1
|
Kataria M, Srivastava E, Arjun K, Kumar S, Gupta I, Jayadeva. A novel coarsened graph learning method for scalable single-cell data analysis. Comput Biol Med 2025; 188:109873. [PMID: 39999493 DOI: 10.1016/j.compbiomed.2025.109873] [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: 10/21/2024] [Revised: 01/26/2025] [Accepted: 02/12/2025] [Indexed: 02/27/2025]
Abstract
The emergence of single-cell technologies, including flow and mass cytometry, as well as single-cell RNA sequencing, has revolutionized the study of cellular heterogeneity, generating vast datasets rich in biological insights. Despite the effectiveness of graph-based analyses in deciphering the complexities of these datasets, managing large-scale graph representations of single-cell data remains computationally challenging. Coarsening has been employed to tackle this difficulty. However, current coarsening techniques such as Cytocoarsening, Heavy Edge Matching (HEM), and Locally Variable Edges (LVE) often suffer from slow processing speeds and limited adaptability. To address these challenges, we propose a novel approach utilizing Feature-Aware Graph Coarsening via Hashing (FACH), which integrates locality-sensitive hashing for scalable and efficient single-cell data analysis. This method directly extracts informative, low-dimensional cell representations from raw single-cell RNA sequencing and mass cytometry data, significantly improving processing speed while preserving essential data features. We demonstrate its effectiveness in downstream tasks, such as scalable graph neural network training on coarsened single-cell data, highlighting its ability to retain crucial biological features and enable efficient, accurate analyses. Our method directly extracts informative, low-dimensional cell representations from raw single-cell RNA sequencing and mass cytometry data, significantly improving processing speed and preserving critical biological features, such as transcriptional signatures and network topology. It reduces computational time by at least 50% compared to existing methods and achieves superior classification accuracy, such as 88.1% on the Baron Human dataset, underscoring its efficiency and precision in large-scale single-cell analysis.
Collapse
Affiliation(s)
- Mohit Kataria
- Yardi School of Artificial Intelligence, Indian Institute of Technology (IIT) Delhi, New Delhi, India.
| | - Ekta Srivastava
- Department Electrical Engineering, Indian Institute of Technology (IIT) Delhi, New Delhi, India
| | - Kumar Arjun
- Department of Mathematics, Indian Institute of Technology (IIT) Delhi, New Delhi, India
| | - Sandeep Kumar
- Yardi School of Artificial Intelligence, Indian Institute of Technology (IIT) Delhi, New Delhi, India; Department Electrical Engineering, Indian Institute of Technology (IIT) Delhi, New Delhi, India; Bharti School of Telecommunication Technology and Management, Indian Institute of Technology (IIT) Delhi, New Delhi, India
| | - Ishaan Gupta
- Department of Biochemical Engineering and Biotechnology, Indian Institute of Technology (IIT) Delhi, New Delhi, India
| | - Jayadeva
- Yardi School of Artificial Intelligence, Indian Institute of Technology (IIT) Delhi, New Delhi, India; Department Electrical Engineering, Indian Institute of Technology (IIT) Delhi, New Delhi, India
| |
Collapse
|
2
|
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
|
3
|
Sherif ZA, Ogunwobi OO, Ressom HW. Mechanisms and technologies in cancer epigenetics. Front Oncol 2025; 14:1513654. [PMID: 39839798 PMCID: PMC11746123 DOI: 10.3389/fonc.2024.1513654] [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: 10/18/2024] [Accepted: 12/04/2024] [Indexed: 01/23/2025] Open
Abstract
Cancer's epigenetic landscape, a labyrinthine tapestry of molecular modifications, has long captivated researchers with its profound influence on gene expression and cellular fate. This review discusses the intricate mechanisms underlying cancer epigenetics, unraveling the complex interplay between DNA methylation, histone modifications, chromatin remodeling, and non-coding RNAs. We navigate through the tumultuous seas of epigenetic dysregulation, exploring how these processes conspire to silence tumor suppressors and unleash oncogenic potential. The narrative pivots to cutting-edge technologies, revolutionizing our ability to decode the epigenome. From the granular insights of single-cell epigenomics to the holistic view offered by multi-omics approaches, we examine how these tools are reshaping our understanding of tumor heterogeneity and evolution. The review also highlights emerging techniques, such as spatial epigenomics and long-read sequencing, which promise to unveil the hidden dimensions of epigenetic regulation. Finally, we probed the transformative potential of CRISPR-based epigenome editing and computational analysis to transmute raw data into biological insights. This study seeks to synthesize a comprehensive yet nuanced understanding of the contemporary landscape and future directions of cancer epigenetic research.
Collapse
Affiliation(s)
- Zaki A. Sherif
- Department of Biochemistry & Molecular Biology, Howard University College of Medicine, Washington, DC, United States
| | - Olorunseun O. Ogunwobi
- Department of Biochemistry & Molecular Biology, Michigan State University, East Lansing, MI, United States
| | - Habtom W. Ressom
- Department of Oncology, Georgetown University Medical Center, Washington, DC, United States
| |
Collapse
|
4
|
Cheng W, Yin C, Yu S, Chen X, Hong N, Jin W. scMMO-atlas: a single cell multimodal omics atlas and portal for exploring fine cell heterogeneity and cell dynamics. Nucleic Acids Res 2025; 53:D1186-D1194. [PMID: 39315707 PMCID: PMC11701702 DOI: 10.1093/nar/gkae821] [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: 07/18/2024] [Revised: 08/27/2024] [Accepted: 09/10/2024] [Indexed: 09/25/2024] Open
Abstract
Single-cell multimodal sequencing parallelly captures multiple modalities of the same cell, providing unparalleled insights into cell heterogeneity and cell dynamics. For example, joint profiling of chromatin accessibility and transcriptome from the same single cell (scATAC + RNA) identified new cell subsets within the well-defined clusters. However, lack of single-cell multimodal omics (scMMO) database has led to data fragmentation, seriously hindering access, utilization and mining of scMMO data. Here, we constructed a scMMO atlas by collecting and integrating various scMMO data, then constructed scMMO database and portal called scMMO-atlas (https://www.biosino.org/scMMO-atlas/). scMMO-atlas includes scATAC + RNA (ISSAAS-seq, SNARE-seq, paired-seq, sci-CAR, scCARE-seq, 10X Multiome and so on), scRNA + protein, scATAC + protein and scTri-modal omics data, with 3 168 824 cells from 27 cell tissues/organs. scMMO-atlas offered an interactive portal for visualization and featured analysis for each modality and the integrated data. Integrated analysis of scATAC + RNA data of mouse cerebral cortex in scMMO-atlas identified more cell subsets compared with unimodal omics data. Among these new cell subsets, there is an early astrocyte subset highly expressed Grm3, called Astro-Grm3. Furthermore, we identified Ex-L6-Tle4-Nrf1, a progenitor of Ex-L6-Tle4, indicating the statistical power provided by the big data in scMMO-atlas. In summary, scMMO-atlas offers cell atlas, database and portal to facilitate data utilization and biological insight.
Collapse
Affiliation(s)
- Wenwen Cheng
- School of Life Sciences, Southern University of Science and Technology, Shenzhen 518055 Guangdong, China
- CAS Key Laboratory of Computational Biology, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Changhui Yin
- School of Life Sciences, Southern University of Science and Technology, Shenzhen 518055 Guangdong, China
- CAS Key Laboratory of Computational Biology, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Shiya Yu
- CAS Key Laboratory of Computational Biology, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Xi Chen
- School of Life Sciences, Southern University of Science and Technology, Shenzhen 518055 Guangdong, China
| | - Ni Hong
- School of Life Sciences, Southern University of Science and Technology, Shenzhen 518055 Guangdong, China
- CAS Key Laboratory of Computational Biology, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Wenfei Jin
- CAS Key Laboratory of Computational Biology, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| |
Collapse
|
5
|
Nakajima S, Tsuchiya H, Fujio K. Unraveling immune cell heterogeneity in autoimmune arthritis: insights from single-cell RNA sequencing. Immunol Med 2024; 47:217-229. [PMID: 39120105 DOI: 10.1080/25785826.2024.2388343] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Accepted: 07/31/2024] [Indexed: 08/10/2024] Open
Abstract
Single-cell RNA sequencing (scRNA-seq) has transformed our understanding of immune-mediated arthritis, which comprises rheumatoid arthritis and spondyloarthritis. This review outlines the key findings and advancements in scRNA-seq studies focused on the pathogenesis of autoimmune arthritis and its clinical application. In rheumatoid arthritis, scRNA-seq has elucidated the heterogeneity among synovial fibroblasts and immune cell subsets in inflammatory sites, offering insights into disease mechanisms and the differences in treatment responses. Various studies have identified distinct synovial fibroblast subpopulations, such as THY1+ inflammatory and THY1- destructive fibroblasts. Furthermore, scRNA-seq has revealed diverse T cell profiles in the synovium, including peripheral helper T cells and clonally expanded CD8+ T cells, shedding light on potential therapeutic targets and predictive markers of treatment response. Similarly, in spondyloarthritis, particularly psoriatic arthritis and ankylosing spondylitis, scRNA-seq studies have identified distinct cellular profiles associated with disease pathology. Challenges such as cost and sample size limitations persist, but collaborative efforts and utilization of public databases hold promise for overcoming these obstacles. Overall, scRNA-seq emerges as a powerful tool for dissecting cellular heterogeneity and driving precision medicine in immune-mediated arthritis.
Collapse
Affiliation(s)
- Sotaro Nakajima
- Department of Allergy and Rheumatology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Haruka Tsuchiya
- Department of Allergy and Rheumatology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Keishi Fujio
- Department of Allergy and Rheumatology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
6
|
Shang Y, Wang Z, Xi L, Wang Y, Liu M, Feng Y, Wang J, Wu Q, Xiang X, Chen M, Ding Y. Droplet-based single-cell sequencing: Strategies and applications. Biotechnol Adv 2024; 77:108454. [PMID: 39271031 DOI: 10.1016/j.biotechadv.2024.108454] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Revised: 08/22/2024] [Accepted: 09/10/2024] [Indexed: 09/15/2024]
Abstract
Notable advancements in single-cell omics technologies have not only addressed longstanding challenges but also enabled unprecedented studies of cellular heterogeneity with unprecedented resolution and scale. These strides have led to groundbreaking insights into complex biological systems, paving the way for a more profound comprehension of human biology and diseases. The droplet microfluidic technology has become a crucial component in many single-cell sequencing workflows in terms of throughput, cost-effectiveness, and automation. Utilizing a microfluidic chip to encapsulate and profile individual cells within droplets has significantly improved single-cell research. Therefore, this review aims to comprehensively elaborate the droplet microfluidics-assisted omics methods from a single-cell perspective. The strategies for using droplet microfluidics in the realms of genomics, epigenomics, transcriptomics, and proteomics analyses are first introduced. On this basis, the focus then turns to the latest applications of this technology in different sequencing patterns, including mono- and multi-omics. Finally, the challenges and further perspectives of droplet-based single-cell sequencing in both foundational research and commercial applications are discussed.
Collapse
Affiliation(s)
- Yuting Shang
- Department of Food Science & Engineering, College of Life Science and Technology, Jinan University, Guangzhou 510632, China
| | - Zhengzheng Wang
- Department of Food Science & Engineering, College of Life Science and Technology, Jinan University, Guangzhou 510632, China
| | - Liqing Xi
- Department of Food Science & Engineering, College of Life Science and Technology, Jinan University, Guangzhou 510632, China
| | - Yantao Wang
- Department of Food Science & Engineering, College of Life Science and Technology, Jinan University, Guangzhou 510632, China
| | - Meijing Liu
- Department of Food Science & Engineering, College of Life Science and Technology, Jinan University, Guangzhou 510632, China
| | - Ying Feng
- Department of Food Science & Engineering, College of Life Science and Technology, Jinan University, Guangzhou 510632, China
| | - Juan Wang
- College of Food Science, South China Agricultural University, Guangzhou 510432, China
| | - Qingping Wu
- National Health Commission Science and Technology Innovation Platform for Nutrition and Safety of Microbial Food, Guangdong Provincial Key Laboratory of Microbial Safety and Health, State Key Laboratory of Applied Microbiology Southern China, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou 510070, China
| | - Xinran Xiang
- Jiangsu Key Laboratory of Huaiyang Food Safety and Nutrition Function Evaluation, Jiangsu Collaborative Innovation Center of Regional Modern Agriculture & Environmental Protection, Jiangsu Key Laboratory for Eco-Agricultural Biotechnology Around Hongze Lake, School of Life Science, Huaiyin Normal University, Huai'an 223300, China; Fujian Key Laboratory of Aptamers Technology, Fuzhou General Clinical Medical School (the 900th Hospital), Fujian Medical University, Fuzhou 350001, China.
| | - Moutong Chen
- National Health Commission Science and Technology Innovation Platform for Nutrition and Safety of Microbial Food, Guangdong Provincial Key Laboratory of Microbial Safety and Health, State Key Laboratory of Applied Microbiology Southern China, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou 510070, China.
| | - Yu Ding
- Department of Food Science & Engineering, College of Life Science and Technology, Jinan University, Guangzhou 510632, China.
| |
Collapse
|
7
|
Kumar V, Beck S. Single-cell multimodal chromatin profiles revealing epigenetic regulations of cells in hepatocellular carcinoma. CLINICAL AND TRANSLATIONAL DISCOVERY 2024; 4:e70002. [PMID: 39444758 PMCID: PMC11495655 DOI: 10.1002/ctd2.70002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Accepted: 09/06/2024] [Indexed: 10/25/2024]
Abstract
AbstractHepatocellular carcinoma (HCC), with its increasing prevalence globally, is emerging as a major health challenge. Hepatocellular carcinoma cells exhibit both significant homogeneity and heterogeneity, governed by complex epigenomic regulations. Recently, numerous single‐cell unimodal techniques have been used to study HCC at various levelswhich have already revealed a complex interplay at genomic, transcriptomic, spatial and epigenomic levels. However, the use of single‐cell multimodal techniques combining different unimodal layers in HCC remains quite limited, necessitating further studies focusing on these methods to uncover novel markers, and mechanisms at epigenetic levels. In this commentary, we highlight how integrating multimodal approaches with epigenetic modifications can provide new insights into HCC and foster future therapeutic advancements.
Collapse
Affiliation(s)
- Vikas Kumar
- Center for Aging Research, Department of Dermatology, Chobanian & Avedisian School of Medicine, Boston University, Boston, USA
| | - Samuel Beck
- Center for Aging Research, Department of Dermatology, Chobanian & Avedisian School of Medicine, Boston University, Boston, USA
| |
Collapse
|
8
|
Wang Z, Luo P, Xiao M, Wang B, Liu T, Sun X. Recover then aggregate: unified cross-modal deep clustering with global structural information for single-cell data. Brief Bioinform 2024; 25:bbae485. [PMID: 39356327 PMCID: PMC11445907 DOI: 10.1093/bib/bbae485] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Revised: 08/24/2024] [Accepted: 09/13/2024] [Indexed: 10/03/2024] Open
Abstract
Single-cell cross-modal joint clustering has been extensively utilized to investigate the tumor microenvironment. Although numerous approaches have been suggested, accurate clustering remains the main challenge. First, the gene expression matrix frequently contains numerous missing values due to measurement limitations. The majority of existing clustering methods treat it as a typical multi-modal dataset without further processing. Few methods conduct recovery before clustering and do not sufficiently engage with the underlying research, leading to suboptimal outcomes. Additionally, the existing cross-modal information fusion strategy does not ensure consistency of representations across different modes, potentially leading to the integration of conflicting information, which could degrade performance. To address these challenges, we propose the 'Recover then Aggregate' strategy and introduce the Unified Cross-Modal Deep Clustering model. Specifically, we have developed a data augmentation technique based on neighborhood similarity, iteratively imposing rank constraints on the Laplacian matrix, thus updating the similarity matrix and recovering dropout events. Concurrently, we integrate cross-modal features and employ contrastive learning to align modality-specific representations with consistent ones, enhancing the effective integration of diverse modal information. Comprehensive experiments on five real-world multi-modal datasets have demonstrated this method's superior effectiveness in single-cell clustering tasks.
Collapse
Affiliation(s)
- Ziyi Wang
- Department of Surgical Oncology and General Surgery, First Hospital of China Medical University, Shenyang 110001, PR China
- Section of Esophageal and Mediastinal Oncology, Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China
- Department of Thoracic Surgery, The First Hospital of China Medical University, No.155 North Nanjing Street, Shenyang 110001, People’s Republic of China
| | - Peng Luo
- Department of Thoracic Surgery, Xinqiao Hospital, Army Medical University, Chongqing 400038, China
| | - Mingming Xiao
- Department of Pathology, People’s Hospital of China Medical University (Liaoning Provincial People’s Hospital), Shenyang, Liaoning Province 110015, People’s Republic of China
| | - Boyang Wang
- Electrical and Computer Engineering, University of Illinois at Chicago, Chicago, IL 60607, United States
| | - Tianyu Liu
- Computer Science and Engineering, University of California, Riverside, Riverside, CA 92521, United States
| | - Xiangyu Sun
- Cancer Hospital of China Medical University, Liaoning Cancer Hospital and Institute, Shenyang 110042, Liaoning, China
- Cancer Hospital of Dalian University of Technology, Shenyang, Liaoning Province 110042, China
| |
Collapse
|
9
|
Xie L, Gong X, Yang K, Huang Y, Zhang S, Shen L, Sun Y, Wu D, Ye C, Zhu QH, Fan L. Technology-enabled great leap in deciphering plant genomes. NATURE PLANTS 2024; 10:551-566. [PMID: 38509222 DOI: 10.1038/s41477-024-01655-6] [Citation(s) in RCA: 20] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/22/2023] [Accepted: 02/20/2024] [Indexed: 03/22/2024]
Abstract
Plant genomes provide essential and vital basic resources for studying many aspects of plant biology and applications (for example, breeding). From 2000 to 2020, 1,144 genomes of 782 plant species were sequenced. In the past three years (2021-2023), 2,373 genomes of 1,031 plant species, including 793 newly sequenced species, have been assembled, representing a great leap. The 2,373 newly assembled genomes, of which 63 are telomere-to-telomere assemblies and 921 have been generated in pan-genome projects, cover the major phylogenetic clades. Substantial advances in read length, throughput, accuracy and cost-effectiveness have notably simplified the achievement of high-quality assemblies. Moreover, the development of multiple software tools using different algorithms offers the opportunity to generate more complete and complex assemblies. A database named N3: plants, genomes, technologies has been developed to accommodate the metadata associated with the 3,517 genomes that have been sequenced from 1,575 plant species since 2000. We also provide an outlook for emerging opportunities in plant genome sequencing.
Collapse
Affiliation(s)
- Lingjuan Xie
- Institute of Crop Sciences & Institute of Bioinformatics, Zhejiang University, Hangzhou, China
- Hainan Institute of Zhejiang University, Yazhou Bay, Shanya, China
| | - Xiaojiao Gong
- Institute of Crop Sciences & Institute of Bioinformatics, Zhejiang University, Hangzhou, China
| | - Kun Yang
- Institute of Crop Sciences & Institute of Bioinformatics, Zhejiang University, Hangzhou, China
| | - Yujie Huang
- Institute of Crop Sciences & Institute of Bioinformatics, Zhejiang University, Hangzhou, China
| | - Shiyu Zhang
- Institute of Crop Sciences & Institute of Bioinformatics, Zhejiang University, Hangzhou, China
| | - Leti Shen
- Hainan Institute of Zhejiang University, Yazhou Bay, Shanya, China
| | - Yanqing Sun
- Institute of Crop Sciences & Institute of Bioinformatics, Zhejiang University, Hangzhou, China
| | - Dongya Wu
- Institute of Crop Sciences & Institute of Bioinformatics, Zhejiang University, Hangzhou, China
| | - Chuyu Ye
- Institute of Crop Sciences & Institute of Bioinformatics, Zhejiang University, Hangzhou, China
| | - Qian-Hao Zhu
- CSIRO Agriculture and Food, Black Mountain Laboratories, Canberra, Australia
| | - Longjiang Fan
- Institute of Crop Sciences & Institute of Bioinformatics, Zhejiang University, Hangzhou, China.
- Hainan Institute of Zhejiang University, Yazhou Bay, Shanya, China.
| |
Collapse
|
10
|
Jogi HR, Smaraki N, Nayak SS, Rajawat D, Kamothi DJ, Panigrahi M. Single cell RNA-seq: a novel tool to unravel virus-host interplay. Virusdisease 2024; 35:41-54. [PMID: 38817399 PMCID: PMC11133279 DOI: 10.1007/s13337-024-00859-w] [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: 12/07/2023] [Accepted: 02/12/2024] [Indexed: 06/01/2024] Open
Abstract
Over the last decade, single cell RNA sequencing (scRNA-seq) technology has caught the momentum of being a vital revolutionary tool to unfold cellular heterogeneity by high resolution assessment. It evades the inadequacies of conventional sequencing technology which was able to detect only average expression level among cell populations. In the era of twenty-first century, several epidemic and pandemic viruses have emerged. Being an intracellular entity, viruses totally rely on host. Complex virus-host dynamics result when the virus tend to obtain factors from host cell required for its replication and establishment of infection. As a prevailing tool, scRNA-seq is able to understand virus-host interplay by comprehensive transcriptome profiling. Because of technological and methodological advancement, this technology is capable to recognize viral genome and host cell response heterogeneity. Further development in analytical methods with multiomics approach and increased availability of accessible scRNA-seq datasets will improve the understanding of viral pathogenesis that can be helpful for development of novel antiviral therapeutic strategies.
Collapse
Affiliation(s)
- Harsh Rajeshbhai Jogi
- Division of Veterinary Microbiology, Indian Veterinary Research Institute, Izatnagar, Bareilly, UP 243122 India
| | - Nabaneeta Smaraki
- Division of Veterinary Microbiology, Indian Veterinary Research Institute, Izatnagar, Bareilly, UP 243122 India
| | - Sonali Sonejita Nayak
- Division of Animal Genetics, ICAR-Indian Veterinary Research Institute, Izatnagar, Bareilly, UP 243122 India
| | - Divya Rajawat
- Division of Animal Genetics, ICAR-Indian Veterinary Research Institute, Izatnagar, Bareilly, UP 243122 India
| | - Dhaval J. Kamothi
- Division of Pharmacology and Toxicology, ICAR-Indian Veterinary Research Institute, Izatnagar, Bareilly, UP 243122 India
| | - Manjit Panigrahi
- Division of Animal Genetics, ICAR-Indian Veterinary Research Institute, Izatnagar, Bareilly, UP 243122 India
| |
Collapse
|
11
|
Chen X, Wu AR. Special Mini-Issue: Quantitative methods to decipher cellular heterogeneity - from single-cell to spatial omic methods. Biophys Rev 2024; 16:11-12. [PMID: 38495439 PMCID: PMC10937863 DOI: 10.1007/s12551-024-01180-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Accepted: 01/18/2024] [Indexed: 03/19/2024] Open
Abstract
In this mini-issue, we have a collection of eight reviews that discuss various advanced topics on the investigation of cellular heterogeneity. These reviews highlight the latest developments in technologies that capture and assess biology at single cell resolution, as well as approaches for cellular measurements with spatial information. Challenges and opportunities to develop future innovations and approaches are also presented.
Collapse
Affiliation(s)
- Xi Chen
- Department of Systems Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen, China
| | - Angela Ruohao Wu
- Division of Life Science, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong S.A.R China
- Department of Chemical and Biological Engineering, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong S.A.R China
| |
Collapse
|
12
|
Abstract
After introducing the winner of this year's Michèle Auger Award for Young Scientists' Independent Research, this Editorial for Volume 16 Issue 1 then describes the Issue contents. The Editorial concludes by providing a look into what lies ahead for 2024.
Collapse
Affiliation(s)
- Damien Hall
- WPI Nano Life Science Institute. Kanazawa University, Kakumamachi, Kanazawa, Ishikawa 920-1164 Japan
| |
Collapse
|
13
|
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
|