, Los Angeles, CA 90095, USA; Institute for Quantitative and Computational Biosciences, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Zachary Hemminger
- Department of Chemistry and Biochemistry, University of California, Los Angeles, Los Angeles, CA 90095, USA; Institute for Quantitative and Computational Biosciences, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Roy Wollman
- Department of Chemistry and Biochemistry, University of California, Los Angeles, Los Angeles, CA 90095, USA; Institute for Quantitative and Computational Biosciences, University of California, Los Angeles, Los Angeles, CA 90095, USA; Department of Integrative Biology and Physiology, University of California, Los Angeles, Los Angeles, CA 90095, USA.
| |
Collapse
|
46
|
Bassiouni R, Gibbs LD, Craig DW, Carpten JD, McEachron TA. Applicability of spatial transcriptional profiling to cancer research. Mol Cell 2021; 81:1631-1639. [PMID: 33826920 PMCID: PMC8052283 DOI: 10.1016/j.molcel.2021.03.016] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Revised: 02/22/2021] [Accepted: 03/10/2021] [Indexed: 12/21/2022]
Abstract
Spatial transcriptional profiling provides gene expression information within the important anatomical context of tissue architecture. This approach is well suited to characterizing solid tumors, which develop within a complex landscape of malignant cells, immune cells, and stroma. In a single assay, spatial transcriptional profiling can interrogate the role of spatial relationships among these cell populations as well as reveal spatial patterns of relevant oncogenic genetic events. The broad utility of this approach is reflected in the array of strategies that have been developed for its implementation as well as in the recent commercial development of several profiling platforms. The flexibility to apply these technologies to both hypothesis-driven and discovery-driven studies allows widespread applicability in research settings. This review discusses available technologies for spatial transcriptional profiling and several applications for their use in cancer research.
Collapse
Affiliation(s)
- Rania Bassiouni
- Department of Translational Genomics, Keck School of Medicine, University of Southern California, 1450 Biggy Street, Los Angeles, CA 90033, USA
| | - Lee D Gibbs
- Department of Translational Genomics, Keck School of Medicine, University of Southern California, 1450 Biggy Street, Los Angeles, CA 90033, USA
| | - David W Craig
- Department of Translational Genomics, Keck School of Medicine, University of Southern California, 1450 Biggy Street, Los Angeles, CA 90033, USA
| | - John D Carpten
- Department of Translational Genomics, Keck School of Medicine, University of Southern California, 1450 Biggy Street, Los Angeles, CA 90033, USA
| | - Troy A McEachron
- Department of Translational Genomics, Keck School of Medicine, University of Southern California, 1450 Biggy Street, Los Angeles, CA 90033, USA; Pediatric Oncology Branch, National Cancer Institute, 10 Center Drive, Bethesda, MD 20892, USA.
| |
Collapse
|
47
|
Compressed sensing for highly efficient imaging transcriptomics. Nat Biotechnol 2021; 39:936-942. [PMID: 33859401 DOI: 10.1038/s41587-021-00883-x] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2019] [Accepted: 03/11/2021] [Indexed: 12/18/2022]
Abstract
Recent methods for spatial imaging of tissue samples can identify up to ~100 individual proteins1-3 or RNAs4-10 at single-cell resolution. However, the number of proteins or genes that can be studied in these approaches is limited by long imaging times. Here we introduce Composite In Situ Imaging (CISI), a method that leverages structure in gene expression across both cells and tissues to limit the number of imaging cycles needed to obtain spatially resolved gene expression maps. CISI defines gene modules that can be detected using composite measurements from imaging probes for subsets of genes. The data are then decompressed to recover expression values for individual genes. CISI further reduces imaging time by not relying on spot-level resolution, enabling lower magnification acquisition, and is overall about 500-fold more efficient than current methods. Applying CISI to 12 mouse brain sections, we accurately recovered the spatial abundance of 37 individual genes from 11 composite measurements covering 180 mm2 and 476,276 cells.
Collapse
|
48
|
Tao S, Wang Y, Peng J, Zhao Y, He X, Yu X, Liu Q, Jin S, Xu F. Whole-Brain Mapping the Direct Inputs of Dorsal and Ventral CA1 Projection Neurons. Front Neural Circuits 2021; 15:643230. [PMID: 33935658 PMCID: PMC8079783 DOI: 10.3389/fncir.2021.643230] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Accepted: 03/01/2021] [Indexed: 11/13/2022] Open
Abstract
The CA1, an important subregion of the hippocampus, is anatomically and functionally heterogeneous in the dorsal and ventral hippocampus. Here, to dissect the distinctions between the dorsal (dCA1) and ventral CA1 (vCA1) in anatomical connections, we systematically analyzed the direct inputs to dCA1 and vCA1 projection neurons (PNs) with the rabies virus-mediated retrograde trans-monosynaptic tracing system in Thy1-Cre mice. Our mapping results revealed that the input proportions and distributions of dCA1 and vCA1 PNs varied significantly. Inside the hippocampal region, dCA1 and vCA1 PNs shared the same upstream brain regions, but with distinctive distribution patterns along the rostrocaudal axis. The intrahippocampal inputs to the dCA1 and vCA1 exhibited opposite trends, decreasing and increasing gradually along the dorsoventral axis, respectively. For extrahippocampal inputs, dCA1 and vCA1 shared some monosynaptic projections from certain regions such as pallidum, striatum, hypothalamus, and thalamus. However, vCA1, not dCA1, received innervations from the subregions of olfactory areas and amygdala nuclei. Characterization of the direct input networks of dCA1 and vCA1 PNs may provide a structural basis to understand the differential functions of dCA1 and vCA1.
Collapse
Affiliation(s)
- Sijue Tao
- Wuhan National Laboratory for Optoelectronics, Huazhong University of Science and Technology, Wuhan, China.,State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, Key Laboratory of Magnetic Resonance in Biological Systems, Wuhan Center for Magnetic Resonance, Wuhan Institute of Physics and Mathematics, Innovation Academy for Precision Measurement Science and Technology, Chinese Academy of Sciences, Wuhan, China
| | - Yihang Wang
- State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, Key Laboratory of Magnetic Resonance in Biological Systems, Wuhan Center for Magnetic Resonance, Wuhan Institute of Physics and Mathematics, Innovation Academy for Precision Measurement Science and Technology, Chinese Academy of Sciences, Wuhan, China
| | - Jundan Peng
- State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, Key Laboratory of Magnetic Resonance in Biological Systems, Wuhan Center for Magnetic Resonance, Wuhan Institute of Physics and Mathematics, Innovation Academy for Precision Measurement Science and Technology, Chinese Academy of Sciences, Wuhan, China
| | - Yang Zhao
- State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, Key Laboratory of Magnetic Resonance in Biological Systems, Wuhan Center for Magnetic Resonance, Wuhan Institute of Physics and Mathematics, Innovation Academy for Precision Measurement Science and Technology, Chinese Academy of Sciences, Wuhan, China
| | - Xiaobin He
- State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, Key Laboratory of Magnetic Resonance in Biological Systems, Wuhan Center for Magnetic Resonance, Wuhan Institute of Physics and Mathematics, Innovation Academy for Precision Measurement Science and Technology, Chinese Academy of Sciences, Wuhan, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Xuefeng Yu
- Materials and Interfaces Center, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Qing Liu
- State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, Key Laboratory of Magnetic Resonance in Biological Systems, Wuhan Center for Magnetic Resonance, Wuhan Institute of Physics and Mathematics, Innovation Academy for Precision Measurement Science and Technology, Chinese Academy of Sciences, Wuhan, China.,University of Chinese Academy of Sciences, Beijing, China.,Shenzhen Key Lab of Neuropsychiatric Modulation, Guangdong Provincial Key Laboratory of Brain Connectome and Behavior, CAS Key Laboratory of Brain Connectome and Manipulation, The Brain Cognition and Brain Disease Institute (BCBDI), Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China.,Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen, China
| | - Sen Jin
- Materials and Interfaces Center, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Fuqiang Xu
- Wuhan National Laboratory for Optoelectronics, Huazhong University of Science and Technology, Wuhan, China.,State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, Key Laboratory of Magnetic Resonance in Biological Systems, Wuhan Center for Magnetic Resonance, Wuhan Institute of Physics and Mathematics, Innovation Academy for Precision Measurement Science and Technology, Chinese Academy of Sciences, Wuhan, China.,University of Chinese Academy of Sciences, Beijing, China.,Shenzhen Key Lab of Neuropsychiatric Modulation, Guangdong Provincial Key Laboratory of Brain Connectome and Behavior, CAS Key Laboratory of Brain Connectome and Manipulation, The Brain Cognition and Brain Disease Institute (BCBDI), Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China.,Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen, China.,Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China
| |
Collapse
|
49
|
Santa-Marinha L, Castanho I, Silva RR, Bravo FV, Miranda AM, Meira T, Morais-Ribeiro R, Marques F, Xu Y, Point du Jour K, Wenk M, Chan RB, Di Paolo G, Pinto V, Oliveira TG. Phospholipase D1 Ablation Disrupts Mouse Longitudinal Hippocampal Axis Organization and Functioning. Cell Rep 2021; 30:4197-4208.e6. [PMID: 32209478 DOI: 10.1016/j.celrep.2020.02.102] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2019] [Revised: 01/29/2020] [Accepted: 02/27/2020] [Indexed: 01/01/2023] Open
Abstract
Phosphatidic acid (PA) is a signaling lipid involved in the modulation of synaptic structure and functioning. Based on previous work showing a decreasing PA gradient along the longitudinal axis of the rodent hippocampus, we asked whether the dorsal hippocampus (DH) and the ventral hippocampus (VH) are differentially affected by PA modulation. Here, we show that phospholipase D1 (PLD1) is a major hippocampal PA source, compared to PLD2, and that PLD1 ablation affects predominantly the lipidome of the DH. Moreover, Pld1 knockout (KO) mice show specific deficits in novel object recognition and social interaction and disruption in the DH-VH dendritic arborization differentiation in CA1/CA3 pyramidal neurons. Also, Pld1 KO animals present reduced long-term depression (LTD) induction and reduced GluN2A and SNAP-25 protein levels in the DH. Overall, we observe that PLD1-derived PA reduction leads to differential lipid signatures along the longitudinal hippocampal axis, predominantly affecting DH organization and functioning.
Collapse
Affiliation(s)
- Luísa Santa-Marinha
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Campus Gualtar, 4710-057 Braga, Portugal; ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Isabel Castanho
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Campus Gualtar, 4710-057 Braga, Portugal; ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Rita Ribeiro Silva
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Campus Gualtar, 4710-057 Braga, Portugal; ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Francisca Vaz Bravo
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Campus Gualtar, 4710-057 Braga, Portugal; ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - André Miguel Miranda
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Campus Gualtar, 4710-057 Braga, Portugal; ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Torcato Meira
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Campus Gualtar, 4710-057 Braga, Portugal; ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Rafaela Morais-Ribeiro
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Campus Gualtar, 4710-057 Braga, Portugal; ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Fernanda Marques
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Campus Gualtar, 4710-057 Braga, Portugal; ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Yimeng Xu
- Department of Pathology and Cell Biology, Columbia University Medical Center, New York, NY 10032, USA
| | - Kimberly Point du Jour
- Department of Pathology and Cell Biology, Columbia University Medical Center, New York, NY 10032, USA; Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University Medical Center, New York, NY 10032, USA
| | - Markus Wenk
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117596, Singapore
| | - Robin Barry Chan
- Department of Pathology and Cell Biology, Columbia University Medical Center, New York, NY 10032, USA; Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University Medical Center, New York, NY 10032, USA
| | - Gilbert Di Paolo
- Department of Pathology and Cell Biology, Columbia University Medical Center, New York, NY 10032, USA; Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University Medical Center, New York, NY 10032, USA
| | - Vítor Pinto
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Campus Gualtar, 4710-057 Braga, Portugal; ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Tiago Gil Oliveira
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Campus Gualtar, 4710-057 Braga, Portugal; ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal.
| |
Collapse
|
50
|
Lee Y, Bogdanoff D, Wang Y, Hartoularos GC, Woo JM, Mowery CT, Nisonoff HM, Lee DS, Sun Y, Lee J, Mehdizadeh S, Cantlon J, Shifrut E, Ngyuen DN, Roth TL, Song YS, Marson A, Chow ED, Ye CJ. XYZeq: Spatially resolved single-cell RNA sequencing reveals expression heterogeneity in the tumor microenvironment. SCIENCE ADVANCES 2021; 7:7/17/eabg4755. [PMID: 33883145 PMCID: PMC8059935 DOI: 10.1126/sciadv.abg4755] [Citation(s) in RCA: 69] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Accepted: 03/04/2021] [Indexed: 05/07/2023]
Abstract
Single-cell RNA sequencing (scRNA-seq) of tissues has revealed remarkable heterogeneity of cell types and states but does not provide information on the spatial organization of cells. To better understand how individual cells function within an anatomical space, we developed XYZeq, a workflow that encodes spatial metadata into scRNA-seq libraries. We used XYZeq to profile mouse tumor models to capture spatially barcoded transcriptomes from tens of thousands of cells. Analyses of these data revealed the spatial distribution of distinct cell types and a cell migration-associated transcriptomic program in tumor-associated mesenchymal stem cells (MSCs). Furthermore, we identify localized expression of tumor suppressor genes by MSCs that vary with proximity to the tumor core. We demonstrate that XYZeq can be used to map the transcriptome and spatial localization of individual cells in situ to reveal how cell composition and cell states can be affected by location within complex pathological tissue.
Collapse
Affiliation(s)
- Youjin Lee
- Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA 94143, USA.
- Diabetes Center, University of California, San Francisco, San Francisco, CA 94143, USA
- Innovative Genomics Institute, University of California, Berkeley, Berkeley, CA 94720, USA
- J. David Gladstone Institutes, San Francisco, CA 94158, USA
| | - Derek Bogdanoff
- Department of Biochemistry and Biophysics, University of California, San Francisco, San Francisco, CA 94158, USA
- Center for Advanced Technology, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Yutong Wang
- Graduate Group in Biostatistics, University of California, Berkeley, CA 94720, USA
- Center for Computational Biology, University of California, Berkeley, CA 94720, USA
| | - George C Hartoularos
- Graduate Program in Biological and Medical Informatics, University of California, San Francisco, San Francisco, CA 94158, USA
- Division of Rheumatology, Department of Medicine, University of California, San Francisco, CA 94143, USA
- Institute for Human Genetics, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Jonathan M Woo
- Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA 94143, USA
- Diabetes Center, University of California, San Francisco, San Francisco, CA 94143, USA
- Innovative Genomics Institute, University of California, Berkeley, Berkeley, CA 94720, USA
- J. David Gladstone Institutes, San Francisco, CA 94158, USA
| | - Cody T Mowery
- Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA 94143, USA
- Diabetes Center, University of California, San Francisco, San Francisco, CA 94143, USA
- Innovative Genomics Institute, University of California, Berkeley, Berkeley, CA 94720, USA
- J. David Gladstone Institutes, San Francisco, CA 94158, USA
- Medical Scientist Training Program, University of California, San Francisco, San Francisco, CA 94143, USA
- Biomedical Sciences Graduate Program, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Hunter M Nisonoff
- Center for Computational Biology, University of California, Berkeley, CA 94720, USA
| | - David S Lee
- Innovative Genomics Institute, University of California, Berkeley, Berkeley, CA 94720, USA
- Division of Rheumatology, Department of Medicine, University of California, San Francisco, CA 94143, USA
- Institute for Human Genetics, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Yang Sun
- Innovative Genomics Institute, University of California, Berkeley, Berkeley, CA 94720, USA
- Division of Rheumatology, Department of Medicine, University of California, San Francisco, CA 94143, USA
- Institute for Human Genetics, University of California, San Francisco, San Francisco, CA 94143, USA
| | - James Lee
- Division of Hematology and Oncology, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Sadaf Mehdizadeh
- Diabetes Center, University of California, San Francisco, San Francisco, CA 94143, USA
| | | | - Eric Shifrut
- Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA 94143, USA
- Diabetes Center, University of California, San Francisco, San Francisco, CA 94143, USA
- Innovative Genomics Institute, University of California, Berkeley, Berkeley, CA 94720, USA
- J. David Gladstone Institutes, San Francisco, CA 94158, USA
| | - David N Ngyuen
- Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA 94143, USA
- Diabetes Center, University of California, San Francisco, San Francisco, CA 94143, USA
- Innovative Genomics Institute, University of California, Berkeley, Berkeley, CA 94720, USA
- J. David Gladstone Institutes, San Francisco, CA 94158, USA
- Department of Medicine, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Theodore L Roth
- Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA 94143, USA
- Diabetes Center, University of California, San Francisco, San Francisco, CA 94143, USA
- Innovative Genomics Institute, University of California, Berkeley, Berkeley, CA 94720, USA
- Medical Scientist Training Program, University of California, San Francisco, San Francisco, CA 94143, USA
- Biomedical Sciences Graduate Program, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Yun S Song
- Computer Science Division, University of California, Berkeley, CA 94720, USA
- Department of Statistics, University of California, Berkeley, CA 94720, USA
- Chan Zuckerberg Biohub, San Francisco, CA 94158, USA
| | - Alexander Marson
- Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA 94143, USA.
- Diabetes Center, University of California, San Francisco, San Francisco, CA 94143, USA
- Innovative Genomics Institute, University of California, Berkeley, Berkeley, CA 94720, USA
- J. David Gladstone Institutes, San Francisco, CA 94158, USA
- Department of Medicine, University of California, San Francisco, San Francisco, CA 94143, USA
- Chan Zuckerberg Biohub, San Francisco, CA 94158, USA
- UCSF Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA 94158, USA
- Parker Institute for Cancer Immunotherapy, University of California, San Francisco, San Francisco, CA 94129, USA
- Institute for Human Genetics, University of California, San Francisco, San Francisco, CA 94143, USA
- Gladstone-UCSF Institute of Genomic Immunology, San Francisco, CA 94158, USA
| | - Eric D Chow
- Department of Biochemistry and Biophysics, University of California, San Francisco, San Francisco, CA 94158, USA.
- Center for Advanced Technology, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Chun Jimmie Ye
- Division of Rheumatology, Department of Medicine, University of California, San Francisco, CA 94143, USA.
- Chan Zuckerberg Biohub, San Francisco, CA 94158, USA
- Parker Institute for Cancer Immunotherapy, University of California, San Francisco, San Francisco, CA 94129, USA
- Institute for Human Genetics, University of California, San Francisco, San Francisco, CA 94143, USA
- Gladstone-UCSF Institute of Genomic Immunology, San Francisco, CA 94158, USA
- Institute of Computational Health Sciences, University of California, San Francisco, San Francisco, CA 94143, USA
- Department of Epidemiology and Biostatistics, University of California, San Francisco, San Francisco, CA 94158, USA
| |
Collapse
|