1
|
Zheng M, Bao N, Wang Z, Song C, Jin Y. Alternative splicing in autism spectrum disorder: Recent insights from mechanisms to therapy. Asian J Psychiatr 2025; 108:104501. [PMID: 40273800 DOI: 10.1016/j.ajp.2025.104501] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/19/2025] [Revised: 04/11/2025] [Accepted: 04/12/2025] [Indexed: 04/26/2025]
Abstract
Alternative splicing (AS) is a vital and highly dynamic RNA regulatory mechanism that allows a single gene to generate multiple mRNA and protein isoforms. Dysregulation of AS has been identified as a key contributor to the pathogenesis of autism spectrum disorders (ASD). A comprehensive understanding of aberrant splicing mechanisms and their functional consequences in ASD can help uncover the molecular basis of the disorder and facilitate the development of therapeutic strategies. This review focuses on the major aberrant splicing events and key splicing regulators associated with ASD, highlighting their roles in linking defective splicing to ASD pathogenesis. In addition, a discussion of how emerging technologies, such as long-read sequencing, single-cell sequencing, spatial transcriptomics and CRISPR-Cas systems are offering novel insights into the role and mechanisms of AS in ASD is presented. Finally, the RNA splicing-based therapeutic strategies are evaluated, emphasizing their potential to address unmet clinical needs in ASD treatment.
Collapse
Affiliation(s)
- Mixue Zheng
- Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China.
| | - Nengcheng Bao
- MOE Laboratory of Biosystems Homeostasis & Protection and Innovation Center for Cell Signaling Network, College of Life Sciences, Zhejiang University, Hangzhou 310058, China.
| | - Zhechao Wang
- MOE Laboratory of Biosystems Homeostasis & Protection and Innovation Center for Cell Signaling Network, College of Life Sciences, Zhejiang University, Hangzhou 310058, China; School of Life Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China.
| | - Chao Song
- Department of Developmental and Behavioral Pediatrics, the Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Centre for Child Health, Hangzhou 310052, China.
| | - Yongfeng Jin
- MOE Laboratory of Biosystems Homeostasis & Protection and Innovation Center for Cell Signaling Network, College of Life Sciences, Zhejiang University, Hangzhou 310058, China.
| |
Collapse
|
2
|
Singh M, Louie RHY, Samir J, Field MA, Milthorpe C, Adikari T, Mackie J, Roper E, Faulks M, Jackson KJL, Calcino A, Hardy MY, Blombery P, Amos TG, Deveson IW, Wende HV, Floor SN, Read SA, Shek D, Guerin A, Ma CS, Tangye SG, Di Sabatino A, Lenti MV, Pasini A, Ciccocioppo R, Ahlenstiel G, Suan D, Tye-Din JA, Goodnow CC, Luciani F. Expanded T cell clones with lymphoma driver somatic mutations accumulate in refractory celiac disease. Sci Transl Med 2025; 17:eadp6812. [PMID: 40367192 DOI: 10.1126/scitranslmed.adp6812] [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: 04/07/2024] [Accepted: 03/31/2025] [Indexed: 05/16/2025]
Abstract
Intestinal inflammation continues in a subset of patients with celiac disease despite a gluten-free diet. Here, by applying multi-omic single-cell analysis to duodenal biopsies, we found that low-grade malignancies with lymphoma driver mutations in patients with refractory celiac disease type 2 (RCD2) are comprised by surface CD3-negative (sCD3-) lymphocytes stalled at an innate lymphoid cell (ILC)-progenitor T cell stage undergoing extensive TRA, TRB, and TRD TCR recombination. In people with refractory celiac disease type 1 (RCD1), a disease currently lacking explanation, we identified sCD3+ T cells with lymphoma driver mutations in 6 of 10 individuals with RCD1 and in one of the patients with active, recently diagnosed celiac disease. Furthermore, the mutant T cells formed large TCRαβ clones and displayed inflammatory and cytotoxic molecular profiles. Thus, accumulation of lymphoma driver-mutated T cells and sCD3- progenitors may contribute to chronic, nonresponsive celiac disease.
Collapse
Affiliation(s)
- Mandeep Singh
- Garvan Institute of Medical Research, Darlinghurst, NSW 2010, Australia
- School of Clinical Medicine, Faculty of Medicine & Health, UNSW Sydney, Sydney, NSW 2052, Australia
| | - Raymond H Y Louie
- School of Computer Science and Engineering, UNSW Sydney, Sydney, NSW 2052, Australia
- School of Medical Sciences, UNSW Sydney, Sydney, NSW 2052, Australia
| | - Jerome Samir
- School of Medical Sciences, UNSW Sydney, Sydney, NSW 2052, Australia
| | - Matthew A Field
- Garvan Institute of Medical Research, Darlinghurst, NSW 2010, Australia
- Australian Institute of Tropical Health and Medicine and Centre for Tropical Bioinformatics and Molecular Biology, James Cook University, Smithfield, QLD 4878, Australia
| | - Claire Milthorpe
- Garvan Institute of Medical Research, Darlinghurst, NSW 2010, Australia
| | - Thiruni Adikari
- School of Medical Sciences, UNSW Sydney, Sydney, NSW 2052, Australia
| | - Joseph Mackie
- Garvan Institute of Medical Research, Darlinghurst, NSW 2010, Australia
- School of Clinical Medicine, Faculty of Medicine & Health, UNSW Sydney, Sydney, NSW 2052, Australia
| | - Ellise Roper
- Garvan Institute of Medical Research, Darlinghurst, NSW 2010, Australia
| | - Megan Faulks
- Garvan Institute of Medical Research, Darlinghurst, NSW 2010, Australia
| | | | - Andrew Calcino
- Australian Institute of Tropical Health and Medicine and Centre for Tropical Bioinformatics and Molecular Biology, James Cook University, Smithfield, QLD 4878, Australia
| | - Melinda Y Hardy
- Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia
- Department of Medical Biology, University of Melbourne, Parkville, VIC 3010, Australia
| | - Piers Blombery
- Peter MacCallum Cancer Centre and University of Melbourne, Melbourne, VIC 3000, Australia
- University of Melbourne, Melbourne, VIC 3010, Australia
| | - Timothy G Amos
- Garvan Institute of Medical Research, Darlinghurst, NSW 2010, Australia
| | - Ira W Deveson
- Garvan Institute of Medical Research, Darlinghurst, NSW 2010, Australia
- School of Clinical Medicine, Faculty of Medicine & Health, UNSW Sydney, Sydney, NSW 2052, Australia
| | - Helen Vander Wende
- Department of Cell and Tissue Biology, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Stephen N Floor
- Department of Cell and Tissue Biology, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Scott A Read
- Westmead Institute for Medical Research, University of Sydney, Westmead, NSW 2145, Australia
- Blacktown Medical School, Western Sydney University, Blacktown, NSW 2148, Australia
- Blacktown Hospital, Blacktown, NSW 2148, Australia
| | - Dmitri Shek
- Westmead Institute for Medical Research, University of Sydney, Westmead, NSW 2145, Australia
- Blacktown Medical School, Western Sydney University, Blacktown, NSW 2148, Australia
- Blacktown Hospital, Blacktown, NSW 2148, Australia
| | - Antoine Guerin
- Garvan Institute of Medical Research, Darlinghurst, NSW 2010, Australia
- School of Clinical Medicine, Faculty of Medicine & Health, UNSW Sydney, Sydney, NSW 2052, Australia
| | - Cindy S Ma
- Garvan Institute of Medical Research, Darlinghurst, NSW 2010, Australia
- School of Clinical Medicine, Faculty of Medicine & Health, UNSW Sydney, Sydney, NSW 2052, Australia
| | - Stuart G Tangye
- Garvan Institute of Medical Research, Darlinghurst, NSW 2010, Australia
- School of Clinical Medicine, Faculty of Medicine & Health, UNSW Sydney, Sydney, NSW 2052, Australia
| | - Antonio Di Sabatino
- Department of Internal Medicine and Medical Therapeutics, University of Pavia, Pavia 27100, Italy
- First Department of Internal Medicine, Fondazione IRCCS Policlinico San Matteo, Pavia 27100, Italy
| | - Marco V Lenti
- Department of Internal Medicine and Medical Therapeutics, University of Pavia, Pavia 27100, Italy
- First Department of Internal Medicine, Fondazione IRCCS Policlinico San Matteo, Pavia 27100, Italy
| | - Alessandra Pasini
- Department of Internal Medicine and Medical Therapeutics, University of Pavia, Pavia 27100, Italy
- First Department of Internal Medicine, Fondazione IRCCS Policlinico San Matteo, Pavia 27100, Italy
| | - Rachele Ciccocioppo
- Gastroenterology Unit, Department of Medicine, University of Verona and AOUI Verona, Policlinico GB Rossi, Verona 37134, Italy
| | - Golo Ahlenstiel
- Westmead Institute for Medical Research, University of Sydney, Westmead, NSW 2145, Australia
- Blacktown Medical School, Western Sydney University, Blacktown, NSW 2148, Australia
- Blacktown Hospital, Blacktown, NSW 2148, Australia
| | - Dan Suan
- Garvan Institute of Medical Research, Darlinghurst, NSW 2010, Australia
- School of Clinical Medicine, Faculty of Medicine & Health, UNSW Sydney, Sydney, NSW 2052, Australia
| | - Jason A Tye-Din
- Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia
- Gastroenterology Department, Royal Melbourne Hospital, Parkville, VIC 3050, Australia
| | - Christopher C Goodnow
- Garvan Institute of Medical Research, Darlinghurst, NSW 2010, Australia
- Cellular Genomics Futures Institute and School of Biomedical Sciences, UNSW Sydney, Sydney, NSW 2052, Australia
| | - Fabio Luciani
- Garvan Institute of Medical Research, Darlinghurst, NSW 2010, Australia
- School of Medical Sciences, UNSW Sydney, Sydney, NSW 2052, Australia
| |
Collapse
|
3
|
Sutanto H, Pradana FR, Adytia GJ, Ansharullah BA, Waitupu A, Bramantono B, Fetarayani D. Memory T Cells in Respiratory Virus Infections: Protective Potential and Persistent Vulnerabilities. Med Sci (Basel) 2025; 13:48. [PMID: 40407543 PMCID: PMC12101432 DOI: 10.3390/medsci13020048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2025] [Revised: 04/19/2025] [Accepted: 04/25/2025] [Indexed: 05/26/2025] Open
Abstract
Respiratory virus infections, such as those caused by influenza viruses, respiratory syncytial virus (RSV), and coronaviruses, pose a significant global health burden. While the immune system's adaptive components, including memory T cells, are critical for recognizing and combating these pathogens, recurrent infections and variable disease outcomes persist. Memory T cells are a key element of long-term immunity, capable of responding swiftly upon re-exposure to pathogens. They play diverse roles, including cross-reactivity to conserved viral epitopes and modulation of inflammatory responses. However, the protective efficacy of these cells is influenced by several factors, including viral evolution, host age, and immune system dynamics. This review explores the dichotomy of memory T cells in respiratory virus infections: their potential to confer robust protection and the limitations that allow for breakthrough infections. Understanding the underlying mechanisms governing the formation, maintenance, and functional deployment of memory T cells in respiratory mucosa is critical for improving immunological interventions. We highlight recent advances in vaccine strategies aimed at bolstering T cell-mediated immunity and discuss the challenges posed by viral immune evasion. Addressing these gaps in knowledge is pivotal for designing effective therapeutics and vaccines to mitigate the global burden of respiratory viruses.
Collapse
Affiliation(s)
- Henry Sutanto
- Internal Medicine Study Program, Department of Internal Medicine, Faculty of Medicine, Universitas Airlangga, Surabaya 60132, Indonesia; (H.S.); (F.R.P.); (G.J.A.); (B.A.A.); (A.W.)
- Department of Internal Medicine, Dr. Soetomo General Academic Hospital, Surabaya 60286, Indonesia
| | - Febrian Ramadhan Pradana
- Internal Medicine Study Program, Department of Internal Medicine, Faculty of Medicine, Universitas Airlangga, Surabaya 60132, Indonesia; (H.S.); (F.R.P.); (G.J.A.); (B.A.A.); (A.W.)
- Department of Internal Medicine, Dr. Soetomo General Academic Hospital, Surabaya 60286, Indonesia
| | - Galih Januar Adytia
- Internal Medicine Study Program, Department of Internal Medicine, Faculty of Medicine, Universitas Airlangga, Surabaya 60132, Indonesia; (H.S.); (F.R.P.); (G.J.A.); (B.A.A.); (A.W.)
- Department of Internal Medicine, Dr. Soetomo General Academic Hospital, Surabaya 60286, Indonesia
| | - Bagus Aditya Ansharullah
- Internal Medicine Study Program, Department of Internal Medicine, Faculty of Medicine, Universitas Airlangga, Surabaya 60132, Indonesia; (H.S.); (F.R.P.); (G.J.A.); (B.A.A.); (A.W.)
- Department of Internal Medicine, Dr. Soetomo General Academic Hospital, Surabaya 60286, Indonesia
| | - Alief Waitupu
- Internal Medicine Study Program, Department of Internal Medicine, Faculty of Medicine, Universitas Airlangga, Surabaya 60132, Indonesia; (H.S.); (F.R.P.); (G.J.A.); (B.A.A.); (A.W.)
- Department of Internal Medicine, Dr. Soetomo General Academic Hospital, Surabaya 60286, Indonesia
| | - Bramantono Bramantono
- Department of Internal Medicine, Dr. Soetomo General Academic Hospital, Surabaya 60286, Indonesia
- Division of Tropical and Infectious Diseases, Department of Internal Medicine, Faculty of Medicine, Universitas Airlangga, Surabaya 60132, Indonesia
| | - Deasy Fetarayani
- Department of Internal Medicine, Dr. Soetomo General Academic Hospital, Surabaya 60286, Indonesia
- Division of Allergy and Clinical Immunology, Department of Internal Medicine, Faculty of Medicine, Universitas Airlangga, Surabaya 60132, Indonesia
| |
Collapse
|
4
|
Beaulaurier J, Ly L, Duty JA, Tyer C, Stevens C, Hung CT, Sookdeo A, Drong AW, Kowdle S, Guzman-Solis A, Tortorella D, Turner DJ, Juul S, Hickey S, Lee B. De novo antibody identification in human blood from full-length single B cell transcriptomics and matching haplotype-resolved germline assemblies. Genome Res 2025; 35:929-941. [PMID: 40118521 PMCID: PMC12047243 DOI: 10.1101/gr.279392.124] [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: 03/21/2024] [Accepted: 02/12/2025] [Indexed: 03/23/2025]
Abstract
Immunoglobulin (IGH, IGK, IGL) loci in the human genome are highly polymorphic regions that encode the building blocks of the light and heavy chain IG proteins that dimerize to form antibodies. The processes of V(D)J recombination and somatic hypermutation in B cells are responsible for creating an enormous reservoir of highly specific antibodies capable of binding a vast array of possible antigens. However, the antibody repertoire is fundamentally limited by the set of variable (V), diversity (D), and joining (J) alleles present in the germline IG loci. To better understand how the germline IG haplotypes contribute to the expressed antibody repertoire, we combined genome sequencing of the germline IG loci with single-cell transcriptome sequencing of B cells from the same donor. Sequencing and assembly of the germline IG loci captured the IGH locus in a single fully phased contig where the maternal and paternal contributions to the germline V, D, and J repertoire can be fully resolved. The B cells were collected following a measles, mumps, and rubella (MMR) vaccination, resulting in a population of cells that were activated in response to this specific immune challenge. Single-cell, full-length transcriptome sequencing of these B cells results in whole transcriptome characterization of each cell, as well as highly accurate consensus sequences for the somatically rearranged and hypermutated light and heavy chain IG transcripts. A subset of antibodies synthesized based on their consensus heavy and light chain transcript sequences demonstrate binding to measles antigens and neutralization of authentic measles virus.
Collapse
Affiliation(s)
- John Beaulaurier
- Oxford Nanopore Technologies, Inc., New York, New York 10013, USA
| | - Lynn Ly
- Oxford Nanopore Technologies, Inc., New York, New York 10013, USA
| | - J Andrew Duty
- Icahn School of Medicine at Mount Sinai, New York, New York 10029, USA
| | - Carly Tyer
- Oxford Nanopore Technologies, Inc., New York, New York 10013, USA
| | - Christian Stevens
- Icahn School of Medicine at Mount Sinai, New York, New York 10029, USA
| | - Chuan-Tien Hung
- Icahn School of Medicine at Mount Sinai, New York, New York 10029, USA
| | - Akash Sookdeo
- Oxford Nanopore Technologies, Inc., New York, New York 10013, USA
| | - Alex W Drong
- Oxford Nanopore Technologies, Inc., New York, New York 10013, USA
| | - Shreyas Kowdle
- Icahn School of Medicine at Mount Sinai, New York, New York 10029, USA
| | - Axel Guzman-Solis
- Icahn School of Medicine at Mount Sinai, New York, New York 10029, USA
| | | | - Daniel J Turner
- Oxford Nanopore Technologies, Inc., New York, New York 10013, USA
| | - Sissel Juul
- Oxford Nanopore Technologies, Inc., New York, New York 10013, USA
| | - Scott Hickey
- Oxford Nanopore Technologies, Inc., New York, New York 10013, USA;
| | - Benhur Lee
- Icahn School of Medicine at Mount Sinai, New York, New York 10029, USA
| |
Collapse
|
5
|
Monzó C, Liu T, Conesa A. Transcriptomics in the era of long-read sequencing. Nat Rev Genet 2025:10.1038/s41576-025-00828-z. [PMID: 40155769 DOI: 10.1038/s41576-025-00828-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/20/2025] [Indexed: 04/01/2025]
Abstract
Transcriptome sequencing revolutionized the analysis of gene expression, providing an unbiased approach to gene detection and quantification that enabled the discovery of novel isoforms, alternative splicing events and fusion transcripts. However, although short-read sequencing technologies have surpassed the limited dynamic range of previous technologies such as microarrays, they have limitations, for example, in resolving full-length transcripts and complex isoforms. Over the past 5 years, long-read sequencing technologies have matured considerably, with improvements in instrumentation and analytical methods, enabling their application to RNA sequencing (RNA-seq). Benchmarking studies are beginning to identify the strengths and limitations of long-read RNA-seq, although there remains a need for comprehensive resources to guide newcomers through the intricacies of this approach. In this Review, we provide a comprehensive overview of the long-read RNA-seq workflow, from library preparation and sequencing challenges to core data processing, downstream analyses and emerging developments. We present an extensive inventory of experimental and analytical methods and discuss current challenges and prospects.
Collapse
Affiliation(s)
- Carolina Monzó
- Institute for Integrative Systems Biology, Spanish National Research Council, Paterna, Valencia, Spain.
| | - Tianyuan Liu
- Institute for Integrative Systems Biology, Spanish National Research Council, Paterna, Valencia, Spain
| | - Ana Conesa
- Institute for Integrative Systems Biology, Spanish National Research Council, Paterna, Valencia, Spain.
| |
Collapse
|
6
|
Li Y, Liu Y, Xie Y, Wang Y, Wang J, Wang H, Xia L, Xie D. Long-read RNA sequencing enables full-length chimeric transcript annotation of transposable elements in lung adenocarcinoma. BMC Cancer 2025; 25:482. [PMID: 40089719 PMCID: PMC11909889 DOI: 10.1186/s12885-025-13888-5] [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: 05/29/2024] [Accepted: 03/07/2025] [Indexed: 03/17/2025] Open
Abstract
BACKGROUND Transposable elements (TEs), which constitute nearly half of the human genome, have long been regarded as genomic "dark matter". However, their reactivation in tumor cells, resulting in the production of TE-chimeric transcripts (TCTs), has emerged as a potential driver of cancer progression. The complexity and full extent of these transcripts remain elusive, largely due to the limitations of short-read next-generation sequencing technologies. These methods have struggled to comprehensively capture the diversity and structure of TCTs, particularly those involving short interspersed nuclear elements (SINEs) or closely co-transcribed TEs. METHODS Leveraging full-length cDNA sequencing technology based on nanopore sequencing platform, we developed a customized pipeline for identifying and quantifying TCTs in 19 lung adenocarcinoma (LUAD) cell lines. The short-read RNA-seq dataset from a LUAD corhort (~ 200 tumor samples) was employed to validate the identified TCTs and explore their association with tumor progression. To assess the functional roles of a specific TCTs, cell migration and cell proliferation assays were performed. RESULTS We uncovered 208 unique TCT candidates in the LUAD cell lines. Our approach allowed for the identification of cryptic promoters and terminators within non-transposing TEs. Notably, we identified a chimeric transcript involving MIR_HKDC1, which appears to play a significant role in the progression of LUAD. Furthermore, the expression of these TCTs were associated with poor clinical outcomes in a cohort of LUAD patients, suggesting their potential as novel biomarkers for both LUAD progression and prognosis. CONCLUSIONS Our study underscores the application of long-read sequencing to unravel the complex landscape of TCTs in LUAD. We provide a comprehensive characterization of TCTs in LUAD, exploring their potential regulatory roles in cancer progression. These findings contribute to a deeper understanding of the genomic intricacies underlying cancer, and offer new directions for the development of targeted therapies and personalized treatment strategies for LUAD. This research highlights the potential of TCTs as both biomarkers and therapeutic targets in the oncogenesis, offering new insights into the interplay between transposable elements and gene regulation in cancer.
Collapse
Affiliation(s)
- Yang Li
- Laboratory of Omics Technology and Bioinformatics, Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Yahui Liu
- Laboratory of Omics Technology and Bioinformatics, Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Yingxin Xie
- West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Yaxuan Wang
- West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, 610041, China
| | - Jing Wang
- Laboratory of Omics Technology and Bioinformatics, Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Huan Wang
- Laboratory of Omics Technology and Bioinformatics, Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Lin Xia
- Laboratory of Omics Technology and Bioinformatics, Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China.
| | - Dan Xie
- Laboratory of Omics Technology and Bioinformatics, Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China.
| |
Collapse
|
7
|
Abdullah L, Emiliani FE, Vaidya CM, Stuart H, Musial SC, Kolling FW, Obar JJ, Rosato PC, Ackerman ME, Song L, McKenna A, Huang YH. The endogenous antigen-specific CD8 + T cell repertoire is composed of unbiased and biased clonotypes with differential fate commitments. Immunity 2025; 58:601-615.e9. [PMID: 40020673 PMCID: PMC11903169 DOI: 10.1016/j.immuni.2025.02.001] [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: 09/05/2023] [Revised: 07/24/2024] [Accepted: 02/03/2025] [Indexed: 03/03/2025]
Abstract
Generating balanced populations of CD8+ effector and memory T cells is necessary for immediate and durable immunity to infections and cancer. Yet, a definitive understanding of how a diverse CD8+ T cell repertoire differentiates remains unclear. We identified several hundred T cell receptor (TCR) clonotypes that constitute the polyclonal response against a single antigen and found that a majority of TCR clonotypes were highly biased toward memory or effector fates. TCR-intrinsic biases were not stochastic and were dominant over environmental cues. Differential gene expression analysis of memory- or effector-biased TCR clonotypes showed bifurcation of differential fates at the early effector stage. Additionally, phylogenetic analysis revealed that memory-biased clonotypes retain their fate preferences in subclonal populations but effector-biased subclones can switch to a memory fate. Our study highlights that the polyclonal CD8+ T cell response is a composite of unbiased and biased clonotypes with varying capacity to incorporate environmental cues in their cell fate decisions.
Collapse
Affiliation(s)
- Leena Abdullah
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Lebanon, NH 03756, USA
| | - Francesco E Emiliani
- Department of Molecular and Systems Biology, Geisel School of Medicine at Dartmouth, Lebanon, NH 03756, USA
| | - Chinmay M Vaidya
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Lebanon, NH 03756, USA; Thayer School of Engineering, Dartmouth College, Hanover, NH 03755, USA
| | - Hannah Stuart
- Department of Molecular and Systems Biology, Geisel School of Medicine at Dartmouth, Lebanon, NH 03756, USA
| | - Shawn C Musial
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Lebanon, NH 03756, USA
| | | | - Joshua J Obar
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Lebanon, NH 03756, USA
| | - Pamela C Rosato
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Lebanon, NH 03756, USA; Dartmouth Cancer Center, Lebanon, NH 03756, USA
| | - Margaret E Ackerman
- Thayer School of Engineering, Dartmouth College, Hanover, NH 03755, USA; Dartmouth Cancer Center, Lebanon, NH 03756, USA
| | - Li Song
- Department of Biomedical Data Science, Geisel School of Medicine at Dartmouth, Lebanon, NH 03756, USA; Dartmouth Cancer Center, Lebanon, NH 03756, USA
| | - Aaron McKenna
- Department of Molecular and Systems Biology, Geisel School of Medicine at Dartmouth, Lebanon, NH 03756, USA; Dartmouth Cancer Center, Lebanon, NH 03756, USA
| | - Yina H Huang
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Lebanon, NH 03756, USA; Dartmouth Cancer Center, Lebanon, NH 03756, USA; Department of Pathology and Laboratory Medicine, Dartmouth Health, Lebanon, NH 03756, USA.
| |
Collapse
|
8
|
Young C, Singh M, Jackson KJL, Field MA, Peters TJ, Angioletti-Uberti S, Frenkel D, Ravishankar S, Gupta M, Wang JJ, Agapiou D, Faulks ML, Al-Eryani G, Luciani F, Gordon TP, Reed JH, Danta M, Carr A, Kelleher AD, Dore GJ, Matthews G, Brink R, Bull RA, Suan D, Goodnow CC. A triad of somatic mutagenesis converges in self-reactive B cells to cause a virus-induced autoimmune disease. Immunity 2025; 58:412-430.e10. [PMID: 39818208 DOI: 10.1016/j.immuni.2024.12.011] [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: 01/30/2024] [Revised: 09/22/2024] [Accepted: 12/18/2024] [Indexed: 01/18/2025]
Abstract
The unexplained association between infection and autoimmune disease is strongest for hepatitis C virus-induced cryoglobulinemic vasculitis (HCV-cryovas). To analyze its origins, we traced the evolution of pathogenic rheumatoid factor (RF) autoantibodies in four HCV-cryovas patients by deep single-cell multi-omic analysis, revealing three sources of B cell somatic mutation converged to drive the accumulation of a large disease-causing clone. A method for quantifying low-affinity binding revealed recurring antibody variable domain combinations created by V(D)J recombination that bound self-immunoglobulin G (IgG) but not viral E2 antigen. Whole-genome sequencing revealed thousands of somatic mutations, numerically comparable to chronic lymphocytic leukemia and normal memory B cells, but with 1-2 corresponding to driver mutations found recurrently in B cell leukemia and lymphoma. V(D)J hypermutation created autoantibodies with compromised solubility in complex with self-IgG. In this virus-induced autoimmune disease, infection promotes a catastrophic confluence of somatic mutagenesis in the descendants of a single B cell.
Collapse
Affiliation(s)
- Clara Young
- Garvan Institute of Medical Research, Darlinghurst, NSW, Australia; St Vincent's Clinical School, UNSW Sydney, Sydney, NSW, Australia
| | - Mandeep Singh
- Garvan Institute of Medical Research, Darlinghurst, NSW, Australia; St Vincent's Clinical School, UNSW Sydney, Sydney, NSW, Australia
| | | | - Matt A Field
- Garvan Institute of Medical Research, Darlinghurst, NSW, Australia; Australian Institute of Tropical Health and Medicine and Centre for Tropical Bioinformatics and Molecular Biology, Smithfield, Cairns, QLD, Australia; Menzies School of Health Research, Darwin, NT, Australia
| | - Timothy J Peters
- Garvan Institute of Medical Research, Darlinghurst, NSW, Australia; St Vincent's Clinical School, UNSW Sydney, Sydney, NSW, Australia
| | | | - Daan Frenkel
- Yusuf Hamied Department of Chemistry, University of Cambridge, Cambridge, UK
| | | | - Money Gupta
- School of Biomedical Sciences, UNSW Sydney, Sydney, NSW, Australia; The Kirby Institute, UNSW Sydney, Sydney, NSW, Australia
| | - Jing J Wang
- Department of Immunology, Flinders University and SA Pathology, Bedford Park, Adelaide, SA, Australia
| | - David Agapiou
- The Kirby Institute, UNSW Sydney, Sydney, NSW, Australia
| | - Megan L Faulks
- Garvan Institute of Medical Research, Darlinghurst, NSW, Australia
| | | | - Fabio Luciani
- School of Biomedical Sciences, UNSW Sydney, Sydney, NSW, Australia; The Kirby Institute, UNSW Sydney, Sydney, NSW, Australia
| | - Tom P Gordon
- Department of Immunology, Flinders University and SA Pathology, Bedford Park, Adelaide, SA, Australia
| | - Joanne H Reed
- Garvan Institute of Medical Research, Darlinghurst, NSW, Australia; Westmead Institute for Medical Research, Westmead, Sydney, NSW, Australia
| | - Mark Danta
- St Vincent's Clinical School, UNSW Sydney, Sydney, NSW, Australia
| | - Andrew Carr
- Immunology and HIV Unit, St Vincent's Hospital, Sydney, NSW, Australia
| | - Anthony D Kelleher
- The Kirby Institute, UNSW Sydney, Sydney, NSW, Australia; Immunology and HIV Unit, St Vincent's Hospital, Sydney, NSW, Australia
| | - Gregory J Dore
- St Vincent's Clinical School, UNSW Sydney, Sydney, NSW, Australia; The Kirby Institute, UNSW Sydney, Sydney, NSW, Australia
| | - Gail Matthews
- St Vincent's Clinical School, UNSW Sydney, Sydney, NSW, Australia; The Kirby Institute, UNSW Sydney, Sydney, NSW, Australia
| | - Robert Brink
- Garvan Institute of Medical Research, Darlinghurst, NSW, Australia; St Vincent's Clinical School, UNSW Sydney, Sydney, NSW, Australia
| | - Rowena A Bull
- School of Biomedical Sciences, UNSW Sydney, Sydney, NSW, Australia; The Kirby Institute, UNSW Sydney, Sydney, NSW, Australia
| | - Dan Suan
- Garvan Institute of Medical Research, Darlinghurst, NSW, Australia; St Vincent's Clinical School, UNSW Sydney, Sydney, NSW, Australia.
| | - Christopher C Goodnow
- Garvan Institute of Medical Research, Darlinghurst, NSW, Australia; St Vincent's Clinical School, UNSW Sydney, Sydney, NSW, Australia.
| |
Collapse
|
9
|
Xu Z, Qu HQ, Chan J, Mu S, Kao C, Hakonarson H, Wang K. Single-Cell Omics for Transcriptome CHaracterization (SCOTCH): isoform-level characterization of gene expression through long-read single-cell RNA sequencing. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.04.29.590597. [PMID: 38746128 PMCID: PMC11092450 DOI: 10.1101/2024.04.29.590597] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/16/2024]
Abstract
Recent development involving long-read single-cell transcriptome sequencing (lr-scRNA-Seq) represents a significant leap forward in single-cell genomics. With the recent introduction of R10 flowcells by Oxford Nanopore, we propose that previous computational methods designed to handle high sequencing error rates are less relevant, and that the traditional approach using short reads to compile "barcode space" (candidate barcode list) to de-multiplex long reads are no longer necessary. Instead, computational methods should now shift focus on harnessing the unique benefits of long reads to analyze transcriptome complexity. In this context, we introduce a comprehensive suite of computational methods named Single-Cell Omics for Transcriptome CHaracterization (SCOTCH). SCOTCH supports both Nanopore and PacBio sequencing platforms, and is compatible with single-cell library preparation protocols from both 10X Genomics and Parse Biosciences. Through a sub-exon identification strategy with dynamic thresholding and read mapping scores, SCOTCH precisely aligns reads to known isoforms and discover novel isoforms, efficiently addressing ambiguous mapping challenges commonly encountered in long-read single-cell data. Comprehensive simulations and real data analyses across multiple platforms (including 10X Genomics and Parse Bioscience, paired with Illumina or Nanopore sequencing technologies with R9 and R10 flowcells, as well as PacBio sequencing) demonstrated that SCOTCH outperforms existing methods in mapping accuracy, quantification accuracy and novel isoform detection, while also uncovering novel biological insights on transcriptome complexity at the single-cell level.
Collapse
Affiliation(s)
- Zhuoran Xu
- Graduate Group in Genomics and Computational Biology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104, USA
- Raymond G. Perelman Center for Cellular and Molecular Therapeutics, Children’s Hospital of Philadelphia, Philadelphia, PA, 19104, USA
| | - Hui-Qi Qu
- The Center for Applied Genomics, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania, 19104, USA
| | - Joe Chan
- Raymond G. Perelman Center for Cellular and Molecular Therapeutics, Children’s Hospital of Philadelphia, Philadelphia, PA, 19104, USA
| | - Shizhuo Mu
- Raymond G. Perelman Center for Cellular and Molecular Therapeutics, Children’s Hospital of Philadelphia, Philadelphia, PA, 19104, USA
| | - Charlly Kao
- The Center for Applied Genomics, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania, 19104, USA
| | - Hakon Hakonarson
- The Center for Applied Genomics, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania, 19104, USA
- Department of Pediatrics, The Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, 19104, USA
| | - Kai Wang
- Raymond G. Perelman Center for Cellular and Molecular Therapeutics, Children’s Hospital of Philadelphia, Philadelphia, PA, 19104, USA
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| |
Collapse
|
10
|
Woolley CR, Chariker JH, Rouchka EC, Ford EE, Hudson E, Rasche KM, Whitley CS, Vanwinkle Z, Casella CR, Smith ML, Mitchell TC. Full-length mRNA sequencing resolves novel variation in 5' UTR length for genes expressed during human CD4 T-cell activation. Immunogenetics 2025; 77:14. [PMID: 39904916 PMCID: PMC11794378 DOI: 10.1007/s00251-025-01371-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2024] [Accepted: 01/23/2025] [Indexed: 02/06/2025]
Abstract
Isoform sequencing (Iso-Seq) uses long-read technology to produce highly accurate full-length reads of mRNA transcripts. Visualization of individual mRNA molecules can reveal new details of transcript variation within understudied portions of mRNA, such as the 5' untranslated region (UTR). Differential 5' UTRs may contain motifs, upstream open reading frames (uORFs), and secondary structures that can serve to regulate translation or further indicate changes in promoter usage, where transcriptional control may impact protein expression levels. To begin to explore isoform variation during T-cell activation, we generated the first Iso-Seq reference transcriptome of activated human CD4 T cells. Within this dataset, we discovered many novel splice- and end-variant transcripts. Remarkably, one in every eight genes expressed in our dataset was found to have a notable proportion of transcripts with 5' UTR lengthened by over 100 bp compared to the longest corresponding UTR within the Gencode dataset. Among these end-variant transcripts, two novel isoforms were identified for CXCR5, a chemokine receptor associated with T follicular helper cell (Tfh) function and differentiation. When investigated in a model cell system, these lengthened UTR conferred reduced transcript stability and, for one of these isoforms, short uORFs introduced by the added length altered protein expression kinetics. This study highlights instances in which current reference databases are incomplete relative to the information obtained by long-read sequencing of intact mRNA. Iso-Seq is thus a promising approach to better understanding the plasticity of promoter usage, alternative splicing, and UTR sequences that influence RNA stability and translation efficiency.
Collapse
Affiliation(s)
- Cassandra R Woolley
- Department of Microbiology and Immunology, University of Louisville School of Medicine, Louisville, KY, USA
| | - Julia H Chariker
- Department of Neuroscience Training, University of Louisville School of Medicine, KY, Louisville, USA
| | - Eric C Rouchka
- Department of Biochemistry and Molecular Genetics, University of Louisville School of Medicine, Louisville, KY, USA
- KY INBRE Bioinformatics Core, University of Louisville School of Medicine, Louisville, KY, USA
| | - Easton E Ford
- Department of Biochemistry and Molecular Genetics, University of Louisville School of Medicine, Louisville, KY, USA
| | - Elizabeth Hudson
- Department of Biochemistry and Molecular Genetics, University of Louisville School of Medicine, Louisville, KY, USA
| | - Kamille M Rasche
- Department of Microbiology and Immunology, University of Louisville School of Medicine, Louisville, KY, USA
| | - Caleb S Whitley
- Department of Microbiology and Immunology, University of Louisville School of Medicine, Louisville, KY, USA
| | - Zachary Vanwinkle
- Department of Biochemistry and Molecular Genetics, University of Louisville School of Medicine, Louisville, KY, USA
| | - Carolyn R Casella
- Department of Microbiology and Immunology, University of Louisville School of Medicine, Louisville, KY, USA
| | - Melissa L Smith
- Department of Biochemistry and Molecular Genetics, University of Louisville School of Medicine, Louisville, KY, USA
| | - Thomas C Mitchell
- Department of Microbiology and Immunology, University of Louisville School of Medicine, Louisville, KY, USA.
| |
Collapse
|
11
|
Smith MA. Evolving Perspectives on Immune Repertoire Profiling: Challenges and Opportunities in the Era of Long-Read Sequencing. Clin Chem 2025; 71:232-234. [PMID: 39723649 DOI: 10.1093/clinchem/hvae219] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2024] [Accepted: 11/26/2024] [Indexed: 12/28/2024]
Affiliation(s)
- Martin A Smith
- Ramaciotti Centre for Genomics, School of Biotechnology and Biomolecular Sciences, Faculty of Science, UNSW Sydney, Kensington, Australia
- RNA Institute, UNSW Sydney, Kensington, Australia
- Department of Biochemistry and Molecular Medicine, Faculty of Medicine, Université de Montréal, Montreal, Canada
- Cancer and Immune Diseases Axis, CHU Sainte-Justine Research Centre, Montreal, Canada
| |
Collapse
|
12
|
Goss K, Horwitz EM. Single-cell multiomics to advance cell therapy. Cytotherapy 2025; 27:137-145. [PMID: 39530970 DOI: 10.1016/j.jcyt.2024.10.009] [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: 09/13/2024] [Revised: 10/21/2024] [Accepted: 10/21/2024] [Indexed: 11/16/2024]
Abstract
Single-cell RNA-sequencing (scRNAseq) was first introduced in 2009 and has evolved with many technological advancements over the last decade. Not only are there several scRNAseq platforms differing in many aspects, but there are also a large number of computational pipelines available for downstream analyses which are being developed at an exponential rate. Such computational data appear in many scientific publications in virtually every field of study; thus, investigators should be able to understand and interpret data in this rapidly evolving field. Here, we discuss key differences in scRNAseq platforms, crucial steps in scRNAseq experiments, standard downstream analyses and introduce newly developed multimodal approaches. We then discuss how single-cell omics has been applied to advance the field of cell therapy.
Collapse
Affiliation(s)
- Kyndal Goss
- Marcus Center for Advanced Cellular Therapy, Children's Healthcare of Atlanta, Atlanta, Georgia, USA; Aflac Cancer & Blood Disorders Center, Children's Healthcare of Atlanta, Atlanta, Georgia, USA; Graduate Division of Biology and Biomedical Sciences, Emory University Laney Graduate School, Atlanta, Georgia, USA
| | - Edwin M Horwitz
- Marcus Center for Advanced Cellular Therapy, Children's Healthcare of Atlanta, Atlanta, Georgia, USA; Aflac Cancer & Blood Disorders Center, Children's Healthcare of Atlanta, Atlanta, Georgia, USA; Department of Pediatrics, Emory University School of Medicine, Atlanta, Georgia, USA; Graduate Division of Biology and Biomedical Sciences, Emory University Laney Graduate School, Atlanta, Georgia, USA.
| |
Collapse
|
13
|
Pentimalli TM, Karaiskos N, Rajewsky N. Challenges and Opportunities in the Clinical Translation of High-Resolution Spatial Transcriptomics. ANNUAL REVIEW OF PATHOLOGY 2025; 20:405-432. [PMID: 39476415 DOI: 10.1146/annurev-pathmechdis-111523-023417] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/25/2025]
Abstract
Pathology has always been fueled by technological advances. Histology powered the study of tissue architecture at single-cell resolution and remains a cornerstone of clinical pathology today. In the last decade, next-generation sequencing has become informative for the targeted treatment of many diseases, demonstrating the importance of genome-scale molecular information for personalized medicine. Today, revolutionary developments in spatial transcriptomics technologies digitalize gene expression at subcellular resolution in intact tissue sections, enabling the computational analysis of cell types, cellular phenotypes, and cell-cell communication in routinely collected and archival clinical samples. Here we review how such molecular microscopes work, highlight their potential to identify disease mechanisms and guide personalized therapies, and provide guidance for clinical study design. Finally, we discuss remaining challenges to the swift translation of high-resolution spatial transcriptomics technologies and how integration of multimodal readouts and deep learning approaches is bringing us closer to a holistic understanding of tissue biology and pathology.
Collapse
Affiliation(s)
- Tancredi Massimo Pentimalli
- Charité - Universitätsmedizin Berlin, Berlin, Germany
- Laboratory for Systems Biology of Regulatory Elements, Berlin Institute for Medical Systems Biology, Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany; , ,
| | - Nikos Karaiskos
- Laboratory for Systems Biology of Regulatory Elements, Berlin Institute for Medical Systems Biology, Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany; , ,
| | - Nikolaus Rajewsky
- Laboratory for Systems Biology of Regulatory Elements, Berlin Institute for Medical Systems Biology, Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany; , ,
- German Center for Cardiovascular Research (DZHK), Berlin, Germany
- Charité - Universitätsmedizin Berlin, Berlin, Germany
- German Cancer Consortium (DKTK), Berlin, Germany
- National Center for Tumor Diseases, Berlin, Germany
- NeuroCure Cluster of Excellence, Berlin, Germany
| |
Collapse
|
14
|
Safavi A, Samir J, Singh M, Bonomi M, Louie RY, Micklethwaite K, Luciani F. Identification of clonally expanded γδ T-cell populations during CAR-T cell therapy. Immunol Cell Biol 2025; 103:60-72. [PMID: 39500484 DOI: 10.1111/imcb.12834] [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: 04/30/2024] [Revised: 08/27/2024] [Accepted: 10/17/2024] [Indexed: 01/02/2025]
Abstract
Anti-CD19 Chimeric Antigen Receptor (CAR)-T cell therapies have shown promise for treating B cell malignancies, but the clinical outcome is influenced by both the CAR-T product and the patient's immune system. The role of γδ T cells in the context of CAR-T cell therapy remains poorly understood. This study investigates the transcriptional heterogeneity, clonal expansion and dynamics of γδ T cells in patients undergoing anti-CD19 CAR-T cell therapy. Longitudinal single cell multi-omics analysis was performed on γδ T cells from four patients receiving anti-CD19 CAR-T cell therapy. Single cell RNA-seq, antibody-based protein profiling (AbSeq) and full-length TCRγδ sequences revealed clonally expanded populations displaying plasticity in T cell differentiation, and temporal dynamics of large clones, suggesting ongoing expansion and differentiation. Clonally expanded γδ T cells had heterogeneous gene expression profiles, occupying seven transcriptionally distinct clusters. Analysis of chemokine markers indicated cluster-specific homing tendencies of circulating γδ T cells to peripheral tissues. We found unexpectedly high frequencies of Vδ1 and Vδ3 cells in the blood with distinct gene and protein expression profiles. This analysis provides insights into the dynamic and heterogeneous nature of γδ T cells following anti-CD19 CAR-T cell therapy, contributing valuable information for optimizing CAR-T cell therapies in B cell malignancies.
Collapse
Affiliation(s)
- Arman Safavi
- School of Biomedical Sciences, UNSW Sydney, Sydney, NSW, Australia
| | - Jerome Samir
- School of Biomedical Sciences, UNSW Sydney, Sydney, NSW, Australia
| | - Mandeep Singh
- Garvan Institute of Medical Research, Sydney, NSW, Australia
| | - Martina Bonomi
- School of Biomedical Sciences, UNSW Sydney, Sydney, NSW, Australia
| | - Raymond Yip Louie
- School of Computer Science and Engineering, UNSW Sydney, Sydney, NSW, Australia
| | - Kenneth Micklethwaite
- NSW Health Pathology Blood Transplant and Cell Therapies Laboratory - ICPMR Westmead, Sydney, NSW, Australia
- Blood Transplant and Cell Therapies Program, Department of Haematology, Westmead Hospital, Sydney, NSW, Australia
- Westmead Institute for Medical Research, Sydney, NSW, Australia
- Sydney Medical School, The University of Sydney, Sydney, NSW, Australia
| | - Fabio Luciani
- School of Biomedical Sciences, UNSW Sydney, Sydney, NSW, Australia
- Garvan Institute of Medical Research, Sydney, NSW, Australia
- Cellular Genomics Future Institute, UNSW Sydney, Sydney, NSW, Australia
| |
Collapse
|
15
|
Hu H, Zhou F, Ma X, Brokstad KA, Kolmar L, Girardot C, Benes V, Cox RJ, Merten CA. Targeted barcoding of variable antibody domains and individual transcriptomes of the human B-cell repertoire using Link-Seq. PNAS NEXUS 2025; 4:pgaf006. [PMID: 39867668 PMCID: PMC11759286 DOI: 10.1093/pnasnexus/pgaf006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Accepted: 12/18/2024] [Indexed: 01/28/2025]
Abstract
Here, we present Link-Seq, a highly efficient droplet microfluidic method for combined sequencing of antibody-encoding genes and the transcriptome of individual B cells at large scale. The method is based on 3' barcoding of the transcriptome and subsequent single-molecule PCR in droplets, which freely shift the barcode along specific gene regions, such as the antibody heavy- and light-chain genes. Using the immune repertoire of COVID-19 patients and healthy donors as a model system, we obtain up to 91.7% correctly paired immunoglobulin heavy and light chains. Furthermore, we map the V(D)J usage and obtain sensitivities comparable with the current gold-standard 10× Genomics commercial systems while offering full flexibility in experimental setup and significant cost savings. A further unique feature of Link-Seq is the possibility of barcoding multiple target genes in a site-specific manner. Based on the open character of the platform and its conceptual advantages, we expect Link-Seq to become a versatile tool for single-cell analysis, especially for applications requiring additional processing steps that cannot be implemented on commercially available platforms.
Collapse
Affiliation(s)
- Hongxing Hu
- Institute of Bioengineering, School of Engineering, École Polytechnique Fédérale de Lausanne (EPFL), 1015 Lausanne, Switzerland
- Genome Biology Unit, European Molecular Biology Laboratory (EMBL), Heidelberg, 69117 Germany
| | - Fan Zhou
- Department of Clinical Sciences, Influenza Centre, University of Bergen, Bergen, N5021, Norway
| | - Xiaoli Ma
- Institute of Bioengineering, School of Engineering, École Polytechnique Fédérale de Lausanne (EPFL), 1015 Lausanne, Switzerland
| | - Karl Albert Brokstad
- Department of Clinical Sciences, Influenza Centre, University of Bergen, Bergen, N5021, Norway
- Department of Safety, Chemistry and Biomedical Laboratory Sciences, Western Norway University of Applied Sciences (HVL), Bergen, N5020, Norway
| | - Leonie Kolmar
- Institute of Bioengineering, School of Engineering, École Polytechnique Fédérale de Lausanne (EPFL), 1015 Lausanne, Switzerland
| | - Charles Girardot
- Genome Biology Unit, European Molecular Biology Laboratory (EMBL), Heidelberg, 69117 Germany
| | - Vladimir Benes
- Genome Biology Unit, European Molecular Biology Laboratory (EMBL), Heidelberg, 69117 Germany
| | - Rebecca J Cox
- Department of Clinical Sciences, Influenza Centre, University of Bergen, Bergen, N5021, Norway
- Department of Microbiology, Haukeland University Hospital, Bergen, N5021, Norway
| | - Christoph A Merten
- Institute of Bioengineering, School of Engineering, École Polytechnique Fédérale de Lausanne (EPFL), 1015 Lausanne, Switzerland
| |
Collapse
|
16
|
Jin Y, Xing J, Dai C, Jin L, Zhang W, Tao Q, Hou M, Li Z, Yang W, Feng Q, Wang H, Yu Q. NK cell exhaustion in Wilson's disease revealed by single-cell RNA sequencing predicts the prognosis of cholecystitis. eLife 2024; 13:RP98867. [PMID: 39854622 PMCID: PMC11684787 DOI: 10.7554/elife.98867] [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] [Indexed: 01/26/2025] Open
Abstract
Metabolic abnormalities associated with liver disease have a significant impact on the risk and prognosis of cholecystitis. However, the underlying mechanism remains to be elucidated. Here, we investigated this issue using Wilson's disease (WD) as a model, which is a genetic disorder characterized by impaired mitochondrial function and copper metabolism. Our retrospective clinical study found that WD patients have a significantly higher incidence of cholecystitis and a poorer prognosis. The hepatic immune cell landscape using single-cell RNA sequencing showed that the tissue immune microenvironment is altered in WD, mainly a major change in the constitution and function of the innate immune system. Exhaustion of natural killer (NK) cells is the fundamental factor, supported by the upregulated expression of inhibitory receptors and the downregulated expression of cytotoxic molecules, which was verified in clinical samples. Further bioinformatic analysis confirmed a positive correlation between NK cell exhaustion and poor prognosis in cholecystitis and other inflammatory diseases. The study demonstrated dysfunction of liver immune cells triggered by specific metabolic abnormalities in WD, with a focus on the correlation between NK cell exhaustion and poor healing of cholecystitis, providing new insights into the improvement of inflammatory diseases by assessing immune cell function.
Collapse
Affiliation(s)
- Yong Jin
- Cancer Research Center, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of ChinaHefeiChina
| | - Jiayu Xing
- Cancer Research Center, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of ChinaHefeiChina
| | - Chenyu Dai
- Department of Cadre Cardiology, The First Affiliated Hospital of Anhui University of Chinese MedicineHefeiChina
| | - Lei Jin
- Department of General Surgery, The First Affiliated Hospital of Anhui University of Chinese MedicineHefeiChina
- Institute of Chinese Medicine Surgery, Anhui Academy of Chinese MedicineHefeiChina
| | - Wanying Zhang
- Cancer Research Center, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of ChinaHefeiChina
| | - Qianqian Tao
- Department of General Surgery, The First Affiliated Hospital of Anhui University of Chinese MedicineHefeiChina
- Institute of Chinese Medicine Surgery, Anhui Academy of Chinese MedicineHefeiChina
| | - Mei Hou
- Cancer Research Center, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of ChinaHefeiChina
| | - Ziyi Li
- Department of General Surgery, The First Affiliated Hospital of Anhui University of Chinese MedicineHefeiChina
- Institute of Chinese Medicine Surgery, Anhui Academy of Chinese MedicineHefeiChina
| | - Wen Yang
- Cancer Research Center, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of ChinaHefeiChina
- International Co-operation Laboratory on Signal Transduction, Eastern Hepatobiliary Surgery Hospital, Second Military Medical UniversityShanghaiChina
- National Center for Liver Cancer, Second Military Medical UniversityShanghaiChina
| | - Qiyu Feng
- Cancer Research Center, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of ChinaHefeiChina
| | - Hongyang Wang
- Cancer Research Center, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of ChinaHefeiChina
- International Co-operation Laboratory on Signal Transduction, Eastern Hepatobiliary Surgery Hospital, Second Military Medical UniversityShanghaiChina
- National Center for Liver Cancer, Second Military Medical UniversityShanghaiChina
| | - Qingsheng Yu
- Department of Cadre Cardiology, The First Affiliated Hospital of Anhui University of Chinese MedicineHefeiChina
- Department of General Surgery, The First Affiliated Hospital of Anhui University of Chinese MedicineHefeiChina
| |
Collapse
|
17
|
Zhang T, Li H, Jiang M, Hou H, Gao Y, Li Y, Wang F, Wang J, Peng K, Liu YX. Nanopore sequencing: flourishing in its teenage years. J Genet Genomics 2024; 51:1361-1374. [PMID: 39293510 DOI: 10.1016/j.jgg.2024.09.007] [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: 07/18/2024] [Revised: 09/09/2024] [Accepted: 09/10/2024] [Indexed: 09/20/2024]
Abstract
Over the past decade, nanopore sequencing has experienced significant advancements and changes, transitioning from an initially emerging technology to a significant instrument in the field of genomic sequencing. However, as advancements in next-generation sequencing technology persist, nanopore sequencing also improves. This paper reviews the developments, applications, and outlook on nanopore sequencing technology. Currently, nanopore sequencing supports both DNA and RNA sequencing, making it widely applicable in areas such as telomere-to-telomere (T2T) genome assembly, direct RNA sequencing (DRS), and metagenomics. The openness and versatility of nanopore sequencing have established it as a preferred option for an increasing number of research teams, signaling a transformative influence on life science research. As the nanopore sequencing technology advances, it provides a faster, more cost-effective approach with extended read lengths, demonstrating the significant potential for complex genome assembly, pathogen detection, environmental monitoring, and human disease research, offering a fresh perspective in sequencing technologies.
Collapse
Affiliation(s)
- Tianyuan Zhang
- Genome Analysis Laboratory of the Ministry of Agriculture and Rural Affairs, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen, Guangdong 518120, China; Wuhan Benagen Technology Co., Ltd, Wuhan, Hubei 430000, China
| | - Hanzhou Li
- Wuhan Benagen Technology Co., Ltd, Wuhan, Hubei 430000, China
| | - Mian Jiang
- Wuhan Benagen Technology Co., Ltd, Wuhan, Hubei 430000, China
| | - Huiyu Hou
- Genome Analysis Laboratory of the Ministry of Agriculture and Rural Affairs, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen, Guangdong 518120, China
| | - Yunyun Gao
- Genome Analysis Laboratory of the Ministry of Agriculture and Rural Affairs, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen, Guangdong 518120, China
| | - Yali Li
- Wuhan Benagen Technology Co., Ltd, Wuhan, Hubei 430000, China
| | - Fuhao Wang
- Wuhan Benagen Technology Co., Ltd, Wuhan, Hubei 430000, China
| | - Jun Wang
- Wuhan Benagen Technology Co., Ltd, Wuhan, Hubei 430000, China
| | - Kai Peng
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, College of Veterinary Medicine, Yangzhou University, Yangzhou, Jiangsu 225000, China
| | - Yong-Xin Liu
- Genome Analysis Laboratory of the Ministry of Agriculture and Rural Affairs, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen, Guangdong 518120, China.
| |
Collapse
|
18
|
Belchikov N, Hsu J, Li XJ, Jarroux J, Hu W, Joglekar A, Tilgner HU. Understanding isoform expression by pairing long-read sequencing with single-cell and spatial transcriptomics. Genome Res 2024; 34:1735-1746. [PMID: 39567235 DOI: 10.1101/gr.279640.124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2024]
Abstract
RNA isoform diversity, produced via alternative splicing, and alternative usage of transcription start and poly(A) sites, results in varied transcripts being derived from the same gene. Distinct isoforms can play important biological roles, including by changing the sequences or expression levels of protein products. The first single-cell approaches to RNA sequencing-and later, spatial approaches-which are now widely used for the identification of differentially expressed genes, rely on short reads and offer the ability to transcriptomically compare different cell types but are limited in their ability to measure differential isoform expression. More recently, long-read sequencing methods have been combined with single-cell and spatial technologies in order to characterize isoform expression. In this review, we provide an overview of the emergence of single-cell and spatial long-read sequencing and discuss the challenges associated with the implementation of these technologies and interpretation of these data. We discuss the opportunities they offer for understanding the relationships between the distinct variable elements of transcript molecules and highlight some of the ways in which they have been used to characterize isoforms' roles in development and pathology. Single-nucleus long-read sequencing, a special case of the single-cell approach, is also discussed. We attempt to cover both the limitations of these technologies and their significant potential for expanding our still-limited understanding of the biological roles of RNA isoforms.
Collapse
Affiliation(s)
- Natan Belchikov
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, New York 10065, USA
- Center for Neurogenetics, Weill Cornell Medicine, New York, New York 10021, USA
- Physiology, Biophysics, and Systems Biology Program, Weill Cornell Medicine, New York, New York 10065, USA
| | - Justine Hsu
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, New York 10065, USA
- Center for Neurogenetics, Weill Cornell Medicine, New York, New York 10021, USA
| | - Xiang Jennie Li
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, New York 10065, USA
- Center for Neurogenetics, Weill Cornell Medicine, New York, New York 10021, USA
- Computational Biology Master's Program, Weill Cornell Medicine, New York, New York 10065, USA
| | - Julien Jarroux
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, New York 10065, USA
- Center for Neurogenetics, Weill Cornell Medicine, New York, New York 10021, USA
| | - Wen Hu
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, New York 10065, USA
- Center for Neurogenetics, Weill Cornell Medicine, New York, New York 10021, USA
| | - Anoushka Joglekar
- New York Genome Center, New York, New York 10013, USA
- Department of Biomedical Informatics, Columbia University, New York, New York 10032, USA
| | - Hagen U Tilgner
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, New York 10065, USA;
- Center for Neurogenetics, Weill Cornell Medicine, New York, New York 10021, USA
| |
Collapse
|
19
|
Iyer SV, Goodwin S, McCombie WR. Leveraging the power of long reads for targeted sequencing. Genome Res 2024; 34:1701-1718. [PMID: 39567237 PMCID: PMC11610587 DOI: 10.1101/gr.279168.124] [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: 03/15/2024] [Accepted: 10/01/2024] [Indexed: 11/22/2024]
Abstract
Long-read sequencing technologies have improved the contiguity and, as a result, the quality of genome assemblies by generating reads long enough to span and resolve complex or repetitive regions of the genome. Several groups have shown the power of long reads in detecting thousands of genomic and epigenomic features that were previously missed by short-read sequencing approaches. While these studies demonstrate how long reads can help resolve repetitive and complex regions of the genome, they also highlight the throughput and coverage requirements needed to accurately resolve variant alleles across large populations using these platforms. At the time of this review, whole-genome long-read sequencing is more expensive than short-read sequencing on the highest throughput short-read instruments; thus, achieving sufficient coverage to detect low-frequency variants (such as somatic variation) in heterogenous samples remains challenging. Targeted sequencing, on the other hand, provides the depth necessary to detect these low-frequency variants in heterogeneous populations. Here, we review currently used and recently developed targeted sequencing strategies that leverage existing long-read technologies to increase the resolution with which we can look at nucleic acids in a variety of biological contexts.
Collapse
Affiliation(s)
- Shruti V Iyer
- Cold Spring Harbor Laboratory, Cold Spring Harbor, New York 11724, USA
| | - Sara Goodwin
- Cold Spring Harbor Laboratory, Cold Spring Harbor, New York 11724, USA
| | | |
Collapse
|
20
|
Shi Q, Zhang Q, Shao M. Transcriptome Assembly at Single-Cell Resolution with Beaver. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.11.04.621958. [PMID: 39574665 PMCID: PMC11580954 DOI: 10.1101/2024.11.04.621958] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/02/2024]
Abstract
Emerging single-cell RNA sequencing techniques (scRNA-seq) has enabled the study of cellular transcriptome heterogeneity, yet accurate reconstruction of full-length transcripts at single-cell resolution remains challenging due to high dropout rates and sparse coverage. While meta-assembly approaches offer promising solutions by integrating information across multiple cells, current methods struggle to balance consensus assembly with cell-specific transcriptional signatures. Here, we present Beaver, a cell-specific transcript assembler designed for short-read scRNA-seq data. Beaver implements a transcript fragment graph to organize individual assemblies and designs an efficient dynamic programming algorithm that searches for candidate full-length transcripts from the graph. Beaver in-corporates two random forest models trained on 51 meticulously engineered features that accurately estimate the likelihood of each candidate transcript being expressed in individual cells. Our experiments, performed using both real and simulated Smart-seq3 scRNA-seq data, firmly show that Beaver substantially outperforms existing meta-assemblers and single-sample assemblers. At the same level of sensitivity, Beaver achieved 32.0%-64.6%, 13.5%-36.6%, and 9.8%-36.3% higher precision in average compared to meta-assemblers Aletsch, TransMeta, and PsiCLASS, respectively, with similar improvements over single-sample assemblers Scallop2 (10.1%-43.6%) and StringTie2 (24.3%-67.0%). Beaver is freely available at https://github.com/Shao-Group/beaver . Scripts that reproduce the experimental results of this manuscript are available at https://github.com/Shao-Group/beaver-test .
Collapse
Affiliation(s)
- Qian Shi
- Department of Computer Science and Engineering, School of Electrical Engineering and Computer Science, The Pennsylvania State University, University Park, PA 16802, USA
| | - Qimin Zhang
- Department of Computer Science and Engineering, School of Electrical Engineering and Computer Science, The Pennsylvania State University, University Park, PA 16802, USA
| | - Mingfu Shao
- Department of Computer Science and Engineering, School of Electrical Engineering and Computer Science, The Pennsylvania State University, University Park, PA 16802, USA
- Huck Institutes of the Life Sciences, The Pennsylvania State University, University Park, PA 16802, USA
| |
Collapse
|
21
|
Kumari P, Kaur M, Dindhoria K, Ashford B, Amarasinghe SL, Thind AS. Advances in long-read single-cell transcriptomics. Hum Genet 2024; 143:1005-1020. [PMID: 38787419 PMCID: PMC11485027 DOI: 10.1007/s00439-024-02678-x] [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: 11/20/2023] [Accepted: 05/07/2024] [Indexed: 05/25/2024]
Abstract
Long-read single-cell transcriptomics (scRNA-Seq) is revolutionizing the way we profile heterogeneity in disease. Traditional short-read scRNA-Seq methods are limited in their ability to provide complete transcript coverage, resolve isoforms, and identify novel transcripts. The scRNA-Seq protocols developed for long-read sequencing platforms overcome these limitations by enabling the characterization of full-length transcripts. Long-read scRNA-Seq techniques initially suffered from comparatively poor accuracy compared to short read scRNA-Seq. However, with improvements in accuracy, accessibility, and cost efficiency, long-reads are gaining popularity in the field of scRNA-Seq. This review details the advances in long-read scRNA-Seq, with an emphasis on library preparation protocols and downstream bioinformatics analysis tools.
Collapse
Affiliation(s)
- Pallawi Kumari
- Institute of Microbial Technology, Council of Scientific and Industrial Research, Chandigarh, India
| | - Manmeet Kaur
- Institute of Microbial Technology, Council of Scientific and Industrial Research, Chandigarh, India
| | - Kiran Dindhoria
- Institute of Microbial Technology, Council of Scientific and Industrial Research, Chandigarh, India
| | - Bruce Ashford
- Illawarra Shoalhaven Local Health District (ISLHD), NSW Health, Wollongong, NSW, Australia
| | - Shanika L Amarasinghe
- Monash Biomedical Discovery Institute, Monash University, Clayton, VIC, 3800, Australia
- Walter and Eliza Hall Institute of Medical Research, 1G, Royal Parade, Parkville, VIC, 3025, Australia
| | - Amarinder Singh Thind
- Illawarra Shoalhaven Local Health District (ISLHD), NSW Health, Wollongong, NSW, Australia.
- The School of Chemistry and Molecular Bioscience (SCMB), University of Wollongong, Loftus St, Wollongong, NSW, 2500, Australia.
| |
Collapse
|
22
|
Aucouturier C, Soirat N, Castéra L, Bertrand D, Atkinson A, Lavolé T, Goardon N, Quesnelle C, Levilly J, Barbachou S, Legros A, Caron O, Crivelli L, Denizeau P, Berthet P, Ricou A, Boulouard F, Vaur D, Krieger S, Leman R. Fine mapping of RNA isoform diversity using an innovative targeted long-read RNA sequencing protocol with novel dedicated bioinformatics pipeline. BMC Genomics 2024; 25:909. [PMID: 39350015 PMCID: PMC11440762 DOI: 10.1186/s12864-024-10741-0] [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: 04/30/2024] [Accepted: 08/28/2024] [Indexed: 10/04/2024] Open
Abstract
BACKGROUND Solving the structure of mRNA transcripts is a major challenge for both research and molecular diagnostic purposes. Current approaches based on short-read RNA sequencing and RT-PCR techniques cannot fully explore the complexity of transcript structure. The emergence of third-generation long-read sequencing addresses this problem by solving this sequence directly. However, genes with low expression levels are difficult to study with the whole transcriptome sequencing approach. To fix this technical limitation, we propose a novel method to capture transcripts of a gene panel using a targeted enrichment approach suitable for Pacific Biosciences and Oxford Nanopore Technologies platforms. RESULTS We designed a set of probes to capture transcripts of a panel of genes involved in hereditary breast and ovarian cancer syndrome. We present SOSTAR (iSofOrmS annoTAtoR), a versatile pipeline to assemble, quantify and annotate isoforms from long read sequencing using a new tool specially designed for this application. The significant enrichment of transcripts by our capture protocol, together with the SOSTAR annotation, allowed the identification of 1,231 unique transcripts within the gene panel from the eight patients sequenced. The structure of these transcripts was annotated with a resolution of one base relative to a reference transcript. All major alternative splicing events of the BRCA1 and BRCA2 genes described in the literature were found. Complex splicing events such as pseudoexons were correctly annotated. SOSTAR enabled the identification of abnormal transcripts in the positive controls. In addition, a case of unexplained inheritance in a family with a history of breast and ovarian cancer was solved by identifying an SVA retrotransposon in intron 13 of the BRCA1 gene. CONCLUSIONS We have validated a new protocol for the enrichment of transcripts of interest using probes adapted to the ONT and PacBio platforms. This protocol allows a complete description of the alternative structures of transcripts, the estimation of their expression and the identification of aberrant transcripts in a single experiment. This proof-of-concept opens new possibilities for RNA structure exploration in both research and molecular diagnostics.
Collapse
Affiliation(s)
- Camille Aucouturier
- Laboratoire de biologie et de génétique du cancer, Département de Biopathologie, Centre François Baclesse, Caen, 14000, France
- Cancer and Brain Genomics, FHU G4 Genomics, Inserm U1245, Normandie University, Rouen, 76183, France
- Normandie Univ, UNICAEN, Caen, 14000, France
| | - Nicolas Soirat
- Laboratoire de biologie et de génétique du cancer, Département de Biopathologie, Centre François Baclesse, Caen, 14000, France
- Cancer and Brain Genomics, FHU G4 Genomics, Inserm U1245, Normandie University, Rouen, 76183, France
- SeqOne Genomics, Montpellier, 34000, France
| | - Laurent Castéra
- Laboratoire de biologie et de génétique du cancer, Département de Biopathologie, Centre François Baclesse, Caen, 14000, France
- Cancer and Brain Genomics, FHU G4 Genomics, Inserm U1245, Normandie University, Rouen, 76183, France
| | | | - Alexandre Atkinson
- Laboratoire de biologie et de génétique du cancer, Département de Biopathologie, Centre François Baclesse, Caen, 14000, France
| | - Thibaut Lavolé
- Laboratoire de biologie et de génétique du cancer, Département de Biopathologie, Centre François Baclesse, Caen, 14000, France
| | - Nicolas Goardon
- Laboratoire de biologie et de génétique du cancer, Département de Biopathologie, Centre François Baclesse, Caen, 14000, France
- Cancer and Brain Genomics, FHU G4 Genomics, Inserm U1245, Normandie University, Rouen, 76183, France
| | - Céline Quesnelle
- Laboratoire de biologie et de génétique du cancer, Département de Biopathologie, Centre François Baclesse, Caen, 14000, France
| | - Julien Levilly
- Laboratoire de biologie et de génétique du cancer, Département de Biopathologie, Centre François Baclesse, Caen, 14000, France
| | - Sosthène Barbachou
- Laboratoire de biologie et de génétique du cancer, Département de Biopathologie, Centre François Baclesse, Caen, 14000, France
| | - Angelina Legros
- Laboratoire de biologie et de génétique du cancer, Département de Biopathologie, Centre François Baclesse, Caen, 14000, France
| | - Olivier Caron
- Département Médecine Oncologique, Institut Gustave Roussy, Villejuif, France
| | - Louise Crivelli
- Service d'Oncogénétique, Centre Eugène Marquis, Rennes, France
| | - Philippe Denizeau
- Service de génétique clinique, Centre Hospitalier Universitaire Rennes, Rennes, France
| | - Pascaline Berthet
- Service d'Oncogénétique, Département de Biopathologie, Centre François Baclesse, Caen, 14000, France
| | - Agathe Ricou
- Laboratoire de biologie et de génétique du cancer, Département de Biopathologie, Centre François Baclesse, Caen, 14000, France
- Cancer and Brain Genomics, FHU G4 Genomics, Inserm U1245, Normandie University, Rouen, 76183, France
| | - Flavie Boulouard
- Laboratoire de biologie et de génétique du cancer, Département de Biopathologie, Centre François Baclesse, Caen, 14000, France
- Cancer and Brain Genomics, FHU G4 Genomics, Inserm U1245, Normandie University, Rouen, 76183, France
| | - Dominique Vaur
- Laboratoire de biologie et de génétique du cancer, Département de Biopathologie, Centre François Baclesse, Caen, 14000, France
- Cancer and Brain Genomics, FHU G4 Genomics, Inserm U1245, Normandie University, Rouen, 76183, France
| | - Sophie Krieger
- Laboratoire de biologie et de génétique du cancer, Département de Biopathologie, Centre François Baclesse, Caen, 14000, France
- Cancer and Brain Genomics, FHU G4 Genomics, Inserm U1245, Normandie University, Rouen, 76183, France
- Normandie Univ, UNICAEN, Caen, 14000, France
| | - Raphael Leman
- Laboratoire de biologie et de génétique du cancer, Département de Biopathologie, Centre François Baclesse, Caen, 14000, France.
- Cancer and Brain Genomics, FHU G4 Genomics, Inserm U1245, Normandie University, Rouen, 76183, France.
| |
Collapse
|
23
|
Morgan DM, Zhang YJ, Kim JH, Murillo M, Singh S, Loschko J, Surendran N, Sekulovic O, Feng E, Shi S, Irvine DJ, Patil SU, Kanevsky I, Chorro L, Christopher Love J. Full-length single-cell BCR sequencing paired with RNA sequencing reveals convergent responses to pneumococcal vaccination. Commun Biol 2024; 7:1208. [PMID: 39341987 PMCID: PMC11438910 DOI: 10.1038/s42003-024-06823-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Accepted: 09/02/2024] [Indexed: 10/01/2024] Open
Abstract
Single-cell RNA sequencing (scRNA-seq) can resolve transcriptional features from individual cells, but scRNA-seq techniques capable of resolving the variable regions of B cell receptors (BCRs) remain limited, especially from widely-used 3'-barcoded libraries. Here, we report a method that can recover paired, full-length variable region sequences of BCRs from 3'-barcoded scRNA-seq libraries. We first verify this method (B3E-seq) can produce accurate, full-length BCR sequences. We then apply this method to profile B cell responses elicited against the capsular polysaccharide of Streptococcus pneumoniae serotype 3 (ST3) by glycoconjugate vaccines in five infant rhesus macaques. We identify BCR features associated with specificity for the ST3 antigen which are present in multiple vaccinated monkeys, indicating a convergent response to vaccination. These results demonstrate the utility of our method to resolve key features of the B cell repertoire and profile antigen-specific responses elicited by vaccination.
Collapse
Affiliation(s)
- Duncan M Morgan
- Koch Institute for Integrative Cancer Research, MIT, Cambridge, MA, USA
- Department of Chemical Engineering, MIT, Cambridge, MA, USA
| | - Yiming J Zhang
- Koch Institute for Integrative Cancer Research, MIT, Cambridge, MA, USA
- Department of Biological Engineering, MIT, Cambridge, MA, USA
| | - Jin-Hwan Kim
- Vaccine Research and Development Pfizer Inc. Pearl River, New York, NY, USA
| | - MaryAnn Murillo
- Vaccine Research and Development Pfizer Inc. Pearl River, New York, NY, USA
| | - Suddham Singh
- Vaccine Research and Development Pfizer Inc. Pearl River, New York, NY, USA
| | - Jakob Loschko
- Vaccine Research and Development Pfizer Inc. Pearl River, New York, NY, USA
- Deerfield Management, New York, NY, USA
| | - Naveen Surendran
- Vaccine Research and Development Pfizer Inc. Pearl River, New York, NY, USA
| | - Ognjen Sekulovic
- Vaccine Research and Development Pfizer Inc. Pearl River, New York, NY, USA
| | - Ellie Feng
- Koch Institute for Integrative Cancer Research, MIT, Cambridge, MA, USA
- Department of Biological Engineering, MIT, Cambridge, MA, USA
| | - Shuting Shi
- Koch Institute for Integrative Cancer Research, MIT, Cambridge, MA, USA
- Department of Chemical Engineering, MIT, Cambridge, MA, USA
| | - Darrell J Irvine
- Koch Institute for Integrative Cancer Research, MIT, Cambridge, MA, USA
- Department of Biological Engineering, MIT, Cambridge, MA, USA
- Howard Hughes Medical Institute, Chevy Chase, MD, USA
- Department of Materials Science and Engineering, MIT, Cambridge, MA, USA
| | - Sarita U Patil
- Center for Immunology and Inflammatory Diseases, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Isis Kanevsky
- Vaccine Research and Development Pfizer Inc. Pearl River, New York, NY, USA
| | - Laurent Chorro
- Vaccine Research and Development Pfizer Inc. Pearl River, New York, NY, USA
- Regeneron, Tarrytown, NY, USA
| | - J Christopher Love
- Koch Institute for Integrative Cancer Research, MIT, Cambridge, MA, USA.
- Department of Chemical Engineering, MIT, Cambridge, MA, USA.
| |
Collapse
|
24
|
Liu B, Hu S, Wang X. Applications of single-cell technologies in drug discovery for tumor treatment. iScience 2024; 27:110486. [PMID: 39171294 PMCID: PMC11338156 DOI: 10.1016/j.isci.2024.110486] [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] [Indexed: 08/23/2024] Open
Abstract
Single-cell technologies have been known as advanced and powerful tools to study tumor biological systems at the single-cell resolution and are playing increasingly critical roles in multiple stages of drug discovery and development. Specifically, single-cell technologies can promote the discovery of drug targets, help high-throughput screening at single-cell level, and contribute to pharmacokinetic studies of anti-tumor drugs. Emerging single-cell analysis technologies have been developed to further integrating multidimensional single-cell molecular features, expanding the scale of single-cell data, profiling phenotypic impact of genes in single cell, and providing full-length coverage single-cell sequencing. In this review, we systematically summarized the applications of single-cell technologies in various sections of drug discovery for tumor treatment, including target identification, high-throughput drug screening, and pharmacokinetic evaluation and highlighted emerging single-cell technologies in providing in-depth understanding of tumor biology. Single-cell-technology-based drug discovery is expected to further optimize therapeutic strategies and improve clinical outcomes of tumor patients.
Collapse
Affiliation(s)
- Bingyu Liu
- Department of Hematology, Shandong Provincial Hospital, Shandong University, Jinan, Shandong 250021, China
| | - Shunfeng Hu
- Department of Hematology, Shandong Provincial Hospital, Shandong University, Jinan, Shandong 250021, China
- Department of Hematology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250021, China
| | - Xin Wang
- Department of Hematology, Shandong Provincial Hospital, Shandong University, Jinan, Shandong 250021, China
- Department of Hematology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250021, China
- Taishan Scholars Program of Shandong Province, Jinan, Shandong 250021, China
| |
Collapse
|
25
|
Gong B, Li D, Łabaj PP, Pan B, Novoradovskaya N, Thierry-Mieg D, Thierry-Mieg J, Chen G, Bergstrom Lucas A, LoCoco JS, Richmond TA, Tseng E, Kusko R, Happe S, Mercer TR, Pabón-Peña C, Salmans M, Tilgner HU, Xiao W, Johann DJ, Jones W, Tong W, Mason CE, Kreil DP, Xu J. Targeted DNA-seq and RNA-seq of Reference Samples with Short-read and Long-read Sequencing. Sci Data 2024; 11:892. [PMID: 39152166 PMCID: PMC11329654 DOI: 10.1038/s41597-024-03741-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: 03/05/2024] [Accepted: 08/05/2024] [Indexed: 08/19/2024] Open
Abstract
Next-generation sequencing (NGS) has revolutionized genomic research by enabling high-throughput, cost-effective genome and transcriptome sequencing accelerating personalized medicine for complex diseases, including cancer. Whole genome/transcriptome sequencing (WGS/WTS) provides comprehensive insights, while targeted sequencing is more cost-effective and sensitive. In comparison to short-read sequencing, which still dominates the field due to high speed and cost-effectiveness, long-read sequencing can overcome alignment limitations and better discriminate similar sequences from alternative transcripts or repetitive regions. Hybrid sequencing combines the best strengths of different technologies for a more comprehensive view of genomic/transcriptomic variations. Understanding each technology's strengths and limitations is critical for translating cutting-edge technologies into clinical applications. In this study, we sequenced DNA and RNA libraries of reference samples using various targeted DNA and RNA panels and the whole transcriptome on both short-read and long-read platforms. This study design enables a comprehensive analysis of sequencing technologies, targeting protocols, and library preparation methods. Our expanded profiling landscape establishes a reference point for assessing current sequencing technologies, facilitating informed decision-making in genomic research and precision medicine.
Collapse
Affiliation(s)
- Binsheng Gong
- Division of Bioinformatics and Biostatistics, National Center for Toxicological Research, US Food and Drug Administration, Jefferson, AR, 72079, USA
| | - Dan Li
- Division of Bioinformatics and Biostatistics, National Center for Toxicological Research, US Food and Drug Administration, Jefferson, AR, 72079, USA
| | - Paweł P Łabaj
- Małopolska Centre of Biotechnology, Jagiellonian University, Krakow, Poland
- Bioinformatics Research, Institute of Molecular Biotechnology, Boku University Vienna, Vienna, Austria
| | - Bohu Pan
- Division of Bioinformatics and Biostatistics, National Center for Toxicological Research, US Food and Drug Administration, Jefferson, AR, 72079, USA
| | | | - Danielle Thierry-Mieg
- National Center for Biotechnology Information, National Library of Medicine, National Institutes of Health, 8600 Rockville Pike, Bethesda, MD, 20894, USA
| | - Jean Thierry-Mieg
- National Center for Biotechnology Information, National Library of Medicine, National Institutes of Health, 8600 Rockville Pike, Bethesda, MD, 20894, USA
| | - Guangchun Chen
- Department of Immunology, Genomics and Microarray Core Facility, University of Texas Southwestern Medical Center, 5323 Harry Hine Blvd., Dallas, TX, 75390, USA
| | - Anne Bergstrom Lucas
- Agilent Technologies, Inc., 5301 Stevens Creek Blvd., Santa Clara, CA, 95051, USA
| | | | - Todd A Richmond
- Market & Application Development Bioinformatics, Roche Sequencing Solutions Inc., 4300 Hacienda Dr., Pleasanton, CA, 94588, USA
| | | | - Rebecca Kusko
- Cellino Bio, 750 Main Street, Cambridge, MA, 02143, USA
| | - Scott Happe
- Agilent Technologies, Inc., 1834 State Hwy 71 West, Cedar Creek, TX, 78612, USA
| | - Timothy R Mercer
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, St Lucia, QLD, Australia
| | - Carlos Pabón-Peña
- Agilent Technologies, Inc., 5301 Stevens Creek Blvd., Santa Clara, CA, 95051, USA
| | | | - Hagen U Tilgner
- Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA
- Center for Neurogenetics, Weill Cornell Medicine, New York, NY, USA
| | - Wenzhong Xiao
- Stanford Genome Technology Center, Stanford University, Palo Alto, CA, 94304, USA
- Massachusetts General Hospital, Harvard Medical School, Boston, MA, 02114, USA
| | - Donald J Johann
- Winthrop P Rockefeller Cancer Institute, University of Arkansas for Medical Sciences, 4301W Markham St., Little Rock, AR, 72205, USA
| | - Wendell Jones
- Q squared Solutions Genomics, 2400 Elis Road, Durham, NC, 27703, USA
| | - Weida Tong
- Division of Bioinformatics and Biostatistics, National Center for Toxicological Research, US Food and Drug Administration, Jefferson, AR, 72079, USA
| | - Christopher E Mason
- Department of Physiology and Biophysics, Weill Cornell Medicine, Cornell University, New York, NY, 10065, USA.
- The HRH Prince Alwaleed Bin Talal Bin Abdulaziz Alsaud Institute for Computational Biomedicine, Weill Cornell Medicine, New York, NY, USA.
- The WorldQuant Initiative for Quantitative Prediction, Weill Cornell Medicine, New York, NY, USA.
| | - David P Kreil
- Bioinformatics Research, Institute of Molecular Biotechnology, Boku University Vienna, Vienna, Austria.
| | - Joshua Xu
- Division of Bioinformatics and Biostatistics, National Center for Toxicological Research, US Food and Drug Administration, Jefferson, AR, 72079, USA.
| |
Collapse
|
26
|
Byrne A, Le D, Sereti K, Menon H, Vaidya S, Patel N, Lund J, Xavier-Magalhães A, Shi M, Liang Y, Sterne-Weiler T, Modrusan Z, Stephenson W. Single-cell long-read targeted sequencing reveals transcriptional variation in ovarian cancer. Nat Commun 2024; 15:6916. [PMID: 39134520 PMCID: PMC11319652 DOI: 10.1038/s41467-024-51252-6] [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: 07/27/2023] [Accepted: 07/31/2024] [Indexed: 08/15/2024] Open
Abstract
Single-cell RNA sequencing predominantly employs short-read sequencing to characterize cell types, states and dynamics; however, it is inadequate for comprehensive characterization of RNA isoforms. Long-read sequencing technologies enable single-cell RNA isoform detection but are hampered by lower throughput and unintended sequencing of artifacts. Here we develop Single-cell Targeted Isoform Long-Read Sequencing (scTaILoR-seq), a hybridization capture method which targets over a thousand genes of interest, improving the median number of on-target transcripts per cell by 29-fold. We use scTaILoR-seq to identify and quantify RNA isoforms from ovarian cancer cell lines and primary tumors, yielding 10,796 single-cell transcriptomes. Using long-read variant calling we reveal associations of expressed single nucleotide variants (SNVs) with alternative transcript structures. Phasing of SNVs across transcripts enables the measurement of allelic imbalance within distinct cell populations. Overall, scTaILoR-seq is a long-read targeted RNA sequencing method and analytical framework for exploring transcriptional variation at single-cell resolution.
Collapse
Affiliation(s)
- Ashley Byrne
- Department of Proteomic and Genomic Technologies, Genentech, South San Francisco, CA, USA
| | - Daniel Le
- Department of Proteomic and Genomic Technologies, Genentech, South San Francisco, CA, USA
| | - Kostianna Sereti
- Department of Discovery Oncology, Genentech, South San Francisco, CA, USA
| | - Hari Menon
- Department of Proteomic and Genomic Technologies, Genentech, South San Francisco, CA, USA
| | - Samir Vaidya
- Department of Proteomic and Genomic Technologies, Genentech, South San Francisco, CA, USA
| | - Neha Patel
- Department of Proteomic and Genomic Technologies, Genentech, South San Francisco, CA, USA
| | - Jessica Lund
- Department of Proteomic and Genomic Technologies, Genentech, South San Francisco, CA, USA
| | - Ana Xavier-Magalhães
- Department of Proteomic and Genomic Technologies, Genentech, South San Francisco, CA, USA
| | - Minyi Shi
- Department of Proteomic and Genomic Technologies, Genentech, South San Francisco, CA, USA
| | - Yuxin Liang
- Department of Proteomic and Genomic Technologies, Genentech, South San Francisco, CA, USA
| | - Timothy Sterne-Weiler
- Department of Discovery Oncology, Genentech, South San Francisco, CA, USA
- Department of Oncology Bioinformatics, Genentech, South San Francisco, CA, USA
| | - Zora Modrusan
- Department of Proteomic and Genomic Technologies, Genentech, South San Francisco, CA, USA.
| | - William Stephenson
- Department of Proteomic and Genomic Technologies, Genentech, South San Francisco, CA, USA.
| |
Collapse
|
27
|
Yu T, Ren Z, Gao X, Li G, Han R. Generating barcodes for nanopore sequencing data with PRO. FUNDAMENTAL RESEARCH 2024; 4:785-794. [PMID: 39660352 PMCID: PMC11630701 DOI: 10.1016/j.fmre.2024.04.014] [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: 06/30/2023] [Revised: 02/20/2024] [Accepted: 04/09/2024] [Indexed: 12/12/2024] Open
Abstract
DNA barcodes, short and unique DNA sequences, play a crucial role in sample identification when processing many samples simultaneously, which helps reduce experimental costs. Nevertheless, the low quality of long-read sequencing makes it difficult to identify barcodes accurately, which poses significant challenges for the design of barcodes for large numbers of samples in a single sequencing run. Here, we present a comprehensive study of the generation of barcodes and develop a tool, PRO, that can be used for selecting optimal barcode sets and demultiplexing. We formulate the barcode design problem as a combinatorial problem and prove that finding the optimal largest barcode set in a given DNA sequence space in which all sequences have the same length is theoretically NP-complete. For practical applications, we developed the novel method PRO by introducing the probability divergence between two DNA sequences to expand the capacity of barcode kits while ensuring demultiplexing accuracy. Specifically, the maximum size of the barcode kits designed by PRO is 2,292, which keeps the length of barcodes the same as that of the official ones used by Oxford Nanopore Technologies (ONT). We validated the performance of PRO on a simulated nanopore dataset with high error rates. The demultiplexing accuracy of PRO reached 98.29% for a barcode kit of size 2,922, 4.31% higher than that of Guppy, the official demultiplexing tool. When the size of the barcode kit generated by PRO is the same as the official size provided by ONT, both tools show superior and comparable demultiplexing accuracy.
Collapse
Affiliation(s)
- Ting Yu
- Research Center for Mathematics and Interdisciplinary Sciences, Frontiers Science Center for Nonlinear Expectations (Ministry of Education), Shandong University, Shandong 266000, China
| | - Zitong Ren
- Research Center for Mathematics and Interdisciplinary Sciences, Frontiers Science Center for Nonlinear Expectations (Ministry of Education), Shandong University, Shandong 266000, China
| | - Xin Gao
- Computer, Electrical and Mathematical Sciences and Engineering Division & Computational Bioscience Research Center, King Abdullah University of Science and Technology, Thuwal 23955, Saudi Arabia
| | - Guojun Li
- Research Center for Mathematics and Interdisciplinary Sciences, Frontiers Science Center for Nonlinear Expectations (Ministry of Education), Shandong University, Shandong 266000, China
| | - Renmin Han
- Research Center for Mathematics and Interdisciplinary Sciences, Frontiers Science Center for Nonlinear Expectations (Ministry of Education), Shandong University, Shandong 266000, China
| |
Collapse
|
28
|
Bilous M, Hérault L, Gabriel AA, Teleman M, Gfeller D. Building and analyzing metacells in single-cell genomics data. Mol Syst Biol 2024; 20:744-766. [PMID: 38811801 PMCID: PMC11220014 DOI: 10.1038/s44320-024-00045-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2024] [Revised: 05/03/2024] [Accepted: 05/08/2024] [Indexed: 05/31/2024] Open
Abstract
The advent of high-throughput single-cell genomics technologies has fundamentally transformed biological sciences. Currently, millions of cells from complex biological tissues can be phenotypically profiled across multiple modalities. The scaling of computational methods to analyze and visualize such data is a constant challenge, and tools need to be regularly updated, if not redesigned, to cope with ever-growing numbers of cells. Over the last few years, metacells have been introduced to reduce the size and complexity of single-cell genomics data while preserving biologically relevant information and improving interpretability. Here, we review recent studies that capitalize on the concept of metacells-and the many variants in nomenclature that have been used. We further outline how and when metacells should (or should not) be used to analyze single-cell genomics data and what should be considered when analyzing such data at the metacell level. To facilitate the exploration of metacells, we provide a comprehensive tutorial on the construction and analysis of metacells from single-cell RNA-seq data ( https://github.com/GfellerLab/MetacellAnalysisTutorial ) as well as a fully integrated pipeline to rapidly build, visualize and evaluate metacells with different methods ( https://github.com/GfellerLab/MetacellAnalysisToolkit ).
Collapse
Affiliation(s)
- Mariia Bilous
- Department of Oncology, Ludwig Institute for Cancer Research Lausanne, University of Lausanne, 1011, Lausanne, Switzerland
- Agora Cancer Research Centre, 1011, Lausanne, Switzerland
- Swiss Cancer Center Leman (SCCL), Lausanne, Switzerland
- Swiss Institute of Bioinformatics (SIB), 1015, Lausanne, Switzerland
| | - Léonard Hérault
- Department of Oncology, Ludwig Institute for Cancer Research Lausanne, University of Lausanne, 1011, Lausanne, Switzerland
- Agora Cancer Research Centre, 1011, Lausanne, Switzerland
- Swiss Cancer Center Leman (SCCL), Lausanne, Switzerland
- Swiss Institute of Bioinformatics (SIB), 1015, Lausanne, Switzerland
| | - Aurélie Ag Gabriel
- Department of Oncology, Ludwig Institute for Cancer Research Lausanne, University of Lausanne, 1011, Lausanne, Switzerland
- Agora Cancer Research Centre, 1011, Lausanne, Switzerland
- Swiss Cancer Center Leman (SCCL), Lausanne, Switzerland
- Swiss Institute of Bioinformatics (SIB), 1015, Lausanne, Switzerland
| | - Matei Teleman
- Department of Oncology, Ludwig Institute for Cancer Research Lausanne, University of Lausanne, 1011, Lausanne, Switzerland
- Agora Cancer Research Centre, 1011, Lausanne, Switzerland
- Swiss Cancer Center Leman (SCCL), Lausanne, Switzerland
- Swiss Institute of Bioinformatics (SIB), 1015, Lausanne, Switzerland
| | - David Gfeller
- Department of Oncology, Ludwig Institute for Cancer Research Lausanne, University of Lausanne, 1011, Lausanne, Switzerland.
- Agora Cancer Research Centre, 1011, Lausanne, Switzerland.
- Swiss Cancer Center Leman (SCCL), Lausanne, Switzerland.
- Swiss Institute of Bioinformatics (SIB), 1015, Lausanne, Switzerland.
| |
Collapse
|
29
|
Kim GD, Shin SI, Jung SW, An H, Choi SY, Eun M, Jun CD, Lee S, Park J. Cell Type- and Age-Specific Expression of lncRNAs across Kidney Cell Types. J Am Soc Nephrol 2024; 35:870-885. [PMID: 38621182 PMCID: PMC11230714 DOI: 10.1681/asn.0000000000000354] [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: 12/06/2023] [Accepted: 04/08/2024] [Indexed: 04/17/2024] Open
Abstract
Key Points
We constructed a single-cell long noncoding RNA atlas of various tissues, including normal and aged kidneys.We identified age- and cell type–specific expression changes of long noncoding RNAs in kidney cells.
Background
Accumulated evidence demonstrates that long noncoding RNAs (lncRNAs) regulate cell differentiation and homeostasis, influencing kidney aging and disease. Despite their versatility, the function of lncRNA remains poorly understood because of the lack of a reference map of lncRNA transcriptome in various cell types.
Methods
In this study, we used a targeted single-cell RNA sequencing method to enrich and characterize lncRNAs in individual cells. We applied this method to various mouse tissues, including normal and aged kidneys.
Results
Through tissue-specific clustering analysis, we identified cell type–specific lncRNAs that showed a high correlation with known cell-type marker genes. Furthermore, we constructed gene regulatory networks to explore the functional roles of differentially expressed lncRNAs in each cell type. In the kidney, we observed dynamic expression changes of lncRNAs during aging, with specific changes in glomerular cells. These cell type– and age-specific expression patterns of lncRNAs suggest that lncRNAs may have a potential role in regulating cellular processes, such as immune response and energy metabolism, during kidney aging.
Conclusions
Our study sheds light on the comprehensive landscape of lncRNA expression and function and provides a valuable resource for future analysis of lncRNAs (https://gist-fgl.github.io/sc-lncrna-atlas/).
Collapse
Affiliation(s)
- Gyeong Dae Kim
- School of Life Sciences, Gwangju Institute of Science and Technology (GIST), Gwangju, Republic of Korea
| | - So-I Shin
- School of Life Sciences, Gwangju Institute of Science and Technology (GIST), Gwangju, Republic of Korea
| | - Su Woong Jung
- School of Life Sciences, Gwangju Institute of Science and Technology (GIST), Gwangju, Republic of Korea
- Division of Nephrology, Department of Internal Medicine, Kyung Hee University, Seoul, Republic of Korea
| | - Hyunsu An
- School of Life Sciences, Gwangju Institute of Science and Technology (GIST), Gwangju, Republic of Korea
| | - Sin Young Choi
- School of Life Sciences, Gwangju Institute of Science and Technology (GIST), Gwangju, Republic of Korea
| | - Minho Eun
- School of Life Sciences, Gwangju Institute of Science and Technology (GIST), Gwangju, Republic of Korea
| | - Chang-Duk Jun
- School of Life Sciences, Gwangju Institute of Science and Technology (GIST), Gwangju, Republic of Korea
| | - Sangho Lee
- Division of Nephrology, Department of Internal Medicine, Kyung Hee University, Seoul, Republic of Korea
| | - Jihwan Park
- School of Life Sciences, Gwangju Institute of Science and Technology (GIST), Gwangju, Republic of Korea
| |
Collapse
|
30
|
Wu X, Lu M, Yun D, Gao S, Sun F. Long-read single-cell sequencing reveals the transcriptional landscape of spermatogenesis in obstructive azoospermia and Sertoli cell-only patients. QJM 2024; 117:422-435. [PMID: 38192002 DOI: 10.1093/qjmed/hcae009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Revised: 12/16/2023] [Indexed: 01/10/2024] Open
Abstract
BACKGROUND High-throughput single-cell RNA sequencing (scRNA-seq) is widely used in spermatogenesis. However, it only reveals short reads in germ and somatic cells, limiting the discovery of novel transcripts and genes. AIM This study shows the long-read transcriptional landscape of spermatogenesis in obstructive azoospermia (OA) and Sertoli cell-only patients. DESIGN Single cells were isolated from testicular biopsies of OA and non-obstructive azoospermia (NOA) patients. Cell culture was identified by comparing PacBio long-read single-cell sequencing (OA n = 3, NOA n = 3) with short-read scRNA-seq (OA n = 6, NOA n = 6). Ten germ cell types and eight somatic cell types were classified based on known markers. METHODS PacBio long-read single-cell sequencing, short-read scRNA-seq, polymerase chain reaction. RESULTS A total of 130 426 long-read transcripts (100 517 novel transcripts and 29 909 known transcripts) and 49 508 long-read transcripts (26 002 novel transcripts and 23 506 known transcripts) have been detected in OA and NOA patients, respectively. Moreover, 36 373 and 1642 new genes are identified in OA and NOA patients, respectively. Importantly, specific expressions of long-read transcripts were detected in germ and stomatic cells during normal spermatogenesis. CONCLUSION We have identified total full-length transcripts in OA and NOA, and new genes were found. Furthermore, specific expressed full-length transcripts were detected, and the genomic structure of transcripts was mapped in different cell types. These findings may provide valuable information on human spermatogenesis and the treatment of male infertility.
Collapse
Affiliation(s)
- X Wu
- Department of Urology and Andrology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - M Lu
- Department of Urology and Andrology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - D Yun
- Institute of Reproductive Medicine, Medical School of Nantong University, Nantong, Jiangsu, China
| | - S Gao
- Institute of Reproductive Medicine, Medical School of Nantong University, Nantong, Jiangsu, China
| | - F Sun
- Department of Urology and Andrology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| |
Collapse
|
31
|
Gupta P, O’Neill H, Wolvetang E, Chatterjee A, Gupta I. Advances in single-cell long-read sequencing technologies. NAR Genom Bioinform 2024; 6:lqae047. [PMID: 38774511 PMCID: PMC11106032 DOI: 10.1093/nargab/lqae047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Revised: 04/18/2024] [Accepted: 04/29/2024] [Indexed: 05/24/2024] Open
Abstract
With an increase in accuracy and throughput of long-read sequencing technologies, they are rapidly being assimilated into the single-cell sequencing pipelines. For transcriptome sequencing, these techniques provide RNA isoform-level information in addition to the gene expression profiles. Long-read sequencing technologies not only help in uncovering complex patterns of cell-type specific splicing, but also offer unprecedented insights into the origin of cellular complexity and thus potentially new avenues for drug development. Additionally, single-cell long-read DNA sequencing enables high-quality assemblies, structural variant detection, haplotype phasing, resolving high-complexity regions, and characterization of epigenetic modifications. Given that significant progress has primarily occurred in single-cell RNA isoform sequencing (scRiso-seq), this review will delve into these advancements in depth and highlight the practical considerations and operational challenges, particularly pertaining to downstream analysis. We also aim to offer a concise introduction to complementary technologies for single-cell sequencing of the genome, epigenome and epitranscriptome. We conclude by identifying certain key areas of innovation that may drive these technologies further and foster more widespread application in biomedical science.
Collapse
Affiliation(s)
- Pallavi Gupta
- University of Queensland – IIT Delhi Research Academy, Hauz Khas, New Delhi 110016, India
- Australian Institute of Bioengineering and Nanotechnology (AIBN), The University of Queensland, St Lucia, QLD 4072, Australia
- Department of Biochemical Engineering and Biotechnology, Indian Institute of Technology Delhi, Hauz Khas, New Delhi 110016, India
| | - Hannah O’Neill
- Department of Pathology, Dunedin School of Medicine, University of Otago, 58 Hanover Street, Dunedin 9054, New Zealand
| | - Ernst J Wolvetang
- Australian Institute of Bioengineering and Nanotechnology (AIBN), The University of Queensland, St Lucia, QLD 4072, Australia
| | - Aniruddha Chatterjee
- Department of Pathology, Dunedin School of Medicine, University of Otago, 58 Hanover Street, Dunedin 9054, New Zealand
| | - Ishaan Gupta
- Department of Biochemical Engineering and Biotechnology, Indian Institute of Technology Delhi, Hauz Khas, New Delhi 110016, India
| |
Collapse
|
32
|
Joglekar A, Hu W, Zhang B, Narykov O, Diekhans M, Marrocco J, Balacco J, Ndhlovu LC, Milner TA, Fedrigo O, Jarvis ED, Sheynkman G, Korkin D, Ross ME, Tilgner HU. Single-cell long-read sequencing-based mapping reveals specialized splicing patterns in developing and adult mouse and human brain. Nat Neurosci 2024; 27:1051-1063. [PMID: 38594596 PMCID: PMC11156538 DOI: 10.1038/s41593-024-01616-4] [Citation(s) in RCA: 26] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2023] [Accepted: 03/07/2024] [Indexed: 04/11/2024]
Abstract
RNA isoforms influence cell identity and function. However, a comprehensive brain isoform map was lacking. We analyze single-cell RNA isoforms across brain regions, cell subtypes, developmental time points and species. For 72% of genes, full-length isoform expression varies along one or more axes. Splicing, transcription start and polyadenylation sites vary strongly between cell types, influence protein architecture and associate with disease-linked variation. Additionally, neurotransmitter transport and synapse turnover genes harbor cell-type variability across anatomical regions. Regulation of cell-type-specific splicing is pronounced in the postnatal day 21-to-postnatal day 28 adolescent transition. Developmental isoform regulation is stronger than regional regulation for the same cell type. Cell-type-specific isoform regulation in mice is mostly maintained in the human hippocampus, allowing extrapolation to the human brain. Conversely, the human brain harbors additional cell-type specificity, suggesting gain-of-function isoforms. Together, this detailed single-cell atlas of full-length isoform regulation across development, anatomical regions and species reveals an unappreciated degree of isoform variability across multiple axes.
Collapse
Affiliation(s)
- Anoushka Joglekar
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA
- Center for Neurogenetics, Weill Cornell Medicine, New York, NY, USA
- New York Genome Center, New York, NY, USA
| | - Wen Hu
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA
- Center for Neurogenetics, Weill Cornell Medicine, New York, NY, USA
| | - Bei Zhang
- Spatial Genomics, Inc., Pasadena, CA, USA
| | - Oleksandr Narykov
- Bioinformatics and Computational Biology Program, Worcester Polytechnic Institute, Worcester, MA, USA
- Computer Science Department, Worcester Polytechnic Institute, Worcester, MA, USA
- Data Science Program, Worcester Polytechnic Institute, Worcester, MA, USA
| | - Mark Diekhans
- UC Genomics Institute, University of California, Santa Cruz, Santa Cruz, CA, USA
| | - Jordan Marrocco
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA
- Department of Biology, Touro University, New York, NY, USA
- Laboratory of Neuroendocrinology, The Rockefeller University, New York, NY, USA
| | - Jennifer Balacco
- Vertebrate Genome Lab, The Rockefeller University, New York, NY, USA
| | - Lishomwa C Ndhlovu
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA
- Department of Medicine, Division of Infectious Diseases, Weill Cornell Medicine, New York, NY, USA
| | - Teresa A Milner
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA
| | - Olivier Fedrigo
- Vertebrate Genome Lab, The Rockefeller University, New York, NY, USA
| | - Erich D Jarvis
- Vertebrate Genome Lab, The Rockefeller University, New York, NY, USA
- Laboratory of Neurogenetics of Language, The Rockefeller University, New York, NY, USA
- Howard Hughes Medical Institute, Chevy Chase, MD, USA
| | - Gloria Sheynkman
- Department of Molecular Physiology and Biological Physics, University of Virginia, Charlottesville, VA, USA
- Department of Biochemistry and Molecular Genetics, University of Virginia, Charlottesville, VA, USA
- Center for Public Health Genomics, University of Virginia, Charlottesville, VA, USA
- UVA Comprehensive Cancer Center, University of Virginia, Charlottesville, VA, USA
| | - Dmitry Korkin
- Bioinformatics and Computational Biology Program, Worcester Polytechnic Institute, Worcester, MA, USA
- Computer Science Department, Worcester Polytechnic Institute, Worcester, MA, USA
- Data Science Program, Worcester Polytechnic Institute, Worcester, MA, USA
| | - M Elizabeth Ross
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA
- Center for Neurogenetics, Weill Cornell Medicine, New York, NY, USA
| | - Hagen U Tilgner
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA.
- Center for Neurogenetics, Weill Cornell Medicine, New York, NY, USA.
| |
Collapse
|
33
|
Camerini E, Amsen D, Kater AP, Peters FS. The complexities of T-cell dysfunction in chronic lymphocytic leukemia. Semin Hematol 2024; 61:163-171. [PMID: 38782635 DOI: 10.1053/j.seminhematol.2024.04.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Revised: 03/13/2024] [Accepted: 04/09/2024] [Indexed: 05/25/2024]
Abstract
Chronic lymphocytic leukemia (CLL) is a B-cell malignancy characterized by profound alterations and defects in the T-cell compartment. This observation has gained renewed interest as T-cell treatment strategies, which are successfully applied in more aggressive B-cell malignancies, have yielded disappointing results in CLL. Despite ongoing efforts to understand and address the observed T-cell defects, the exact mechanisms and nature underlying this dysfunction remain largely unknown. In this review, we examine the supporting signals from T cells to CLL cells in the lymph node niche, summarize key findings on T-cell functional defects, delve into potential underlying causes, and explore novel strategies for reversing these deficiencies. Our goal is to identify strategies aimed at resolving CLL-induced T-cell dysfunction which, in the future, will enhance the efficacy of autologous T-cell-based therapies for CLL patients.
Collapse
Affiliation(s)
- Elena Camerini
- Department of Experimental Immunology, Amsterdam UMC, Amsterdam, The Netherlands; Department of Hematology, Amsterdam UMC, Amsterdam, The Netherlands
| | - Derk Amsen
- Department of Experimental Immunology, Amsterdam UMC, Amsterdam, The Netherlands; Landsteiner Laboratory for Blood Cell Research at Sanquin, Amsterdam, The Netherlands
| | - Arnon P Kater
- Department of Hematology, Amsterdam UMC, Amsterdam, The Netherlands.
| | - Fleur S Peters
- Department of Experimental Immunology, Amsterdam UMC, Amsterdam, The Netherlands; Department of Hematology, Amsterdam UMC, Amsterdam, The Netherlands
| |
Collapse
|
34
|
Huang S, Shi W, Li S, Fan Q, Yang C, Cao J, Wu L. Advanced sequencing-based high-throughput and long-read single-cell transcriptome analysis. LAB ON A CHIP 2024; 24:2601-2621. [PMID: 38669201 DOI: 10.1039/d4lc00105b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/28/2024]
Abstract
Cells are the fundamental building blocks of living systems, exhibiting significant heterogeneity. The transcriptome connects the cellular genotype and phenotype, and profiling single-cell transcriptomes is critical for uncovering distinct cell types, states, and the interplay between cells in development, health, and disease. Nevertheless, single-cell transcriptome analysis faces daunting challenges due to the low abundance and diverse nature of RNAs in individual cells, as well as their heterogeneous expression. The advent and continuous advancements of next-generation sequencing (NGS) and third-generation sequencing (TGS) technologies have solved these problems and facilitated the high-throughput, sensitive, full-length, and rapid profiling of single-cell RNAs. In this review, we provide a broad introduction to current methodologies for single-cell transcriptome sequencing. First, state-of-the-art advancements in high-throughput and full-length single-cell RNA sequencing (scRNA-seq) platforms using NGS are reviewed. Next, TGS-based long-read scRNA-seq methods are summarized. Finally, a brief conclusion and perspectives for comprehensive single-cell transcriptome analysis are discussed.
Collapse
Affiliation(s)
- Shanqing Huang
- Discipline of Intelligent Instrument and Equipment, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen 361005, China
| | - Weixiong Shi
- Discipline of Intelligent Instrument and Equipment, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen 361005, China
| | - Shiyu Li
- Discipline of Intelligent Instrument and Equipment, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen 361005, China
| | - Qian Fan
- Institute of Molecular Medicine, Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China.
| | - Chaoyong Yang
- Institute of Molecular Medicine, Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China.
- Discipline of Intelligent Instrument and Equipment, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen 361005, China
| | - Jiao Cao
- Institute of Molecular Medicine, Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China.
| | - Lingling Wu
- Institute of Molecular Medicine, Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China.
| |
Collapse
|
35
|
Guérin A, Moncada-Vélez M, Jackson K, Ogishi M, Rosain J, Mancini M, Langlais D, Nunez A, Webster S, Goyette J, Khan T, Marr N, Avery DT, Rao G, Waterboer T, Michels B, Neves E, Iracema Morais C, London J, Mestrallet S, Quartier dit Maire P, Neven B, Rapaport F, Seeleuthner Y, Lev A, Simon AJ, Montoya J, Barel O, Gómez-Rodríguez J, Orrego JC, L’Honneur AS, Soudée C, Rojas J, Velez AC, Sereti I, Terrier B, Marin N, García LF, Abel L, Boisson-Dupuis S, Reis J, Marinho A, Lisco A, Faria E, Goodnow CC, Vasconcelos J, Béziat V, Ma CS, Somech R, Casanova JL, Bustamante J, Franco JL, Tangye SG. Helper T cell immunity in humans with inherited CD4 deficiency. J Exp Med 2024; 221:e20231044. [PMID: 38557723 PMCID: PMC10983808 DOI: 10.1084/jem.20231044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Revised: 01/04/2024] [Accepted: 01/31/2024] [Indexed: 04/04/2024] Open
Abstract
CD4+ T cells are vital for host defense and immune regulation. However, the fundamental role of CD4 itself remains enigmatic. We report seven patients aged 5-61 years from five families of four ancestries with autosomal recessive CD4 deficiency and a range of infections, including recalcitrant warts and Whipple's disease. All patients are homozygous for rare deleterious CD4 variants impacting expression of the canonical CD4 isoform. A shorter expressed isoform that interacts with LCK, but not HLA class II, is affected by only one variant. All patients lack CD4+ T cells and have increased numbers of TCRαβ+CD4-CD8- T cells, which phenotypically and transcriptionally resemble conventional Th cells. Finally, patient CD4-CD8- αβ T cells exhibit intact responses to HLA class II-restricted antigens and promote B cell differentiation in vitro. Thus, compensatory development of Th cells enables patients with inherited CD4 deficiency to acquire effective cellular and humoral immunity against an unexpectedly large range of pathogens. Nevertheless, CD4 is indispensable for protective immunity against at least human papillomaviruses and Trophyrema whipplei.
Collapse
Affiliation(s)
- Antoine Guérin
- Garvan Institute of Medical Research, Darlinghurst, Australia
- Faculty of Medicine and Health, School of Clinical Medicine, University of New South Wales Sydney, Sydney, Australia
| | - Marcela Moncada-Vélez
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, NY, USA
- Primary Immunodeficiencies Group, Department of Microbiology and Parasitology, School of Medicine, University of Antioquia UdeA, Medellin, Colombia
| | | | - Masato Ogishi
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, NY, USA
| | - Jérémie Rosain
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, NY, USA
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM U1163, Necker Hospital for Sick Children, Paris, France
- Paris Cité University, Imagine Institute, Paris, France
- Study Center for Primary Immunodeficiencies, Necker Hospital for Sick Children, Assistance Publique–Hôpitaux de Paris, Paris, France
| | - Mathieu Mancini
- Department of Human Genetics, McGill University, Montreal, Canada
- Department of Microbiology and Immunology, McGill University, Montreal, Canada
- Dahdaleh Institute of Genomic Medicine, McGill Research Centre on Complex Traits, McGill University, Montreal, Canada
| | - David Langlais
- Department of Human Genetics, McGill University, Montreal, Canada
- Department of Microbiology and Immunology, McGill University, Montreal, Canada
- Dahdaleh Institute of Genomic Medicine, McGill Research Centre on Complex Traits, McGill University, Montreal, Canada
| | - Andrea Nunez
- Department of Molecular Medicine, School of Biomedical Sciences, University of New South Wales, Sydney, Australia
| | - Samantha Webster
- Department of Molecular Medicine, School of Biomedical Sciences, University of New South Wales, Sydney, Australia
| | - Jesse Goyette
- Department of Molecular Medicine, School of Biomedical Sciences, University of New South Wales, Sydney, Australia
| | - Taushif Khan
- Department of Human Immunology, Sidra Medicine, Doha, Qatar
- The Jackson Laboratory, Farmington, CT, USA
| | - Nico Marr
- Department of Human Immunology, Sidra Medicine, Doha, Qatar
- College of Health and Life Sciences, Hamad Bin Khalifa University, Doha, Qatar
| | - Danielle T. Avery
- Garvan Institute of Medical Research, Darlinghurst, Australia
- Faculty of Medicine and Health, School of Clinical Medicine, University of New South Wales Sydney, Sydney, Australia
| | - Geetha Rao
- Garvan Institute of Medical Research, Darlinghurst, Australia
- Faculty of Medicine and Health, School of Clinical Medicine, University of New South Wales Sydney, Sydney, Australia
| | - Tim Waterboer
- Division of Infections and Cancer Epidemiology, German Cancer Research Center, Heidelberg, Germany
| | - Birgitta Michels
- Division of Infections and Cancer Epidemiology, German Cancer Research Center, Heidelberg, Germany
| | - Esmeralda Neves
- Immunology Department—Pathology, University Hospital Center of Porto, Porto, Portugal
- Unit for Multidisciplinary Research in Biomedicine, Institute of Biomedical Sciences Abel Salazar, University of Porto, Porto, Portugal
| | - Cátia Iracema Morais
- Immunology Department—Pathology, University Hospital Center of Porto, Porto, Portugal
- Unit for Multidisciplinary Research in Biomedicine, Institute of Biomedical Sciences Abel Salazar, University of Porto, Porto, Portugal
| | - Jonathan London
- Service of Internal Medicine, Diaconesse-Croix Saint Simon Hospital, Paris, France
| | - Stéphanie Mestrallet
- Department of Internal Medicine and Infectious Diseases, Manchester Hospital, Charleville-Mézières, France
| | - Pierre Quartier dit Maire
- Pediatric Immunology-Hematology and Rheumatology Unit, Necker Hospital for Sick Children, Paris, France
| | - Bénédicte Neven
- Pediatric Immunology-Hematology and Rheumatology Unit, Necker Hospital for Sick Children, Paris, France
| | - Franck Rapaport
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, NY, USA
| | - Yoann Seeleuthner
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM U1163, Necker Hospital for Sick Children, Paris, France
- Paris Cité University, Imagine Institute, Paris, France
| | - Atar Lev
- Department of Pediatrics and Immunology Service, Edmond and Lily Safra Children's Hospital, Sheba Medical Center, Tel Aviv School of Medicine, Tel Aviv, Israel
| | - Amos J. Simon
- Department of Pediatrics and Immunology Service, Edmond and Lily Safra Children's Hospital, Sheba Medical Center, Tel Aviv School of Medicine, Tel Aviv, Israel
| | - Jorge Montoya
- San Vicente de Paul University Hospital, Medellin, Colombia
| | - Ortal Barel
- The Genomic Unit, Sheba Cancer Research Center, Sheba Medical Center, Ramat Gan, Israel
| | - Julio Gómez-Rodríguez
- National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, USA
| | - Julio C. Orrego
- Primary Immunodeficiencies Group, Department of Microbiology and Parasitology, School of Medicine, University of Antioquia UdeA, Medellin, Colombia
| | - Anne-Sophie L’Honneur
- Department of Virology, Paris Cité University and Cochin Hospital, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Camille Soudée
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM U1163, Necker Hospital for Sick Children, Paris, France
- Paris Cité University, Imagine Institute, Paris, France
| | - Jessica Rojas
- Primary Immunodeficiencies Group, Department of Microbiology and Parasitology, School of Medicine, University of Antioquia UdeA, Medellin, Colombia
| | - Alejandra C. Velez
- Primary Immunodeficiencies Group, Department of Microbiology and Parasitology, School of Medicine, University of Antioquia UdeA, Medellin, Colombia
| | - Irini Sereti
- National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Benjamin Terrier
- Department of Internal Medicine, Cochin Hospital, Assistance Publique–Hôpitaux de Paris, Paris Cité University, Paris, France
| | - Nancy Marin
- Cellular Immunology and Immunogenetics Group, University of Antioquia UdeA, Medellin, Colombia
| | - Luis F. García
- Cellular Immunology and Immunogenetics Group, University of Antioquia UdeA, Medellin, Colombia
| | - Laurent Abel
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, NY, USA
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM U1163, Necker Hospital for Sick Children, Paris, France
- Paris Cité University, Imagine Institute, Paris, France
| | - Stéphanie Boisson-Dupuis
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, NY, USA
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM U1163, Necker Hospital for Sick Children, Paris, France
- Paris Cité University, Imagine Institute, Paris, France
| | - Joel Reis
- Dermatology Service, University Hospital Center of Porto, Porto, Portugal
| | - Antonio Marinho
- School of Medicine and Biomedical Sciences, University of Porto, Porto, Portugal
- Department of Clinical Immunology, University Hospital Center of Porto, Porto, Portugal
| | - Andrea Lisco
- National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, USA
| | - Emilia Faria
- Allergy and Clinical Immunology Department, University Hospital Center of Coimbra, Coimbra, Portugal
| | - Christopher C. Goodnow
- Garvan Institute of Medical Research, Darlinghurst, Australia
- Faculty of Medicine and Health, School of Clinical Medicine, University of New South Wales Sydney, Sydney, Australia
| | - Julia Vasconcelos
- Immunology Department—Pathology, University Hospital Center of Porto, Porto, Portugal
- Unit for Multidisciplinary Research in Biomedicine, Institute of Biomedical Sciences Abel Salazar, University of Porto, Porto, Portugal
| | - Vivien Béziat
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, NY, USA
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM U1163, Necker Hospital for Sick Children, Paris, France
- Paris Cité University, Imagine Institute, Paris, France
| | - Cindy S. Ma
- Garvan Institute of Medical Research, Darlinghurst, Australia
- Faculty of Medicine and Health, School of Clinical Medicine, University of New South Wales Sydney, Sydney, Australia
| | - Raz Somech
- Department of Pediatrics and Immunology Service, Edmond and Lily Safra Children's Hospital, Sheba Medical Center, Tel Aviv School of Medicine, Tel Aviv, Israel
| | - Jean-Laurent Casanova
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, NY, USA
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM U1163, Necker Hospital for Sick Children, Paris, France
- Paris Cité University, Imagine Institute, Paris, France
- Howard Hughes Medical Institute, New York, NY, USA
- Department of Pediatrics, Necker Hospital for Sick Children, Assistance Publique–Hôpitaux de Paris, Paris, France
| | - Jacinta Bustamante
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, NY, USA
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM U1163, Necker Hospital for Sick Children, Paris, France
- Paris Cité University, Imagine Institute, Paris, France
- Study Center for Primary Immunodeficiencies, Necker Hospital for Sick Children, Assistance Publique–Hôpitaux de Paris, Paris, France
| | - Jose Luis Franco
- Primary Immunodeficiencies Group, Department of Microbiology and Parasitology, School of Medicine, University of Antioquia UdeA, Medellin, Colombia
| | - Stuart G. Tangye
- Garvan Institute of Medical Research, Darlinghurst, Australia
- Faculty of Medicine and Health, School of Clinical Medicine, University of New South Wales Sydney, Sydney, Australia
| |
Collapse
|
36
|
Pardo-Palacios FJ, Arzalluz-Luque A, Kondratova L, Salguero P, Mestre-Tomás J, Amorín R, Estevan-Morió E, Liu T, Nanni A, McIntyre L, Tseng E, Conesa A. SQANTI3: curation of long-read transcriptomes for accurate identification of known and novel isoforms. Nat Methods 2024; 21:793-797. [PMID: 38509328 PMCID: PMC11093726 DOI: 10.1038/s41592-024-02229-2] [Citation(s) in RCA: 51] [Impact Index Per Article: 51.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Accepted: 03/01/2024] [Indexed: 03/22/2024]
Abstract
SQANTI3 is a tool designed for the quality control, curation and annotation of long-read transcript models obtained with third-generation sequencing technologies. Leveraging its annotation framework, SQANTI3 calculates quality descriptors of transcript models, junctions and transcript ends. With this information, potential artifacts can be identified and replaced with reliable sequences. Furthermore, the integrated functional annotation feature enables subsequent functional iso-transcriptomics analyses.
Collapse
Affiliation(s)
- Francisco J Pardo-Palacios
- Institute for Integrative Systems Biology, Spanish National Research Council, Paterna, Valencia, Spain
- Department of Applied Statistics and Operational Research, and Quality, Universitat Politècnica de València, Valencia, Valencia, Spain
| | - Angeles Arzalluz-Luque
- Institute for Integrative Systems Biology, Spanish National Research Council, Paterna, Valencia, Spain
- Department of Applied Statistics and Operational Research, and Quality, Universitat Politècnica de València, Valencia, Valencia, Spain
| | - Liudmyla Kondratova
- Horticultural Sciences Department, University of Florida, Gainesville, FL, USA
- Genetics Institute, University of Florida, Gainesville, FL, USA
| | - Pedro Salguero
- Department of Applied Statistics and Operational Research, and Quality, Universitat Politècnica de València, Valencia, Valencia, Spain
| | - Jorge Mestre-Tomás
- Institute for Integrative Systems Biology, Spanish National Research Council, Paterna, Valencia, Spain
| | - Rocío Amorín
- Genetics Institute, University of Florida, Gainesville, FL, USA
- Department of Microbiology and Cell Science, University of Florida, Gainesville, FL, USA
| | - Eva Estevan-Morió
- Institute for Integrative Systems Biology, Spanish National Research Council, Paterna, Valencia, Spain
| | - Tianyuan Liu
- Institute for Integrative Systems Biology, Spanish National Research Council, Paterna, Valencia, Spain
| | - Adalena Nanni
- Department of Molecular Genetics and Microbiology, University of Florida, Gainesville, FL, USA
| | - Lauren McIntyre
- Genetics Institute, University of Florida, Gainesville, FL, USA
- Department of Molecular Genetics and Microbiology, University of Florida, Gainesville, FL, USA
| | | | - Ana Conesa
- Institute for Integrative Systems Biology, Spanish National Research Council, Paterna, Valencia, Spain.
| |
Collapse
|
37
|
Nishimura K, Saika W, Inoue D. Minor introns impact on hematopoietic malignancies. Exp Hematol 2024; 132:104173. [PMID: 38309573 DOI: 10.1016/j.exphem.2024.104173] [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: 11/26/2023] [Revised: 12/25/2023] [Accepted: 01/03/2024] [Indexed: 02/05/2024]
Abstract
In the intricate orchestration of the central dogma, pre-mRNA splicing plays a crucial role in the post-transcriptional process that transforms DNA into mature mRNA. Widely acknowledged as a pivotal RNA processing step, it significantly influences gene expression and alters the functionality of gene product proteins. Although U2-dependent spliceosomes efficiently manage the removal of over 99% of introns, a distinct subset of essential genes undergo splicing with a different intron type, denoted as minor introns, using U12-dependent spliceosomes. Mutations in spliceosome component genes are now recognized as prevalent genetic abnormalities in cancer patients, especially those with hematologic malignancies. Despite the relative rarity of minor introns, genes containing them are evolutionarily conserved and play crucial roles in functions such as the RAS-MAPK pathway. Disruptions in U12-type minor intron splicing caused by mutations in snRNA or its regulatory components significantly contribute to cancer progression. Notably, recurrent mutations associated with myelodysplastic syndrome (MDS) in the minor spliceosome component ZRSR2 underscore its significance. Examination of ZRSR2-mutated MDS cells has revealed that only a subset of minor spliceosome-dependent genes, such as LZTR1, consistently exhibit missplicing. Recent technological advancements have uncovered insights into minor introns, raising inquiries beyond current understanding. This review comprehensively explores the importance of minor intron regulation, the molecular implications of minor (U12-type) spliceosomal mutations and cis-regulatory regions, and the evolutionary progress of studies on minor, aiming to provide a sophisticated understanding of their intricate role in cancer biology.
Collapse
Affiliation(s)
- Koutarou Nishimura
- Department of Hematology-Oncology, Institute of Biomedical Research and Innovation, Foundation for Biomedical Research and Innovation at Kobe, Kobe, Hyogo, Japan.
| | - Wataru Saika
- Department of Hematology-Oncology, Institute of Biomedical Research and Innovation, Foundation for Biomedical Research and Innovation at Kobe, Kobe, Hyogo, Japan; Department of Hematology, Shiga University of Medical Science, Ōtsu, Shiga, Japan
| | - Daichi Inoue
- Department of Hematology-Oncology, Institute of Biomedical Research and Innovation, Foundation for Biomedical Research and Innovation at Kobe, Kobe, Hyogo, Japan.
| |
Collapse
|
38
|
Kim HS, Kweon J, Kim Y. Recent advances in CRISPR-based functional genomics for the study of disease-associated genetic variants. Exp Mol Med 2024; 56:861-869. [PMID: 38556550 PMCID: PMC11058232 DOI: 10.1038/s12276-024-01212-3] [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: 07/31/2023] [Revised: 01/15/2024] [Accepted: 01/30/2024] [Indexed: 04/02/2024] Open
Abstract
Advances in sequencing technology have greatly increased our ability to gather genomic data, yet understanding the impact of genetic mutations, particularly variants of uncertain significance (VUSs), remains a challenge in precision medicine. The CRISPR‒Cas system has emerged as a pivotal tool for genome engineering, enabling the precise incorporation of specific genetic variations, including VUSs, into DNA to facilitate their functional characterization. Additionally, the integration of CRISPR‒Cas technology with sequencing tools allows the high-throughput evaluation of mutations, transforming uncertain genetic data into actionable insights. This allows researchers to comprehensively study the functional consequences of point mutations, paving the way for enhanced understanding and increasing application to precision medicine. This review summarizes the current genome editing tools utilizing CRISPR‒Cas systems and their combination with sequencing tools for functional genomics, with a focus on point mutations.
Collapse
Affiliation(s)
- Heon Seok Kim
- Department of Life Science, College of Natural Sciences, Hanyang University, Seoul, Republic of Korea
- Hanyang Institute of Bioscience and Biotechnology, Hanyang University, Seoul, Republic of Korea
- Hanyang Institute of Advanced BioConvergence, Hanyang University, Seongdong-gu, Seoul, Republic of Korea
| | - Jiyeon Kweon
- Department of Cell and Genetic Engineering, Asan Medical Institute of Convergence Science and Technology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Yongsub Kim
- Department of Cell and Genetic Engineering, Asan Medical Institute of Convergence Science and Technology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea.
- Stem Cell Immunomodulation Research Center, University of Ulsan College of Medicine, Seoul, Republic of Korea.
| |
Collapse
|
39
|
Yuan CU, Quah FX, Hemberg M. Single-cell and spatial transcriptomics: Bridging current technologies with long-read sequencing. Mol Aspects Med 2024; 96:101255. [PMID: 38368637 DOI: 10.1016/j.mam.2024.101255] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Revised: 01/30/2024] [Accepted: 02/07/2024] [Indexed: 02/20/2024]
Abstract
Single-cell technologies have transformed biomedical research over the last decade, opening up new possibilities for understanding cellular heterogeneity, both at the genomic and transcriptomic level. In addition, more recent developments of spatial transcriptomics technologies have made it possible to profile cells in their tissue context. In parallel, there have been substantial advances in sequencing technologies, and the third generation of methods are able to produce reads that are tens of kilobases long, with error rates matching the second generation short reads. Long reads technologies make it possible to better map large genome rearrangements and quantify isoform specific abundances. This further improves our ability to characterize functionally relevant heterogeneity. Here, we show how researchers have begun to combine single-cell, spatial transcriptomics, and long-read technologies, and how this is resulting in powerful new approaches to profiling both the genome and the transcriptome. We discuss the achievements so far, and we highlight remaining challenges and opportunities.
Collapse
Affiliation(s)
- Chengwei Ulrika Yuan
- Department of Biochemistry, University of Cambridge, Cambridge, UK; Wellcome Sanger Institute, Wellcome Genome Campus, Cambridge, UK
| | - Fu Xiang Quah
- Department of Biochemistry, University of Cambridge, Cambridge, UK
| | - Martin Hemberg
- Gene Lay Institute, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
40
|
Beaulaurier J, Ly L, Duty JA, Tyer C, Stevens C, Hung CT, Sookdeo A, Drong AW, Kowdle S, Turner DJ, Juul S, Hickey S, Lee B. De novo antibody discovery in human blood from full-length single B cell transcriptomics and matching haplotyped-resolved germline assemblies. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.26.586834. [PMID: 38585716 PMCID: PMC10996687 DOI: 10.1101/2024.03.26.586834] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/09/2024]
Abstract
Immunoglobulin (IGH, IGK, IGL) loci in the human genome are highly polymorphic regions that encode the building blocks of the light and heavy chain IG proteins that dimerize to form antibodies. The processes of V(D)J recombination and somatic hypermutation in B cells are responsible for creating an enormous reservoir of highly specific antibodies capable of binding a vast array of possible antigens. However, the antibody repertoire is fundamentally limited by the set of variable (V), diversity (D), and joining (J) alleles present in the germline IG loci. To better understand how the germline IG haplotypes contribute to the expressed antibody repertoire, we combined genome sequencing of the germline IG loci with single-cell transcriptome sequencing of B cells from the same donor. Sequencing and assembly of the germline IG loci captured the IGH locus in a single fully-phased contig where the maternal and paternal contributions to the germline V, D, and J repertoire can be fully resolved. The B cells were collected following a measles, mumps, and rubella (MMR) vaccination, resulting in a population of cells that were activated in response to this specific immune challenge. Single-cell, full-length transcriptome sequencing of these B cells resulted in whole transcriptome characterization of each cell, as well as highly-accurate consensus sequences for the somatically rearranged and hypermutated light and heavy chain IG transcripts. A subset of antibodies synthesized based on their consensus heavy and light chain transcript sequences demonstrated binding to measles antigens and neutralization of measles live virus.
Collapse
|
41
|
Abdullah L, Emiliani FE, Vaidya CM, Stuart H, Kolling FW, Ackerman ME, Song L, McKenna A, Huang YH. Hierarchal single-cell lineage tracing reveals differential fate commitment of CD8 T-cell clones in response to acute infection. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.21.586160. [PMID: 38585810 PMCID: PMC10996474 DOI: 10.1101/2024.03.21.586160] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/09/2024]
Abstract
Generating balanced populations of CD8 effector and memory T cells is necessary for immediate and durable immunity to infections and cancer. Yet, a definitive understanding of CD8 differentiation remains unclear. We used CARLIN, a processive lineage recording mouse model with single-cell RNA-seq and TCR-seq to track endogenous antigen-specific CD8 T cells during acute viral infection. We identified a diverse repertoire of expanded T-cell clones represented by seven transcriptional states. TCR enrichment analysis revealed differential memory- or effector-fate biases within clonal populations. Shared Vb segments and amino acid motifs were found within biased categories despite high TCR diversity. Using single-cell CARLIN barcode-seq we tracked multi-generational clones and found that unlike unbiased or memory-biased clones, which stably retain their fate profiles, effector-biased clones could adopt memory- or effector-bias within subclones. Collectively, our study demonstrates that a heterogenous T-cell repertoire specific for a shared antigen is composed of clones with distinct TCR-intrinsic fate-biases.
Collapse
Affiliation(s)
- Leena Abdullah
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Lebanon, NH 03756, USA
| | - Francesco E. Emiliani
- Department of Molecular and Systems Biology, Geisel School of Medicine at Dartmouth, Lebanon, NH 03756, USA
| | - Chinmay M. Vaidya
- Thayer School of Engineering, Dartmouth College, Hanover, NH 03755, USA
| | - Hannah Stuart
- Department of Molecular and Systems Biology, Geisel School of Medicine at Dartmouth, Lebanon, NH 03756, USA
| | | | - Margaret E. Ackerman
- Thayer School of Engineering, Dartmouth College, Hanover, NH 03755, USA
- Dartmouth Cancer Center, Lebanon, NH 03756, USA
| | - Li Song
- Department of Biomedical Data Science, Geisel School of Medicine at Dartmouth, Lebanon, NH 03756, USA
- Dartmouth Cancer Center, Lebanon, NH 03756, USA
| | - Aaron McKenna
- Department of Molecular and Systems Biology, Geisel School of Medicine at Dartmouth, Lebanon, NH 03756, USA
- Dartmouth Cancer Center, Lebanon, NH 03756, USA
| | - Yina H. Huang
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Lebanon, NH 03756, USA
- Dartmouth Cancer Center, Lebanon, NH 03756, USA
- Department of Pathology and Laboratory Medicine, Dartmouth Health, Lebanon, NH 03756, USA
| |
Collapse
|
42
|
Jones EF, Haldar A, Oza VH, Lasseigne BN. Quantifying transcriptome diversity: a review. Brief Funct Genomics 2024; 23:83-94. [PMID: 37225889 PMCID: PMC11484519 DOI: 10.1093/bfgp/elad019] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Revised: 04/14/2023] [Accepted: 05/05/2023] [Indexed: 05/26/2023] Open
Abstract
Following the central dogma of molecular biology, gene expression heterogeneity can aid in predicting and explaining the wide variety of protein products, functions and, ultimately, heterogeneity in phenotypes. There is currently overlapping terminology used to describe the types of diversity in gene expression profiles, and overlooking these nuances can misrepresent important biological information. Here, we describe transcriptome diversity as a measure of the heterogeneity in (1) the expression of all genes within a sample or a single gene across samples in a population (gene-level diversity) or (2) the isoform-specific expression of a given gene (isoform-level diversity). We first overview modulators and quantification of transcriptome diversity at the gene level. Then, we discuss the role alternative splicing plays in driving transcript isoform-level diversity and how it can be quantified. Additionally, we overview computational resources for calculating gene-level and isoform-level diversity for high-throughput sequencing data. Finally, we discuss future applications of transcriptome diversity. This review provides a comprehensive overview of how gene expression diversity arises, and how measuring it determines a more complete picture of heterogeneity across proteins, cells, tissues, organisms and species.
Collapse
Affiliation(s)
- Emma F Jones
- The Department of Cell, Developmental and Integrative Biology, Heersink School of Medicine, The University of Alabama at Birmingham, Birmingham, AL, USA
| | - Anisha Haldar
- The Department of Cell, Developmental and Integrative Biology, Heersink School of Medicine, The University of Alabama at Birmingham, Birmingham, AL, USA
| | - Vishal H Oza
- The Department of Cell, Developmental and Integrative Biology, Heersink School of Medicine, The University of Alabama at Birmingham, Birmingham, AL, USA
| | - Brittany N Lasseigne
- The Department of Cell, Developmental and Integrative Biology, Heersink School of Medicine, The University of Alabama at Birmingham, Birmingham, AL, USA
| |
Collapse
|
43
|
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
|
44
|
Hoffmann M, Willruth LL, Dietrich A, Lee HK, Knabl L, Trummer N, Baumbach J, Furth PA, Hennighausen L, List M. Blood transcriptomics analysis offers insights into variant-specific immune response to SARS-CoV-2. Sci Rep 2024; 14:2808. [PMID: 38307916 PMCID: PMC10837437 DOI: 10.1038/s41598-024-53117-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: 11/03/2023] [Accepted: 01/28/2024] [Indexed: 02/04/2024] Open
Abstract
Bulk RNA sequencing (RNA-seq) of blood is typically used for gene expression analysis in biomedical research but is still rarely used in clinical practice. In this study, we propose that RNA-seq should be considered a diagnostic tool, as it offers not only insights into aberrant gene expression and splicing but also delivers additional readouts on immune cell type composition as well as B-cell and T-cell receptor (BCR/TCR) repertoires. We demonstrate that RNA-seq offers insights into a patient's immune status via integrative analysis of RNA-seq data from patients infected with various SARS-CoV-2 variants (in total 196 samples with up to 200 million reads sequencing depth). We compare the results of computational cell-type deconvolution methods (e.g., MCP-counter, xCell, EPIC, quanTIseq) to complete blood count data, the current gold standard in clinical practice. We observe varying levels of lymphocyte depletion and significant differences in neutrophil levels between SARS-CoV-2 variants. Additionally, we identify B and T cell receptor (BCR/TCR) sequences using the tools MiXCR and TRUST4 to show that-combined with sequence alignments and BLASTp-they could be used to classify a patient's disease. Finally, we investigated the sequencing depth required for such analyses and concluded that 10 million reads per sample is sufficient. In conclusion, our study reveals that computational cell-type deconvolution and BCR/TCR methods using bulk RNA-seq analyses can supplement missing CBC data and offer insights into immune responses, disease severity, and pathogen-specific immunity, all achievable with a sequencing depth of 10 million reads per sample.
Collapse
Affiliation(s)
- Markus Hoffmann
- Data Science in Systems Biomedicine, TUM School of Life Sciences, Technical University of Munich, Freising, Germany.
- Institute for Advanced Study, Technical University of Munich, Lichtenbergstrasse 2 a, 85748, Garching, Germany.
- National Institute of Diabetes, Digestive, and Kidney Diseases, Bethesda, MD, 20892, USA.
| | - Lina-Liv Willruth
- Data Science in Systems Biomedicine, TUM School of Life Sciences, Technical University of Munich, Freising, Germany
| | - Alexander Dietrich
- Data Science in Systems Biomedicine, TUM School of Life Sciences, Technical University of Munich, Freising, Germany
| | - Hye Kyung Lee
- National Institute of Diabetes, Digestive, and Kidney Diseases, Bethesda, MD, 20892, USA
| | | | - Nico Trummer
- Data Science in Systems Biomedicine, TUM School of Life Sciences, Technical University of Munich, Freising, Germany
| | - Jan Baumbach
- Chair of Computational Systems Biology, University of Hamburg, Hamburg, Germany
- Computational BioMedicine Lab, University of Southern Denmark, Odense, Denmark
| | - Priscilla A Furth
- Institute for Advanced Study, Technical University of Munich, Lichtenbergstrasse 2 a, 85748, Garching, Germany
- National Institute of Diabetes, Digestive, and Kidney Diseases, Bethesda, MD, 20892, USA
- Departments of Oncology & Medicine, Georgetown University, Washington, DC, USA
| | - Lothar Hennighausen
- Institute for Advanced Study, Technical University of Munich, Lichtenbergstrasse 2 a, 85748, Garching, Germany
- National Institute of Diabetes, Digestive, and Kidney Diseases, Bethesda, MD, 20892, USA
| | - Markus List
- Data Science in Systems Biomedicine, TUM School of Life Sciences, Technical University of Munich, Freising, Germany.
| |
Collapse
|
45
|
Karaoğlanoğlu F, Orabi B, Flannigan R, Chauve C, Hach F. TKSM: highly modular, user-customizable, and scalable transcriptomic sequencing long-read simulator. Bioinformatics 2024; 40:btae051. [PMID: 38273664 PMCID: PMC10868325 DOI: 10.1093/bioinformatics/btae051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Revised: 01/10/2024] [Accepted: 01/23/2024] [Indexed: 01/27/2024] Open
Abstract
MOTIVATION Transcriptomic long-read (LR) sequencing is an increasingly cost-effective technology for probing various RNA features. Numerous tools have been developed to tackle various transcriptomic sequencing tasks (e.g. isoform and gene fusion detection). However, the lack of abundant gold-standard datasets hinders the benchmarking of such tools. Therefore, the simulation of LR sequencing is an important and practical alternative. While the existing LR simulators aim to imitate the sequencing machine noise and to target specific library protocols, they lack some important library preparation steps (e.g. PCR) and are difficult to modify to new and changing library preparation techniques (e.g. single-cell LRs). RESULTS We present TKSM, a modular and scalable LR simulator, designed so that each RNA modification step is targeted explicitly by a specific module. This allows the user to assemble a simulation pipeline as a combination of TKSM modules to emulate a specific sequencing design. Additionally, the input/output of all the core modules of TKSM follows the same simple format (Molecule Description Format) allowing the user to easily extend TKSM with new modules targeting new library preparation steps. AVAILABILITY AND IMPLEMENTATION TKSM is available as an open source software at https://github.com/vpc-ccg/tksm.
Collapse
Affiliation(s)
- Fatih Karaoğlanoğlu
- Computing Science Department, Simon Fraser University, Burnaby, BC V5A 1S6, Canada
| | - Baraa Orabi
- Department of Computer Science, the University of British Columbia, Vancouver, BC V6T 1Z4, Canada
| | - Ryan Flannigan
- Department of Urologic Sciences, the University of British Columbia, Vancouver, BC V5Z 1M9, Canada
- Vancouver Prostate Centre, Vancouver, BC V6H 3Z6, Canada
| | - Cedric Chauve
- Department of Mathematics, Simon Fraser University, Burnaby, BC V5A 1S6, Canada
| | - Faraz Hach
- Department of Computer Science, the University of British Columbia, Vancouver, BC V6T 1Z4, Canada
- Department of Urologic Sciences, the University of British Columbia, Vancouver, BC V5Z 1M9, Canada
- Vancouver Prostate Centre, Vancouver, BC V6H 3Z6, Canada
| |
Collapse
|
46
|
Chang JT, Liu LB, Wang PG, An J. Single-cell RNA sequencing to understand host-virus interactions. Virol Sin 2024; 39:1-8. [PMID: 38008383 PMCID: PMC10877424 DOI: 10.1016/j.virs.2023.11.009] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Accepted: 11/23/2023] [Indexed: 11/28/2023] Open
Abstract
Single-cell RNA sequencing (scRNA-seq) has allowed for the profiling of host and virus transcripts and host-virus interactions at single-cell resolution. This review summarizes the existing scRNA-seq technologies together with their strengths and weaknesses. The applications of scRNA-seq in various virological studies are discussed in depth, which broaden the understanding of the immune atlas, host-virus interactions, and immune repertoire. scRNA-seq can be widely used for virology in the near future to better understand the pathogenic mechanisms and discover more effective therapeutic strategies.
Collapse
Affiliation(s)
- Jia-Tong Chang
- Department of Microbiology, School of Basic Medical Sciences, Capital Medical University, Beijing 100069, China
| | - Li-Bo Liu
- Department of Microbiology, School of Basic Medical Sciences, Capital Medical University, Beijing 100069, China
| | - Pei-Gang Wang
- Department of Microbiology, School of Basic Medical Sciences, Capital Medical University, Beijing 100069, China.
| | - Jing An
- Department of Microbiology, School of Basic Medical Sciences, Capital Medical University, Beijing 100069, China.
| |
Collapse
|
47
|
Addala V, Newell F, Pearson JV, Redwood A, Robinson BW, Creaney J, Waddell N. Computational immunogenomic approaches to predict response to cancer immunotherapies. Nat Rev Clin Oncol 2024; 21:28-46. [PMID: 37907723 DOI: 10.1038/s41571-023-00830-6] [Citation(s) in RCA: 26] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/03/2023] [Indexed: 11/02/2023]
Abstract
Cancer immunogenomics is an emerging field that bridges genomics and immunology. The establishment of large-scale genomic collaborative efforts along with the development of new single-cell transcriptomic techniques and multi-omics approaches have enabled characterization of the mutational and transcriptional profiles of many cancer types and helped to identify clinically actionable alterations as well as predictive and prognostic biomarkers. Researchers have developed computational approaches and machine learning algorithms to accurately obtain clinically useful information from genomic and transcriptomic sequencing data from bulk tissue or single cells and explore tumours and their microenvironment. The rapid growth in sequencing and computational approaches has resulted in the unmet need to understand their true potential and limitations in enabling improvements in the management of patients with cancer who are receiving immunotherapies. In this Review, we describe the computational approaches currently available to analyse bulk tissue and single-cell sequencing data from cancer, stromal and immune cells, as well as how best to select the most appropriate tool to address various clinical questions and, ultimately, improve patient outcomes.
Collapse
Affiliation(s)
- Venkateswar Addala
- Cancer Program, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia.
- Faculty of Medicine, The University of Queensland, Brisbane, Queensland, Australia.
| | - Felicity Newell
- Cancer Program, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - John V Pearson
- Cancer Program, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - Alec Redwood
- National Centre for Asbestos Related Diseases, University of Western Australia, Perth, Western Australia, Australia
- Institute of Respiratory Health, Perth, Western Australia, Australia
- School of Biomedical Science, University of Western Australia, Perth, Western Australia, Australia
| | - Bruce W Robinson
- National Centre for Asbestos Related Diseases, University of Western Australia, Perth, Western Australia, Australia
- Institute of Respiratory Health, Perth, Western Australia, Australia
- Department of Respiratory Medicine, Sir Charles Gairdner Hospital, Perth, Western Australia, Australia
- Medical School, University of Western Australia, Perth, Western Australia, Australia
| | - Jenette Creaney
- National Centre for Asbestos Related Diseases, University of Western Australia, Perth, Western Australia, Australia
- Institute of Respiratory Health, Perth, Western Australia, Australia
- School of Biomedical Science, University of Western Australia, Perth, Western Australia, Australia
- Department of Respiratory Medicine, Sir Charles Gairdner Hospital, Perth, Western Australia, Australia
| | - Nicola Waddell
- Cancer Program, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia.
- Faculty of Medicine, The University of Queensland, Brisbane, Queensland, Australia.
| |
Collapse
|
48
|
Sidiropoulos DN, Ho WJ, Jaffee EM, Kagohara LT, Fertig EJ. Systems immunology spanning tumors, lymph nodes, and periphery. CELL REPORTS METHODS 2023; 3:100670. [PMID: 38086385 PMCID: PMC10753389 DOI: 10.1016/j.crmeth.2023.100670] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/06/2023] [Revised: 10/20/2023] [Accepted: 11/17/2023] [Indexed: 12/21/2023]
Abstract
The immune system defines a complex network of tissues and cell types that orchestrate responses across the body in a dynamic manner. The local and systemic interactions between immune and cancer cells contribute to disease progression. Lymphocytes are activated in lymph nodes, traffic through the periphery, and impact cancer progression through their interactions with tumor cells. As a result, therapeutic response and resistance are mediated across tissues, and a comprehensive understanding of lymphocyte dynamics requires a systems-level approach. In this review, we highlight experimental and computational methods that can leverage the study of leukocyte trafficking through an immunomics lens and reveal how adaptive immunity shapes cancer.
Collapse
Affiliation(s)
- Dimitrios N Sidiropoulos
- Johns Hopkins University School of Medicine, Baltimore, MD, USA; Johns Hopkins Convergence Institute, Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD, USA; Johns Hopkins Bloomberg Kimmel Institute for Immunotherapy, Johns Hopkins Medicine, Baltimore, MD, USA; Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins Medicine, Baltimore, MD, USA
| | - Won Jin Ho
- Johns Hopkins Convergence Institute, Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD, USA; Johns Hopkins Bloomberg Kimmel Institute for Immunotherapy, Johns Hopkins Medicine, Baltimore, MD, USA; Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins Medicine, Baltimore, MD, USA
| | - Elizabeth M Jaffee
- Johns Hopkins Convergence Institute, Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD, USA; Johns Hopkins Bloomberg Kimmel Institute for Immunotherapy, Johns Hopkins Medicine, Baltimore, MD, USA; Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins Medicine, Baltimore, MD, USA
| | - Luciane T Kagohara
- Johns Hopkins Convergence Institute, Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD, USA; Johns Hopkins Bloomberg Kimmel Institute for Immunotherapy, Johns Hopkins Medicine, Baltimore, MD, USA; Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins Medicine, Baltimore, MD, USA.
| | - Elana J Fertig
- Johns Hopkins Convergence Institute, Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD, USA; Johns Hopkins Bloomberg Kimmel Institute for Immunotherapy, Johns Hopkins Medicine, Baltimore, MD, USA; Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins Medicine, Baltimore, MD, USA; Department of Applied Mathematics and Statistics, Johns Hopkins University, Baltimore, MD, USA; Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
49
|
Zhu B, Wang Y, Ku LT, van Dijk D, Zhang L, Hafler DA, Zhao H. scNAT: a deep learning method for integrating paired single-cell RNA and T cell receptor sequencing profiles. Genome Biol 2023; 24:292. [PMID: 38111007 PMCID: PMC10726524 DOI: 10.1186/s13059-023-03129-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: 05/30/2023] [Accepted: 11/27/2023] [Indexed: 12/20/2023] Open
Abstract
Many deep learning-based methods have been proposed to handle complex single-cell data. Deep learning approaches may also prove useful to jointly analyze single-cell RNA sequencing (scRNA-seq) and single-cell T cell receptor sequencing (scTCR-seq) data for novel discoveries. We developed scNAT, a deep learning method that integrates paired scRNA-seq and scTCR-seq data to represent data in a unified latent space for downstream analysis. We demonstrate that scNAT is capable of removing batch effects, and identifying cell clusters and a T cell migration trajectory from blood to cerebrospinal fluid in multiple sclerosis.
Collapse
Affiliation(s)
- Biqing Zhu
- Program of Computational Biology and Bioinformatics, Yale University, New Haven, CT, 06511, USA
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, USA, MD , 20815
| | - Yuge Wang
- Department of Biostatistics, School of Public Health, Yale University, New Haven, CT, 06511, USA
| | - Li-Ting Ku
- Department of Biostatistics, School of Public Health, Yale University, New Haven, CT, 06511, USA
| | - David van Dijk
- Department of Internal Medicine, Yale School of Medicine, New Haven, CT, 06511, USA
- Department of Computer Science, Yale University, New Haven, CT, 06511, USA
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, USA, MD , 20815
| | - Le Zhang
- Department of Neuroscience, School of Medicine, Yale University, New Haven, CT, 06511, USA
- Department of Immunobiology, School of Medicine, Yale University, New Haven, CT, 06511, USA
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, USA, MD , 20815
| | - David A Hafler
- Department of Neurology, School of Medicine, Yale University, New Haven, CT, 06511, USA
- Department of Immunobiology, School of Medicine, Yale University, New Haven, CT, 06511, USA
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, USA, MD , 20815
| | - Hongyu Zhao
- Program of Computational Biology and Bioinformatics, Yale University, New Haven, CT, 06511, USA.
- Department of Biostatistics, School of Public Health, Yale University, New Haven, CT, 06511, USA.
| |
Collapse
|
50
|
Zhang C, Fang Y, Chen W, Chen Z, Zhang Y, Xie Y, Chen W, Xie Z, Guo M, Wang J, Tan C, Wang H, Tang C. Improving the RNA velocity approach with single-cell RNA lifecycle (nascent, mature and degrading RNAs) sequencing technologies. Nucleic Acids Res 2023; 51:e112. [PMID: 37941145 PMCID: PMC10711548 DOI: 10.1093/nar/gkad969] [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: 04/05/2023] [Revised: 09/27/2023] [Accepted: 10/14/2023] [Indexed: 11/10/2023] Open
Abstract
We presented an experimental method called FLOUR-seq, which combines BD Rhapsody and nanopore sequencing to detect the RNA lifecycle (including nascent, mature, and degrading RNAs) in cells. Additionally, we updated our HIT-scISOseq V2 to discover a more accurate RNA lifecycle using 10x Chromium and Pacbio sequencing. Most importantly, to explore how single-cell full-length RNA sequencing technologies could help improve the RNA velocity approach, we introduced a new algorithm called 'Region Velocity' to more accurately configure cellular RNA velocity. We applied this algorithm to study spermiogenesis and compared the performance of FLOUR-seq with Pacbio-based HIT-scISOseq V2. Our findings demonstrated that 'Region Velocity' is more suitable for analyzing single-cell full-length RNA data than traditional RNA velocity approaches. These novel methods could be useful for researchers looking to discover full-length RNAs in single cells and comprehensively monitor RNA lifecycle in cells.
Collapse
Affiliation(s)
| | | | - Weitian Chen
- BGI, Shenzhen 518000, China
- BGI Education Center, University of Chinese Academy of Sciences, Shenzhen 518083, China
| | | | - Ying Zhang
- Guangdong Provincial Reproductive Science Institute (Guangdong Provincial Fertility Hospital), Guangzhou, China; NHC Key Laboratory of Male Reproduction and Genetics, Guangzhou, China
| | | | | | | | | | | | | | | | | |
Collapse
|