1
|
Liu X, Xia J, Shao W, Li X, Yuan D, Xie J, Zhang L, Tang Y, Zhao H, Wu P. Adhesion-Related Pathways and Functional Polarization of Astrocytes in Traumatic Brain Injury: Insights from Single-cell RNA Sequencing. Neuromolecular Med 2025; 27:30. [PMID: 40287916 DOI: 10.1007/s12017-025-08858-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2025] [Accepted: 04/20/2025] [Indexed: 04/29/2025]
Abstract
Traumatic brain injury (TBI) induces profound functional heterogeneity in astrocytes, yet the regulatory mechanisms underlying this diversity remain poorly understood. In this study, we analyzed single-cell RNA sequencing data from the cortex and hippocampus of TBI mouse models to characterize astrocyte subtypes and their functional dynamics. We identified two major reactive subtypes: A1 astrocytes, enriched in inflammatory response, synaptic regulation, and neurodegenerative disease-related pathways; and A2 astrocytes, enriched in lipid metabolism, extracellular matrix (ECM) remodeling, and phagosome formation pathways. These functional differences were consistently observed across datasets with varying injury severities. Notably, adhesion-related pathways-including gap junctions, adherens junctions, and calcium-dependent adhesion-showed significant subtype-specific expression patterns and temporal shifts. Pseudotime trajectory analysis further suggested a potential transition between A1 and A2 states, accompanied by dynamic regulation of adhesion-related genes. Our findings highlight the complex and context-dependent roles of astrocytes in TBI and propose cell adhesion as a key modulator of astrocyte functional polarization.
Collapse
Affiliation(s)
- Xiaoyan Liu
- Institute for Traffic Medicine, Daping Hospital, Army Medical University, Chongqing, 400042, China
- Chongqing Key Laboratory of Traffic Injury and Vehicle Ergonomics, Chongqing, 400042, China
| | - Ji Xia
- Department of Neurosurgery, Daping Hospital and Institute Research of Surgery, Army Medical University, Chongqing, 400042, China
| | - Wenjing Shao
- Department of Anesthesiology, Chongqing Huamei Plastic Surgery Hospital, Chongqing, 400015, China
| | - Xiaoming Li
- Institute for Traffic Medicine, Daping Hospital, Army Medical University, Chongqing, 400042, China
- Chongqing Key Laboratory of Traffic Injury and Vehicle Ergonomics, Chongqing, 400042, China
| | - Danfeng Yuan
- Institute for Traffic Medicine, Daping Hospital, Army Medical University, Chongqing, 400042, China
- Chongqing Key Laboratory of Traffic Injury and Vehicle Ergonomics, Chongqing, 400042, China
| | - Jingru Xie
- Institute for Traffic Medicine, Daping Hospital, Army Medical University, Chongqing, 400042, China
- Chongqing Key Laboratory of Traffic Injury and Vehicle Ergonomics, Chongqing, 400042, China
| | - Liang Zhang
- Institute for Traffic Medicine, Daping Hospital, Army Medical University, Chongqing, 400042, China
- Chongqing Key Laboratory of Traffic Injury and Vehicle Ergonomics, Chongqing, 400042, China
| | - Yuqian Tang
- Institute for Traffic Medicine, Daping Hospital, Army Medical University, Chongqing, 400042, China
- Chongqing Key Laboratory of Traffic Injury and Vehicle Ergonomics, Chongqing, 400042, China
| | - Hui Zhao
- Institute for Traffic Medicine, Daping Hospital, Army Medical University, Chongqing, 400042, China
- Chongqing Key Laboratory of Traffic Injury and Vehicle Ergonomics, Chongqing, 400042, China
| | - Pengfei Wu
- Institute for Traffic Medicine, Daping Hospital, Army Medical University, Chongqing, 400042, China.
- Chongqing Key Laboratory of Traffic Injury and Vehicle Ergonomics, Chongqing, 400042, China.
| |
Collapse
|
2
|
Thorp EB, Ananthakrishnan A, Lantz CW. Decoding immune cell interactions during cardiac allograft vasculopathy: insights derived from bioinformatic strategies. Front Cardiovasc Med 2025; 12:1568528. [PMID: 40342971 PMCID: PMC12058854 DOI: 10.3389/fcvm.2025.1568528] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2025] [Accepted: 04/04/2025] [Indexed: 05/11/2025] Open
Abstract
Chronic allograft vasculopathy (CAV) is a major cause of late graft failure in heart transplant recipients, characterized by progressive intimal thickening and diffuse narrowing of the coronary arteries. Unlike atherosclerosis, CAV exhibits a distinct cellular composition and lesion distribution, yet its pathogenesis remains incompletely understood. A major challenge in CAV research has been the limited application of advanced "-omics" technologies, which have revolutionized the study of other vascular diseases. Recent advancements in single-cell and spatial transcriptomics, proteomics, and metabolomics have begun to uncover the complex immune-endothelial-stromal interactions driving CAV progression. Notably, single-cell RNA sequencing has identified previously unrecognized immune cell populations and signaling pathways implicated in endothelial injury and vascular remodeling after heart transplantation. Despite these breakthroughs, studies applying these technologies to CAV remain sparse, limiting the translation of these insights into clinical practice. This review aims to bridge this gap by summarizing recent findings from single-cell and multi-omic approaches, highlighting key discoveries, and discussing their implications for understanding CAV pathogenesis.
Collapse
Affiliation(s)
- Edward B. Thorp
- Department of Pathology, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| | - Aparnaa Ananthakrishnan
- Department of Pathology, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| | - Connor W. Lantz
- Department of Surgery, Comprehensive Transplant Center, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| |
Collapse
|
3
|
Li Y, Liu X, Guo L, Han K, Fang S, Wan X, Wang D, Xu X, Jiang L, Fan G, Xu M. SpaGRN: Investigating spatially informed regulatory paths for spatially resolved transcriptomics data. Cell Syst 2025; 16:101243. [PMID: 40179878 DOI: 10.1016/j.cels.2025.101243] [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: 11/27/2023] [Revised: 08/30/2024] [Accepted: 03/07/2025] [Indexed: 04/05/2025]
Abstract
Cells spatially organize into distinct cell types or functional domains through localized gene regulatory networks. However, current spatially resolved transcriptomics analyses fail to integrate spatial constraints and proximal cell influences, limiting the mechanistic understanding of tissue organization. Here, we introduce SpaGRN, a statistical framework that reconstructs cell-type- or functional-domain-specific, dynamic, and spatial regulons by coupling intracellular spatial regulatory causality with extracellular signaling path information. Benchmarking across synthetic and real datasets demonstrates SpaGRN's superior precision over state-of-the-art tools in identifying context-dependent regulons. Applied to diverse spatially resolved transcriptomics platforms (Stereo-seq, STARmap, MERFISH, CosMx, Slide-seq, and 10x Visium), complex cancerous samples, and 3D datasets of developing Drosophila embryos and larvae, SpaGRN not only provides a versatile toolkit for decoding receptor-mediated spatial regulons but also reveals spatiotemporal regulatory mechanisms underlying organogenesis and inflammation.
Collapse
Affiliation(s)
- Yao Li
- BGI Research, Sanya 572025, China; BGI Research, Qingdao 266555, China
| | | | - Lidong Guo
- BGI Research, Qingdao 266555, China; College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Kai Han
- BGI Research, Qingdao 266555, China
| | - Shuangsang Fang
- BGI Research, Beijing 102601, China; BGI Research, Shenzhen 518083, China
| | - Xinjiang Wan
- CAS Key Laboratory of Marine Ecology and Environmental Sciences, and Center of Deep Sea Research Institute of Oceanology, Chinese Academy of Sciences, Qingdao 266071, China
| | | | - Xun Xu
- BGI Research, Wuhan 430074, China; State Key Laboratory of Genome and Multi-omics Technologies, BGI Research, Shenzhen 518083, China
| | - Ling Jiang
- State Key Laboratory of Materials-Oriented Chemical Engineering, Nanjing Tech University, Nanjing 211816, China.
| | - Guangyi Fan
- BGI Research, Sanya 572025, China; BGI Research, Qingdao 266555, China; BGI Research, Shenzhen 518083, China; State Key Laboratory of Genome and Multi-omics Technologies, BGI Research, Shenzhen 518083, China.
| | - Mengyang Xu
- BGI Research, Sanya 572025, China; BGI Research, Qingdao 266555, China; BGI Research, Shenzhen 518083, China; State Key Laboratory of Genome and Multi-omics Technologies, BGI Research, Shenzhen 518083, China.
| |
Collapse
|
4
|
Kleino I, Perk M, Sousa AGG, Linden M, Mathlin J, Giesel D, Frolovaite P, Pietilä S, Junttila S, Suomi T, Elo LL. CellRomeR: an R package for clustering cell migration phenotypes from microscopy data. BIOINFORMATICS ADVANCES 2025; 5:vbaf069. [PMID: 40330627 PMCID: PMC12052403 DOI: 10.1093/bioadv/vbaf069] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/19/2024] [Revised: 03/06/2025] [Accepted: 04/02/2025] [Indexed: 05/08/2025]
Abstract
Motivation The analysis of cell migration using time-lapse microscopy typically focuses on track characteristics for classification and statistical evaluation of migration behaviour. However, considerable heterogeneity can be seen in cell morphology and microscope signal intensity features within the migrating cell populations. Results To utilize this information in cell migration analysis, we introduce here an R package CellRomeR, designed for the phenotypic clustering of cells based on their morphological and motility features from microscopy images. Utilizing machine learning techniques and building on an iterative clustering projection method, CellRomeR offers a new approach to identify heterogeneity in cell populations. The clustering of cells along the migration tracks allows association of distinct cellular phenotypes with different cell migration types and detection of migration patterns associated with stable and unstable cell phenotypes. The user-friendly interface of CellRomeR and multiple visualization options facilitate an in-depth understanding of cellular behaviour, addressing previous challenges in clustering cell trajectories using microscope cell tracking data. Availability and implementation CellRomeR is available as an R package from https://github.com/elolab/CellRomeR.
Collapse
Affiliation(s)
- Iivari Kleino
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, Turku, FI-20520, Finland
| | - Mats Perk
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, Turku, FI-20520, Finland
| | - António G G Sousa
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, Turku, FI-20520, Finland
| | - Markus Linden
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, Turku, FI-20520, Finland
| | - Julia Mathlin
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, Turku, FI-20520, Finland
- Institute of Biomedicine, University of Turku, Turku, FI-20014, Finland
| | - Daniel Giesel
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, Turku, FI-20520, Finland
| | - Paulina Frolovaite
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, Turku, FI-20520, Finland
| | - Sami Pietilä
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, Turku, FI-20520, Finland
| | - Sini Junttila
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, Turku, FI-20520, Finland
| | - Tomi Suomi
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, Turku, FI-20520, Finland
| | - Laura L Elo
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, Turku, FI-20520, Finland
- Institute of Biomedicine, University of Turku, Turku, FI-20014, Finland
| |
Collapse
|
5
|
Cheng Y, Peng H, Chen Q, Xu L, Qin L. Machine learning-based transcriptmics analysis reveals BMX, GRB10, and GADD45A as crucial biomarkers and therapeutic targets in sepsis. Front Pharmacol 2025; 16:1576467. [PMID: 40230692 PMCID: PMC11994739 DOI: 10.3389/fphar.2025.1576467] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2025] [Accepted: 03/18/2025] [Indexed: 04/16/2025] Open
Abstract
Sepsis is a life-threatening condition characterized by a dysregulated host response to infection, resulting in high mortality rates and complex clinical management. This study leverages transcriptomics and machine learning (ML) to identify critical biomarkers and therapeutic targets in sepsis. Analyzing microarray data from the Gene Expression Omnibus (GEO) datasets GSE28750, GSE26440, GSE13205, and GSE9960, we discovered three pivotal biomarkers that BMX (bone marrow tyrosine kinase gene on chromosome X), GRB10 (growth factor receptor bound protein 10), and GADD45A (growth arrest and DNA damage inducible alpha), exhibiting exceptional diagnostic accuracy (AUC >0.9). Functional enrichment analyses revealed that these genes play key roles in reactive oxygen species metabolism and immune response regulation. Specifically, GADD45A was positively correlated with eosinophils and inversely associated with activated NK cells, CD8 T cells, and activated memory CD4 T cells. BMX showed positive correlations with eosinophils, mast cells, and neutrophils, while GRB10 was linked to eosinophils and M2 macrophages. Additionally, we constructed a comprehensive mRNA-miRNA-lncRNA regulatory network, identifying key interactions that may drive sepsis pathogenesis. Molecular docking and dynamics simulations validated Bendroflumethiazide, Cianidanol, and Hexamidine as promising therapeutic agents targeting these biomarkers. In conclusion, this integrated approach provides profound insights into the molecular mechanisms underlying sepsis, pinpointing BMX, GRB10, and GADD45A as pivotal biomarkers and therapeutic targets. These findings significantly enhance our understanding of sepsis pathophysiology and lay the groundwork for developing personalized diagnostic and therapeutic strategies aimed at improving patient outcomes.
Collapse
Affiliation(s)
- Yanwei Cheng
- Department of Emergency, Henan Provincial People’s Hospital, People’s Hospital of Zhengzhou University, People’s Hospital of Henan University, Zhengzhou, China
| | - Haoran Peng
- Department of Neurology, People’s Hospital of Henan University, Henan Provincial People’s Hospital, Zhengzhou, Henan, China
| | - Qiao Chen
- Nursing Department, Air Force Medical Center, PLA, Beijing, China
| | - Lijun Xu
- Department of Emergency, Henan Provincial People’s Hospital, People’s Hospital of Zhengzhou University, People’s Hospital of Henan University, Zhengzhou, China
| | - Lijie Qin
- Department of Emergency, Henan Provincial People’s Hospital, People’s Hospital of Zhengzhou University, People’s Hospital of Henan University, Zhengzhou, China
| |
Collapse
|
6
|
Du MRM, Wang C, Law CW, Amann-Zalcenstein D, Anttila CJA, Ling L, Hickey PF, Sargeant CJ, Chen Y, Ioannidis LJ, Rajasekhar P, Yip RKH, Rogers KL, Hansen DS, Bowden R, Ritchie ME. Benchmarking spatial transcriptomics technologies with the multi-sample SpatialBenchVisium dataset. Genome Biol 2025; 26:77. [PMID: 40156041 PMCID: PMC11954323 DOI: 10.1186/s13059-025-03543-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Accepted: 03/13/2025] [Indexed: 04/01/2025] Open
Abstract
BACKGROUND Spatial transcriptomics allows gene expression to be measured within complex tissue contexts. Among the array of spatial capture technologies available is 10x Genomics' Visium platform, a popular method which enables transcriptome-wide profiling of tissue sections. Visium offers a range of sample handling and library construction methods which introduces a need for benchmarking to compare data quality and assess how well the technology can recover expected tissue features and biological signatures. RESULTS Here we present SpatialBenchVisium, a unique reference dataset generated from spleen tissue of mice responding to malaria infection spanning several tissue preparation protocols (both fresh frozen and FFPE, with either manual or CytAssist tissue placement). We note better quality control metrics in reference samples prepared using probe-based capture methods, particularly those processed with CytAssist, validating the improvement in data quality produced with the platform. Our analysis of replicate samples extends to explore spatially variable gene detection, the outcomes of clustering and cell deconvolution using matched single-cell RNA-sequencing data and publicly available reference data to identify cell types and tissue regions expected in the spleen. Multi-sample differential expression analysis recovered known gene signatures related to biological sex or gene knockout.
Collapse
Affiliation(s)
- Mei R M Du
- The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, 3052, Australia
| | - Changqing Wang
- The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, 3052, Australia
- Department of Medical Biology, The University of Melbourne, Parkville, VIC, 3010, Australia
| | - Charity W Law
- The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, 3052, Australia
- Department of Medical Biology, The University of Melbourne, Parkville, VIC, 3010, Australia
| | - Daniela Amann-Zalcenstein
- The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, 3052, Australia
- Department of Medical Biology, The University of Melbourne, Parkville, VIC, 3010, Australia
| | - Casey J A Anttila
- The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, 3052, Australia
| | - Ling Ling
- The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, 3052, Australia
| | - Peter F Hickey
- The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, 3052, Australia
- Department of Medical Biology, The University of Melbourne, Parkville, VIC, 3010, Australia
| | - Callum J Sargeant
- The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, 3052, Australia
| | - Yunshun Chen
- The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, 3052, Australia
- Department of Medical Biology, The University of Melbourne, Parkville, VIC, 3010, Australia
| | - Lisa J Ioannidis
- The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, 3052, Australia
- Department of Medical Biology, The University of Melbourne, Parkville, VIC, 3010, Australia
| | - Pradeep Rajasekhar
- The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, 3052, Australia
- Department of Medical Biology, The University of Melbourne, Parkville, VIC, 3010, Australia
| | - Raymond K H Yip
- The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, 3052, Australia
- Department of Medical Biology, The University of Melbourne, Parkville, VIC, 3010, Australia
| | - Kelly L Rogers
- The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, 3052, Australia
- Department of Medical Biology, The University of Melbourne, Parkville, VIC, 3010, Australia
| | - Diana S Hansen
- Department of Microbiology, Monash Biomedicine Discovery Institute, Monash University, Clayton, VIC, 3800, Australia
| | - Rory Bowden
- The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, 3052, Australia
- Department of Medical Biology, The University of Melbourne, Parkville, VIC, 3010, Australia
| | - Matthew E Ritchie
- The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, 3052, Australia.
- Department of Medical Biology, The University of Melbourne, Parkville, VIC, 3010, Australia.
| |
Collapse
|
7
|
Wusiman Z, Zhang AM, Zhang SS, Zhao PP, Kang YT, Zhang Y, Li ZJ, Huo SX. Galangin ameliorates PTU-induced vitiligo in zebrafish and B16F10 cells by increasing melanogenesis through activation of the p38/JNK MAPK pathway. Front Pharmacol 2025; 16:1521097. [PMID: 40129935 PMCID: PMC11931063 DOI: 10.3389/fphar.2025.1521097] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2024] [Accepted: 02/17/2025] [Indexed: 03/26/2025] Open
Abstract
Objective Alpinia officinarum Hance is a traditional herb in Xinjiang for the treatment of vitiligo, and galangin (GA) is a flavonoid isolated from its roots. However, its therapeutic mechanism remains unclear. Methods In this study, 1-phenyl-2-thiourea (PTU) was used to establish a vitiligo model in zebrafish. After successful modeling, different concentrations of GA (1 and 2 μM) were administered, and the distribution of melanin granules was observed by assaying the melanin content, masson-fontana staining and tyrosinase activity. Transcriptomic analysis and molecular docking were used to identify potential GA-related pathways and targets for improving vitiligo. In addition, we evaluated the proliferation of B16F10 cells by PTU induction and also observed cellular melanin distribution using masson-fontana staining. Finally, Western blot was performed to detect the proteins of the relevant pathways. Results The results showed that GA significantly increased melanin production and tyrosinase activity in depigmented zebrafish. In addition, we found that GA decreased ROS and MDA levels and increased the expression of GSH, CAT and T-SOD. In addition, transcriptome analysis indicated that GA likely acts through the mitogen-activated protein kinase (MAPK) signaling pathway. GA has a strong binding affinity for important targets.GA significantly increased the expression of genes such as mapk8b, mapk14a, mapk3, mitf, tyr, tyrp1b, tyrp1a, dct, and oca2, and decreased the expression of genes such as expression of genes such as raf1 and egfr. In addition, GA enhanced the viability of B16F10 cells, increased intracellular melanin content, and increased the expression of proteins such as p38, JNK1/2/3, TYR, MITF, TRP1, TRP2, and so on. Conclusion GA increases melanin production and distribution, improves tyrosinase activity, upregulates the expression of related genes and proteins through activation of MAPK and tyrosine metabolic pathways, downregulates oxidative stress, and then regulates changes in melanin synthesis to improve vitiligo.
Collapse
Affiliation(s)
- Zulipikaer Wusiman
- Uygur Medical Hospital of Xinjiang Uygur Autonomous Region, Urumqi, China
- Key Laboratory of Evidence-Based and Translation, Xinjiang Hospital preparation of Traditional Chinese Medicine, Urumqi, China
| | - Ai-Mei Zhang
- The First Affiliated Hospital of Shihezi University, Shihezi, China
| | - Shu-Shu Zhang
- Uygur Medical Hospital of Xinjiang Uygur Autonomous Region, Urumqi, China
| | - Ping-Ping Zhao
- Uygur Medical Hospital of Xinjiang Uygur Autonomous Region, Urumqi, China
- Key Laboratory of Evidence-Based and Translation, Xinjiang Hospital preparation of Traditional Chinese Medicine, Urumqi, China
| | - Yu-Tong Kang
- Uygur Medical Hospital of Xinjiang Uygur Autonomous Region, Urumqi, China
- Key Laboratory of Evidence-Based and Translation, Xinjiang Hospital preparation of Traditional Chinese Medicine, Urumqi, China
| | - Yun Zhang
- Biology Institute, Qilu University of Technology (Shandong Academy of Sciences), Jinan, China
| | - Zhi-Jian Li
- Uygur Medical Hospital of Xinjiang Uygur Autonomous Region, Urumqi, China
- Key Laboratory of Evidence-Based and Translation, Xinjiang Hospital preparation of Traditional Chinese Medicine, Urumqi, China
| | - Shi-Xia Huo
- Uygur Medical Hospital of Xinjiang Uygur Autonomous Region, Urumqi, China
- Key Laboratory of Evidence-Based and Translation, Xinjiang Hospital preparation of Traditional Chinese Medicine, Urumqi, China
| |
Collapse
|
8
|
Kumaran G, Carroll L, Muirhead N, Bottomley MJ. How Can Spatial Transcriptomic Profiling Advance Our Understanding of Skin Diseases? J Invest Dermatol 2025; 145:522-535. [PMID: 39177547 DOI: 10.1016/j.jid.2024.07.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2024] [Revised: 05/23/2024] [Accepted: 07/04/2024] [Indexed: 08/24/2024]
Abstract
Spatial transcriptomic (ST) profiling is the mapping of gene expression within cell populations with preservation of positional context and represents an exciting new approach to develop our understanding of local and regional influences upon skin biology in health and disease. With the ability to probe from a few hundred transcripts to the entire transcriptome, multiple ST approaches are now widely available. In this paper, we review the ST field and discuss its application to dermatology. Its potential to advance our understanding of skin biology in health and disease is highlighted through the illustrative examples of 3 research areas: cutaneous aging, tumorigenesis, and psoriasis.
Collapse
Affiliation(s)
- Girishkumar Kumaran
- Chinese Academy of Medical Sciences Oxford Institute, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Liam Carroll
- Chinese Academy of Medical Sciences Oxford Institute, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| | | | - Matthew J Bottomley
- Chinese Academy of Medical Sciences Oxford Institute, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom.
| |
Collapse
|
9
|
Krull D, Haynes P, Kesarwani A, Tessier J, Chen BJ, Hunter K, Rodriguez D, Liang Y, Mansfield J, McClain M, Ramos C, Bonnevie E, Anguiano E. A best practices framework for spatial biology studies in drug discovery and development: enabling successful cohort studies using digital spatial profiling. J Histotechnol 2025; 48:7-26. [PMID: 39225147 DOI: 10.1080/01478885.2024.2391683] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Accepted: 08/08/2024] [Indexed: 09/04/2024]
Abstract
The discovery of biomarkers, essential for successful drug development, is often hindered by the limited availability of tissue samples, typically obtained through core needle biopsies. Standard 'omics platforms can consume significant amounts of tissue, forcing scientist to trade off spatial context for high-plex assays, such as genome-wide assays. While bulk gene expression approaches and standard single-cell transcriptomics have been valuable in defining various molecular and cellular mechanisms, they do not retain spatial context. As such, they have limited power in resolving tissue heterogeneity and cell-cell interactions. Current spatial transcriptomics platforms offer limited transcriptome coverage and have low throughput, restricting the number of samples that can be analyzed daily or even weekly. While the Digital Spatial Profiling (DSP) method does not provide single-cell resolution, it presents a significant advancement by enabling scalable whole transcriptome and ultrahigh-plex protein analysis from distinct tissue compartments and structures using a single tissue slide. These capabilities overcome significant constraints in biomarker analysis in solid tissue specimens. These advancements in tissue profiling play a crucial role in deepening our understanding of disease biology and in identifying potential therapeutic targets and biomarkers. To enhance the use of spatial biology tools in drug discovery and development, the DSP Scientific Consortium has created best practices guidelines. These guidelines, built on digital spatial profiling data and expertise, offer a practical framework for designing spatial studies and using current and future spatial biology platforms. The aim is to improve tissue analysis in all research areas supporting drug discovery and development.
Collapse
Affiliation(s)
- David Krull
- Precision Medicine, GlaxoSmithKline, Collegeville, USA
| | - Premi Haynes
- Cancer Immunology & Cell Therapy Thematic Research Center, Bristol Myers Squibb, Seattle, WA, USA
| | | | - Julien Tessier
- Precision Medicine and Computational Biology, Sanofi, Cambridge, USA
| | - Benjamin J Chen
- Translational Medicine, Bristol Myers Squibb, Cambridge, MA, USA
| | - Kelly Hunter
- Molecular Pathology and Histology, ProPath-UK Ltd, Hereford, UK
| | | | - Yan Liang
- Spatial Platforms Product Development and Support, NanoString Technologies Inc, Seattle, USA
| | - Jim Mansfield
- Research Business Development, Visiopharm Corp, Broomfield, USA
| | - Maxine McClain
- Spatial Platforms Product Development and Support, NanoString Technologies Inc, Seattle, USA
| | | | - Edward Bonnevie
- Cancer Immunology & Cell Therapy Thematic Research Center, Bristol Myers Squibb, Seattle, WA, USA
| | - Esperanza Anguiano
- Spatial Platforms Product Development and Support, NanoString Technologies Inc, Seattle, USA
| |
Collapse
|
10
|
Wu Q, Zhang D, Dai S, Liu F, Zhang W, Shen T. Desflurane attenuates renal ischemia-reperfusion injury by modulating ITGB1/CD9 and reducing oxidative stress in tubular epithelial cells. Redox Biol 2025; 80:103490. [PMID: 39854938 PMCID: PMC11804601 DOI: 10.1016/j.redox.2025.103490] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Revised: 12/26/2024] [Accepted: 01/02/2025] [Indexed: 01/27/2025] Open
Abstract
OBJECTIVE Renal ischemia-reperfusion (I/R) injury triggers significant oxidative stress and inflammation, leading to tubular epithelial cell (TEC) damage. This study investigates the protective role of Desflurane (DFE) in renal I/R by modulating the ITGB1/CD9 signaling pathway and mitigating oxidative damage. METHODS Single-cell RNA sequencing (scRNA-seq) and transcriptome analysis identified ITGB1 as a key regulatory gene in TECs during renal I/R. The effects of DFE on ITGB1/CD9 expression were evaluated through in vitro experiments using RT-qPCR, Western blot, and TUNEL assays. A mouse model of renal I/R was employed to assess renal function and oxidative stress markers under DFE treatment. RESULTS DFE reduced ITGB1 and CD9 expression, resulting in decreased TEC apoptosis and enhanced proliferation. In vivo, DFE-treated mice exhibited improved renal function, with significantly lower serum creatinine and blood urea nitrogen (BUN) levels. Additionally, DFE reduced oxidative stress, as indicated by decreased malondialdehyde (MDA) and myeloperoxidase (MPO) activity, alongside increased superoxide dismutase (SOD) activity. CONCLUSION DFE confers renal protection against I/R by modulating the ITGB1/CD9 axis and reducing oxidative stress, offering a promising therapeutic strategy for mitigating kidney damage.
Collapse
Affiliation(s)
- Qiaoling Wu
- Department of Anesthesiology, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, 121001, PR China
| | - Dongbo Zhang
- Department of Anesthesiology, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, 121001, PR China
| | - Siqi Dai
- Department of Anesthesiology, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, 121001, PR China
| | - Feifei Liu
- Department of Anesthesiology, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, 121001, PR China
| | - Wei Zhang
- Department of Anesthesiology, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, 121001, PR China
| | - Tu Shen
- Department of Anesthesiology, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, 121001, PR China.
| |
Collapse
|
11
|
Robinson SD, Filippopoulou C, Besta S, Samuels M, Betrán AL, Abu Ajamieh M, Vella V, Jones W, Giamas G. Spatial biology - unravelling complexity within the glioblastoma microenvironment. Trends Mol Med 2025:S1471-4914(25)00014-0. [PMID: 39934022 DOI: 10.1016/j.molmed.2025.01.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2024] [Revised: 01/21/2025] [Accepted: 01/22/2025] [Indexed: 02/13/2025]
Abstract
The advent and refinement of state-of-the-art spatial biology technologies have facilitated analysis that combines the advantages of high-throughput single cell analysis with techniques that preserve tissue architecture. This combination of cellular phenotyping with retained spatial context provides a much greater understanding of cellular interactions within the tumour microenvironment (TME). For glioblastoma, with its significant intra-tumoural heterogeneity, cellular plasticity, and complex TME, appreciating and understanding these spatial patterns may prove key to improving patient outcomes. This review examines the advances in spatial biology techniques, discusses how these methodologies are being applied to study glioblastoma, and explores how spatial information improves understanding of the TME. Ultimately, it is this spatial context that will accelerate the identification of more effective treatments for glioblastoma.
Collapse
Affiliation(s)
- Stephen D Robinson
- Department of Biochemistry and Biomedicine, School of Life Sciences, University of Sussex, Brighton, BN1 9QG, UK; Sussex Cancer Centre, University Hospitals Sussex NHS Foundation Trust, Brighton, BN2 5BD, UK.
| | - Chrysa Filippopoulou
- International Oncology Institute, The First Affiliated Hospital of Zhejiang Chinese Medical University, Oncology Department of the First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Simoni Besta
- International Oncology Institute, The First Affiliated Hospital of Zhejiang Chinese Medical University, Oncology Department of the First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Mark Samuels
- International Oncology Institute, The First Affiliated Hospital of Zhejiang Chinese Medical University, Oncology Department of the First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Andrea L Betrán
- Department of Biochemistry and Biomedicine, School of Life Sciences, University of Sussex, Brighton, BN1 9QG, UK
| | - Maha Abu Ajamieh
- Department of Biochemistry and Biomedicine, School of Life Sciences, University of Sussex, Brighton, BN1 9QG, UK
| | - Viviana Vella
- Department of Biochemistry and Biomedicine, School of Life Sciences, University of Sussex, Brighton, BN1 9QG, UK
| | - William Jones
- Department of Biochemistry and Biomedicine, School of Life Sciences, University of Sussex, Brighton, BN1 9QG, UK
| | - Georgios Giamas
- Department of Biochemistry and Biomedicine, School of Life Sciences, University of Sussex, Brighton, BN1 9QG, UK; International Oncology Institute, The First Affiliated Hospital of Zhejiang Chinese Medical University, Oncology Department of the First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou 310053, China.
| |
Collapse
|
12
|
Yang T, Zhang N, Yang N. Single-cell sequencing in diabetic retinopathy: progress and prospects. J Transl Med 2025; 23:49. [PMID: 39806376 PMCID: PMC11727737 DOI: 10.1186/s12967-024-06066-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2024] [Accepted: 12/30/2024] [Indexed: 01/16/2025] Open
Abstract
Diabetic retinopathy is a major ocular complication of diabetes, characterized by progressive retinal microvascular damage and significant visual impairment in working-age adults. Traditional bulk RNA sequencing offers overall gene expression profiles but does not account for cellular heterogeneity. Single-cell RNA sequencing overcomes this limitation by providing transcriptomic data at the individual cell level and distinguishing novel cell subtypes, developmental trajectories, and intercellular communications. Researchers can use single-cell sequencing to draw retinal cell atlases and identify the transcriptomic features of retinal cells, enhancing our understanding of the pathogenesis and pathological changes in diabetic retinopathy. Additionally, single-cell sequencing is widely employed to analyze retinal organoids and single extracellular vesicles. Single-cell multi-omics sequencing integrates omics information, whereas stereo-sequencing analyzes gene expression and spatiotemporal data simultaneously. This review discusses the protocols of single-cell sequencing for obtaining single cells from retina and accurate sequencing data. It highlights the applications and advancements of single-cell sequencing in the study of normal retinas and the pathological changes associated with diabetic retinopathy. This underscores the potential of these technologies to deepen our understanding of the pathogenesis of diabetic retinopathy that may lead to the introduction of new therapeutic strategies.
Collapse
Affiliation(s)
- Tianshu Yang
- Department of Ophthalmology, Renmin Hospital of Wuhan University, Jiefang Road, Wuhan, Hubei, 430060, China
| | - Ningzhi Zhang
- Department of Ophthalmology, Renmin Hospital of Wuhan University, Jiefang Road, Wuhan, Hubei, 430060, China
| | - Ning Yang
- Department of Ophthalmology, Renmin Hospital of Wuhan University, Jiefang Road, Wuhan, Hubei, 430060, China.
| |
Collapse
|
13
|
Sisó S, Kavirayani AM, Couto S, Stierstorfer B, Mohanan S, Morel C, Marella M, Bangari DS, Clark E, Schwartz A, Carreira V. Trends and Challenges of the Modern Pathology Laboratory for Biopharmaceutical Research Excellence. Toxicol Pathol 2025; 53:5-20. [PMID: 39673215 DOI: 10.1177/01926233241303898] [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: 12/16/2024]
Abstract
Pathology, a fundamental discipline that bridges basic scientific discovery to the clinic, is integral to successful drug development. Intrinsically multimodal and multidimensional, anatomic pathology continues to be empowered by advancements in molecular and digital technologies enabling the spatial tissue detection of biomolecules such as genes, transcripts, and proteins. Over the past two decades, breakthroughs in spatial molecular biology technologies and advancements in automation and digitization of laboratory processes have enabled the implementation of higher throughput assays and the generation of extensive molecular data sets from tissue sections in biopharmaceutical research and development research units. It is our goal to provide readers with some rationale, advice, and ideas to help establish a modern molecular pathology laboratory to meet the emerging needs of biopharmaceutical research. This manuscript provides (1) a high-level overview of the current state and future vision for excellence in research pathology practice and (2) shared perspectives on how to optimally leverage the expertise of discovery, toxicologic, and translational pathologists to provide effective spatial, molecular, and digital pathology data to support modern drug discovery. It captures insights from the experiences, challenges, and solutions from pathology laboratories of various biopharmaceutical organizations, including their approaches to troubleshooting and adopting new technologies.
Collapse
Affiliation(s)
- Sílvia Sisó
- AbbVie Bioresearch Center, Worcester, Massachusetts, USA
| | | | | | | | | | | | - Mathiew Marella
- Janssen Research & Development, LLC, La Jolla, California, USA
| | | | - Elizabeth Clark
- Boehringer Ingelheim Pharmaceuticals, Inc., Ridgefield, Connecticut, USA
| | | | | |
Collapse
|
14
|
Li X, Huang Z, Bai J, Che A, Zhou J, Yang H. Molecular profiling unveils pyroptosis markers in preterm birth. FASEB J 2024; 38:e70112. [PMID: 39673596 DOI: 10.1096/fj.202302716rr] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2023] [Revised: 08/29/2024] [Accepted: 10/04/2024] [Indexed: 12/16/2024]
Abstract
Through a comprehensive examination of pyroptosis-related differential expressed genes (PRDEGs), this work investigates the molecular complexities of spontaneous preterm birth (SPTB), also known as premature delivery, before the due date. Through the process of merging and correcting batch effects in the GSE120480 and GSE73714 datasets, we were able to identify 36 PRDEGs that exhibited significant expression differentiation in SPTB. Through functional enrichment and pathway analysis, their importance in amino acid transport and cytokine receptor interaction has been highlighted. Among the genes that have emerged as crucial, CEBPA, APOA1, and CEP55 have been identified. The relevance of these molecules was demonstrated using experimental knockdowns, which also suggested that they could be used as molecular biomarkers and therapeutic targets for SPTB.
Collapse
Affiliation(s)
- Xiaoyun Li
- Department of Ultrasound Medicine, Longgang District Maternity & Child Healthcare Hospital of Shenzhen City (Longgang Maternity and Child Institute of Shantou University Medical College), Shenzhen, China
| | - Zhulan Huang
- Department of Ultrasound Medicine, Longgang District Maternity & Child Healthcare Hospital of Shenzhen City (Longgang Maternity and Child Institute of Shantou University Medical College), Shenzhen, China
| | - Jiangtao Bai
- Central Laboratory, Longgang District Maternity & Child Healthcare Hospital of Shenzhen City (Longgang Maternity and Child Institute of Shantou University Medical College), Shenzhen, China
| | - Aiwen Che
- Department of Pathology, Longgang District Maternity & Child Healthcare Hospital of Shenzhen City (Longgang Maternity and Child Institute of Shantou University Medical College), Shenzhen, China
| | - Jinhua Zhou
- Department of Obstetrics, Longgang District Maternity & Child Healthcare Hospital of Shenzhen City (Longgang Maternity and Child Institute of Shantou University Medical College), Shenzhen, China
| | - Hongmei Yang
- Department of Clinical Laboratory, Longgang District Maternity & Child Healthcare Hospital of Shenzhen City (Longgang Maternity and Child Institute of Shantou University Medical College), Shenzhen, China
| |
Collapse
|
15
|
Li X, Dong X, Zhang W, Shi Z, Liu Z, Sa Y, Li L, Ni N, Mei Y. Multi-omics in exploring the pathophysiology of diabetic retinopathy. Front Cell Dev Biol 2024; 12:1500474. [PMID: 39723239 PMCID: PMC11668801 DOI: 10.3389/fcell.2024.1500474] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2024] [Accepted: 11/25/2024] [Indexed: 12/28/2024] Open
Abstract
Diabetic retinopathy (DR) is a leading global cause of vision impairment, with its prevalence increasing alongside the rising rates of diabetes mellitus (DM). Despite the retina's complex structure, the underlying pathology of DR remains incompletely understood. Single-cell RNA sequencing (scRNA-seq) and recent advancements in multi-omics analyses have revolutionized molecular profiling, enabling high-throughput analysis and comprehensive characterization of complex biological systems. This review highlights the significant contributions of scRNA-seq, in conjunction with other multi-omics technologies, to DR research. Integrated scRNA-seq and transcriptomic analyses have revealed novel insights into DR pathogenesis, including alternative transcription start site events, fluctuations in cell populations, altered gene expression profiles, and critical signaling pathways within retinal cells. Furthermore, by integrating scRNA-seq with genetic association studies and multi-omics analyses, researchers have identified novel biomarkers, susceptibility genes, and potential therapeutic targets for DR, emphasizing the importance of specific retinal cell types in disease progression. The integration of scRNA-seq with metabolomics has also been instrumental in identifying specific metabolites and dysregulated pathways associated with DR. It is highly conceivable that the continued synergy between scRNA-seq and other multi-omics approaches will accelerate the discovery of underlying mechanisms and the development of novel therapeutic interventions for DR.
Collapse
Affiliation(s)
- Xinlu Li
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming, China
- Department of Ophthalmology, The Affiliated Hospital of Kunming University of Science and Technology, Kunming, China
- Department of Ophthalmology, The First People’s Hospital of Yunnan Province, Kunming, China
- Medical School, Kunming University of Science and Technology, Kunming, China
| | - XiaoJing Dong
- Department of Ophthalmology, The Affiliated Hospital of Kunming University of Science and Technology, Kunming, China
- Department of Ophthalmology, The First People’s Hospital of Yunnan Province, Kunming, China
- Medical School, Kunming University of Science and Technology, Kunming, China
| | - Wen Zhang
- Medical School, Kunming University of Science and Technology, Kunming, China
| | - Zhizhou Shi
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming, China
| | - Zhongjian Liu
- Institute of Basic and Clinical Medicine, The First People’s Hospital of Yunnan Province, Kunming, China
| | - Yalian Sa
- Institute of Basic and Clinical Medicine, The First People’s Hospital of Yunnan Province, Kunming, China
| | - Li Li
- Institute of Basic and Clinical Medicine, The First People’s Hospital of Yunnan Province, Kunming, China
| | - Ninghua Ni
- Department of Ophthalmology, The Affiliated Hospital of Kunming University of Science and Technology, Kunming, China
- Department of Ophthalmology, The First People’s Hospital of Yunnan Province, Kunming, China
- Medical School, Kunming University of Science and Technology, Kunming, China
| | - Yan Mei
- Department of Ophthalmology, The Affiliated Hospital of Kunming University of Science and Technology, Kunming, China
- Department of Ophthalmology, The First People’s Hospital of Yunnan Province, Kunming, China
- Medical School, Kunming University of Science and Technology, Kunming, China
| |
Collapse
|
16
|
Zhang T, Zhang X, Wu Z, Ren J, Zhao Z, Zhang H, Wang G, Wang T. VGAE-CCI: variational graph autoencoder-based construction of 3D spatial cell-cell communication network. Brief Bioinform 2024; 26:bbae619. [PMID: 39581873 PMCID: PMC11586124 DOI: 10.1093/bib/bbae619] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2024] [Revised: 10/04/2024] [Accepted: 11/12/2024] [Indexed: 11/26/2024] Open
Abstract
Cell-cell communication plays a critical role in maintaining normal biological functions, regulating development and differentiation, and controlling immune responses. The rapid development of single-cell RNA sequencing and spatial transcriptomics sequencing (ST-seq) technologies provides essential data support for in-depth and comprehensive analysis of cell-cell communication. However, ST-seq data often contain incomplete data and systematic biases, which may reduce the accuracy and reliability of predicting cell-cell communication. Furthermore, other methods for analyzing cell-cell communication mainly focus on individual tissue sections, neglecting cell-cell communication across multiple tissue layers, and fail to comprehensively elucidate cell-cell communication networks within three-dimensional tissues. To address the aforementioned issues, we propose VGAE-CCI, a deep learning framework based on the Variational Graph Autoencoder, capable of identifying cell-cell communication across multiple tissue layers. Additionally, this model can be applied to spatial transcriptomics data with missing or partially incomplete data and can clustered cells at single-cell resolution based on spatial encoding information within complex tissues, thereby enabling more accurate inference of cell-cell communication. Finally, we tested our method on six datasets and compared it with other state of art methods for predicting cell-cell communication. Our method outperformed other methods across multiple metrics, demonstrating its efficiency and reliability in predicting cell-cell communication.
Collapse
Affiliation(s)
- Tianjiao Zhang
- College of Computer and Control Engineering, Northeast Forestry University, Harbin 150040, China
| | - Xiang Zhang
- College of Computer and Control Engineering, Northeast Forestry University, Harbin 150040, China
| | - Zhenao Wu
- College of Computer and Control Engineering, Northeast Forestry University, Harbin 150040, China
| | - Jixiang Ren
- College of Computer and Control Engineering, Northeast Forestry University, Harbin 150040, China
| | - Zhongqian Zhao
- College of Computer and Control Engineering, Northeast Forestry University, Harbin 150040, China
| | - Hongfei Zhang
- College of Computer and Control Engineering, Northeast Forestry University, Harbin 150040, China
| | - Guohua Wang
- College of Computer and Control Engineering, Northeast Forestry University, Harbin 150040, China
- Faculty of Computing, Harbin Institute of Technology, Harbin 150001, China
| | - Tao Wang
- School of Computer Science, Northwestern Polytechnical University, Xi'an 710072, China
| |
Collapse
|
17
|
Tasca P, van den Berg BM, Rabelink TJ, Wang G, Heijs B, van Kooten C, de Vries APJ, Kers J. Application of spatial-omics to the classification of kidney biopsy samples in transplantation. Nat Rev Nephrol 2024; 20:755-766. [PMID: 38965417 DOI: 10.1038/s41581-024-00861-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/06/2024] [Indexed: 07/06/2024]
Abstract
Improvement of long-term outcomes through targeted treatment is a primary concern in kidney transplant medicine. Currently, the validation of a rejection diagnosis and subsequent treatment depends on the histological assessment of allograft biopsy samples, according to the Banff classification system. However, the lack of (early) disease-specific tissue markers hinders accurate diagnosis and thus timely intervention. This challenge mainly results from an incomplete understanding of the pathophysiological processes underlying late allograft failure. Integration of large-scale multimodal approaches for investigating allograft biopsy samples might offer new insights into this pathophysiology, which are necessary for the identification of novel therapeutic targets and the development of tailored immunotherapeutic interventions. Several omics technologies - including transcriptomic, proteomic, lipidomic and metabolomic tools (and multimodal data analysis strategies) - can be applied to allograft biopsy investigation. However, despite their successful application in research settings and their potential clinical value, several barriers limit the broad implementation of many of these tools into clinical practice. Among spatial-omics technologies, mass spectrometry imaging, which is under-represented in the transplant field, has the potential to enable multi-omics investigations that might expand the insights gained with current clinical analysis technologies.
Collapse
Affiliation(s)
- Paola Tasca
- Center for Proteomics and Metabolomics, Leiden University Medical Center, Leiden, the Netherlands
- Leiden Transplant Center, Leiden University Medical Center, Leiden, the Netherlands
- Department of Pathology, Leiden University Medical Center, Leiden, the Netherlands
| | - Bernard M van den Berg
- Department of Internal Medicine, Division of Nephrology, Einthoven Laboratory of Vascular and Regenerative Medicine, Leiden University Medical Center, Leiden, the Netherlands
| | - Ton J Rabelink
- Department of Internal Medicine, Division of Nephrology, Einthoven Laboratory of Vascular and Regenerative Medicine, Leiden University Medical Center, Leiden, the Netherlands
- The Novo Nordisk Foundation Center for Stem Cell Medicine (Renew), Leiden University Medical Center, Leiden, the Netherlands
| | - Gangqi Wang
- Department of Internal Medicine, Division of Nephrology, Einthoven Laboratory of Vascular and Regenerative Medicine, Leiden University Medical Center, Leiden, the Netherlands
- The Novo Nordisk Foundation Center for Stem Cell Medicine (Renew), Leiden University Medical Center, Leiden, the Netherlands
| | - Bram Heijs
- Center for Proteomics and Metabolomics, Leiden University Medical Center, Leiden, the Netherlands
- Bruker Daltonics GmbH & Co. KG, Bremen, Germany
| | - Cees van Kooten
- Leiden Transplant Center, Leiden University Medical Center, Leiden, the Netherlands
- Department of Internal Medicine, Division of Nephrology, Einthoven Laboratory of Vascular and Regenerative Medicine, Leiden University Medical Center, Leiden, the Netherlands
| | - Aiko P J de Vries
- Leiden Transplant Center, Leiden University Medical Center, Leiden, the Netherlands.
- Department of Internal Medicine, Division of Nephrology, Einthoven Laboratory of Vascular and Regenerative Medicine, Leiden University Medical Center, Leiden, the Netherlands.
| | - Jesper Kers
- Leiden Transplant Center, Leiden University Medical Center, Leiden, the Netherlands
- Department of Pathology, Leiden University Medical Center, Leiden, the Netherlands
- Department of Pathology, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, the Netherlands
- Center for Analytical Sciences Amsterdam, Van't Hoff Institute for Molecular Sciences, University of Amsterdam, Amsterdam, the Netherlands
| |
Collapse
|
18
|
Pala F, Notarangelo LD, Bosticardo M. Rediscovering the human thymus through cutting-edge technologies. J Exp Med 2024; 221:e20230892. [PMID: 39167072 PMCID: PMC11338284 DOI: 10.1084/jem.20230892] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2024] [Revised: 06/24/2024] [Accepted: 07/30/2024] [Indexed: 08/23/2024] Open
Abstract
Recent technological advances have transformed our understanding of the human thymus. Innovations such as high-resolution imaging, single-cell omics, and organoid cultures, including thymic epithelial cell (TEC) differentiation and culture, and improvements in biomaterials, have further elucidated the thymus architecture, cellular dynamics, and molecular mechanisms underlying T cell development, and have unraveled previously unrecognized levels of stromal cell heterogeneity. These advancements offer unprecedented insights into thymic biology and hold promise for the development of novel therapeutic strategies for immune-related disorders.
Collapse
Affiliation(s)
- Francesca Pala
- Immune Deficiency Genetics Section, Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health , Bethesda, MD, USA
| | - Luigi D Notarangelo
- Immune Deficiency Genetics Section, Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health , Bethesda, MD, USA
| | - Marita Bosticardo
- Immune Deficiency Genetics Section, Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health , Bethesda, MD, USA
| |
Collapse
|
19
|
Hu S, Tang Y, Li X, Li W, Zeng Y, Jiang M, Chen R, Zheng P, Yang L, Song Z, Xie D, Chen Y, Yuan Y. Hsp90aa1/JUN/Ccl2 regulatory axis mediates migration and differentiation of NSPCs, promoting the onset and progression of early post-ischemic stroke epilepsy. Neurobiol Dis 2024; 200:106635. [PMID: 39128813 DOI: 10.1016/j.nbd.2024.106635] [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: 01/27/2024] [Revised: 08/07/2024] [Accepted: 08/08/2024] [Indexed: 08/13/2024] Open
Abstract
Early-onset epilepsy following ischemic stroke is a severe neurological condition, the pathogenesis of which remains incompletely understood. Recent studies suggest that Neural stem/progenitor cells (NSPCs) play a crucial role in the disease process, yet the precise molecular mechanisms regulating NSPCs have not been thoroughly investigated. This study utilized single-cell transcriptome sequencing and bioinformatics analysis to identify disease-related genes, which were subsequently validated in both in vitro and in vivo experiments. The findings revealed that Hsp90aa1 (heat shock protein 90 kDa alpha, class A member 1), Jun proto-oncogene (JUN), and CC Motif Ligation 2 (Ccl2) constitute an important regulatory axis influencing the migration and differentiation of NSPCs, potentially impacting the onset and progression of early-onset epilepsy post-ischemic stroke. Additionally, the expression of Hsp90aa1 was found to influence the likelihood of seizure occurrence and the severity of brain ischemia.
Collapse
Affiliation(s)
- Shuntong Hu
- Department of Neurology, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Yongzhong Tang
- Department of Anesthesiology, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Xiaobo Li
- Department of Neurology, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Wenjun Li
- Department of Neurology, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Yini Zeng
- Department of Neurology, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Mi Jiang
- Department of Neurology, The Third Xiangya Hospital, Central South University, Changsha, China; Department of Neurology, University of Michigan, Ann Arbor, MI, USA
| | - Ru Chen
- Department of Neurology, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Ping Zheng
- Department of Neurosurgery, Shanghai Pudong New Area People's Hospital, Shanghai, China
| | - Liang Yang
- Department of Neurosurgery, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Zhi Song
- Department of Neurology, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Dujie Xie
- Department of Neurology, The Third Xiangya Hospital, Central South University, Changsha, China.
| | - Yiwei Chen
- Department of Neurology, The Third Xiangya Hospital, Central South University, Changsha, China.
| | - Yi Yuan
- Department of Neurology, The Third Xiangya Hospital, Central South University, Changsha, China.
| |
Collapse
|
20
|
Wang N, Hong W, Wu Y, Chen Z, Bai M, Wang W, Zhu J. Next-generation spatial transcriptomics: unleashing the power to gear up translational oncology. MedComm (Beijing) 2024; 5:e765. [PMID: 39376738 PMCID: PMC11456678 DOI: 10.1002/mco2.765] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2024] [Revised: 08/30/2024] [Accepted: 09/03/2024] [Indexed: 10/09/2024] Open
Abstract
The growing advances in spatial transcriptomics (ST) stand as the new frontier bringing unprecedented influences in the realm of translational oncology. This has triggered systemic experimental design, analytical scope, and depth alongside with thorough bioinformatics approaches being constantly developed in the last few years. However, harnessing the power of spatial biology and streamlining an array of ST tools to achieve designated research goals are fundamental and require real-world experiences. We present a systemic review by updating the technical scope of ST across different principal basis in a timeline manner hinting on the generally adopted ST techniques used within the community. We also review the current progress of bioinformatic tools and propose in a pipelined workflow with a toolbox available for ST data exploration. With particular interests in tumor microenvironment where ST is being broadly utilized, we summarize the up-to-date progress made via ST-based technologies by narrating studies categorized into either mechanistic elucidation or biomarker profiling (translational oncology) across multiple cancer types and their ways of deploying the research through ST. This updated review offers as a guidance with forward-looking viewpoints endorsed by many high-resolution ST tools being utilized to disentangle biological questions that may lead to clinical significance in the future.
Collapse
Affiliation(s)
- Nan Wang
- Cosmos Wisdom Biotech Co. LtdHangzhouChina
| | - Weifeng Hong
- Department of Radiation OncologyZhejiang Cancer HospitalHangzhouChina
- Hangzhou Institute of Medicine (HIM)Chinese Academy of SciencesHangzhouChina
- Zhejiang Key Laboratory of Radiation OncologyHangzhouChina
| | - Yixing Wu
- Department of Pulmonary and Critical Care MedicineZhongshan HospitalFudan UniversityShanghaiChina
| | - Zhe‐Sheng Chen
- Department of Pharmaceutical SciencesCollege of Pharmacy and Health SciencesInstitute for BiotechnologySt. John's UniversityQueensNew YorkUSA
| | - Minghua Bai
- Department of Radiation OncologyZhejiang Cancer HospitalHangzhouChina
- Hangzhou Institute of Medicine (HIM)Chinese Academy of SciencesHangzhouChina
- Zhejiang Key Laboratory of Radiation OncologyHangzhouChina
| | | | - Ji Zhu
- Department of Radiation OncologyZhejiang Cancer HospitalHangzhouChina
- Hangzhou Institute of Medicine (HIM)Chinese Academy of SciencesHangzhouChina
- Zhejiang Key Laboratory of Radiation OncologyHangzhouChina
| |
Collapse
|
21
|
Bhat AA, Afzal M, Moglad E, Thapa R, Ali H, Almalki WH, Kazmi I, Alzarea SI, Gupta G, Subramaniyan V. lncRNAs as prognostic markers and therapeutic targets in cuproptosis-mediated cancer. Clin Exp Med 2024; 24:226. [PMID: 39325172 PMCID: PMC11427524 DOI: 10.1007/s10238-024-01491-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2024] [Accepted: 09/16/2024] [Indexed: 09/27/2024]
Abstract
Long non-coding RNAs (lncRNAs) have emerged as crucial regulators in various cellular processes, including cancer progression and stress response. Recent studies have demonstrated that copper accumulation induces a unique form of cell death known as cuproptosis, with lncRNAs playing a key role in regulating cuproptosis-associated pathways. These lncRNAs may trigger cell-specific responses to copper stress, presenting new opportunities as prognostic markers and therapeutic targets. This paper delves into the role of lncRNAs in cuproptosis-mediated cancer, underscoring their potential as biomarkers and targets for innovative therapeutic strategies. A thorough review of scientific literature was conducted, utilizing databases such as PubMed, Google Scholar, and ScienceDirect, with search terms like 'lncRNAs,' 'cuproptosis,' and 'cancer.' Studies were selected based on their relevance to lncRNA regulation of cuproptosis pathways and their implications for cancer prognosis and treatment. The review highlights the significant contribution of lncRNAs in regulating cuproptosis-related genes and pathways, impacting copper metabolism, mitochondrial stress responses, and apoptotic signaling. Specific lncRNAs are potential prognostic markers in breast, lung, liver, ovarian, pancreatic, and gastric cancers. The objective of this article is to explore the role of lncRNAs as potential prognostic markers and therapeutic targets in cancers mediated by cuproptosis.
Collapse
Affiliation(s)
- Asif Ahmad Bhat
- Uttaranchal Institute of Pharmaceutical Sciences, Uttaranchal University, Dehradun, India
| | - Muhammad Afzal
- Department of Pharmaceutical Sciences, Pharmacy Program, Batterjee Medical College, P.O. Box 6231, 21442, Jeddah, Saudi Arabia
| | - Ehssan Moglad
- Department of Pharmaceutics, College of Pharmacy, Prince Sattam Bin Abdulaziz University, 11942, Al Kharj, Saudi Arabia
| | - Riya Thapa
- Uttaranchal Institute of Pharmaceutical Sciences, Uttaranchal University, Dehradun, India
| | - Haider Ali
- Centre for Global Health Research, Saveetha Medical College, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, India
- Department of Pharmacology, Kyrgyz State Medical College, Bishkek, Kyrgyzstan
| | - Waleed Hassan Almalki
- Department of Pharmacology, College of Pharmacy, Umm Al-Qura University, Makkah, Saudi Arabia
| | - Imran Kazmi
- Department of Biochemistry, Faculty of Science, King Abdulaziz University, 21589, Jeddah, Saudi Arabia
| | - Sami I Alzarea
- Department of Pharmacology, College of Pharmacy, Jouf University, 72341, Sakaka, Aljouf, Saudi Arabia
| | - Gaurav Gupta
- Centre for Research Impact & Outcome, Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, 140401, India
- Centre of Medical and Bio-Allied Health Sciences Research, Ajman University, Ajman, United Arab Emirates
| | - Vetriselvan Subramaniyan
- Pharmacology Unit, Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Jalan Lagoon Selatan, 47500, Bandar Sunway, Selangor Darul Ehsan, Malaysia.
- Department of Medical Sciences, School of Medical and Life Sciences, Sunway University, Bandar Sunway, 47500, Subang Jaya, Selangor, Malaysia.
| |
Collapse
|
22
|
Liu X, Ren X. Computational Strategies and Algorithms for Inferring Cellular Composition of Spatial Transcriptomics Data. GENOMICS, PROTEOMICS & BIOINFORMATICS 2024; 22:qzae057. [PMID: 39110523 PMCID: PMC11398939 DOI: 10.1093/gpbjnl/qzae057] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Revised: 07/26/2024] [Accepted: 08/02/2024] [Indexed: 09/15/2024]
Abstract
Spatial transcriptomics technology has been an essential and powerful method for delineating tissue architecture at the molecular level. However, due to the limitations of the current spatial techniques, the cellular information cannot be directly measured but instead spatial spots typically varying from a diameter of 0.2 to 100 µm are characterized. Therefore, it is vital to apply computational strategies for inferring the cellular composition within each spatial spot. The main objective of this review is to summarize the most recent progresses in estimating the exact cellular proportions for each spatial spot, and to prospect the future directions of this field.
Collapse
|
23
|
Unger Avila P, Padvitski T, Leote AC, Chen H, Saez-Rodriguez J, Kann M, Beyer A. Gene regulatory networks in disease and ageing. Nat Rev Nephrol 2024; 20:616-633. [PMID: 38867109 DOI: 10.1038/s41581-024-00849-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/15/2024] [Indexed: 06/14/2024]
Abstract
The precise control of gene expression is required for the maintenance of cellular homeostasis and proper cellular function, and the declining control of gene expression with age is considered a major contributor to age-associated changes in cellular physiology and disease. The coordination of gene expression can be represented through models of the molecular interactions that govern gene expression levels, so-called gene regulatory networks. Gene regulatory networks can represent interactions that occur through signal transduction, those that involve regulatory transcription factors, or statistical models of gene-gene relationships based on the premise that certain sets of genes tend to be coexpressed across a range of conditions and cell types. Advances in experimental and computational technologies have enabled the inference of these networks on an unprecedented scale and at unprecedented precision. Here, we delineate different types of gene regulatory networks and their cell-biological interpretation. We describe methods for inferring such networks from large-scale, multi-omics datasets and present applications that have aided our understanding of cellular ageing and disease mechanisms.
Collapse
Affiliation(s)
- Paula Unger Avila
- Cluster of Excellence on Cellular Stress Responses in Aging-associated Diseases (CECAD), University of Cologne, Cologne, Germany
| | - Tsimafei Padvitski
- Cluster of Excellence on Cellular Stress Responses in Aging-associated Diseases (CECAD), University of Cologne, Cologne, Germany
| | - Ana Carolina Leote
- Cluster of Excellence on Cellular Stress Responses in Aging-associated Diseases (CECAD), University of Cologne, Cologne, Germany
| | - He Chen
- Cluster of Excellence on Cellular Stress Responses in Aging-associated Diseases (CECAD), University of Cologne, Cologne, Germany
- Department II of Internal Medicine, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Julio Saez-Rodriguez
- Faculty of Medicine and Heidelberg University Hospital, Institute for Computational Biomedicine, Heidelberg University, Heidelberg, Germany
| | - Martin Kann
- Department II of Internal Medicine, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
- Center for Molecular Medicine Cologne, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Andreas Beyer
- Cluster of Excellence on Cellular Stress Responses in Aging-associated Diseases (CECAD), University of Cologne, Cologne, Germany.
- Center for Molecular Medicine Cologne, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany.
- Institute for Genetics, Faculty of Mathematics and Natural Sciences, University of Cologne, Cologne, Germany.
| |
Collapse
|
24
|
Song Y, Yang Z, Gao N, Zhang B. MICAL1 promotes the proliferation in acute myeloid leukemia and is associated with clinical prognosis and immune infiltration. Discov Oncol 2024; 15:279. [PMID: 38995414 PMCID: PMC11245461 DOI: 10.1007/s12672-024-01150-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Accepted: 07/08/2024] [Indexed: 07/13/2024] Open
Abstract
Acute myeloid leukemia (AML) is one of the most common hematopoietic malignancies that has a poor prognosis and a high rate of relapse. Dysregulated metabolism plays an important role in AML progression. This study aimed to conduct a comprehensive analysis of MRGs using TCGA and GEO datasets and further explore the potential function of critical MRGs in AML progression. In this study, we identified 17 survival-related differentially expressed MRGs in AML using TCGA and GEO datasets. The 150 AML samples were divided into three molecular subtypes using 17 MRGs, and we found that three molecular subtypes exhibited a different association with ferroptosis, cuproptosis and m6A related genes. Moreover, a prognostic signature that comprised nine MRGs and had good predictive capacity was established by LASSO-Cox stepwise regression analysis. Among the 17 MRGs, our attention focused on MICAL1 which was highly expressed in many types of tumors, including AML and its overexpression was also confirmed in several AML cell lines. We also found that the expression of MICAL1 was associated with several immune cells. Moreover, functional experiments revealed that knockdown of MICAL1 distinctly suppressed the proliferation of AML cells. Overall, this study not only contributes to a deeper understanding of the molecular mechanisms underlying AML but also provides potential targets and prognostic markers for AML treatment. These findings offer robust support for further research into therapeutic strategies and mechanisms related to AML, with the potential to improve the prognosis and quality of life for AML patients. Nevertheless, further research is needed to validate these findings and explore more in-depth molecular mechanisms.
Collapse
Affiliation(s)
- Yinsen Song
- Translational Medicine Research Center (Key Laboratory of Organ Transplantation of Henan Province), The Fifth Clinical Medical College of Henan University of Chinese Medicine (Zhengzhou People's Hospital), Zhengzhou, China
| | - Zhenzhen Yang
- Translational Medicine Research Center (Key Laboratory of Organ Transplantation of Henan Province), The Fifth Clinical Medical College of Henan University of Chinese Medicine (Zhengzhou People's Hospital), Zhengzhou, China
| | - Na Gao
- Translational Medicine Research Center (Key Laboratory of Organ Transplantation of Henan Province), The Fifth Clinical Medical College of Henan University of Chinese Medicine (Zhengzhou People's Hospital), Zhengzhou, China
| | - Bojun Zhang
- Department of Pathogenic Microbiology and Immunology, School of Basic Medical Sciences, Xi'an Jiaotong University, No.76 Yanta West Road, Xi'an, China.
| |
Collapse
|
25
|
Razzouk S. Single-cell sequencing, spatial transcriptome ad periodontitis: Rethink pathogenesis and classification. Oral Dis 2024; 30:2771-2783. [PMID: 37794757 DOI: 10.1111/odi.14761] [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: 05/21/2023] [Revised: 08/02/2023] [Accepted: 09/21/2023] [Indexed: 10/06/2023]
Abstract
OBJECTIVE This narrative review illuminates on the application of single-cell RNA sequencing (scRNA-seq) and spatial transcriptomics (ST) in periodontitis and highlights the probability of relating cell population and gene signatures to the pathogenesis of the disease for a better diagnosis. METHODS An electronic search of the literature in the PubMed database for the keywords, "single cell sequencing" OR "spatial transcriptomics" and "periodontitis" OR "gingiva" OR "oral mucosa" yielded 486 research articles and reviews. After filtering duplicates and careful curation, 22 papers conducted in humans were retained. RESULTS The molecular mechanisms underlying periodontitis are complex and involve the interaction of multiple cells and various gene expressions. Most residing cells in periodontal tissues participate in maintaining homeostasis and health, while in addition to infiltrating immune cells contribute to the fight against the bacterial insult. CONCLUSION scRNA-seq and ST have provided new insights into the cellular and molecular changes associated with periodontitis for a better diagnosis and clinical outcome. New functions of cells and genes are revealed with these techniques; however, no cells or gene signatures are attributed to periodontitis so far.
Collapse
Affiliation(s)
- Sleiman Razzouk
- Department of Periodontology and Implant Dentistry, New York University College of Dentistry, New York, New York, USA
- Private Practice, Beirut, Lebanon
| |
Collapse
|
26
|
Chen R, Nie P, Wang J, Wang GZ. Deciphering brain cellular and behavioral mechanisms: Insights from single-cell and spatial RNA sequencing. WILEY INTERDISCIPLINARY REVIEWS. RNA 2024; 15:e1865. [PMID: 38972934 DOI: 10.1002/wrna.1865] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Revised: 05/05/2024] [Accepted: 05/14/2024] [Indexed: 07/09/2024]
Abstract
The brain is a complex computing system composed of a multitude of interacting neurons. The computational outputs of this system determine the behavior and perception of every individual. Each brain cell expresses thousands of genes that dictate the cell's function and physiological properties. Therefore, deciphering the molecular expression of each cell is of great significance for understanding its characteristics and role in brain function. Additionally, the positional information of each cell can provide crucial insights into their involvement in local brain circuits. In this review, we briefly overview the principles of single-cell RNA sequencing and spatial transcriptomics, the potential issues and challenges in their data processing, and their applications in brain research. We further outline several promising directions in neuroscience that could be integrated with single-cell RNA sequencing, including neurodevelopment, the identification of novel brain microstructures, cognition and behavior, neuronal cell positioning, molecules and cells related to advanced brain functions, sleep-wake cycles/circadian rhythms, and computational modeling of brain function. We believe that the deep integration of these directions with single-cell and spatial RNA sequencing can contribute significantly to understanding the roles of individual cells or cell types in these specific functions, thereby making important contributions to addressing critical questions in those fields. This article is categorized under: RNA Evolution and Genomics > Computational Analyses of RNA RNA in Disease and Development > RNA in Development RNA in Disease and Development > RNA in Disease.
Collapse
Affiliation(s)
- Renrui Chen
- CAS Key Laboratory of Computational Biology, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Pengxing Nie
- CAS Key Laboratory of Computational Biology, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Jing Wang
- CAS Key Laboratory of Computational Biology, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Guang-Zhong Wang
- CAS Key Laboratory of Computational Biology, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| |
Collapse
|
27
|
Atta L, Clifton K, Anant M, Aihara G, Fan J. Gene count normalization in single-cell imaging-based spatially resolved transcriptomics. Genome Biol 2024; 25:153. [PMID: 38867267 PMCID: PMC11167774 DOI: 10.1186/s13059-024-03303-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2023] [Accepted: 06/06/2024] [Indexed: 06/14/2024] Open
Abstract
BACKGROUND Recent advances in imaging-based spatially resolved transcriptomics (im-SRT) technologies now enable high-throughput profiling of targeted genes and their locations in fixed tissues. Normalization of gene expression data is often needed to account for technical factors that may confound underlying biological signals. RESULTS Here, we investigate the potential impact of different gene count normalization methods with different targeted gene panels in the analysis and interpretation of im-SRT data. Using different simulated gene panels that overrepresent genes expressed in specific tissue regions or cell types, we demonstrate how normalization methods based on detected gene counts per cell differentially impact normalized gene expression magnitudes in a region- or cell type-specific manner. We show that these normalization-induced effects may reduce the reliability of downstream analyses including differential gene expression, gene fold change, and spatially variable gene analysis, introducing false positive and false negative results when compared to results obtained from gene panels that are more representative of the gene expression of the tissue's component cell types. These effects are not observed with normalization approaches that do not use detected gene counts for gene expression magnitude adjustment, such as with cell volume or cell area normalization. CONCLUSIONS We recommend using non-gene count-based normalization approaches when feasible and evaluating gene panel representativeness before using gene count-based normalization methods if necessary. Overall, we caution that the choice of normalization method and gene panel may impact the biological interpretation of the im-SRT data.
Collapse
Affiliation(s)
- Lyla Atta
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, 21218, USA
- Center for Computational Biology, Whiting School of Engineering, Johns Hopkins University, Baltimore, MD, 21211, USA
| | - Kalen Clifton
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, 21218, USA
- Center for Computational Biology, Whiting School of Engineering, Johns Hopkins University, Baltimore, MD, 21211, USA
| | - Manjari Anant
- Center for Computational Biology, Whiting School of Engineering, Johns Hopkins University, Baltimore, MD, 21211, USA
- Department of Neuroscience, Johns Hopkins University, Baltimore, MD, 21218, USA
| | - Gohta Aihara
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, 21218, USA
- Center for Computational Biology, Whiting School of Engineering, Johns Hopkins University, Baltimore, MD, 21211, USA
| | - Jean Fan
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, 21218, USA.
- Center for Computational Biology, Whiting School of Engineering, Johns Hopkins University, Baltimore, MD, 21211, USA.
| |
Collapse
|
28
|
Xu H, Lu X, Yu Y, Zhou Y, Qi T, Zheng Y. Elucidating the molecular landscape of tendinitis: the role of inflammasome-related genes and immune interactions. Front Immunol 2024; 15:1393851. [PMID: 38919626 PMCID: PMC11196777 DOI: 10.3389/fimmu.2024.1393851] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Accepted: 05/28/2024] [Indexed: 06/27/2024] Open
Abstract
Tendinitis, characterized by the inflammation of tendons, poses significant challenges in both diagnosis and treatment due to its multifaceted etiology and complex pathophysiology. This study aimed to dissect the molecular mechanisms underlying tendinitis, with a particular focus on inflammasome-related genes and their interactions with the immune system. Through comprehensive gene expression analysis and bioinformatics approaches, we identified distinct expression profiles of inflammasome genes, such as NLRP6, NLRP1, and MEFV, which showed significant correlations with immune checkpoint molecules, indicating a pivotal role in the inflammatory cascade of tendinitis. Additionally, MYD88 and CD36 were found to be closely associated with HLA family molecules, underscoring their involvement in immune response modulation. Contrary to expectations, chemokines exhibited minimal correlation with inflammasome genes, suggesting an unconventional inflammatory pathway in tendinitis. Transcription factors like SP110 and CREB5 emerged as key regulators of inflammasome genes, providing insight into the transcriptional control mechanisms in tendinitis. Furthermore, potential therapeutic targets were identified through the DGidb database, highlighting drugs that could modulate the activity of inflammasome genes, offering new avenues for targeted tendinitis therapy. Our findings elucidate the complex molecular landscape of tendinitis, emphasizing the significant role of inflammasomes and immune interactions, and pave the way for the development of novel diagnostic and therapeutic strategies.
Collapse
Affiliation(s)
- Hongwei Xu
- The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, China
| | - Xiaolang Lu
- The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, China
| | - Yang Yu
- The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, China
| | - Yifei Zhou
- The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, China
| | - Tengfei Qi
- Department of Neurosurgery, Trauma Center, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Yijing Zheng
- The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
29
|
Totty M, Hicks SC, Guo B. SpotSweeper: spatially-aware quality control for spatial transcriptomics. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.06.597765. [PMID: 38895212 PMCID: PMC11185656 DOI: 10.1101/2024.06.06.597765] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/21/2024]
Abstract
Quality control (QC) is a crucial step to ensure the reliability and accuracy of the data obtained from RNA sequencing experiments, including spatially-resolved transcriptomics (SRT). Existing QC approaches for SRT that have been adopted from single-nucleus RNA sequencing (snRNA-seq) methods are confounded by spatial biology and are inappropriate for SRT data. In addition, no methods currently exist for identifying histological tissue artifacts unique to SRT. Here, we introduce SpotSweeper, spatially-aware QC methods for identifying local outliers and regional artifacts in SRT. SpotSweeper evaluates the quality of individual spots relative to their local neighborhood, thus minimizing bias due to biological heterogeneity, and uses multiscale methods to detect regional artifacts. Using SpotSweeper on publicly available data, we identified a consistent set of Visium barcodes/spots as systematically low quality and demonstrate that SpotSweeper accurately identifies two distinct types of regional artifacts, resulting in improved downstream clustering and marker gene detection for spatial domains.
Collapse
Affiliation(s)
- Michael Totty
- Department of Biostatistics, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Stephanie C. Hicks
- Department of Biostatistics, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, USA
- Center for Computational Biology, Johns Hopkins University, Baltimore, MD, USA
- Malone Center for Engineering in Healthcare, Johns Hopkins University, Baltimore, MD, USA
| | - Boyi Guo
- Department of Biostatistics, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| |
Collapse
|
30
|
Cilento MA, Sweeney CJ, Butler LM. Spatial transcriptomics in cancer research and potential clinical impact: a narrative review. J Cancer Res Clin Oncol 2024; 150:296. [PMID: 38850363 PMCID: PMC11162383 DOI: 10.1007/s00432-024-05816-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Accepted: 05/22/2024] [Indexed: 06/10/2024]
Abstract
Spatial transcriptomics (ST) provides novel insights into the tumor microenvironment (TME). ST allows the quantification and illustration of gene expression profiles in the spatial context of tissues, including both the cancer cells and the microenvironment in which they are found. In cancer research, ST has already provided novel insights into cancer metastasis, prognosis, and immunotherapy responsiveness. The clinical precision oncology application of next-generation sequencing (NGS) and RNA profiling of tumors relies on bulk methods that lack spatial context. The ability to preserve spatial information is now possible, as it allows us to capture tumor heterogeneity and multifocality. In this narrative review, we summarize precision oncology, discuss tumor sequencing in the clinic, and review the available ST research methods, including seqFISH, MERFISH (Vizgen), CosMx SMI (NanoString), Xenium (10x), Visium (10x), Stereo-seq (STOmics), and GeoMx DSP (NanoString). We then review the current ST literature with a focus on solid tumors organized by tumor type. Finally, we conclude by addressing an important question: how will spatial transcriptomics ultimately help patients with cancer?
Collapse
Affiliation(s)
- Michael A Cilento
- South Australian Immunogenomics Cancer Institute, The University of Adelaide, Adelaide, SA, Australia.
- South Australian Health and Medical Research Institute, Adelaide, SA, Australia.
- The Queen Elizabeth Hospital, Woodville South, SA, Australia.
| | - Christopher J Sweeney
- South Australian Immunogenomics Cancer Institute, The University of Adelaide, Adelaide, SA, Australia
| | - Lisa M Butler
- South Australian Immunogenomics Cancer Institute, The University of Adelaide, Adelaide, SA, Australia
- South Australian Health and Medical Research Institute, Adelaide, SA, Australia
| |
Collapse
|
31
|
Chen X, Lin J, Wang Y, Zhang W, Xie W, Zheng Z, Wong KC. HE2Gene: image-to-RNA translation via multi-task learning for spatial transcriptomics data. Bioinformatics 2024; 40:btae343. [PMID: 38837395 PMCID: PMC11164830 DOI: 10.1093/bioinformatics/btae343] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2023] [Revised: 05/06/2024] [Accepted: 05/25/2024] [Indexed: 06/07/2024] Open
Abstract
MOTIVATION Tissue context and molecular profiling are commonly used measures in understanding normal development and disease pathology. In recent years, the development of spatial molecular profiling technologies (e.g. spatial resolved transcriptomics) has enabled the exploration of quantitative links between tissue morphology and gene expression. However, these technologies remain expensive and time-consuming, with subsequent analyses necessitating high-throughput pathological annotations. On the other hand, existing computational tools are limited to predicting only a few dozen to several hundred genes, and the majority of the methods are designed for bulk RNA-seq. RESULTS In this context, we propose HE2Gene, the first multi-task learning-based method capable of predicting tens of thousands of spot-level gene expressions along with pathological annotations from H&E-stained images. Experimental results demonstrate that HE2Gene is comparable to state-of-the-art methods and generalizes well on an external dataset without the need for re-training. Moreover, HE2Gene preserves the annotated spatial domains and has the potential to identify biomarkers. This capability facilitates cancer diagnosis and broadens its applicability to investigate gene-disease associations. AVAILABILITY AND IMPLEMENTATION The source code and data information has been deposited at https://github.com/Microbiods/HE2Gene.
Collapse
Affiliation(s)
- Xingjian Chen
- Cutaneous Biology Research Center, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02129, USA
- Department of Computer Science, City University of Hong Kong, Kowloog Tong 999077, Hong Kong SAR
| | - Jiecong Lin
- Molecular Pathology Unit, Center for Cancer Research, Massachusetts General Hospital, Department of Pathology, Harvard Medical School, Boston, MA 02129, USA
- Department of Computer Science, The University of Hong Kong, Pokfulam 999077, Hong Kong SAR
| | - Yuchen Wang
- Department of Computer Science, City University of Hong Kong, Kowloog Tong 999077, Hong Kong SAR
| | - Weitong Zhang
- Department of Computer Science, City University of Hong Kong, Kowloog Tong 999077, Hong Kong SAR
| | - Weidun Xie
- Department of Computer Science, City University of Hong Kong, Kowloog Tong 999077, Hong Kong SAR
| | - Zetian Zheng
- Department of Computer Science, City University of Hong Kong, Kowloog Tong 999077, Hong Kong SAR
| | - Ka-Chun Wong
- Department of Computer Science, City University of Hong Kong, Kowloog Tong 999077, Hong Kong SAR
- Shenzhen Research Institute, City University of Hong Kong, Shenzhen 518057, China
| |
Collapse
|
32
|
Makino M, Shimizu K, Kadota K. Enhanced clustering-based differential expression analysis method for RNA-seq data. MethodsX 2024; 12:102518. [PMID: 38179066 PMCID: PMC10764243 DOI: 10.1016/j.mex.2023.102518] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Accepted: 12/10/2023] [Indexed: 01/06/2024] Open
Abstract
RNA-seq is a tool for measuring gene expression and is commonly used to identify differentially expressed genes (DEGs). Gene clustering has been widely used to classify DEGs with similar expression patterns, but rarely used to identify DEGs themselves. We recently reported that the clustering-based method (called MBCdeg1 and 2) for identifying DEGs has great potential. However, these methods left room for improvement. This study reports on the improvement (named MBCdeg3). We compared a total of six competing methods: three conventional R packages (edgeR, DESeq2, and TCC) and three versions of MBCdeg (i.e., MBCdeg1, 2, and 3) corresponding to three different normalization algorithms. As MBCdeg3 performs well in many simulation scenarios of RNA-seq count data, MBCdeg3 replaces MBCdeg1 and 2 in our previous report. •MBCdeg3 is a method for both identification and classification of DEGs from RNA-seq count data.•MBCdeg3 is available as a function of R, which is common in the field of expression analysis.•MBCdeg3 performs well in a variety of simulation scenarios for RNA-seq count data.
Collapse
Affiliation(s)
- Manon Makino
- Graduate School of Agricultural and Life Sciences, The University of Tokyo, Yayoi 1-1-1, Bunkyo-ku, Tokyo 113-8657, Japan
| | - Kentaro Shimizu
- Graduate School of Agricultural and Life Sciences, The University of Tokyo, Yayoi 1-1-1, Bunkyo-ku, Tokyo 113-8657, Japan
| | - Koji Kadota
- Graduate School of Agricultural and Life Sciences, The University of Tokyo, Yayoi 1-1-1, Bunkyo-ku, Tokyo 113-8657, Japan
- Interfaculty Initiative in Information Studies, The University of Tokyo, Hongo 7-3-1, Bunkyo-ku, Tokyo 113-0033, Japan
- Collaborative Research Institute for Innovative Microbiology, The University of Tokyo, Yayoi 1-1-1, Bunkyo-ku, Tokyo 113-8657, Japan
| |
Collapse
|
33
|
Swain AK, Pandit V, Sharma J, Yadav P. SpatialPrompt: spatially aware scalable and accurate tool for spot deconvolution and domain identification in spatial transcriptomics. Commun Biol 2024; 7:639. [PMID: 38796505 PMCID: PMC11127982 DOI: 10.1038/s42003-024-06349-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Accepted: 05/17/2024] [Indexed: 05/28/2024] Open
Abstract
Efficiently mapping of cell types in situ remains a major challenge in spatial transcriptomics. Most spot deconvolution tools ignore spatial coordinate information and perform extremely slow on large datasets. Here, we introduce SpatialPrompt, a spatially aware and scalable tool for spot deconvolution and domain identification. SpatialPrompt integrates gene expression, spatial location, and single-cell RNA sequencing (scRNA-seq) dataset as reference to accurately infer cell-type proportions of spatial spots. SpatialPrompt uses non-negative ridge regression and graph neural network to efficiently capture local microenvironment information. Our extensive benchmarking analysis on Visium, Slide-seq, and MERFISH datasets demonstrated superior performance of SpatialPrompt over 15 existing tools. On mouse hippocampus dataset, SpatialPrompt achieves spot deconvolution and domain identification within 2 minutes for 50,000 spots. Overall, domain identification using SpatialPrompt was 44 to 150 times faster than existing methods. We build a database housing 40 plus curated scRNA-seq datasets for seamless integration with SpatialPrompt for spot deconvolution.
Collapse
Affiliation(s)
- Asish Kumar Swain
- Department of Bioscience & Bioengineering, Indian Institute of Technology, Jodhpur, Rajasthan, 342030, India
| | - Vrushali Pandit
- Department of Bioscience & Bioengineering, Indian Institute of Technology, Jodhpur, Rajasthan, 342030, India
| | - Jyoti Sharma
- Department of Bioscience & Bioengineering, Indian Institute of Technology, Jodhpur, Rajasthan, 342030, India
| | - Pankaj Yadav
- Department of Bioscience & Bioengineering, Indian Institute of Technology, Jodhpur, Rajasthan, 342030, India.
- School of Artificial Intelligence and Data Science, Indian Institute of Technology, Jodhpur, Rajasthan, 342030, India.
| |
Collapse
|
34
|
Johnson AL, Lopez-Bertoni H. Cellular diversity through space and time: adding new dimensions to GBM therapeutic development. Front Genet 2024; 15:1356611. [PMID: 38774283 PMCID: PMC11106394 DOI: 10.3389/fgene.2024.1356611] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Accepted: 04/15/2024] [Indexed: 05/24/2024] Open
Abstract
The current median survival for glioblastoma (GBM) patients is only about 16 months, with many patients succumbing to the disease in just a matter of months, making it the most common and aggressive primary brain cancer in adults. This poor outcome is, in part, due to the lack of new treatment options with only one FDA-approved treatment in the last decade. Advances in sequencing techniques and transcriptomic analyses have revealed a vast degree of heterogeneity in GBM, from inter-patient diversity to intra-tumoral cellular variability. These cutting-edge approaches are providing new molecular insights highlighting a critical role for the tumor microenvironment (TME) as a driver of cellular plasticity and phenotypic heterogeneity. With this expanded molecular toolbox, the influence of TME factors, including endogenous (e.g., oxygen and nutrient availability and interactions with non-malignant cells) and iatrogenically induced (e.g., post-therapeutic intervention) stimuli, on tumor cell states can be explored to a greater depth. There exists a critical need for interrogating the temporal and spatial aspects of patient tumors at a high, cell-level resolution to identify therapeutically targetable states, interactions and mechanisms. In this review, we discuss advancements in our understanding of spatiotemporal diversity in GBM with an emphasis on the influence of hypoxia and immune cell interactions on tumor cell heterogeneity. Additionally, we describe specific high-resolution spatially resolved methodologies and their potential to expand the impact of pre-clinical GBM studies. Finally, we highlight clinical attempts at targeting hypoxia- and immune-related mechanisms of malignancy and the potential therapeutic opportunities afforded by single-cell and spatial exploration of GBM patient specimens.
Collapse
Affiliation(s)
- Amanda L. Johnson
- Hugo W. Moser Research Institute at Kennedy Krieger, Baltimore, MD, United States
- Department of Neurology, Baltimore, MD, United States
| | - Hernando Lopez-Bertoni
- Hugo W. Moser Research Institute at Kennedy Krieger, Baltimore, MD, United States
- Department of Neurology, Baltimore, MD, United States
- Oncology, Baltimore, MD, United States
- Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins University School of Medicine, Baltimore, MD, United States
| |
Collapse
|
35
|
Kang ZY, Huang QY, Zhen NX, Xuan NX, Zhou QC, Zhao J, Cui W, Zhang ZC, Tian BP. Heterogeneity of immune cells and their communications unveiled by transcriptome profiling in acute inflammatory lung injury. Front Immunol 2024; 15:1382449. [PMID: 38745657 PMCID: PMC11092984 DOI: 10.3389/fimmu.2024.1382449] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Accepted: 04/03/2024] [Indexed: 05/16/2024] Open
Abstract
Background Acute Respiratory Distress Syndrome (ARDS) or its earlier stage Acute lung injury (ALI), is a worldwide health concern that jeopardizes human well-being. Currently, the treatment strategies to mitigate the incidence and mortality of ARDS are severely restricted. This limitation can be attributed, at least in part, to the substantial variations in immunity observed in individuals with this syndrome. Methods Bulk and single cell RNA sequencing from ALI mice and single cell RNA sequencing from ARDS patients were analyzed. We utilized the Seurat program package in R and cellmarker 2.0 to cluster and annotate the data. The differential, enrichment, protein interaction, and cell-cell communication analysis were conducted. Results The mice with ALI caused by pulmonary and extrapulmonary factors demonstrated differential expression including Clec4e, Retnlg, S100a9, Coro1a, and Lars2. We have determined that inflammatory factors have a greater significance in extrapulmonary ALI, while multiple pathways collaborate in the development of pulmonary ALI. Clustering analysis revealed significant heterogeneity in the relative abundance of immune cells in different ALI models. The autocrine action of neutrophils plays a crucial role in pulmonary ALI. Additionally, there was a significant increase in signaling intensity between B cells and M1 macrophages, NKT cells and M1 macrophages in extrapulmonary ALI. The CXCL, CSF3 and MIF, TGFβ signaling pathways play a vital role in pulmonary and extrapulmonary ALI, respectively. Moreover, the analysis of human single-cell revealed DCs signaling to monocytes and neutrophils in COVID-19-associated ARDS is stronger compared to sepsis-related ARDS. In sepsis-related ARDS, CD8+ T and Th cells exhibit more prominent signaling to B-cell nucleated DCs. Meanwhile, both MIF and CXCL signaling pathways are specific to sepsis-related ARDS. Conclusion This study has identified specific gene signatures and signaling pathways in animal models and human samples that facilitate the interaction between immune cells, which could be targeted therapeutically in ARDS patients of various etiologies.
Collapse
Affiliation(s)
- Zhi-ying Kang
- Department of Critical Care Medicine, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Qian-yu Huang
- Department of Critical Care Medicine, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Ning-xin Zhen
- Department of Critical Care Medicine, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Nan-xia Xuan
- Department of Critical Care Medicine, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Qi-chao Zhou
- Department of Critical Care Medicine, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Jie Zhao
- Department of Critical Care Medicine, The First Affiliated Hospital, Ningbo University, Ningbo, Zhejiang, China
| | - Wei Cui
- Department of Critical Care Medicine, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Zhao-cai Zhang
- Department of Critical Care Medicine, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Bao-ping Tian
- Department of Critical Care Medicine, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| |
Collapse
|
36
|
Yu S, Li WV. spVC for the detection and interpretation of spatial gene expression variation. Genome Biol 2024; 25:103. [PMID: 38641849 PMCID: PMC11027374 DOI: 10.1186/s13059-024-03245-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2023] [Accepted: 04/10/2024] [Indexed: 04/21/2024] Open
Abstract
Spatially resolved transcriptomics technologies have opened new avenues for understanding gene expression heterogeneity in spatial contexts. However, existing methods for identifying spatially variable genes often focus solely on statistical significance, limiting their ability to capture continuous expression patterns and integrate spot-level covariates. To address these challenges, we introduce spVC, a statistical method based on a generalized Poisson model. spVC seamlessly integrates constant and spatially varying effects of covariates, facilitating comprehensive exploration of gene expression variability and enhancing interpretability. Simulation and real data applications confirm spVC's accuracy in these tasks, highlighting its versatility in spatial transcriptomics analysis.
Collapse
Affiliation(s)
- Shan Yu
- Department of Statistics, Unversity of Virginia, Charlottesville, 22903, VA, USA.
| | - Wei Vivian Li
- Department of Statistics, University of California, Riverside, 92521, CA, USA.
| |
Collapse
|
37
|
Lu Y, Chen QM, An L. SPADE: spatial deconvolution for domain specific cell-type estimation. Commun Biol 2024; 7:469. [PMID: 38632414 PMCID: PMC11024133 DOI: 10.1038/s42003-024-06172-y] [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: 06/06/2023] [Accepted: 04/10/2024] [Indexed: 04/19/2024] Open
Abstract
Understanding gene expression in different cell types within their spatial context is a key goal in genomics research. SPADE (SPAtial DEconvolution), our proposed method, addresses this by integrating spatial patterns into the analysis of cell type composition. This approach uses a combination of single-cell RNA sequencing, spatial transcriptomics, and histological data to accurately estimate the proportions of cell types in various locations. Our analyses of synthetic data have demonstrated SPADE's capability to discern cell type-specific spatial patterns effectively. When applied to real-life datasets, SPADE provides insights into cellular dynamics and the composition of tumor tissues. This enhances our comprehension of complex biological systems and aids in exploring cellular diversity. SPADE represents a significant advancement in deciphering spatial gene expression patterns, offering a powerful tool for the detailed investigation of cell types in spatial transcriptomics.
Collapse
Affiliation(s)
- Yingying Lu
- Interdisciplinary Program in Statistics and Data Science, University of Arizona, Tucson, AZ, 85721, USA
| | - Qin M Chen
- College of Pharmacy, University of Arizona, Tucson, AZ, 85721, USA
| | - Lingling An
- Interdisciplinary Program in Statistics and Data Science, University of Arizona, Tucson, AZ, 85721, USA.
- Department of Biosystems Engineering, University of Arizona, Tucson, AZ, 85721, USA.
- Department of Epidemiology and Biostatistics, University of Arizona, Tucson, AZ, 85721, USA.
| |
Collapse
|
38
|
de Reus AJEM, Basak O, Dykstra W, van Asperen JV, van Bodegraven EJ, Hol EM. GFAP-isoforms in the nervous system: Understanding the need for diversity. Curr Opin Cell Biol 2024; 87:102340. [PMID: 38401182 DOI: 10.1016/j.ceb.2024.102340] [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: 11/22/2023] [Accepted: 01/30/2024] [Indexed: 02/26/2024]
Abstract
Glial fibrillary acidic protein (GFAP) is an intermediate filament (IF) protein expressed in specific types of glial cells in the nervous system. The expression of GFAP is highly regulated during brain development and in neurological diseases. The presence of distinct GFAP-isoforms in various cell types, developmental stages, and diseases indicates that GFAP (post-)transcriptional regulation has a role in glial cell physiology and pathology. GFAP-isoforms differ in sub-cellular localisation, IF-network assembly properties, and IF-dynamics which results in distinct molecular interactions and mechanical properties of the IF-network. Therefore, GFAP (post-)transcriptional regulation is likely a mechanism by which radial glia, astrocytes, and glioma cells can modulate cellular function.
Collapse
Affiliation(s)
- Alexandra J E M de Reus
- Department of Translational Neuroscience, Brain Center, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Onur Basak
- Department of Translational Neuroscience, Brain Center, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Werner Dykstra
- Department of Translational Neuroscience, Brain Center, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Jessy V van Asperen
- Institut NeuroMyoGène (INMG), Unité Physiopathologie et Génétique du Neurone et du Muscle, Unversité Claude Bernard Lyon 1 CNRS UMR 5261, INSERM U1315, Lyon, France
| | - Emma J van Bodegraven
- Department of Translational Neuroscience, Brain Center, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Elly M Hol
- Department of Translational Neuroscience, Brain Center, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands.
| |
Collapse
|
39
|
Li R, Chen X, Yang X. Navigating the landscapes of spatial transcriptomics: How computational methods guide the way. WILEY INTERDISCIPLINARY REVIEWS. RNA 2024; 15:e1839. [PMID: 38527900 DOI: 10.1002/wrna.1839] [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: 11/23/2023] [Revised: 02/24/2024] [Accepted: 03/04/2024] [Indexed: 03/27/2024]
Abstract
Spatially resolved transcriptomics has been dramatically transforming biological and medical research in various fields. It enables transcriptome profiling at single-cell, multi-cellular, or sub-cellular resolution, while retaining the information of geometric localizations of cells in complex tissues. The coupling of cell spatial information and its molecular characteristics generates a novel multi-modal high-throughput data source, which poses new challenges for the development of analytical methods for data-mining. Spatial transcriptomic data are often highly complex, noisy, and biased, presenting a series of difficulties, many unresolved, for data analysis and generation of biological insights. In addition, to keep pace with the ever-evolving spatial transcriptomic experimental technologies, the existing analytical theories and tools need to be updated and reformed accordingly. In this review, we provide an overview and discussion of the current computational approaches for mining of spatial transcriptomics data. Future directions and perspectives of methodology design are proposed to stimulate further discussions and advances in new analytical models and algorithms. This article is categorized under: RNA Methods > RNA Analyses in Cells RNA Evolution and Genomics > Computational Analyses of RNA RNA Export and Localization > RNA Localization.
Collapse
Affiliation(s)
- Runze Li
- MOE Key Laboratory of Bioinformatics, Center for Synthetic & Systems Biology, School of Life Sciences, Tsinghua University, Beijing, China
| | - Xu Chen
- MOE Key Laboratory of Bioinformatics, Center for Synthetic & Systems Biology, School of Life Sciences, Tsinghua University, Beijing, China
| | - Xuerui Yang
- MOE Key Laboratory of Bioinformatics, Center for Synthetic & Systems Biology, School of Life Sciences, Tsinghua University, Beijing, China
| |
Collapse
|
40
|
Yan K, Liu Q, Huang R, Jiang Y, Bian Z, Li S, Li L, Shen F, Tsuneyama K, Zhang Q, Lian Z, Guan H, Xu B. Spatial transcriptomics reveals prognosis-associated cellular heterogeneity in the papillary thyroid carcinoma microenvironment. Clin Transl Med 2024; 14:e1594. [PMID: 38426403 PMCID: PMC10905537 DOI: 10.1002/ctm2.1594] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Revised: 01/28/2024] [Accepted: 02/05/2024] [Indexed: 03/02/2024] Open
Abstract
BACKGROUND Papillary thyroid carcinoma (PTC) is the most common malignant endocrine tumour, and its incidence and prevalence are increasing considerably. Cellular heterogeneity in the tumour microenvironment is important for PTC prognosis. Spatial transcriptomics is a powerful technique for cellular heterogeneity study. METHODS In conjunction with a clinical pathologist identification method, spatial transcriptomics was employed to characterise the spatial location and RNA profiles of PTC-associated cells within the tissue sections. The spatial RNA-clinical signature genes for each cell type were extracted and applied to outlining the distribution regions of specific cells on the entire section. The cellular heterogeneity of each cell type was further revealed by ContourPlot analysis, monocle analysis, trajectory analysis, ligand-receptor analysis and Gene Ontology enrichment analysis. RESULTS The spatial distribution region of tumour cells, typical and atypical follicular cells (FCs and AFCs) and immune cells were accurately and comprehensively identified in all five PTC tissue sections. AFCs were identified as a transitional state between FCs and tumour cells, exhibiting a higher resemblance to the latter. Three tumour foci were shared among all patients out of the 13 observed. Notably, tumour foci No. 2 displayed elevated expression levels of genes associated with lower relapse-free survival in PTC patients. We discovered key ligand-receptor interactions, including LAMB3-ITGA2, FN1-ITGA3 and FN1-SDC4, involved in the transition of PTC cells from FCs to AFCs and eventually to tumour cells. High expression of these patterns correlated with reduced relapse-free survival. In the tumour immune microenvironment, reduced interaction between myeloid-derived TGFB1 and TGFBR1 in tumour focus No. 2 contributed to tumourigenesis and increased heterogeneity. The spatial RNA-clinical analysis method developed here revealed prognosis-associated cellular heterogeneity in the PTC microenvironment. CONCLUSIONS The occurrence of tumour foci No. 2 and three enhanced ligand-receptor interactions in the AFC area/tumour foci reduced the relapse-free survival of PTC patients, potentially leading to improved prognostic strategies and targeted therapies for PTC patients.
Collapse
Affiliation(s)
- Kai Yan
- Guangdong Cardiovascular InstituteGuangdong Provincial People's HospitalGuangdong Academy of Medical SciencesGuangzhouChina
| | - Qing‐Zhi Liu
- Chronic Disease LaboratoryInstitutes for Life SciencesSouth China University of TechnologyGuangzhouChina
| | - Rong‐Rong Huang
- Guangdong Cardiovascular InstituteGuangdong Provincial People's HospitalGuangdong Academy of Medical SciencesGuangzhouChina
| | - Yi‐Hua Jiang
- Guangdong Cardiovascular InstituteGuangdong Provincial People's HospitalGuangdong Academy of Medical SciencesGuangzhouChina
- Guangdong Provincial Key Laboratory of Artificial Intelligence in Medical Image Analysis and ApplicationGuangzhouChina
| | - Zhen‐Hua Bian
- School of Biomedical Sciences and EngineeringSouth China University of TechnologyGuangzhou International CampusGuangzhouChina
| | - Si‐Jin Li
- Department of Thyroid SurgeryGuangzhou First People's HospitalSouth China University of TechnologyGuangzhouChina
| | - Liang Li
- Medical Research InstituteGuangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences)Southern Medical UniversityGuangzhouChina
| | - Fei Shen
- Department of Thyroid SurgeryGuangzhou First People's HospitalSouth China University of TechnologyGuangzhouChina
| | - Koichi Tsuneyama
- Department of Pathology and Laboratory MedicineInstitute of Biomedical SciencesTokushima University Graduate SchoolTokushimaJapan
| | - Qing‐Ling Zhang
- Department of PathologyGuangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences)Southern Medical UniversityGuangzhouChina
| | - Zhe‐Xiong Lian
- Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences)Southern Medical UniversityGuangzhouChina
| | - Haixia Guan
- Department of EndocrinologyGuangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences)Southern Medical UniversityGuangzhouChina
| | - Bo Xu
- Department of Thyroid SurgeryGuangzhou First People's HospitalSouth China University of TechnologyGuangzhouChina
| |
Collapse
|
41
|
Hartman A, Satija R. Comparative analysis of multiplexed in situ gene expression profiling technologies. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.11.575135. [PMID: 38260366 PMCID: PMC10802595 DOI: 10.1101/2024.01.11.575135] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/24/2024]
Abstract
The burgeoning interest in in situ multiplexed gene expression profiling technologies has opened new avenues for understanding cellular behavior and interactions. In this study, we present a comparative benchmark analysis of six in situ gene expression profiling methods, including both commercially available and academically developed methods, using publicly accessible mouse brain datasets. We find that standard sensitivity metrics, such as the number of unique molecules detected per cell, are not directly comparable across datasets due to substantial differences in the incidence of off-target molecular artifacts impacting specificity. To address these challenges, we explored various potential sources of molecular artifacts, developed novel metrics to control for them, and utilized these metrics to evaluate and compare different in situ technologies. Finally, we demonstrate how molecular false positives can seriously confound spatially-aware differential expression analysis, requiring caution in the interpretation of downstream results. Our analysis provides guidance for the selection, processing, and interpretation of in situ spatial technologies.
Collapse
|
42
|
Kiessling P, Kuppe C. Spatial multi-omics: novel tools to study the complexity of cardiovascular diseases. Genome Med 2024; 16:14. [PMID: 38238823 PMCID: PMC10795303 DOI: 10.1186/s13073-024-01282-y] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2023] [Accepted: 01/02/2024] [Indexed: 01/22/2024] Open
Abstract
Spatial multi-omic studies have emerged as a promising approach to comprehensively analyze cells in tissues, enabling the joint analysis of multiple data modalities like transcriptome, epigenome, proteome, and metabolome in parallel or even the same tissue section. This review focuses on the recent advancements in spatial multi-omics technologies, including novel data modalities and computational approaches. We discuss the advancements in low-resolution and high-resolution spatial multi-omics methods which can resolve up to 10,000 of individual molecules at subcellular level. By applying and integrating these techniques, researchers have recently gained valuable insights into the molecular circuits and mechanisms which govern cell biology along the cardiovascular disease spectrum. We provide an overview of current data analysis approaches, with a focus on data integration of multi-omic datasets, highlighting strengths and weaknesses of various computational pipelines. These tools play a crucial role in analyzing and interpreting spatial multi-omics datasets, facilitating the discovery of new findings, and enhancing translational cardiovascular research. Despite nontrivial challenges, such as the need for standardization of experimental setups, data analysis, and improved computational tools, the application of spatial multi-omics holds tremendous potential in revolutionizing our understanding of human disease processes and the identification of novel biomarkers and therapeutic targets. Exciting opportunities lie ahead for the spatial multi-omics field and will likely contribute to the advancement of personalized medicine for cardiovascular diseases.
Collapse
Affiliation(s)
- Paul Kiessling
- Department of Nephrology, Rheumatology, and Clinical Immunology, University Hospital RWTH Aachen, Aachen, Germany
| | - Christoph Kuppe
- Department of Nephrology, Rheumatology, and Clinical Immunology, University Hospital RWTH Aachen, Aachen, Germany.
| |
Collapse
|
43
|
Chen Y, Yang S, Yu K, Zhang J, Wu M, Zheng Y, Zhu Y, Dai J, Wang C, Zhu X, Dai Y, Sun Y, Wu T, Wang S. Spatial omics: An innovative frontier in aging research. Ageing Res Rev 2024; 93:102158. [PMID: 38056503 DOI: 10.1016/j.arr.2023.102158] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Revised: 11/25/2023] [Accepted: 12/01/2023] [Indexed: 12/08/2023]
Abstract
Disentangling the impact of aging on health and disease has become critical as population aging progresses rapidly. Studying aging at the molecular level is complicated by the diverse aging profiles and dynamics. However, the examination of cellular states within aging tissues in situ is hampered by the lack of high-resolution spatial data. Emerging spatial omics technologies facilitate molecular and spatial analysis of tissues, providing direct access to precise information on various functional regions and serving as a favorable tool for unraveling the heterogeneity of aging. In this review, we summarize the recent advances in spatial omics application in multi-organ aging research, which has enhanced the understanding of aging mechanisms from multiple standpoints. We also discuss the main challenges in spatial omics research to date, the opportunities for further developing the technology, and the potential applications of spatial omics in aging and aging-related diseases.
Collapse
Affiliation(s)
- Ying Chen
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; National Clinical Research Center for Obstetrical and Gynecological Diseases, Wuhan, China; Ministry of Education, Key Laboratory of Cancer Invasion and Metastasis, Wuhan, China
| | - Shuhao Yang
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; National Clinical Research Center for Obstetrical and Gynecological Diseases, Wuhan, China; Ministry of Education, Key Laboratory of Cancer Invasion and Metastasis, Wuhan, China
| | - Kaixu Yu
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jinjin Zhang
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; National Clinical Research Center for Obstetrical and Gynecological Diseases, Wuhan, China; Ministry of Education, Key Laboratory of Cancer Invasion and Metastasis, Wuhan, China
| | - Meng Wu
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; National Clinical Research Center for Obstetrical and Gynecological Diseases, Wuhan, China; Ministry of Education, Key Laboratory of Cancer Invasion and Metastasis, Wuhan, China
| | - Yongqiang Zheng
- State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Centre, Sun Yat-sen University, Guangzhou, China
| | - Yun Zhu
- Department of Internal Medicine, Southern Illinois University School of Medicine, 801 N. Rutledge, P.O. Box 19628, Springfield, IL 62702, USA
| | - Jun Dai
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; National Clinical Research Center for Obstetrical and Gynecological Diseases, Wuhan, China; Ministry of Education, Key Laboratory of Cancer Invasion and Metastasis, Wuhan, China
| | - Chunyan Wang
- College of Science & Engineering Jinan University, Guangzhou, China
| | - Xiaoran Zhu
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; National Clinical Research Center for Obstetrical and Gynecological Diseases, Wuhan, China; Ministry of Education, Key Laboratory of Cancer Invasion and Metastasis, Wuhan, China
| | - Yun Dai
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; National Clinical Research Center for Obstetrical and Gynecological Diseases, Wuhan, China; Ministry of Education, Key Laboratory of Cancer Invasion and Metastasis, Wuhan, China
| | - Yunhong Sun
- Hubei Key Laboratory of Food Nutrition and Safety, MOE Key Lab of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Tong Wu
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; National Clinical Research Center for Obstetrical and Gynecological Diseases, Wuhan, China; Ministry of Education, Key Laboratory of Cancer Invasion and Metastasis, Wuhan, China.
| | - Shixuan Wang
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; National Clinical Research Center for Obstetrical and Gynecological Diseases, Wuhan, China; Ministry of Education, Key Laboratory of Cancer Invasion and Metastasis, Wuhan, China.
| |
Collapse
|
44
|
Pan L, Mou T, Huang Y, Hong W, Yu M, Li X. Ursa: A Comprehensive Multiomics Toolbox for High-Throughput Single-Cell Analysis. Mol Biol Evol 2023; 40:msad267. [PMID: 38091963 PMCID: PMC10752348 DOI: 10.1093/molbev/msad267] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Revised: 09/08/2023] [Accepted: 11/03/2023] [Indexed: 12/28/2023] Open
Abstract
The burgeoning amount of single-cell data has been accompanied by revolutionary changes to computational methods to map, quantify, and analyze the outputs of these cutting-edge technologies. Many are still unable to reap the benefits of these advancements due to the lack of bioinformatics expertise. To address this issue, we present Ursa, an automated single-cell multiomics R package containing 6 automated single-cell omics and spatial transcriptomics workflows. Ursa allows scientists to carry out post-quantification single or multiomics analyses in genomics, transcriptomics, epigenetics, proteomics, and immunomics at the single-cell level. It serves as a 1-stop analytic solution by providing users with outcomes to quality control assessments, multidimensional analyses such as dimension reduction and clustering, and extended analyses such as pseudotime trajectory and gene-set enrichment analyses. Ursa aims bridge the gap between those with bioinformatics expertise and those without by providing an easy-to-use bioinformatics package for scientists in hoping to accelerate their research potential. Ursa is freely available at https://github.com/singlecellomics/ursa.
Collapse
Affiliation(s)
- Lu Pan
- Institute of Environmental Medicine, Karolinska Institutet, Solna 171 65, Sweden
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Solna 171 65, Sweden
| | - Tian Mou
- School of Biomedical Engineering, Shenzhen University, Shenzhen, Guangdong 518060, China
| | - Yue Huang
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Solna 171 65, Sweden
| | - Weifeng Hong
- Department of Radiation Oncology, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Min Yu
- Department of General Surgery, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong 510515, China
| | - Xuexin Li
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Solna 171 65, Sweden
- Department of General Surgery, The Fourth Affiliated Hospital, China Medical University, Shenyang 110032, China
| |
Collapse
|
45
|
Kodous AS, Balaiah M, Ramanathan P. Single cell RNA sequencing – a valuable tool for cancer immunotherapy: a mini review. ONCOLOGIE 2023; 25:635-639. [DOI: 10.1515/oncologie-2023-0244] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2025]
Abstract
Abstract
Single-cell RNA sequencing (scRNA-seq) technology has made great strides in research over the last decade. Data analysis has been aided by developments in bioinformatics tools and artificial intelligence, allowing biological and clinical researchers to get a deeper understanding of the different cell clusters and their dynamics within tumours. Combining conventional treatment modalities like chemotherapy and radiation with immunotherapy is a growing trend in cancer treatment. Hence, knowledge of the tumour microenvironment and the effect of each treatment modality on the TME, at a single cell level can provide treating clinicians with better clues for patient stratification and prognostication. With this knowledge, immunotherapy could become successful in treating a wide range of cancers, opening the path for the creation of even more effective treatment strategies. Despite the widespread availability of scRNA-seq technology, computational analysis and data interpretation are still challenges. Worldwide, such challenges are being addressed by various researchers, strengthening the contribution of this technology towards cancer elimination. In this mini-review, we primarily focus on the technique, its workflow, and the computational aspects of scRNA technology, along with an overview of the current challenges in the analysis and interpretation of the data generated.
Collapse
Affiliation(s)
- Ahmad S. Kodous
- Department of Molecular Oncology , Cancer Institute (WIA) , Chennai , Tamil Nadu , India
- Radiation Biology Department , National Centre for Radiation Research and Technology (NCRRT), Egyptian Atomic Energy Authority (EAEA) , Cairo , Egypt
| | - Meenakumari Balaiah
- Department of Molecular Oncology , Cancer Institute (WIA) , Chennai , Tamil Nadu , India
| | - Priya Ramanathan
- Department of Molecular Oncology , Cancer Institute (WIA) , Chennai , Tamil Nadu , India
| |
Collapse
|
46
|
Robles-Remacho A, Sanchez-Martin RM, Diaz-Mochon JJ. Spatial Transcriptomics: Emerging Technologies in Tissue Gene Expression Profiling. Anal Chem 2023; 95:15450-15460. [PMID: 37814884 PMCID: PMC10603609 DOI: 10.1021/acs.analchem.3c02029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Accepted: 09/21/2023] [Indexed: 10/11/2023]
Abstract
In this Perspective, we discuss the current status and advances in spatial transcriptomics technologies, which allow high-resolution mapping of gene expression in intact cell and tissue samples. Spatial transcriptomics enables the creation of high-resolution maps of gene expression patterns within their native spatial context, adding an extra layer of information to the bulk sequencing data. Spatial transcriptomics has expanded significantly in recent years and is making a notable impact on a range of fields, including tissue architecture, developmental biology, cancer, and neurodegenerative and infectious diseases. The latest advancements in spatial transcriptomics have resulted in the development of highly multiplexed methods, transcriptomic-wide analysis, and single-cell resolution utilizing diverse technological approaches. In this Perspective, we provide a detailed analysis of the molecular foundations behind the main spatial transcriptomics technologies, including methods based on microdissection, in situ sequencing, single-molecule FISH, spatial capturing, selection of regions of interest, and single-cell or nuclei dissociation. We contextualize the detection and capturing efficiency, strengths, limitations, tissue compatibility, and applications of these techniques as well as provide information on data analysis. In addition, this Perspective discusses future directions and potential applications of spatial transcriptomics, highlighting the importance of the continued development to promote widespread adoption of these techniques within the research community.
Collapse
Affiliation(s)
- Agustín Robles-Remacho
- GENYO.
Centre for Genomics and Oncological Research, Pfizer/University of Granada/Andalusian Regional Government, PTS Granada, Avenida de la Ilustracion,
114. 18016 Granada, Spain
- Department
of Medicinal and Organic Chemistry, School of Pharmacy, University of Granada, Campus Cartuja s/n, 18071 Granada, Spain
- Instituto
de Investigación Biosanitaria ibs.GRANADA, 18012 Granada, Spain
| | - Rosario M. Sanchez-Martin
- GENYO.
Centre for Genomics and Oncological Research, Pfizer/University of Granada/Andalusian Regional Government, PTS Granada, Avenida de la Ilustracion,
114. 18016 Granada, Spain
- Department
of Medicinal and Organic Chemistry, School of Pharmacy, University of Granada, Campus Cartuja s/n, 18071 Granada, Spain
- Instituto
de Investigación Biosanitaria ibs.GRANADA, 18012 Granada, Spain
| | - Juan J. Diaz-Mochon
- GENYO.
Centre for Genomics and Oncological Research, Pfizer/University of Granada/Andalusian Regional Government, PTS Granada, Avenida de la Ilustracion,
114. 18016 Granada, Spain
- Department
of Medicinal and Organic Chemistry, School of Pharmacy, University of Granada, Campus Cartuja s/n, 18071 Granada, Spain
- Instituto
de Investigación Biosanitaria ibs.GRANADA, 18012 Granada, Spain
| |
Collapse
|
47
|
Mukhaleva E, Ma N, van der Velden WJC, Gogoshin G, Branciamore S, Bhattacharya S, Rodin AS, Vaidehi N. Bayesian network models identify co-operative GPCR:G protein interactions that contribute to G protein coupling. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.10.09.561618. [PMID: 37873104 PMCID: PMC10592737 DOI: 10.1101/2023.10.09.561618] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/25/2023]
Abstract
Cooperative interactions in protein-protein interfaces demonstrate the interdependency or the linked network-like behavior of interface interactions and their effect on the coupling of proteins. Cooperative interactions also could cause ripple or allosteric effects at a distance in protein-protein interfaces. Although they are critically important in protein-protein interfaces it is challenging to determine which amino acid pair interactions are cooperative. In this work we have used Bayesian network modeling, an interpretable machine learning method, combined with molecular dynamics trajectories to identify the residue pairs that show high cooperativity and their allosteric effect in the interface of G protein-coupled receptor (GPCR) complexes with G proteins. Our results reveal a strong co-dependency in the formation of interface GPCR:G protein contacts. This observation indicates that cooperativity of GPCR:G protein interactions is necessary for the coupling and selectivity of G proteins and is thus critical for receptor function. We have identified subnetworks containing polar and hydrophobic interactions that are common among multiple GPCRs coupling to different G protein subtypes (Gs, Gi and Gq). These common subnetworks along with G protein-specific subnetworks together confer selectivity to the G protein coupling. This work underscores the potential of data-driven Bayesian network modeling in elucidating the intricate dependencies and selectivity determinants in GPCR:G protein complexes, offering valuable insights into the dynamic nature of these essential cellular signaling components.
Collapse
Affiliation(s)
- Elizaveta Mukhaleva
- Department of Computational and Quantitative Medicine, Beckman Research Institute of the City of Hope, Duarte, CA 91010
- Irell and Manella Graduate School of Biological Sciences, Beckman Research Institute of the City of Hope, Duarte, CA 91010
| | - Ning Ma
- Department of Computational and Quantitative Medicine, Beckman Research Institute of the City of Hope, Duarte, CA 91010
| | - Wijnand J. C. van der Velden
- Department of Computational and Quantitative Medicine, Beckman Research Institute of the City of Hope, Duarte, CA 91010
| | - Grigoriy Gogoshin
- Department of Computational and Quantitative Medicine, Beckman Research Institute of the City of Hope, Duarte, CA 91010
| | - Sergio Branciamore
- Department of Computational and Quantitative Medicine, Beckman Research Institute of the City of Hope, Duarte, CA 91010
- Irell and Manella Graduate School of Biological Sciences, Beckman Research Institute of the City of Hope, Duarte, CA 91010
| | - Supriyo Bhattacharya
- Department of Computational and Quantitative Medicine, Beckman Research Institute of the City of Hope, Duarte, CA 91010
| | - Andrei S. Rodin
- Department of Computational and Quantitative Medicine, Beckman Research Institute of the City of Hope, Duarte, CA 91010
- Irell and Manella Graduate School of Biological Sciences, Beckman Research Institute of the City of Hope, Duarte, CA 91010
| | - Nagarajan Vaidehi
- Department of Computational and Quantitative Medicine, Beckman Research Institute of the City of Hope, Duarte, CA 91010
- Irell and Manella Graduate School of Biological Sciences, Beckman Research Institute of the City of Hope, Duarte, CA 91010
| |
Collapse
|
48
|
Zhou Y, Jiang X, Wang X, Huang J, Li T, Jin H, He J. Promise of spatially resolved omics for tumor research. J Pharm Anal 2023; 13:851-861. [PMID: 37719191 PMCID: PMC10499658 DOI: 10.1016/j.jpha.2023.07.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2022] [Revised: 07/01/2023] [Accepted: 07/06/2023] [Indexed: 09/19/2023] Open
Abstract
Tumors are spatially heterogeneous tissues that comprise numerous cell types with intricate structures. By interacting with the microenvironment, tumor cells undergo dynamic changes in gene expression and metabolism, resulting in spatiotemporal variations in their capacity for proliferation and metastasis. In recent years, the rapid development of histological techniques has enabled efficient and high-throughput biomolecule analysis. By preserving location information while obtaining a large number of gene and molecular data, spatially resolved metabolomics (SRM) and spatially resolved transcriptomics (SRT) approaches can offer new ideas and reliable tools for the in-depth study of tumors. This review provides a comprehensive introduction and summary of the fundamental principles and research methods used for SRM and SRT techniques, as well as a review of their applications in cancer-related fields.
Collapse
Affiliation(s)
- Yanhe Zhou
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China
| | - Xinyi Jiang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China
| | - Xiangyi Wang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China
| | - Jianpeng Huang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China
| | - Tong Li
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China
| | - Hongtao Jin
- New Drug Safety Evaluation Center, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, China
- NMPA Key Laboratory for Safety Research and Evaluation of Innovative Drug, Beijing, 10050, China
| | - Jiuming He
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China
- NMPA Key Laboratory for Safety Research and Evaluation of Innovative Drug, Beijing, 10050, China
| |
Collapse
|
49
|
Krieger KL, Mann EK, Lee KJ, Bolterstein E, Jebakumar D, Ittmann MM, Dal Zotto VL, Shaban M, Sreekumar A, Gassman NR. Spatial mapping of the DNA adducts in cancer. DNA Repair (Amst) 2023; 128:103529. [PMID: 37390674 PMCID: PMC10330576 DOI: 10.1016/j.dnarep.2023.103529] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Revised: 06/19/2023] [Accepted: 06/21/2023] [Indexed: 07/02/2023]
Abstract
DNA adducts and strand breaks are induced by various exogenous and endogenous agents. Accumulation of DNA damage is implicated in many disease processes, including cancer, aging, and neurodegeneration. The continuous acquisition of DNA damage from exogenous and endogenous stressors coupled with defects in DNA repair pathways contribute to the accumulation of DNA damage within the genome and genomic instability. While mutational burden offers some insight into the level of DNA damage a cell may have experienced and subsequently repaired, it does not quantify DNA adducts and strand breaks. Mutational burden also infers the identity of the DNA damage. With advances in DNA adduct detection and quantification methods, there is an opportunity to identify DNA adducts driving mutagenesis and correlate with a known exposome. However, most DNA adduct detection methods require isolation or separation of the DNA and its adducts from the context of the nuclei. Mass spectrometry, comet assays, and other techniques precisely quantify lesion types but lose the nuclear context and even tissue context of the DNA damage. The growth in spatial analysis technologies offers a novel opportunity to leverage DNA damage detection with nuclear and tissue context. However, we lack a wealth of techniques capable of detecting DNA damage in situ. Here, we review the limited existing in situ DNA damage detection methods and examine their potential to offer spatial analysis of DNA adducts in tumors or other tissues. We also offer a perspective on the need for spatial analysis of DNA damage in situ and highlight Repair Assisted Damage Detection (RADD) as an in situ DNA adduct technique with the potential to integrate with spatial analysis and the challenges to be addressed.
Collapse
Affiliation(s)
- Kimiko L Krieger
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA; Center for Translational Metabolism and Health Disparities (C-TMH), Baylor College of Medicine, Houston, TX 77030, USA
| | - Elise K Mann
- Department of Physiology and Cell Biology, College of Medicine, University of South Alabama, Mobile, AL 36688, USA; Mitchell Cancer Institute, University of South Alabama, Mobile, AL 36604, USA
| | - Kevin J Lee
- Department of Physiology and Cell Biology, College of Medicine, University of South Alabama, Mobile, AL 36688, USA; Mitchell Cancer Institute, University of South Alabama, Mobile, AL 36604, USA
| | - Elyse Bolterstein
- Department of Biology, Northeastern Illinois University, Chicago, IL 60625, USA
| | - Deborah Jebakumar
- Department of Anatomic Pathology, Baylor Scott & White Medical Center, Temple, TX 76508, USA; Texas A&M College of Medicine, Temple, TX 76508, USA
| | - Michael M Ittmann
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, TX 77030, USA; Human Tissue Acquisition & Pathology Shared Resource, Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX 77030, USA
| | - Valeria L Dal Zotto
- Department of Pathology, College of Medicine, University of South Alabama, Mobile, AL 36688, USA
| | - Mohamed Shaban
- Department of Electrical and Computer Engineering, University of South Alabama, Mobile, AL 36688, USA
| | - Arun Sreekumar
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA; Center for Translational Metabolism and Health Disparities (C-TMH), Baylor College of Medicine, Houston, TX 77030, USA; Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX 77030, USA; Verna and Marrs McLean Department of Biochemistry and Molecular Biology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Natalie R Gassman
- Department of Pharmacology and Toxicology, University of Alabama at Birmingham, Birmingham, AL 35294, USA.
| |
Collapse
|
50
|
Lyu P, Li Y, Wen X, Cao H. JUMP: replicability analysis of high-throughput experiments with applications to spatial transcriptomic studies. Bioinformatics 2023; 39:btad366. [PMID: 37279733 PMCID: PMC10279524 DOI: 10.1093/bioinformatics/btad366] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Revised: 05/26/2023] [Accepted: 06/02/2023] [Indexed: 06/08/2023] Open
Abstract
MOTIVATION Replicability is the cornerstone of scientific research. The current statistical method for high-dimensional replicability analysis either cannot control the false discovery rate (FDR) or is too conservative. RESULTS We propose a statistical method, JUMP, for the high-dimensional replicability analysis of two studies. The input is a high-dimensional paired sequence of p-values from two studies and the test statistic is the maximum of p-values of the pair. JUMP uses four states of the p-value pairs to indicate whether they are null or non-null. Conditional on the hidden states, JUMP computes the cumulative distribution function of the maximum of p-values for each state to conservatively approximate the probability of rejection under the composite null of replicability. JUMP estimates unknown parameters and uses a step-up procedure to control FDR. By incorporating different states of composite null, JUMP achieves a substantial power gain over existing methods while controlling the FDR. Analyzing two pairs of spatially resolved transcriptomic datasets, JUMP makes biological discoveries that otherwise cannot be obtained by using existing methods. AVAILABILITY AND IMPLEMENTATION An R package JUMP implementing the JUMP method is available on CRAN (https://CRAN.R-project.org/package=JUMP).
Collapse
Affiliation(s)
- Pengfei Lyu
- Department of Statistics, Florida State University, 600 W College AVE, Tallahassee, FL 32306, United States
| | - Yan Li
- School of Mathematics, Jilin University, 2699 Qianjin ST, Changchun, Jilin 130012, China
| | - Xiaoquan Wen
- Department of Biostatistics, University of Michigan, Ann Arbor, MI 48109, United States
| | - Hongyuan Cao
- Department of Statistics, Florida State University, 600 W College AVE, Tallahassee, FL 32306, United States
- School of Mathematics, Jilin University, 2699 Qianjin ST, Changchun, Jilin 130012, China
| |
Collapse
|