1
|
Lederer AR, Leonardi M, Talamanca L, Bobrovskiy DM, Herrera A, Droin C, Khven I, Carvalho HJF, Valente A, Dominguez Mantes A, Mulet Arabí P, Pinello L, Naef F, La Manno G. Statistical inference with a manifold-constrained RNA velocity model uncovers cell cycle speed modulations. Nat Methods 2024:10.1038/s41592-024-02471-8. [PMID: 39482463 DOI: 10.1038/s41592-024-02471-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Accepted: 09/15/2024] [Indexed: 11/03/2024]
Abstract
Across biological systems, cells undergo coordinated changes in gene expression, resulting in transcriptome dynamics that unfold within a low-dimensional manifold. While low-dimensional dynamics can be extracted using RNA velocity, these algorithms can be fragile and rely on heuristics lacking statistical control. Moreover, the estimated vector field is not dynamically consistent with the traversed gene expression manifold. To address these challenges, we introduce a Bayesian model of RNA velocity that couples velocity field and manifold estimation in a reformulated, unified framework, identifying the parameters of an explicit dynamical system. Focusing on the cell cycle, we implement VeloCycle to study gene regulation dynamics on one-dimensional periodic manifolds and validate its ability to infer cell cycle periods using live imaging. We also apply VeloCycle to reveal speed differences in regionally defined progenitors and Perturb-seq gene knockdowns. Overall, VeloCycle expands the single-cell RNA sequencing analysis toolkit with a modular and statistically consistent RNA velocity inference framework.
Collapse
Affiliation(s)
- Alex R Lederer
- Laboratory of Brain Development and Biological Data Science, Brain Mind Institute, Faculty of Life Sciences, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Maxine Leonardi
- Laboratory of Computational and Systems Biology, Institute of Bioengineering, Faculty of Life Sciences, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Lorenzo Talamanca
- Laboratory of Computational and Systems Biology, Institute of Bioengineering, Faculty of Life Sciences, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Daniil M Bobrovskiy
- Laboratory of Brain Development and Biological Data Science, Brain Mind Institute, Faculty of Life Sciences, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Antonio Herrera
- Laboratory of Brain Development and Biological Data Science, Brain Mind Institute, Faculty of Life Sciences, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Colas Droin
- Laboratory of Computational and Systems Biology, Institute of Bioengineering, Faculty of Life Sciences, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Irina Khven
- Laboratory of Brain Development and Biological Data Science, Brain Mind Institute, Faculty of Life Sciences, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Hugo J F Carvalho
- Laboratory of Computational and Systems Biology, Institute of Bioengineering, Faculty of Life Sciences, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Alessandro Valente
- Laboratory of Brain Development and Biological Data Science, Brain Mind Institute, Faculty of Life Sciences, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Albert Dominguez Mantes
- Laboratory of Brain Development and Biological Data Science, Brain Mind Institute, Faculty of Life Sciences, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
- Laboratory of Bioimage Analysis and Computational Microscopy, Institute of Bioengineering, Faculty of Life Sciences, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Pau Mulet Arabí
- Laboratory of Computational and Systems Biology, Institute of Bioengineering, Faculty of Life Sciences, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Luca Pinello
- Molecular Pathology Unit, Massachusetts General Research Institute, Charlestown, MA, USA
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Charlestown, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Felix Naef
- Laboratory of Computational and Systems Biology, Institute of Bioengineering, Faculty of Life Sciences, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland.
| | - Gioele La Manno
- Laboratory of Brain Development and Biological Data Science, Brain Mind Institute, Faculty of Life Sciences, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland.
| |
Collapse
|
2
|
Parker ME, Mehta NU, Liao TC, Tomaszewski WH, Snyder SA, Busch J, Ciofani M. Restriction of innate Tγδ17 cell plasticity by an AP-1 regulatory axis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.15.618522. [PMID: 39463970 PMCID: PMC11507935 DOI: 10.1101/2024.10.15.618522] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 10/29/2024]
Abstract
IL-17-producing γδ T (Tγδ17) cells are innate-like mediators of intestinal barrier immunity. While Th17 cell and ILC3 plasticity have been extensively studied, the mechanisms governing Tγδ17 cell effector flexibility remain undefined. Here, we combined type 3 fate-mapping with single cell ATAC/RNA-seq multiome profiling to define the cellular features and regulatory networks underlying Tγδ17 cell plasticity. During homeostasis, Tγδ17 cell effector identity was stable across tissues, including for intestinal T-bet + Tγδ17 cells that restrained IFNγ production. However, S. typhimurium infection induced intestinal Vγ6 + Tγδ17 cell conversion into type 1 effectors, with loss of IL-17A production and partial RORγt downregulation. Multiome analysis revealed a trajectory along Vγ6 + Tγδ17 effector conversion, with TIM-3 marking ex-Tγδ17 cells with enhanced type 1 functionality. Lastly, we characterized and validated a critical AP-1 regulatory axis centered around JunB and Fosl2 that controls Vγ6 + Tγδ17 cell plasticity by stabilizing type 3 identity and restricting type 1 effector conversion.
Collapse
|
3
|
Bai Z, Zhang D, Gao Y, Tao B, Zhang D, Bao S, Enninful A, Wang Y, Li H, Su G, Tian X, Zhang N, Xiao Y, Liu Y, Gerstein M, Li M, Xing Y, Lu J, Xu ML, Fan R. Spatially exploring RNA biology in archival formalin-fixed paraffin-embedded tissues. Cell 2024:S0092-8674(24)01019-5. [PMID: 39353436 DOI: 10.1016/j.cell.2024.09.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2024] [Revised: 07/29/2024] [Accepted: 09/03/2024] [Indexed: 10/04/2024]
Abstract
The capability to spatially explore RNA biology in formalin-fixed paraffin-embedded (FFPE) tissues holds transformative potential for histopathology research. Here, we present pathology-compatible deterministic barcoding in tissue (Patho-DBiT) by combining in situ polyadenylation and computational innovation for spatial whole transcriptome sequencing, tailored to probe the diverse RNA species in clinically archived FFPE samples. It permits spatial co-profiling of gene expression and RNA processing, unveiling region-specific splicing isoforms, and high-sensitivity transcriptomic mapping of clinical tumor FFPE tissues stored for 5 years. Furthermore, genome-wide single-nucleotide RNA variants can be captured to distinguish malignant subclones from non-malignant cells in human lymphomas. Patho-DBiT also maps microRNA regulatory networks and RNA splicing dynamics, decoding their roles in spatial tumorigenesis. Single-cell level Patho-DBiT dissects the spatiotemporal cellular dynamics driving tumor clonal architecture and progression. Patho-DBiT stands poised as a valuable platform to unravel rich RNA biology in FFPE tissues to aid in clinical pathology evaluation.
Collapse
Affiliation(s)
- Zhiliang Bai
- Department of Biomedical Engineering, Yale University, New Haven, CT 06520, USA.
| | - Dingyao Zhang
- Department of Genetics, Yale University School of Medicine, New Haven, CT 06520, USA; Program in Computational Biology and Bioinformatics, Yale University, New Haven, CT 06520, USA
| | - Yan Gao
- Center for Computational and Genomic Medicine, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Bo Tao
- Department of Pathology, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Daiwei Zhang
- Department of Biostatistics, Epidemiology and Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Shuozhen Bao
- Department of Biomedical Engineering, Yale University, New Haven, CT 06520, USA
| | - Archibald Enninful
- Department of Biomedical Engineering, Yale University, New Haven, CT 06520, USA
| | - Yadong Wang
- Department of Genetics, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Haikuo Li
- Department of Biomedical Engineering, Yale University, New Haven, CT 06520, USA
| | - Graham Su
- Department of Biomedical Engineering, Yale University, New Haven, CT 06520, USA
| | - Xiaolong Tian
- Department of Biomedical Engineering, Yale University, New Haven, CT 06520, USA
| | - Ningning Zhang
- Department of Genetics, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Yang Xiao
- Department of Biomedical Engineering, Columbia University, New York, NY 10027, USA
| | - Yang Liu
- Department of Pathology, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Mark Gerstein
- Program in Computational Biology and Bioinformatics, Yale University, New Haven, CT 06520, USA
| | - Mingyao Li
- Department of Biostatistics, Epidemiology and Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA.
| | - Yi Xing
- Center for Computational and Genomic Medicine, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA; Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA.
| | - Jun Lu
- Department of Genetics, Yale University School of Medicine, New Haven, CT 06520, USA; Yale Stem Cell Center and Yale Cancer Center, Yale University School of Medicine, New Haven, CT 06520, USA.
| | - Mina L Xu
- Department of Pathology, Yale University School of Medicine, New Haven, CT 06520, USA.
| | - Rong Fan
- Department of Biomedical Engineering, Yale University, New Haven, CT 06520, USA; Department of Pathology, Yale University School of Medicine, New Haven, CT 06520, USA; Yale Stem Cell Center and Yale Cancer Center, Yale University School of Medicine, New Haven, CT 06520, USA; Human and Translational Immunology, Yale University School of Medicine, New Haven, CT 06520, USA.
| |
Collapse
|
4
|
Luo C, Xu H, Yu Z, Liu D, Zhong D, Zhou S, Zhang B, Zhan J, Sun F. Meiotic chromatin-associated HSF5 is indispensable for pachynema progression and male fertility. Nucleic Acids Res 2024; 52:10255-10275. [PMID: 39162221 PMCID: PMC11417359 DOI: 10.1093/nar/gkae701] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2024] [Revised: 07/04/2024] [Accepted: 08/07/2024] [Indexed: 08/21/2024] Open
Abstract
Pachynema progression contributes to the completion of prophase I. Nevertheless, the regulation of this significant meiotic process remains poorly understood. In this study, we identified a novel testis-specific protein HSF5, which regulates pachynema progression during male meiosis in a manner dependent on chromatin-binding. Deficiency of HSF5 results in meiotic arrest and male infertility, characterized as unconventional pachynema arrested at the mid-to-late stage, with extensive spermatocyte apoptosis. Our scRNA-seq data confirmed consistent expressional alterations of certain driver genes (Sycp1, Msh4, Meiob, etc.) crucial for pachynema progression in Hsf5-/- individuals. HSF5 was revealed to primarily bind to promoter regions of such key divers by CUT&Tag analysis. Also, our results demonstrated that HSF5 biologically interacted with SMARCA5, SMARCA4 and SMARCE1, and it could function as a transcription factor for pachynema progression during meiosis. Therefore, our study underscores the importance of the chromatin-associated HSF5 for the differentiation of spermatocytes, improving the protein regulatory network of the pachynema progression.
Collapse
Affiliation(s)
- Chunhai Luo
- Department of Urology & Andrology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310016, China
| | - Haoran Xu
- Department of Urology & Andrology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310016, China
| | - Ziqi Yu
- Department of Urology & Andrology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310016, China
| | - Dalin Liu
- Department of Urology & Andrology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310016, China
| | - Danyang Zhong
- Department of General Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310016, China
| | - Shumin Zhou
- Department of Urology & Andrology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310016, China
| | - Beibei Zhang
- Department of Urology & Andrology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310016, China
| | - Junfeng Zhan
- Department of Urology & Andrology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310016, China
| | - Fei Sun
- Department of Urology & Andrology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310016, China
| |
Collapse
|
5
|
Liu Y, Kim YS, Xue X, Miao Y, Kobayashi N, Sun S, Yan RZ, Yang Q, Pourquié O, Fu J. A human pluripotent stem cell-based somitogenesis model using microfluidics. Cell Stem Cell 2024; 31:1113-1126.e6. [PMID: 38981471 DOI: 10.1016/j.stem.2024.06.004] [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: 09/10/2023] [Revised: 03/04/2024] [Accepted: 06/04/2024] [Indexed: 07/11/2024]
Abstract
Emerging human pluripotent stem cell (hPSC)-based embryo models are useful for studying human embryogenesis. Particularly, there are hPSC-based somitogenesis models using free-floating culture that recapitulate somite formation. Somitogenesis in vivo involves intricately orchestrated biochemical and biomechanical events. However, none of the current somitogenesis models controls biochemical gradients or biomechanical signals in the culture, limiting their applicability to untangle complex biochemical-biomechanical interactions that drive somitogenesis. Herein, we develop a human somitogenesis model by confining hPSC-derived presomitic mesoderm (PSM) tissues in microfabricated trenches. Exogenous microfluidic morphogen gradients imposed on the PSM tissues cause axial patterning and trigger spontaneous rostral-to-caudal somite formation. A mechanical theory is developed to explain the size dependency between somites and the PSM. The microfluidic somitogenesis model is further exploited to reveal regulatory roles of cellular and tissue biomechanics in somite formation. This study presents a useful microengineered, hPSC-based model for understanding the biochemical and biomechanical events that guide somite formation.
Collapse
Affiliation(s)
- Yue Liu
- Department of Mechanical Engineering, University of Michigan, Ann Arbor, MI 48109, USA.
| | - Yung Su Kim
- Department of Mechanical Engineering, University of Michigan, Ann Arbor, MI 48109, USA
| | - Xufeng Xue
- Department of Mechanical Engineering, University of Michigan, Ann Arbor, MI 48109, USA
| | - Yuchuan Miao
- Department of Genetics, Harvard Medical School, Boston, MA 02115, USA; Department of Pathology, Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Norio Kobayashi
- Department of Mechanical Engineering, University of Michigan, Ann Arbor, MI 48109, USA
| | - Shiyu Sun
- Department of Mechanical Engineering, University of Michigan, Ann Arbor, MI 48109, USA
| | - Robin Zhexuan Yan
- Department of Mechanical Engineering, University of Michigan, Ann Arbor, MI 48109, USA
| | - Qiong Yang
- Department of Biophysics, University of Michigan, Ann Arbor, MI 48109, USA; Department of Physics, University of Michigan, Ann Arbor, MI 48109, USA
| | - Olivier Pourquié
- Department of Genetics, Harvard Medical School, Boston, MA 02115, USA; Department of Pathology, Brigham and Women's Hospital, Boston, MA 02115, USA; Harvard Stem Cell Institute, Harvard University, Cambridge, MA 02138, USA
| | - Jianping Fu
- Department of Mechanical Engineering, University of Michigan, Ann Arbor, MI 48109, USA; Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109, USA; Department of Cell & Developmental Biology, University of Michigan Medical School, Ann Arbor, MI 48109, USA.
| |
Collapse
|
6
|
Weiler P, Lange M, Klein M, Pe'er D, Theis F. CellRank 2: unified fate mapping in multiview single-cell data. Nat Methods 2024; 21:1196-1205. [PMID: 38871986 PMCID: PMC11239496 DOI: 10.1038/s41592-024-02303-9] [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: 07/18/2023] [Accepted: 05/09/2024] [Indexed: 06/15/2024]
Abstract
Single-cell RNA sequencing allows us to model cellular state dynamics and fate decisions using expression similarity or RNA velocity to reconstruct state-change trajectories; however, trajectory inference does not incorporate valuable time point information or utilize additional modalities, whereas methods that address these different data views cannot be combined or do not scale. Here we present CellRank 2, a versatile and scalable framework to study cellular fate using multiview single-cell data of up to millions of cells in a unified fashion. CellRank 2 consistently recovers terminal states and fate probabilities across data modalities in human hematopoiesis and endodermal development. Our framework also allows combining transitions within and across experimental time points, a feature we use to recover genes promoting medullary thymic epithelial cell formation during pharyngeal endoderm development. Moreover, we enable estimating cell-specific transcription and degradation rates from metabolic-labeling data, which we apply to an intestinal organoid system to delineate differentiation trajectories and pinpoint regulatory strategies.
Collapse
Affiliation(s)
- Philipp Weiler
- Institute of Computational Biology, Department of Computational Health, Helmholtz Munich, Munich, Germany
- School of Computation, Information and Technology, Technical University of Munich, Munich, Germany
| | - Marius Lange
- Institute of Computational Biology, Department of Computational Health, Helmholtz Munich, Munich, Germany
- School of Computation, Information and Technology, Technical University of Munich, Munich, Germany
- Department of Biosystems Science and Engineering, ETH Zürich, Basel, Switzerland
| | - Michal Klein
- Institute of Computational Biology, Department of Computational Health, Helmholtz Munich, Munich, Germany
- Machine Learning Research, Apple, Paris, France
| | - Dana Pe'er
- Computational and Systems Biology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Howard Hughes Medical Institute, Chevy Chase, MD, USA
| | - Fabian Theis
- Institute of Computational Biology, Department of Computational Health, Helmholtz Munich, Munich, Germany.
- School of Computation, Information and Technology, Technical University of Munich, Munich, Germany.
- TUM School of Life Sciences Weihenstephan, Technical University of Munich, Munich, Germany.
| |
Collapse
|
7
|
Xavier AM, Lin Q, Kang CJ, Cheadle L. A single-cell transcriptomic atlas of sensory-dependent gene expression in developing mouse visual cortex. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.25.600673. [PMID: 38979325 PMCID: PMC11230371 DOI: 10.1101/2024.06.25.600673] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/10/2024]
Abstract
Sensory experience drives the refinement and maturation of neural circuits during postnatal brain development through molecular mechanisms that remain to be fully elucidated. One likely mechanism involves the sensory-dependent expression of genes that encode direct mediators of circuit remodeling within developing cells. However, while studies in adult systems have begun to uncover crucial roles for sensory-induced genes in modifying circuit connectivity, the gene programs induced by brain cells in response to sensory experience during development remain to be fully characterized. Here, we present a single-nucleus RNA-sequencing dataset describing the transcriptional responses of cells in mouse visual cortex to sensory deprivation or sensory stimulation during a developmental window when visual input is necessary for circuit refinement. We sequenced 118,529 individual nuclei across sixteen neuronal and non-neuronal cortical cell types isolated from control, sensory deprived, and sensory stimulated mice, identifying 1,268 unique sensory-induced genes within the developing brain. To demonstrate the utility of this resource, we compared the architecture and ontology of sensory-induced gene programs between cell types, annotated transcriptional induction and repression events based upon RNA velocity, and discovered Neurexin and Neuregulin signaling networks that underlie cell-cell interactions via CellChat . We find that excitatory neurons, especially layer 2/3 pyramidal neurons, are highly sensitive to sensory stimulation, and that the sensory-induced genes in these cells are poised to strengthen synapse-to-nucleus crosstalk by heightening protein serine/threonine kinase activity. Altogether, we expect this dataset to significantly broaden our understanding of the molecular mechanisms through which sensory experience shapes neural circuit wiring in the developing brain.
Collapse
|
8
|
Zhou P, Bocci F, Li T, Nie Q. Spatial transition tensor of single cells. Nat Methods 2024; 21:1053-1062. [PMID: 38755322 PMCID: PMC11166574 DOI: 10.1038/s41592-024-02266-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Accepted: 04/02/2024] [Indexed: 05/18/2024]
Abstract
Spatial transcriptomics and messenger RNA splicing encode extensive spatiotemporal information for cell states and transitions. The current lineage-inference methods either lack spatial dynamics for state transition or cannot capture different dynamics associated with multiple cell states and transition paths. Here we present spatial transition tensor (STT), a method that uses messenger RNA splicing and spatial transcriptomes through a multiscale dynamical model to characterize multistability in space. By learning a four-dimensional transition tensor and spatial-constrained random walk, STT reconstructs cell-state-specific dynamics and spatial state transitions via both short-time local tensor streamlines between cells and long-time transition paths among attractors. Benchmarking and applications of STT on several transcriptome datasets via multiple technologies on epithelial-mesenchymal transitions, blood development, spatially resolved mouse brain and chicken heart development, indicate STT's capability in recovering cell-state-specific dynamics and their associated genes not seen using existing methods. Overall, STT provides a consistent multiscale description of single-cell transcriptome data across multiple spatiotemporal scales.
Collapse
Affiliation(s)
- Peijie Zhou
- Department of Mathematics, University of California, Irvine, Irvine, CA, USA
- Center for Machine Learning Research, Peking University, Beijing, China
- AI for Science Institute, Beijing, China
- National Engineering Laboratory for Big Data Analysis and Applications, Beijing, China
| | - Federico Bocci
- Department of Mathematics, University of California, Irvine, Irvine, CA, USA
| | - Tiejun Li
- LMAM and School of Mathematical Sciences, Peking University, Beijing, China
| | - Qing Nie
- Department of Mathematics, University of California, Irvine, Irvine, CA, USA.
- Department of Cell and Developmental Biology, University of California, Irvine, Irvine, CA, USA.
| |
Collapse
|
9
|
Simard S, Matosin N, Mechawar N. Adult Hippocampal Neurogenesis in the Human Brain: Updates, Challenges, and Perspectives. Neuroscientist 2024:10738584241252581. [PMID: 38757781 DOI: 10.1177/10738584241252581] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/18/2024]
Abstract
The existence of neurogenesis in the adult human hippocampus has been under considerable debate within the past three decades due to the diverging conclusions originating mostly from immunohistochemistry studies. While some of these reports conclude that hippocampal neurogenesis in humans occurs throughout physiologic aging, others indicate that this phenomenon ends by early childhood. More recently, some groups have adopted next-generation sequencing technologies to characterize with more acuity the extent of this phenomenon in humans. Here, we review the current state of research on adult hippocampal neurogenesis in the human brain with an emphasis on the challenges and limitations of using immunohistochemistry and next-generation sequencing technologies for its study.
Collapse
Affiliation(s)
- Sophie Simard
- McGill Group for Suicide Studies, Douglas Mental Health University Institute, McGill University, Montréal, Canada
| | - Natalie Matosin
- School of Medical Sciences, Faculty of Medicine and Health, University of Sydney, Sydney, Australia
| | - Naguib Mechawar
- McGill Group for Suicide Studies, Douglas Mental Health University Institute, McGill University, Montréal, Canada
- Department of Psychiatry, McGill University, Montréal, Canada
| |
Collapse
|
10
|
Maizels RJ, Snell DM, Briscoe J. Reconstructing developmental trajectories using latent dynamical systems and time-resolved transcriptomics. Cell Syst 2024; 15:411-424.e9. [PMID: 38754365 DOI: 10.1016/j.cels.2024.04.004] [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: 09/19/2023] [Revised: 02/01/2024] [Accepted: 04/17/2024] [Indexed: 05/18/2024]
Abstract
The snapshot nature of single-cell transcriptomics presents a challenge for studying the dynamics of cell fate decisions. Metabolic labeling and splicing can provide temporal information at single-cell level, but current methods have limitations. Here, we present a framework that overcomes these limitations: experimentally, we developed sci-FATE2, an optimized method for metabolic labeling with increased data quality, which we used to profile 45,000 embryonic stem (ES) cells differentiating into neural tube identities. Computationally, we developed a two-stage framework for dynamical modeling: VelvetVAE, a variational autoencoder (VAE) for velocity inference that outperforms all other tools tested, and VelvetSDE, a neural stochastic differential equation (nSDE) framework for simulating trajectory distributions. These recapitulate underlying dataset distributions and capture features such as decision boundaries between alternative fates and fate-specific gene expression. These methods recast single-cell analyses from descriptions of observed data to models of the dynamics that generated them, providing a framework for investigating developmental fate decisions.
Collapse
Affiliation(s)
- Rory J Maizels
- The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK; University College, London, UK
| | - Daniel M Snell
- The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
| | - James Briscoe
- The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK.
| |
Collapse
|
11
|
Brümmer A, Bergmann S. Disentangling genetic effects on transcriptional and post-transcriptional gene regulation through integrating exon and intron expression QTLs. Nat Commun 2024; 15:3786. [PMID: 38710690 DOI: 10.1038/s41467-024-48244-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: 05/04/2023] [Accepted: 04/23/2024] [Indexed: 05/08/2024] Open
Abstract
Expression quantitative trait loci (eQTL) studies typically consider exon expression of genes and discard intronic RNA sequencing reads despite their information on RNA metabolism. Here, we quantify genetic effects on exon and intron levels of genes and their ratio in lymphoblastoid cell lines, revealing thousands of cis-QTLs of each type. While genetic effects are often shared between cis-QTL types, 7814 (47%) are not detected as top cis-QTLs at exon levels. We show that exon levels preferentially capture genetic effects on transcriptional regulation, while exon-intron-ratios better detect those on co- and post-transcriptional processes. Considering all cis-QTL types substantially increases (by 71%) the number of colocalizing variants identified by genome-wide association studies (GWAS). It further allows dissecting the potential gene regulatory processes underlying GWAS associations, suggesting comparable contributions by transcriptional (50%) and co- and post-transcriptional regulation (46%) to complex traits. Overall, integrating intronic RNA sequencing reads in eQTL studies expands our understanding of genetic effects on gene regulatory processes.
Collapse
Affiliation(s)
- Anneke Brümmer
- Department of Computational Biology, University of Lausanne, Lausanne, Switzerland.
- Swiss Institute of Bioinformatics, Lausanne, Switzerland.
- Bioinformatics Competence Center, University of Lausanne, Lausanne, Switzerland.
| | - Sven Bergmann
- Department of Computational Biology, University of Lausanne, Lausanne, Switzerland.
- Swiss Institute of Bioinformatics, Lausanne, Switzerland.
- Department of Integrative Biomedical Sciences, University of Cape Town, Cape Town, South Africa.
| |
Collapse
|
12
|
Wang K, Hou L, Wang X, Zhai X, Lu Z, Zi Z, Zhai W, He X, Curtis C, Zhou D, Hu Z. PhyloVelo enhances transcriptomic velocity field mapping using monotonically expressed genes. Nat Biotechnol 2024; 42:778-789. [PMID: 37524958 DOI: 10.1038/s41587-023-01887-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Accepted: 06/28/2023] [Indexed: 08/02/2023]
Abstract
Single-cell RNA sequencing (scRNA-seq) is a powerful approach for studying cellular differentiation, but accurately tracking cell fate transitions can be challenging, especially in disease conditions. Here we introduce PhyloVelo, a computational framework that estimates the velocity of transcriptomic dynamics by using monotonically expressed genes (MEGs) or genes with expression patterns that either increase or decrease, but do not cycle, through phylogenetic time. Through integration of scRNA-seq data with lineage information, PhyloVelo identifies MEGs and reconstructs a transcriptomic velocity field. We validate PhyloVelo using simulated data and Caenorhabditis elegans ground truth data, successfully recovering linear, bifurcated and convergent differentiations. Applying PhyloVelo to seven lineage-traced scRNA-seq datasets, generated using CRISPR-Cas9 editing, lentiviral barcoding or immune repertoire profiling, demonstrates its high accuracy and robustness in inferring complex lineage trajectories while outperforming RNA velocity. Additionally, we discovered that MEGs across tissues and organisms share similar functions in translation and ribosome biogenesis.
Collapse
Affiliation(s)
- Kun Wang
- CAS Key Laboratory of Quantitative Engineering Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
- School of Mathematical Sciences, Xiamen University, Xiamen, China
| | - Liangzhen Hou
- CAS Key Laboratory of Quantitative Engineering Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
- Faculty of Health Sciences, University of Macau, Taipa, Macau, China
| | - Xin Wang
- CAS Key Laboratory of Quantitative Engineering Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Xiangwei Zhai
- MOE Key Laboratory of Gene Function and Regulation, State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-Sen University, Guangzhou, China
| | - Zhaolian Lu
- CAS Key Laboratory of Quantitative Engineering Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Zhike Zi
- CAS Key Laboratory of Quantitative Engineering Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Weiwei Zhai
- CAS Key Laboratory of Zoological Systematics and Evolution, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- Center for Excellence in Animal Evolution and Genetics, Chinese Academy of Sciences, Kunming, China
| | - Xionglei He
- MOE Key Laboratory of Gene Function and Regulation, State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-Sen University, Guangzhou, China
| | - Christina Curtis
- Department of Medicine, Division of Oncology, Stanford University School of Medicine, Stanford, CA, USA
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA
- Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA, USA
| | - Da Zhou
- School of Mathematical Sciences, Xiamen University, Xiamen, China.
- National Institute for Data Science in Health and Medicine, Xiamen University, Xiamen, China.
| | - Zheng Hu
- CAS Key Laboratory of Quantitative Engineering Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China.
| |
Collapse
|
13
|
Gao CF, Vaikuntanathan S, Riesenfeld SJ. Dissection and integration of bursty transcriptional dynamics for complex systems. Proc Natl Acad Sci U S A 2024; 121:e2306901121. [PMID: 38669186 PMCID: PMC11067469 DOI: 10.1073/pnas.2306901121] [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: 04/26/2023] [Accepted: 03/06/2024] [Indexed: 04/28/2024] Open
Abstract
RNA velocity estimation is a potentially powerful tool to reveal the directionality of transcriptional changes in single-cell RNA-sequencing data, but it lacks accuracy, absent advanced metabolic labeling techniques. We developed an approach, TopicVelo, that disentangles simultaneous, yet distinct, dynamics by using a probabilistic topic model, a highly interpretable form of latent space factorization, to infer cells and genes associated with individual processes, thereby capturing cellular pluripotency or multifaceted functionality. Focusing on process-associated cells and genes enables accurate estimation of process-specific velocities via a master equation for a transcriptional burst model accounting for intrinsic stochasticity. The method obtains a global transition matrix by leveraging cell topic weights to integrate process-specific signals. In challenging systems, this method accurately recovers complex transitions and terminal states, while our use of first-passage time analysis provides insights into transient transitions. These results expand the limits of RNA velocity, empowering future studies of cell fate and functional responses.
Collapse
Affiliation(s)
- Cheng Frank Gao
- Department of Chemistry, University of Chicago, Chicago, IL60637
| | - Suriyanarayanan Vaikuntanathan
- Department of Chemistry, University of Chicago, Chicago, IL60637
- Institute for Biophysical Dynamics, University of Chicago, Chicago, IL60637
| | - Samantha J. Riesenfeld
- Institute for Biophysical Dynamics, University of Chicago, Chicago, IL60637
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL60637
- Department of Medicine, University of Chicago, Chicago, IL60637
- Committee on Immunology, Biological Sciences Division, University of Chicago, Chicago, IL60637
| |
Collapse
|
14
|
Maizels RJ. A dynamical perspective: moving towards mechanism in single-cell transcriptomics. Philos Trans R Soc Lond B Biol Sci 2024; 379:20230049. [PMID: 38432314 PMCID: PMC10909508 DOI: 10.1098/rstb.2023.0049] [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: 07/10/2023] [Accepted: 10/31/2023] [Indexed: 03/05/2024] Open
Abstract
As the field of single-cell transcriptomics matures, research is shifting focus from phenomenological descriptions of cellular phenotypes to a mechanistic understanding of the gene regulation underneath. This perspective considers the value of capturing dynamical information at single-cell resolution for gaining mechanistic insight; reviews the available technologies for recording and inferring temporal information in single cells; and explores whether better dynamical resolution is sufficient to adequately capture the causal relationships driving complex biological systems. This article is part of a discussion meeting issue 'Causes and consequences of stochastic processes in development and disease'.
Collapse
Affiliation(s)
- Rory J. Maizels
- The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
- University College London, London WC1E 6BT, UK
| |
Collapse
|
15
|
Afzali AM, Nirschl L, Sie C, Pfaller M, Ulianov O, Hassler T, Federle C, Petrozziello E, Kalluri SR, Chen HH, Tyystjärvi S, Muschaweckh A, Lammens K, Delbridge C, Büttner A, Steiger K, Seyhan G, Ottersen OP, Öllinger R, Rad R, Jarosch S, Straub A, Mühlbauer A, Grassmann S, Hemmer B, Böttcher JP, Wagner I, Kreutzfeldt M, Merkler D, Pardàs IB, Schmidt Supprian M, Buchholz VR, Heink S, Busch DH, Klein L, Korn T. B cells orchestrate tolerance to the neuromyelitis optica autoantigen AQP4. Nature 2024; 627:407-415. [PMID: 38383779 PMCID: PMC10937377 DOI: 10.1038/s41586-024-07079-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2023] [Accepted: 01/16/2024] [Indexed: 02/23/2024]
Abstract
Neuromyelitis optica is a paradigmatic autoimmune disease of the central nervous system, in which the water-channel protein AQP4 is the target antigen1. The immunopathology in neuromyelitis optica is largely driven by autoantibodies to AQP42. However, the T cell response that is required for the generation of these anti-AQP4 antibodies is not well understood. Here we show that B cells endogenously express AQP4 in response to activation with anti-CD40 and IL-21 and are able to present their endogenous AQP4 to T cells with an AQP4-specific T cell receptor (TCR). A population of thymic B cells emulates a CD40-stimulated B cell transcriptome, including AQP4 (in mice and humans), and efficiently purges the thymic TCR repertoire of AQP4-reactive clones. Genetic ablation of Aqp4 in B cells rescues AQP4-specific TCRs despite sufficient expression of AQP4 in medullary thymic epithelial cells, and B-cell-conditional AQP4-deficient mice are fully competent to raise AQP4-specific antibodies in productive germinal-centre responses. Thus, the negative selection of AQP4-specific thymocytes is dependent on the expression and presentation of AQP4 by thymic B cells. As AQP4 is expressed in B cells in a CD40-dependent (but not AIRE-dependent) manner, we propose that thymic B cells might tolerize against a group of germinal-centre-associated antigens, including disease-relevant autoantigens such as AQP4.
Collapse
Affiliation(s)
- Ali Maisam Afzali
- Institute for Experimental Neuroimmunology, Technical University of Munich School of Medicine and Health, Munich, Germany
- Department of Neurology, Technical University of Munich School of Medicine and Health, Munich, Germany
- Munich Cluster for Systems Neurology, Munich, Germany
| | - Lucy Nirschl
- Institute for Experimental Neuroimmunology, Technical University of Munich School of Medicine and Health, Munich, Germany
| | - Christopher Sie
- Institute for Experimental Neuroimmunology, Technical University of Munich School of Medicine and Health, Munich, Germany
| | - Monika Pfaller
- Institute for Experimental Neuroimmunology, Technical University of Munich School of Medicine and Health, Munich, Germany
| | - Oleksii Ulianov
- Institute for Experimental Neuroimmunology, Technical University of Munich School of Medicine and Health, Munich, Germany
| | - Tobias Hassler
- Biomedical Center (BMC), Institute for Immunology, Faculty of Medicine, Ludwig-Maximilians-University Munich, Planegg-Martinsried, Germany
| | - Christine Federle
- Biomedical Center (BMC), Institute for Immunology, Faculty of Medicine, Ludwig-Maximilians-University Munich, Planegg-Martinsried, Germany
| | - Elisabetta Petrozziello
- Biomedical Center (BMC), Institute for Immunology, Faculty of Medicine, Ludwig-Maximilians-University Munich, Planegg-Martinsried, Germany
| | - Sudhakar Reddy Kalluri
- Department of Neurology, Technical University of Munich School of Medicine and Health, Munich, Germany
| | - Hsin Hsiang Chen
- Institute for Experimental Neuroimmunology, Technical University of Munich School of Medicine and Health, Munich, Germany
| | - Sofia Tyystjärvi
- Institute for Experimental Neuroimmunology, Technical University of Munich School of Medicine and Health, Munich, Germany
| | - Andreas Muschaweckh
- Institute for Experimental Neuroimmunology, Technical University of Munich School of Medicine and Health, Munich, Germany
| | - Katja Lammens
- Department of Biochemistry at the Gene Center, Ludwig-Maximilians-University, Munich, Germany
| | - Claire Delbridge
- Institute of Pathology, Technical University of Munich School of Medicine and Health, Munich, Germany
- Department of Neuropathology, Institute of Pathology, Technical University of Munich School of Medicine and Health, Munich, Germany
| | - Andreas Büttner
- Institute of Forensic Medicine, Rostock University Medical Center, Rostock, Germany
| | - Katja Steiger
- Institute of Pathology, Technical University of Munich School of Medicine and Health, Munich, Germany
| | - Gönül Seyhan
- Institute for Experimental Hematology, TranslaTUM Cancer Center, Technical University of Munich School of Medicine and Health, Munich, Germany
| | - Ole Petter Ottersen
- Division of Anatomy, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
| | - Rupert Öllinger
- Institute of Molecular Oncology and Functional Genomics, TranslaTUM Cancer Center, Technical University of Munich School of Medicine and Health, Munich, Germany
| | - Roland Rad
- Institute of Molecular Oncology and Functional Genomics, TranslaTUM Cancer Center, Technical University of Munich School of Medicine and Health, Munich, Germany
| | - Sebastian Jarosch
- Institute for Medical Microbiology, Immunology and Hygiene, Technical University of Munich School of Medicine and Health, Munich, Germany
| | - Adrian Straub
- Institute for Medical Microbiology, Immunology and Hygiene, Technical University of Munich School of Medicine and Health, Munich, Germany
| | - Anton Mühlbauer
- Institute for Medical Microbiology, Immunology and Hygiene, Technical University of Munich School of Medicine and Health, Munich, Germany
| | - Simon Grassmann
- Immunology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Bernhard Hemmer
- Department of Neurology, Technical University of Munich School of Medicine and Health, Munich, Germany
- Munich Cluster for Systems Neurology, Munich, Germany
| | - Jan P Böttcher
- Institute of Molecular Immunology, Technical University of Munich School of Medicine and Health, Munich, Germany
| | - Ingrid Wagner
- Department of Pathology and Immunology, Division of Clinical Pathology, Geneva Faculty of Medicine, Centre Médical Universitaire, Geneva, Switzerland
| | - Mario Kreutzfeldt
- Department of Pathology and Immunology, Division of Clinical Pathology, Geneva Faculty of Medicine, Centre Médical Universitaire, Geneva, Switzerland
| | - Doron Merkler
- Department of Pathology and Immunology, Division of Clinical Pathology, Geneva Faculty of Medicine, Centre Médical Universitaire, Geneva, Switzerland
| | | | - Marc Schmidt Supprian
- Institute for Experimental Hematology, TranslaTUM Cancer Center, Technical University of Munich School of Medicine and Health, Munich, Germany
| | - Veit R Buchholz
- Institute for Medical Microbiology, Immunology and Hygiene, Technical University of Munich School of Medicine and Health, Munich, Germany
| | - Sylvia Heink
- Institute for Experimental Neuroimmunology, Technical University of Munich School of Medicine and Health, Munich, Germany
| | - Dirk H Busch
- Institute for Medical Microbiology, Immunology and Hygiene, Technical University of Munich School of Medicine and Health, Munich, Germany
- German Center for Infection Research (DZIF), Partner Site Munich, Munich, Germany
| | - Ludger Klein
- Biomedical Center (BMC), Institute for Immunology, Faculty of Medicine, Ludwig-Maximilians-University Munich, Planegg-Martinsried, Germany
| | - Thomas Korn
- Institute for Experimental Neuroimmunology, Technical University of Munich School of Medicine and Health, Munich, Germany.
- Department of Neurology, Technical University of Munich School of Medicine and Health, Munich, Germany.
- Munich Cluster for Systems Neurology, Munich, Germany.
| |
Collapse
|
16
|
Pan X, Zhang X. Studying temporal dynamics of single cells: expression, lineage and regulatory networks. Biophys Rev 2024; 16:57-67. [PMID: 38495440 PMCID: PMC10937865 DOI: 10.1007/s12551-023-01090-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Accepted: 06/27/2023] [Indexed: 03/19/2024] Open
Abstract
Learning how multicellular organs are developed from single cells to different cell types is a fundamental problem in biology. With the high-throughput scRNA-seq technology, computational methods have been developed to reveal the temporal dynamics of single cells from transcriptomic data, from phenomena on cell trajectories to the underlying mechanism that formed the trajectory. There are several distinct families of computational methods including Trajectory Inference (TI), Lineage Tracing (LT), and Gene Regulatory Network (GRN) Inference which are involved in such studies. This review summarizes these computational approaches which use scRNA-seq data to study cell differentiation and cell fate specification as well as the advantages and limitations of different methods. We further discuss how GRNs can potentially affect cell fate decisions and trajectory structures. Supplementary Information The online version contains supplementary material available at 10.1007/s12551-023-01090-5.
Collapse
Affiliation(s)
- Xinhai Pan
- School of Computational Science and Engineering, Georgia Institute of Technology, Atlanta, GA 30332 USA
| | - Xiuwei Zhang
- School of Computational Science and Engineering, Georgia Institute of Technology, Atlanta, GA 30332 USA
| |
Collapse
|
17
|
Lederer AR, Leonardi M, Talamanca L, Herrera A, Droin C, Khven I, Carvalho HJF, Valente A, Mantes AD, Arabí PM, Pinello L, Naef F, Manno GL. Statistical inference with a manifold-constrained RNA velocity model uncovers cell cycle speed modulations. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.18.576093. [PMID: 38328127 PMCID: PMC10849531 DOI: 10.1101/2024.01.18.576093] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/09/2024]
Abstract
Across a range of biological processes, cells undergo coordinated changes in gene expression, resulting in transcriptome dynamics that unfold within a low-dimensional manifold. Single-cell RNA-sequencing (scRNA-seq) only measures temporal snapshots of gene expression. However, information on the underlying low-dimensional dynamics can be extracted using RNA velocity, which models unspliced and spliced RNA abundances to estimate the rate of change of gene expression. Available RNA velocity algorithms can be fragile and rely on heuristics that lack statistical control. Moreover, the estimated vector field is not dynamically consistent with the traversed gene expression manifold. Here, we develop a generative model of RNA velocity and a Bayesian inference approach that solves these problems. Our model couples velocity field and manifold estimation in a reformulated, unified framework, so as to coherently identify the parameters of an autonomous dynamical system. Focusing on the cell cycle, we implemented VeloCycle to study gene regulation dynamics on one-dimensional periodic manifolds and validated using live-imaging its ability to infer actual cell cycle periods. We benchmarked RNA velocity inference with sensitivity analyses and demonstrated one- and multiple-sample testing. We also conducted Markov chain Monte Carlo inference on the model, uncovering key relationships between gene-specific kinetics and our gene-independent velocity estimate. Finally, we applied VeloCycle to in vivo samples and in vitro genome-wide Perturb-seq, revealing regionally-defined proliferation modes in neural progenitors and the effect of gene knockdowns on cell cycle speed. Ultimately, VeloCycle expands the scRNA-seq analysis toolkit with a modular and statistically rigorous RNA velocity inference framework.
Collapse
|
18
|
Li S, Zhang P, Chen W, Ye L, Brannan KW, Le NT, Abe JI, Cooke JP, Wang G. A relay velocity model infers cell-dependent RNA velocity. Nat Biotechnol 2024; 42:99-108. [PMID: 37012448 PMCID: PMC10545816 DOI: 10.1038/s41587-023-01728-5] [Citation(s) in RCA: 16] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Accepted: 02/28/2023] [Indexed: 04/05/2023]
Abstract
RNA velocity provides an approach for inferring cellular state transitions from single-cell RNA sequencing (scRNA-seq) data. Conventional RNA velocity models infer universal kinetics from all cells in an scRNA-seq experiment, resulting in unpredictable performance in experiments with multi-stage and/or multi-lineage transition of cell states where the assumption of the same kinetic rates for all cells no longer holds. Here we present cellDancer, a scalable deep neural network that locally infers velocity for each cell from its neighbors and then relays a series of local velocities to provide single-cell resolution inference of velocity kinetics. In the simulation benchmark, cellDancer shows robust performance in multiple kinetic regimes, high dropout ratio datasets and sparse datasets. We show that cellDancer overcomes the limitations of existing RNA velocity models in modeling erythroid maturation and hippocampus development. Moreover, cellDancer provides cell-specific predictions of transcription, splicing and degradation rates, which we identify as potential indicators of cell fate in the mouse pancreas.
Collapse
Affiliation(s)
- Shengyu Li
- Center for Bioinformatics and Computational Biology, Houston Methodist Research Institute, Houston, TX, USA
- Center for Cardiovascular Regeneration, Houston Methodist Research Institute, Houston, TX, USA
- Center for RNA Therapeutics, Houston Methodist Research Institute, Houston, TX, USA
- Department of Cardiothoracic Surgery, Weill Cornell Medicine, Cornell University, New York, NY, USA
| | - Pengzhi Zhang
- Center for Bioinformatics and Computational Biology, Houston Methodist Research Institute, Houston, TX, USA
- Center for Cardiovascular Regeneration, Houston Methodist Research Institute, Houston, TX, USA
- Center for RNA Therapeutics, Houston Methodist Research Institute, Houston, TX, USA
- Department of Cardiothoracic Surgery, Weill Cornell Medicine, Cornell University, New York, NY, USA
| | - Weiqing Chen
- Center for Bioinformatics and Computational Biology, Houston Methodist Research Institute, Houston, TX, USA
- Department of Physiology, Biophysics & Systems Biology, Weill Cornell Graduate School of Medical Science, Weill Cornell Medicine, Cornell University, Ithaca, NY, USA
| | - Lingqun Ye
- Center for Bioinformatics and Computational Biology, Houston Methodist Research Institute, Houston, TX, USA
- Center for Cardiovascular Regeneration, Houston Methodist Research Institute, Houston, TX, USA
- Center for RNA Therapeutics, Houston Methodist Research Institute, Houston, TX, USA
| | - Kristopher W Brannan
- Center for Cardiovascular Regeneration, Houston Methodist Research Institute, Houston, TX, USA
- Center for RNA Therapeutics, Houston Methodist Research Institute, Houston, TX, USA
- Department of Cardiothoracic Surgery, Weill Cornell Medicine, Cornell University, New York, NY, USA
| | - Nhat-Tu Le
- Center for Cardiovascular Regeneration, Houston Methodist Research Institute, Houston, TX, USA
- Department of Cardiothoracic Surgery, Weill Cornell Medicine, Cornell University, New York, NY, USA
| | - Jun-Ichi Abe
- Department of Cardiology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - John P Cooke
- Center for Cardiovascular Regeneration, Houston Methodist Research Institute, Houston, TX, USA
| | - Guangyu Wang
- Center for Bioinformatics and Computational Biology, Houston Methodist Research Institute, Houston, TX, USA.
- Center for Cardiovascular Regeneration, Houston Methodist Research Institute, Houston, TX, USA.
- Center for RNA Therapeutics, Houston Methodist Research Institute, Houston, TX, USA.
- Department of Cardiothoracic Surgery, Weill Cornell Medicine, Cornell University, New York, NY, USA.
| |
Collapse
|
19
|
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
|
20
|
Kyaw W, Chai RC, Khoo WH, Goldstein LD, Croucher PI, Murray JM, Phan TG. ENTRAIN: integrating trajectory inference and gene regulatory networks with spatial data to co-localize the receptor-ligand interactions that specify cell fate. Bioinformatics 2023; 39:btad765. [PMID: 38113422 PMCID: PMC10752580 DOI: 10.1093/bioinformatics/btad765] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2023] [Revised: 12/07/2023] [Accepted: 12/18/2023] [Indexed: 12/21/2023] Open
Abstract
MOTIVATION Cell fate is commonly studied by profiling the gene expression of single cells to infer developmental trajectories based on expression similarity, RNA velocity, or statistical mechanical properties. However, current approaches do not recover microenvironmental signals from the cellular niche that drive a differentiation trajectory. RESULTS We resolve this with environment-aware trajectory inference (ENTRAIN), a computational method that integrates trajectory inference methods with ligand-receptor pair gene regulatory networks to identify extracellular signals and evaluate their relative contribution towards a differentiation trajectory. The output from ENTRAIN can be superimposed on spatial data to co-localize cells and molecules in space and time to map cell fate potentials to cell-cell interactions. We validate and benchmark our approach on single-cell bone marrow and spatially resolved embryonic neurogenesis datasets to identify known and novel environmental drivers of cellular differentiation. AVAILABILITY AND IMPLEMENTATION ENTRAIN is available as a public package at https://github.com/theimagelab/entrain and can be used on both single-cell and spatially resolved datasets.
Collapse
Affiliation(s)
- Wunna Kyaw
- Precision Immunology Program, Garvan Institute of Medical Research, Darlinghurst, NSW 2010, Australia
- St Vincent’s Healthcare Clinical Campus, Faculty of Medicine and Health, UNSW Sydney, Darlinghurst, NSW 2010, Australia
| | - Ryan C Chai
- St Vincent’s Healthcare Clinical Campus, Faculty of Medicine and Health, UNSW Sydney, Darlinghurst, NSW 2010, Australia
- Cancer Plasticity and Dormancy Program, Garvan Institute of Medical Research, Darlinghurst, NSW 2010Australia
| | - Weng Hua Khoo
- St Vincent’s Healthcare Clinical Campus, Faculty of Medicine and Health, UNSW Sydney, Darlinghurst, NSW 2010, Australia
- Cancer Plasticity and Dormancy Program, Garvan Institute of Medical Research, Darlinghurst, NSW 2010Australia
| | - Leonard D Goldstein
- St Vincent’s Healthcare Clinical Campus, Faculty of Medicine and Health, UNSW Sydney, Darlinghurst, NSW 2010, Australia
- Data Science Platform, Garvan Institute of Medical Research, Darlinghurst, NSW 2010, Australia
| | - Peter I Croucher
- St Vincent’s Healthcare Clinical Campus, Faculty of Medicine and Health, UNSW Sydney, Darlinghurst, NSW 2010, Australia
- Cancer Plasticity and Dormancy Program, Garvan Institute of Medical Research, Darlinghurst, NSW 2010Australia
| | - John M Murray
- School of Mathematics and Statistics, Faculty of Science, UNSW Sydney, Kensington, NSW 2033, Australia
| | - Tri Giang Phan
- Precision Immunology Program, Garvan Institute of Medical Research, Darlinghurst, NSW 2010, Australia
- St Vincent’s Healthcare Clinical Campus, Faculty of Medicine and Health, UNSW Sydney, Darlinghurst, NSW 2010, Australia
| |
Collapse
|
21
|
Li M, Nishimura T, Takeuchi Y, Hongu T, Wang Y, Shiokawa D, Wang K, Hirose H, Sasahara A, Yano M, Ishikawa S, Inokuchi M, Ota T, Tanabe M, Tada KI, Akiyama T, Cheng X, Liu CC, Yamashita T, Sugano S, Uchida Y, Chiba T, Asahara H, Nakagawa M, Sato S, Miyagi Y, Shimamura T, Nagai LAE, Kanai A, Katoh M, Nomura S, Nakato R, Suzuki Y, Tojo A, Voon DC, Ogawa S, Okamoto K, Foukakis T, Gotoh N. FXYD3 functionally demarcates an ancestral breast cancer stem cell subpopulation with features of drug-tolerant persisters. J Clin Invest 2023; 133:e166666. [PMID: 37966117 PMCID: PMC10645391 DOI: 10.1172/jci166666] [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: 11/08/2022] [Accepted: 09/21/2023] [Indexed: 11/16/2023] Open
Abstract
The heterogeneity of cancer stem cells (CSCs) within tumors presents a challenge in therapeutic targeting. To decipher the cellular plasticity that fuels phenotypic heterogeneity, we undertook single-cell transcriptomics analysis in triple-negative breast cancer (TNBC) to identify subpopulations in CSCs. We found a subpopulation of CSCs with ancestral features that is marked by FXYD domain-containing ion transport regulator 3 (FXYD3), a component of the Na+/K+ pump. Accordingly, FXYD3+ CSCs evolve and proliferate, while displaying traits of alveolar progenitors that are normally induced during pregnancy. Clinically, FXYD3+ CSCs were persistent during neoadjuvant chemotherapy, hence linking them to drug-tolerant persisters (DTPs) and identifying them as crucial therapeutic targets. Importantly, FXYD3+ CSCs were sensitive to senolytic Na+/K+ pump inhibitors, such as cardiac glycosides. Together, our data indicate that FXYD3+ CSCs with ancestral features are drivers of plasticity and chemoresistance in TNBC. Targeting the Na+/K+ pump could be an effective strategy to eliminate CSCs with ancestral and DTP features that could improve TNBC prognosis.
Collapse
Affiliation(s)
- Mengjiao Li
- Division of Cancer Cell Biology, Cancer Research Institute, and
| | | | - Yasuto Takeuchi
- Division of Cancer Cell Biology, Cancer Research Institute, and
- Institute for Frontier Science Initiative, Kanazawa University, Kanazawa City, Japan
| | - Tsunaki Hongu
- Division of Cancer Cell Biology, Cancer Research Institute, and
| | - Yuming Wang
- Division of Cancer Cell Biology, Cancer Research Institute, and
| | - Daisuke Shiokawa
- Division of Cancer Differentiation, National Cancer Center Research Institute, Chuo-ku, Tokyo, Japan
| | - Kang Wang
- Department of Oncology-Pathology, Karolinska Institute, Karolinska University Hospital, Stockholm, Sweden
| | - Haruka Hirose
- Division of Systems Biology, Graduate School of Medicine, Nagoya University, Nagoya City, Japan
| | - Asako Sasahara
- Department of Breast and Endocrine Surgery, Graduate School of Medicine, The University of Tokyo, Bunkyo-ku, Tokyo, Japan
| | - Masao Yano
- Department of Surgery, Minami-machida Hospital, Machida City, Tokyo, Japan
| | - Satoko Ishikawa
- Department of Breast Oncology, Kanazawa University Hospital, Kanazawa City, Japan
| | - Masafumi Inokuchi
- Department of Breast Oncology, Kanazawa University Hospital, Kanazawa City, Japan
| | - Tetsuo Ota
- Department of Breast Oncology, Kanazawa University Hospital, Kanazawa City, Japan
| | - Masahiko Tanabe
- Department of Breast and Endocrine Surgery, Graduate School of Medicine, The University of Tokyo, Bunkyo-ku, Tokyo, Japan
| | - Kei-ichiro Tada
- Department of Breast and Endocrine Surgery, Nihon University, Itabashi-ku, Tokyo, Japan
| | - Tetsu Akiyama
- Laboratory of Molecular and Genetic Information, Institute for Quantitative Biosciences, The University of Tokyo, Bunkyo-ku, Tokyo, Japan
| | - Xi Cheng
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Chia-Chi Liu
- North Shore Heart Research Group, Kolling Institute, University of Sydney, Sydney, New South Wales, Australia
| | - Toshinari Yamashita
- Department of Breast and Endocrine Surgery, Kanagawa Cancer Center, Yokohama City, Kanagawa, Japan
| | - Sumio Sugano
- Tokyo Medical and Dental University, Bunkyo-ku, Tokyo, Japan
| | - Yutaro Uchida
- Department of Systems Biomedicine, Tokyo Medical and Dental University, Bunkyo-ku, Tokyo, Japan
| | - Tomoki Chiba
- Department of Systems Biomedicine, Tokyo Medical and Dental University, Bunkyo-ku, Tokyo, Japan
| | - Hiroshi Asahara
- Department of Systems Biomedicine, Tokyo Medical and Dental University, Bunkyo-ku, Tokyo, Japan
| | - Masahiro Nakagawa
- Department of Pathology and Tumor Biology, Graduate School of Medicine, Kyoto University, Sakyo-ku, Kyoto, Japan
| | - Shinya Sato
- Molecular Pathology and Genetics Division, Kanagawa Cancer Center Research Institute, Yokohama City, Kanagawa, Japan
| | - Yohei Miyagi
- Molecular Pathology and Genetics Division, Kanagawa Cancer Center Research Institute, Yokohama City, Kanagawa, Japan
| | - Teppei Shimamura
- Division of Systems Biology, Graduate School of Medicine, Nagoya University, Nagoya City, Japan
| | | | - Akinori Kanai
- Department of Computational Biology and Medical Sciences, Graduate School of Frontier Biosciences
| | - Manami Katoh
- Department of Cardiovascular Medicine, Graduate School of Medicine
- Genome Science Division, Research Center for Advanced Science and Technology
| | - Seitaro Nomura
- Department of Cardiovascular Medicine, Graduate School of Medicine
- Genome Science Division, Research Center for Advanced Science and Technology
- Department of Frontier Cardiovascular Science, Graduate School of Medicine, and
| | - Ryuichiro Nakato
- Laboratory of Computational Genomics, Institute for Quantitative Biosciences
| | - Yutaka Suzuki
- Department of Computational Biology and Medical Sciences, Graduate School of Frontier Biosciences
| | - Arinobu Tojo
- Tokyo Medical and Dental University, Bunkyo-ku, Tokyo, Japan
- Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Dominic C. Voon
- Institute for Frontier Science Initiative, Kanazawa University, Kanazawa City, Japan
- Inflammation and Epithelial Plasticity Unit, Cancer Research Institute, Kanazawa University, Kanazawa City, Japan
| | - Seishi Ogawa
- Department of Pathology and Tumor Biology, Graduate School of Medicine, Kyoto University, Sakyo-ku, Kyoto, Japan
| | - Koji Okamoto
- Division of Cancer Differentiation, National Cancer Center Research Institute, Chuo-ku, Tokyo, Japan
- Advanced Comprehensive Research Organization, Teikyo University, Itabashi-ku, Tokyo, Japan
| | - Theodoros Foukakis
- Department of Oncology-Pathology, Karolinska Institute, Karolinska University Hospital, Stockholm, Sweden
| | - Noriko Gotoh
- Division of Cancer Cell Biology, Cancer Research Institute, and
- Institute for Frontier Science Initiative, Kanazawa University, Kanazawa City, Japan
| |
Collapse
|
22
|
Liu Y, Kim YS, Xue X, Kobayashi N, Sun S, Yang Q, Pourquié O, Fu J. A human pluripotent stem cell-based somitogenesis model using microfluidics. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.10.29.564399. [PMID: 37961125 PMCID: PMC10634932 DOI: 10.1101/2023.10.29.564399] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/15/2023]
Abstract
Emerging human pluripotent stem cell (hPSC)-based embryo models are useful for studying human embryogenesis. Particularly, there are hPSC-based somitogenesis models using free-floating culture that recapitulate somite formation. Somitogenesis in vivo involves intricately orchestrated bio-chemical and -mechanical events. However, none of the current somitogenesis models controls biochemical gradients or biomechanical signals in the culture, limiting their applicability to untangle complex biochemical-biomechanical interactions that drive somitogenesis. Here we report a new human somitogenesis model by confining hPSC-derived presomitic mesoderm (PSM) tissues in microfabricated trenches. Exogenous microfluidic morphogen gradients imposed on PSM cause axial patterning and trigger spontaneous rostral-to-caudal somite formation. A mechanical theory is developed to explain the size dependency between somites and PSM. The microfluidic somitogenesis model is further exploited to reveal regulatory roles of cellular and tissue biomechanics in somite formation. This study presents a useful microengineered, hPSC-based model for understanding the bio-chemical and -mechanical events that guide somite formation.
Collapse
|
23
|
Zheng SC, Stein-O’Brien G, Boukas L, Goff LA, Hansen KD. Pumping the brakes on RNA velocity by understanding and interpreting RNA velocity estimates. Genome Biol 2023; 24:246. [PMID: 37885016 PMCID: PMC10601342 DOI: 10.1186/s13059-023-03065-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Accepted: 09/19/2023] [Indexed: 10/28/2023] Open
Abstract
BACKGROUND RNA velocity analysis of single cells offers the potential to predict temporal dynamics from gene expression. In many systems, RNA velocity has been observed to produce a vector field that qualitatively reflects known features of the system. However, the limitations of RNA velocity estimates are still not well understood. RESULTS We analyze the impact of different steps in the RNA velocity workflow on direction and speed. We consider both high-dimensional velocity estimates and low-dimensional velocity vector fields mapped onto an embedding. We conclude the transition probability method for mapping velocity estimates onto an embedding is effectively interpolating in the embedding space. Our findings reveal a significant dependence of the RNA velocity workflow on smoothing via the k-nearest-neighbors (k-NN) graph of the observed data. This reliance results in considerable estimation errors for both direction and speed in both high- and low-dimensional settings when the k-NN graph fails to accurately represent the true data structure; this is an unknown feature of real data. RNA velocity performs poorly at estimating speed in both low- and high-dimensional spaces, except in very low noise settings. We introduce a novel quality measure that can identify when RNA velocity should not be used. CONCLUSIONS Our findings emphasize the importance of choices in the RNA velocity workflow and highlight critical limitations of data analysis. We advise against over-interpreting expression dynamics using RNA velocity, particularly in terms of speed. Finally, we emphasize that the use of RNA velocity in assessing the correctness of a low-dimensional embedding is circular.
Collapse
Affiliation(s)
- Shijie C. Zheng
- Department of Biostatistics, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD USA
| | - Genevieve Stein-O’Brien
- Department of Genetic Medicine, Johns Hopkins School of Medicine, Baltimore, MD USA
- Department of Neuroscience, Johns Hopkins School of Medicine, Baltimore, MD USA
- Kavli Neurodiscovery Institute, Johns Hopkins University, Baltimore, MD USA
- Quantitative Sciences Division, Department of Oncology, Johns Hopkins School of Medicine, Baltimore, MD USA
| | - Leandros Boukas
- Department of Biostatistics, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD USA
- Department of Genetic Medicine, Johns Hopkins School of Medicine, Baltimore, MD USA
| | - Loyal A. Goff
- Department of Genetic Medicine, Johns Hopkins School of Medicine, Baltimore, MD USA
- Department of Neuroscience, Johns Hopkins School of Medicine, Baltimore, MD USA
- Kavli Neurodiscovery Institute, Johns Hopkins University, Baltimore, MD USA
| | - Kasper D. Hansen
- Department of Biostatistics, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD USA
- Department of Genetic Medicine, Johns Hopkins School of Medicine, Baltimore, MD USA
| |
Collapse
|
24
|
Shi Q, Chen X, Zhang Z. Decoding Human Biology and Disease Using Single-cell Omics Technologies. GENOMICS, PROTEOMICS & BIOINFORMATICS 2023; 21:926-949. [PMID: 37739168 PMCID: PMC10928380 DOI: 10.1016/j.gpb.2023.06.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Revised: 05/22/2023] [Accepted: 06/08/2023] [Indexed: 09/24/2023]
Abstract
Over the past decade, advances in single-cell omics (SCO) technologies have enabled the investigation of cellular heterogeneity at an unprecedented resolution and scale, opening a new avenue for understanding human biology and disease. In this review, we summarize the developments of sequencing-based SCO technologies and computational methods, and focus on considerable insights acquired from SCO sequencing studies to understand normal and diseased properties, with a particular emphasis on cancer research. We also discuss the technological improvements of SCO and its possible contribution to fundamental research of the human, as well as its great potential in clinical diagnoses and personalized therapies of human disease.
Collapse
Affiliation(s)
- Qiang Shi
- Biomedical Pioneering Innovation Center, School of Life Sciences, Peking University, Beijing 100871, China
| | - Xueyan Chen
- Biomedical Pioneering Innovation Center, School of Life Sciences, Peking University, Beijing 100871, China
| | - Zemin Zhang
- Biomedical Pioneering Innovation Center, School of Life Sciences, Peking University, Beijing 100871, China; Peking-Tsinghua Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing 100871, China; Changping Laboratory, Beijing 102206, China.
| |
Collapse
|
25
|
Farrell S, Mani M, Goyal S. Inferring single-cell transcriptomic dynamics with structured latent gene expression dynamics. CELL REPORTS METHODS 2023; 3:100581. [PMID: 37708894 PMCID: PMC10545944 DOI: 10.1016/j.crmeth.2023.100581] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Revised: 06/16/2023] [Accepted: 08/18/2023] [Indexed: 09/16/2023]
Abstract
Gene expression dynamics provide directional information for trajectory inference from single-cell RNA sequencing data. Traditional approaches compute RNA velocity using strict modeling assumptions about transcription and splicing of RNA. This can fail in scenarios where multiple lineages have distinct gene dynamics or where rates of transcription and splicing are time dependent. We present "LatentVelo," an approach to compute a low-dimensional representation of gene dynamics with deep learning. LatentVelo embeds cells into a latent space with a variational autoencoder and models differentiation dynamics on this "dynamics-based" latent space with neural ordinary differential equations. LatentVelo infers a latent regulatory state that controls the dynamics of an individual cell to model multiple lineages. LatentVelo can predict latent trajectories, describing the inferred developmental path for individual cells rather than just local RNA velocity vectors. The dynamics-based embedding batch corrects cell states and velocities, outperforming comparable autoencoder batch correction methods that do not consider gene expression dynamics.
Collapse
Affiliation(s)
- Spencer Farrell
- Department of Physics, University of Toronto, Toronto, ON M5S1A7, Canada.
| | - Madhav Mani
- Department of Engineering Sciences and Applied Mathematics, Northwestern University, Evanston, IL 60208, USA; NSF-Simons Center for Quantitative Biology, Northwestern University, Evanston, IL 60208, USA; Department of Molecular Biosciences, Northwestern University, Evanston, IL 60208, USA
| | - Sidhartha Goyal
- Department of Physics, University of Toronto, Toronto, ON M5S1A7, Canada; Institute of Biomedical Engineering, University of Toronto, Toronto, ON M5S3G9, Canada.
| |
Collapse
|
26
|
Burdziak C, Zhao CJ, Haviv D, Alonso-Curbelo D, Lowe SW, Pe’er D. scKINETICS: inference of regulatory velocity with single-cell transcriptomics data. Bioinformatics 2023; 39:i394-i403. [PMID: 37387147 PMCID: PMC10311321 DOI: 10.1093/bioinformatics/btad267] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/01/2023] Open
Abstract
MOTIVATION Transcriptional dynamics are governed by the action of regulatory proteins and are fundamental to systems ranging from normal development to disease. RNA velocity methods for tracking phenotypic dynamics ignore information on the regulatory drivers of gene expression variability through time. RESULTS We introduce scKINETICS (Key regulatory Interaction NETwork for Inferring Cell Speed), a dynamical model of gene expression change which is fit with the simultaneous learning of per-cell transcriptional velocities and a governing gene regulatory network. Fitting is accomplished through an expectation-maximization approach designed to learn the impact of each regulator on its target genes, leveraging biologically motivated priors from epigenetic data, gene-gene coexpression, and constraints on cells' future states imposed by the phenotypic manifold. Applying this approach to an acute pancreatitis dataset recapitulates a well-studied axis of acinar-to-ductal transdifferentiation whilst proposing novel regulators of this process, including factors with previously appreciated roles in driving pancreatic tumorigenesis. In benchmarking experiments, we show that scKINETICS successfully extends and improves existing velocity approaches to generate interpretable, mechanistic models of gene regulatory dynamics. AVAILABILITY AND IMPLEMENTATION All python code and an accompanying Jupyter notebook with demonstrations are available at http://github.com/dpeerlab/scKINETICS.
Collapse
Affiliation(s)
- Cassandra Burdziak
- Computational and Systems Biology Program, Memorial Sloan Kettering Cancer Center, Sloan Kettering Institute, 408 E 69th Street, New York, NY 10021, United States
| | - Chujun Julia Zhao
- Computational and Systems Biology Program, Memorial Sloan Kettering Cancer Center, Sloan Kettering Institute, 408 E 69th Street, New York, NY 10021, United States
- Department of Biomedical Engineering, Columbia University, 1210 Amsterdam Ave, New York, NY 10027, United States
| | - Doron Haviv
- Computational and Systems Biology Program, Memorial Sloan Kettering Cancer Center, Sloan Kettering Institute, 408 E 69th Street, New York, NY 10021, United States
| | - Direna Alonso-Curbelo
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology (BIST), Carrer de Baldiri Reixac, 10, Barcelona 08028, Spain
- Cancer Biology and Genetics Program, Memorial Sloan Kettering Cancer Center, Sloan Kettering Institute, 408 E 69th Street, New York, NY 10021, United States
| | - Scott W Lowe
- Cancer Biology and Genetics Program, Memorial Sloan Kettering Cancer Center, Sloan Kettering Institute, 408 E 69th Street, New York, NY 10021, United States
- Howard Hughes Medical Institute, 4000 Jones Bridge Road, Chevy Chase, Maryland 20815, United States
| | - Dana Pe’er
- Computational and Systems Biology Program, Memorial Sloan Kettering Cancer Center, Sloan Kettering Institute, 408 E 69th Street, New York, NY 10021, United States
- Howard Hughes Medical Institute, 4000 Jones Bridge Road, Chevy Chase, Maryland 20815, United States
| |
Collapse
|
27
|
Fernández-Moya SM, Ganesh AJ, Plass M. Neural cell diversity in the light of single-cell transcriptomics. Transcription 2023; 14:158-176. [PMID: 38229529 PMCID: PMC10807474 DOI: 10.1080/21541264.2023.2295044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Revised: 10/02/2023] [Accepted: 11/10/2023] [Indexed: 01/18/2024] Open
Abstract
The development of highly parallel and affordable high-throughput single-cell transcriptomics technologies has revolutionized our understanding of brain complexity. These methods have been used to build cellular maps of the brain, its different regions, and catalog the diversity of cells in each of them during development, aging and even in disease. Now we know that cellular diversity is way beyond what was previously thought. Single-cell transcriptomics analyses have revealed that cell types previously considered homogeneous based on imaging techniques differ depending on several factors including sex, age and location within the brain. The expression profiles of these cells have also been exploited to understand which are the regulatory programs behind cellular diversity and decipher the transcriptional pathways driving them. In this review, we summarize how single-cell transcriptomics have changed our view on the cellular diversity in the human brain, and how it could impact the way we study neurodegenerative diseases. Moreover, we describe the new computational approaches that can be used to study cellular differentiation and gain insight into the functions of individual cell populations under different conditions and their alterations in disease.
Collapse
Affiliation(s)
- Sandra María Fernández-Moya
- Gene Regulation of Cell Identity, Regenerative Medicine Program, Bellvitge Institute for Biomedical Research (IDIBELL), Barcelona, L’Hospitalet del Llobregat, Spain
- Program for Advancing Clinical Translation of Regenerative Medicine of Catalonia, P- CMR[C], Barcelona, L’Hospitalet del Llobregat, Spain
| | - Akshay Jaya Ganesh
- Gene Regulation of Cell Identity, Regenerative Medicine Program, Bellvitge Institute for Biomedical Research (IDIBELL), Barcelona, L’Hospitalet del Llobregat, Spain
- Program for Advancing Clinical Translation of Regenerative Medicine of Catalonia, P- CMR[C], Barcelona, L’Hospitalet del Llobregat, Spain
| | - Mireya Plass
- Gene Regulation of Cell Identity, Regenerative Medicine Program, Bellvitge Institute for Biomedical Research (IDIBELL), Barcelona, L’Hospitalet del Llobregat, Spain
- Program for Advancing Clinical Translation of Regenerative Medicine of Catalonia, P- CMR[C], Barcelona, L’Hospitalet del Llobregat, Spain
- Center for Networked Biomedical Research on Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Madrid, Spain
| |
Collapse
|
28
|
He D, Soneson C, Patro R. Understanding and evaluating ambiguity in single-cell and single-nucleus RNA-sequencing. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.01.04.522742. [PMID: 36711921 PMCID: PMC9881993 DOI: 10.1101/2023.01.04.522742] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Recently, a new modification has been proposed by Hjörleifsson and Sullivan et al. to the model used to classify the splicing status of reads (as spliced (mature), unspliced (nascent), or ambiguous) in single-cell and single-nucleus RNA-seq data. Here, we evaluate both the theoretical basis and practical implementation of the proposed method. The proposed method is highly-conservative, and therefore, unlikely to mischaracterize reads as spliced (mature) or unspliced (nascent) when they are not. However, we find that it leaves a large fraction of reads classified as ambiguous, and, in practice, allocates these ambiguous reads in an all-or-nothing manner, and differently between single-cell and single-nucleus RNA-seq data. Further, as implemented in practice, the ambiguous classification is implicit and based on the index against which the reads are mapped, which leads to several drawbacks compared to methods that consider both spliced (mature) and unspliced (nascent) mapping targets simultaneously - for example, the ability to use confidently assigned reads to rescue ambiguous reads based on shared UMIs and gene targets. Nonetheless, we show that these conservative assignment rules can be obtained directly in existing approaches simply by altering the set of targets that are indexed. To this end, we introduce the spliceu reference and show that its use with alevin-fry recapitulates the more conservative proposed classification. We also observe that, on experimental data, and under the proposed allocation rules for ambiguous UMIs, the difference between the proposed classification scheme and existing conventions appears much smaller than previously reported. We demonstrate the use of the new piscem index for mapping simultaneously against spliced (mature) and unspliced (nascent) targets, allowing classification against the full nascent and mature transcriptome in human or mouse in <3GB of memory. Finally, we discuss the potential of incorporating probabilistic evidence into the inference of splicing status, and suggest that it may provide benefits beyond what can be obtained from discrete classification of UMIs as splicing-ambiguous.
Collapse
Affiliation(s)
- Dongze He
- Department of Cell Biology and Molecular Genetics and Center for Bioinformatics and Computational Biology, University of Maryland, College Park, MD, USA
| | - Charlotte Soneson
- Friedrich Miescher Institute for Biomedical Research, Basel, Switzerland
- SIB Swiss Institute of Bioinformatics, Basel, Switzerland
| | - Rob Patro
- Department of Computer Science and Center for Bioinformatics and Computational Biology, University of Maryland, College Park, MD, USA
| |
Collapse
|