1
|
Simard S, Matosin N, Mechawar N. Adult Hippocampal Neurogenesis in the Human Brain: Updates, Challenges, and Perspectives. Neuroscientist 2025; 31:141-158. [PMID: 38757781 DOI: 10.1177/10738584241252581] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/18/2024]
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
|
2
|
Jin Z, Zhou X, Fang Z. DelaySSA: stochastic simulation of biochemical systems and gene regulatory networks with or without time delays. PLoS Comput Biol 2025; 21:e1012919. [PMID: 40198732 PMCID: PMC11977973 DOI: 10.1371/journal.pcbi.1012919] [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: 10/08/2024] [Accepted: 02/26/2025] [Indexed: 04/10/2025] Open
Abstract
Stochastic Simulation Algorithm (SSA) is crucial for modeling biochemical reactions and gene regulatory networks. Traditional SSA is characterized by Markovian property and cannot naturally model systems with time delays. Several algorithms have already been designed to handle delayed reactions, yet few easy-to-use implementations exist. To address these challenges, we have developed DelaySSA, an R package that implements currently available algorithms for SSA with or without delays. Meanwhile, we also provided Matlab and Python versions to support wider applications. We demonstrated its accuracy and validity by simulating two classical models: the Bursty model and Refractory model. We then tested its capability to simulate the RNA Velocity model, where it successfully reproduced both the up- and down-regulation stages in the phase portrait. Finally, we extended its application to simulate a gene regulatory network of lung cancer adeno-to-squamous transition (AST) and qualitatively analyzed its bistability behavior by approximating the Waddington's landscape. Modeling the therapeutic intervention of a SOX2 degrader as a delayed degradation reaction, AST is effectively blocked and reprogrammed back to the adenocarcinoma state, providing a useful clue for targeting drug-resistant AST in the future. Taken together, DelaySSA is a powerful and easy-to-use software suite, facilitating accurate modeling of various kinds of biological systems and broadening the scope of stochastic simulations in systems biology.
Collapse
Affiliation(s)
- Ziyan Jin
- Department of Colorectal Surgery and Oncology of the Second Affiliated Hospital, and Centre of Biomedical Systems and Informatics of Zhejiang University-University of Edinburgh Institute (ZJU-UoE Institute), Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China
| | - Xinyi Zhou
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai, China
| | - Zhaoyuan Fang
- Department of Colorectal Surgery and Oncology of the Second Affiliated Hospital, and Centre of Biomedical Systems and Informatics of Zhejiang University-University of Edinburgh Institute (ZJU-UoE Institute), Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China
- Edinburgh Medical School, College of Medicine and Veterinary Medicine, The University of Edinburgh, Edinburgh, United Kingdom
- Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, Hangzhou, China
- Biomedical and Health Translational Research Center of Zhejiang Province, Haining, China
| |
Collapse
|
3
|
Aivazidis A, Memi F, Kleshchevnikov V, Er S, Clarke B, Stegle O, Bayraktar OA. Cell2fate infers RNA velocity modules to improve cell fate prediction. Nat Methods 2025; 22:698-707. [PMID: 40032996 PMCID: PMC11978503 DOI: 10.1038/s41592-025-02608-3] [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: 08/02/2023] [Accepted: 01/23/2025] [Indexed: 03/05/2025]
Abstract
RNA velocity exploits the temporal information contained in spliced and unspliced RNA counts to infer transcriptional dynamics. Existing velocity models often rely on coarse biophysical simplifications or numerical approximations to solve the underlying ordinary differential equations (ODEs), which can compromise accuracy in challenging settings, such as complex or weak transcription rate changes across cellular trajectories. Here we present cell2fate, a formulation of RNA velocity based on a linearization of the velocity ODE, which allows solving a biophysically more accurate model in a fully Bayesian fashion. As a result, cell2fate decomposes the RNA velocity solutions into modules, providing a biophysical connection between RNA velocity and statistical dimensionality reduction. We comprehensively benchmark cell2fate in real-world settings, demonstrating enhanced interpretability and power to reconstruct complex dynamics and weak dynamical signals in rare and mature cell types. Finally, we apply cell2fate to the developing human brain, where we spatially map RNA velocity modules onto the tissue architecture, connecting the spatial organization of tissues with temporal dynamics of transcription.
Collapse
Affiliation(s)
| | - Fani Memi
- Wellcome Sanger Institute, Cambridge, UK
| | | | - Sezgin Er
- International School of Medicine, Istanbul Medipol University, Istanbul, Turkey
| | - Brian Clarke
- Division of Computational Genomics and Systems Genetics, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Oliver Stegle
- Wellcome Sanger Institute, Cambridge, UK.
- Division of Computational Genomics and Systems Genetics, German Cancer Research Center (DKFZ), Heidelberg, Germany.
- Genome Biology Unit, European Molecular Biology Laboratory, Heidelberg, Germany.
| | | |
Collapse
|
4
|
Bhattacharya S, Tie G, Singh PNP, Malagola E, Eskiocak O, He R, Kraiczy J, Gu W, Perlov Y, Alici-Garipcan A, Beyaz S, Wang TC, Zhou Q, Shivdasani RA. Intestinal secretory differentiation reflects niche-driven phenotypic and epigenetic plasticity of a common signal-responsive terminal cell. Cell Stem Cell 2025:S1934-5909(25)00095-5. [PMID: 40203837 DOI: 10.1016/j.stem.2025.03.005] [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: 09/19/2024] [Revised: 12/27/2024] [Accepted: 03/10/2025] [Indexed: 04/11/2025]
Abstract
Enterocytes and four classic secretory cell types derive from intestinal epithelial stem cells. Based on morphology, location, and canonical markers, goblet and Paneth cells are considered distinct secretory types. Here, we report high overlap in their transcripts and sites of accessible chromatin, in marked contrast to those of their enteroendocrine or tuft cell siblings. Mouse and human goblet and Paneth cells express extraordinary fractions of few antimicrobial genes, which reflect specific responses to local niches. Wnt signaling retains some ATOH1+ secretory cells in crypt bottoms, where the absence of BMP signaling potently induces Paneth features. Cells that migrate away from crypt bottoms encounter BMPs and thereby acquire goblet properties. These phenotypes and underlying accessible cis-elements interconvert in post-mitotic cells. Thus, goblet and Paneth properties represent alternative phenotypic manifestations of a common signal-responsive terminal cell type. These findings reveal exquisite niche-dependent cell plasticity and cis-regulatory dynamics in likely response to antimicrobial needs.
Collapse
Affiliation(s)
- Swarnabh Bhattacharya
- Department of Medical Oncology and Center for Functional Cancer Epigenetics, Dana-Farber Cancer Institute, Boston, MA 02215, USA; Department of Medicine, Harvard Medical School, Boston, MA 02115, USA
| | - Guodong Tie
- Department of Medical Oncology and Center for Functional Cancer Epigenetics, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - Pratik N P Singh
- Department of Medical Oncology and Center for Functional Cancer Epigenetics, Dana-Farber Cancer Institute, Boston, MA 02215, USA; Department of Medicine, Harvard Medical School, Boston, MA 02115, USA
| | - Ermanno Malagola
- Division of Digestive and Liver Diseases, Department of Medicine and Irving Cancer Research Center, Columbia University Medical Center, New York, NY 10032, USA
| | - Onur Eskiocak
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724, USA; Graduate Program in Genetics, State University of New York, Stony Brook, NY 11794, USA
| | - Ruiyang He
- Department of Medical Oncology and Center for Functional Cancer Epigenetics, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - Judith Kraiczy
- Department of Medical Oncology and Center for Functional Cancer Epigenetics, Dana-Farber Cancer Institute, Boston, MA 02215, USA; Department of Medicine, Harvard Medical School, Boston, MA 02115, USA
| | - Wei Gu
- Division of Regenerative Medicine & Hartman Institute for Therapeutic Organ Regeneration, Department of Medicine, Weill Cornell Medicine, New York, NY 10065, USA
| | - Yakov Perlov
- Department of Medical Oncology and Center for Functional Cancer Epigenetics, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | | | - Semir Beyaz
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724, USA
| | - Timothy C Wang
- Division of Digestive and Liver Diseases, Department of Medicine and Irving Cancer Research Center, Columbia University Medical Center, New York, NY 10032, USA
| | - Qiao Zhou
- Division of Regenerative Medicine & Hartman Institute for Therapeutic Organ Regeneration, Department of Medicine, Weill Cornell Medicine, New York, NY 10065, USA
| | - Ramesh A Shivdasani
- Department of Medical Oncology and Center for Functional Cancer Epigenetics, Dana-Farber Cancer Institute, Boston, MA 02215, USA; Department of Medicine, Harvard Medical School, Boston, MA 02115, USA; Harvard Stem Cell Institute, Cambridge, MA 02138, USA.
| |
Collapse
|
5
|
Llera-Oyola J, Pérez-Moraga R, Parras M, Rosón B. How to view the female reproductive tract through single-cell looking glasses. Am J Obstet Gynecol 2025; 232:S21-S43. [PMID: 40253081 DOI: 10.1016/j.ajog.2024.08.040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Revised: 07/04/2024] [Accepted: 08/24/2024] [Indexed: 04/21/2025]
Abstract
Single-cell technologies have emerged as an unprecedented tool for biologists and clinicians, allowing them to assess organs and tissues at the level of individual cells. In the field of women's reproductive biology, single-cell studies have provided insights into the cellular and molecular processes that regulate reproductive and obstetrical functions in health and disease. The knowledge that these studies generate is helping clinicians to improve the understanding and diagnosis of infertility related issues or pregnancy complications and to find new avenues for their treatment. However, navigating the expansive landscape of this type of transcriptomic data analysis represents a pivotal challenge in current research. Single cell RNA sequencing involves isolating cells into droplets, reverse transcribing RNA to generate complementary DNA, with each droplet content uniquely labeled by a barcode. Upon sequencing the complementary DNAs, the barcodes enable the reassignment of sequencing reads to individual droplets, facilitating the reconstruction of the cellular landscape of the sample obtained from a tissue or organ and beyond. Researchers, equipped with the metaphorical 'single-cell glasses,' must adequately choose from a plethora of strategies to dissect and interpret cellular information. Sophisticated algorithms and the decision-making process are often underestimated, resulting in artefactual or cumbersome interpreted results. Computational biologists apply and innovate computational tools designed to process, model, and interpret expansive datasets. The ramifications of their work extend far beyond the realm of data processing; they give shape to the outcome of analyses, playing a pivotal role in drawing meaningful conclusions from the wealth of information garnered. In this review, we describe the wide variety of approaches and analytical steps available with enough detail to gain a concise picture of what a complete examination of a single-cell dataset would be. We commence with a discussion on key points in experimental design, highlighting crucial questions one should consider. Following this, we delve into the various preprocessing and quality control steps essential for any single-cell dataset. The subsequent section offers a detailed guide on constructing a single-cell atlas, exploring nuances such as differential characteristics in visualization and clustering techniques, as well as strategies for assigning identity to cell populations through gene marker annotations. Moving beyond the creation of an atlas, we explore methods for investigating pathological conditions. This involves conducting cell population comparison tests between conditions and analyzing specific cell-to-cell communications and cellular differentiation trajectories in both health and disease scenarios. This work aims to furnish a newcomer researcher and/or clinician with essential guidelines to embark on a single-cell adventure without succumbing to common pitfalls. By bridging the gap between theory and practice, it facilitates the translation of single-cell technologies into clinically relevant applications. Throughout the manuscript, practical examples of its usage in women's reproductive health studies are provided. Various sections delve into specific clinical scenarios, demonstrating how these guidelines can be instrumental in unraveling the molecular landscapes of diseases and physiological processes related to women's reproduction.
Collapse
Affiliation(s)
- Jaime Llera-Oyola
- Carlos Simon Foundation, INCLIVA Health Research Institute, Valencia, Spain
| | - Raúl Pérez-Moraga
- Carlos Simon Foundation, INCLIVA Health Research Institute, Valencia, Spain; R&D Department, Igenomix, Valencia, Spain
| | - Marcos Parras
- Carlos Simon Foundation, INCLIVA Health Research Institute, Valencia, Spain
| | - Beatriz Rosón
- Carlos Simon Foundation, INCLIVA Health Research Institute, Valencia, Spain.
| |
Collapse
|
6
|
Zhang Y, Nomura M, Nishimura K, Zang W, Koike Y, Xiao M, Ito H, Fukumoto M, Tanaka A, Aoyama Y, Saika W, Hasegawa C, Yamazaki H, Takaori-Kondo A, Inoue D. In-depth functional analysis of BRD9 in fetal hematopoiesis reveals context-dependent roles. iScience 2025; 28:112010. [PMID: 40109374 PMCID: PMC11919606 DOI: 10.1016/j.isci.2025.112010] [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: 07/18/2024] [Revised: 10/19/2024] [Accepted: 02/10/2025] [Indexed: 03/22/2025] Open
Abstract
The hierarchical organization of hematopoietic stem cells (HSCs) governing adult hematopoiesis has been extensively investigated. However, the dynamic epigenomic transition from fetal to adult hematopoiesis remains incompletely understood, particularly regarding the involvement of epigenetic factors. In this study, we investigate the roles of BRD9, an essential component of the non-canonical BAF (ncBAF) complex known to govern the fate of adult HSCs, in fetal hematopoiesis. Consistent with observations in adult hematopoiesis, BRD9 loss impairs fetal HSC stemness and disturbs erythroid maturation. Intriguingly, the impact on myeloid lineage was discrepant: BRD9 loss inhibited and promoted myeloid differentiation in fetal and adult models, respectively. Through comprehensive transcriptomic and epigenomic analysis, we elucidate the differential roles of BRD9 in a context- and lineage-dependent manner. Our data uncover how BRD9/ncBAF complex modulates transcription in a stage-specific manner, providing deeper insights into the epigenetic regulation underlying the transition from fetal to adult hematopoiesis.
Collapse
Affiliation(s)
- Yifan Zhang
- Department of Hematology-Oncology, Institute of Biomedical Research and Innovation, Foundation for Biomedical Research and Innovation at Kobe, Kobe, Japan
- Department of Hematology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Masaki Nomura
- Department of Hematology-Oncology, Institute of Biomedical Research and Innovation, Foundation for Biomedical Research and Innovation at Kobe, Kobe, Japan
- Facility for iPS Cell Therapy, CiRA Foundation, Kyoto, Japan
- Department of Cancer Pathology, Graduate School of Medicine and Frontier Biosciences, Osaka University, Suita, Japan
| | - Koutarou Nishimura
- Department of Hematology-Oncology, Institute of Biomedical Research and Innovation, Foundation for Biomedical Research and Innovation at Kobe, Kobe, Japan
- Department of Cancer Pathology, Graduate School of Medicine and Frontier Biosciences, Osaka University, Suita, Japan
| | - Weijia Zang
- Department of Hematology-Oncology, Institute of Biomedical Research and Innovation, Foundation for Biomedical Research and Innovation at Kobe, Kobe, Japan
- Department of Hematology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
- Department of Cancer Pathology, Graduate School of Medicine and Frontier Biosciences, Osaka University, Suita, Japan
| | - Yui Koike
- Department of Hematology-Oncology, Institute of Biomedical Research and Innovation, Foundation for Biomedical Research and Innovation at Kobe, Kobe, Japan
- Department of Cancer Pathology, Graduate School of Medicine and Frontier Biosciences, Osaka University, Suita, Japan
| | - Muran Xiao
- Department of Hematology-Oncology, Institute of Biomedical Research and Innovation, Foundation for Biomedical Research and Innovation at Kobe, Kobe, Japan
| | - Hiromi Ito
- Department of Hematology-Oncology, Institute of Biomedical Research and Innovation, Foundation for Biomedical Research and Innovation at Kobe, Kobe, Japan
- Department of Cancer Pathology, Graduate School of Medicine and Frontier Biosciences, Osaka University, Suita, Japan
| | - Miki Fukumoto
- Department of Hematology-Oncology, Institute of Biomedical Research and Innovation, Foundation for Biomedical Research and Innovation at Kobe, Kobe, Japan
| | - Atsushi Tanaka
- Department of Hematology-Oncology, Institute of Biomedical Research and Innovation, Foundation for Biomedical Research and Innovation at Kobe, Kobe, Japan
| | - Yumi Aoyama
- Department of Hematology-Oncology, Institute of Biomedical Research and Innovation, Foundation for Biomedical Research and Innovation at Kobe, Kobe, Japan
- Department of Hematology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Wataru Saika
- Department of Hematology-Oncology, Institute of Biomedical Research and Innovation, Foundation for Biomedical Research and Innovation at Kobe, Kobe, Japan
- Department of Cancer Pathology, Graduate School of Medicine and Frontier Biosciences, Osaka University, Suita, Japan
- Department of Hematology, Shiga University of Medical Science, Otsu, Japan
| | - Chihiro Hasegawa
- Department of Hematology-Oncology, Institute of Biomedical Research and Innovation, Foundation for Biomedical Research and Innovation at Kobe, Kobe, Japan
- Department of Cancer Pathology, Graduate School of Medicine and Frontier Biosciences, Osaka University, Suita, Japan
- Department of Hematology and Oncology, Graduate School of Medicine, Osaka University, Suita, Japan
| | - Hiromi Yamazaki
- Department of Hematology-Oncology, Institute of Biomedical Research and Innovation, Foundation for Biomedical Research and Innovation at Kobe, Kobe, Japan
- Department of Cancer Pathology, Graduate School of Medicine and Frontier Biosciences, Osaka University, Suita, Japan
| | - Akifumi Takaori-Kondo
- Department of Hematology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Daichi Inoue
- Department of Hematology-Oncology, Institute of Biomedical Research and Innovation, Foundation for Biomedical Research and Innovation at Kobe, Kobe, Japan
- Department of Hematology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
- Department of Cancer Pathology, Graduate School of Medicine and Frontier Biosciences, Osaka University, Suita, Japan
- Institute for Open and Transdisciplinary Research Initiatives, Osaka University, Suita, Japan
| |
Collapse
|
7
|
Feng Y, Liu G, Li H, Cheng L. The landscape of cell lineage tracing. SCIENCE CHINA. LIFE SCIENCES 2025:10.1007/s11427-024-2751-6. [PMID: 40035969 DOI: 10.1007/s11427-024-2751-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Accepted: 09/30/2024] [Indexed: 03/06/2025]
Abstract
Cell fate changes play a crucial role in the processes of natural development, disease progression, and the efficacy of therapeutic interventions. The definition of the various types of cell fate changes, including cell expansion, differentiation, transdifferentiation, dedifferentiation, reprogramming, and state transitions, represents a complex and evolving field of research known as cell lineage tracing. This review will systematically introduce the research history and progress in this field, which can be broadly divided into two parts: prospective tracing and retrospective tracing. The initial section encompasses an array of methodologies pertaining to isotope labeling, transient fluorescent tracers, non-fluorescent transient tracers, non-fluorescent genetic markers, fluorescent protein, genetic marker delivery, genetic recombination, exogenous DNA barcodes, CRISPR-Cas9 mediated DNA barcodes, and base editor-mediated DNA barcodes. The second part of the review covers genetic mosaicism, genomic DNA alteration, TCR/BCR, DNA methylation, and mitochondrial DNA mutation. In the final section, we will address the principal challenges and prospective avenues of enquiry in the field of cell lineage tracing, with a particular focus on the sequencing techniques and mathematical models pertinent to single-cell genetic lineage tracing, and the value of pursuing a more comprehensive investigation at both the spatial and temporal levels in the study of cell lineage tracing.
Collapse
Affiliation(s)
- Ye Feng
- Shanghai YangZhi Rehabilitation Hospital (Shanghai Sunshine Rehabilitation Center), Tongji University School of Medicine, Shanghai, 201619, China.
| | - Guang Liu
- Department of Vascular Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200023, China.
| | - Haiqing Li
- Department of Cardiac Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
| | - Lin Cheng
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
| |
Collapse
|
8
|
Xu Y, Chan MTJ, Yang M, Meng H, Chen CH. Time-resolved single-cell secretion analysis via microfluidics. LAB ON A CHIP 2025; 25:1282-1295. [PMID: 39789982 DOI: 10.1039/d4lc00904e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/12/2025]
Abstract
Revealing how individual cells alter their secretions over time is crucial for understanding their responses to environmental changes. Key questions include: When do cells modify their functions and states? What transitions occur? Insights into the kinetic secretion trajectories of various cell types are essential for unraveling complex biological systems. This review highlights seven microfluidic technologies for time-resolved single-cell secretion analysis: 1. Microwell real-time electrical detection: uses microelectrodes for precise, cell-specific, real-time measurement of secreted molecules. 2. Microwell real-time optical detection: employs advanced optical systems for real-time, multiplexed monitoring of cellular secretions. 3. Microvalve real-time optical detection: dynamically analyzes secretions under controlled in situ stimuli, enabling detailed kinetic studies at the single-cell level. 4. Droplet real-time optical detection: provides superior throughput by generating droplets containing single cells and sensors for high-throughput screening. 5. Microwell time-barcoded optical detection: utilizes sequential barcoding techniques to facilitate scalable assays for tracking multiple secretions over time. 6. Microvalve time-barcoded optical detection: incorporates automated time-barcoding via micro-valves for robust and scalable analysis. 7. Microwell time-barcoded sequencing: captures and labels secretions for sequencing, enabling multidimensional analysis, though currently limited to a few time points and extended intervals. This review specifically addresses the challenges of achieving high-resolution timing measurements with short intervals while maintaining scalability for single-cell screening. Future advancements in microfluidic devices, integrating innovative barcoding technologies, advanced imaging technologies, artificial intelligence-powered decoding and analysis, and automations are anticipated to enable highly sensitive, scalable, high-throughput single-cell dynamic analysis. These developments hold great promise for deepening our understanding of biosystems by exploring single-cell timing responses on a larger scale.
Collapse
Affiliation(s)
- Ying Xu
- Department of Biomedical Engineering, City University of Hong Kong, 83 Tat Chee Avenue, Kowloon Tong, Hong Kong, China.
| | - Mei Tsz Jewel Chan
- Department of Biomedical Engineering, City University of Hong Kong, 83 Tat Chee Avenue, Kowloon Tong, Hong Kong, China.
| | - Ming Yang
- Department of Biomedical Engineering, City University of Hong Kong, 83 Tat Chee Avenue, Kowloon Tong, Hong Kong, China.
| | - Heixu Meng
- Department of Biomedical Engineering, City University of Hong Kong, 83 Tat Chee Avenue, Kowloon Tong, Hong Kong, China.
| | - Chia-Hung Chen
- Department of Biomedical Engineering, City University of Hong Kong, 83 Tat Chee Avenue, Kowloon Tong, Hong Kong, China.
| |
Collapse
|
9
|
Zhu X, Xu M, Portal C, Lin Y, Ferdinand A, Peng T, Morrisey EE, Dlugosz AA, Castellano JM, Lee V, Seykora JT, Wong SY, Iomini C, Millar SE. Identification of Meibomian gland stem cell populations and mechanisms of aging. Nat Commun 2025; 16:1663. [PMID: 39955307 PMCID: PMC11830078 DOI: 10.1038/s41467-025-56907-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2024] [Accepted: 02/05/2025] [Indexed: 02/17/2025] Open
Abstract
Meibomian glands secrete lipid-rich meibum, which prevents tear evaporation. Aging-related Meibomian gland shrinkage may result in part from stem cell exhaustion and is associated with evaporative dry eye disease, a common condition lacking effective treatment. The identities and niche of Meibomian gland stem cells and the signals controlling their activity are poorly defined. Using snRNA-seq, in vivo lineage tracing, ex vivo live imaging, and genetic studies in mice, we identify markers for stem cell populations that maintain distinct regions of the gland and uncover Hedgehog (Hh) signaling as a key regulator of stem cell proliferation. Consistent with this, we show that human Meibomian gland carcinoma exhibits increased Hh signaling. Aged glands display decreased Hh and EGF signaling, deficient innervation, and loss of collagen I in niche fibroblasts, indicating that alterations in both glandular epithelial cells and their surrounding microenvironment contribute to age-related degeneration. These findings suggest new approaches to treat aging-associated Meibomian gland loss.
Collapse
Affiliation(s)
- Xuming Zhu
- Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Institute for Regenerative Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Department of Cell, Developmental and Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Mingang Xu
- Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Institute for Regenerative Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Department of Cell, Developmental and Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Celine Portal
- Department of Ophthalmology, Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD, 21231, USA
- Department of Cell Biology, Johns Hopkins University School of Medicine, Baltimore, MD, 21231, USA
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, 17 rue Moreau, F-75012, Paris, France
| | - Yvonne Lin
- Department of Ophthalmology, Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD, 21231, USA
- Department of Cell Biology, Johns Hopkins University School of Medicine, Baltimore, MD, 21231, USA
- Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Alyssa Ferdinand
- Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Institute for Regenerative Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Department of Cell, Developmental and Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Tien Peng
- Department of Medicine, University of California San Francisco, San Francisco, CA, 94143, USA
| | - Edward E Morrisey
- Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Andrzej A Dlugosz
- Department of Dermatology, University of Michigan Medical School, Ann Arbor, MI, 48109, USA
- Rogel Cancer Center, University of Michigan Medical School, Ann Arbor, MI, 48109, USA
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI, 48109, USA
| | - Joseph M Castellano
- Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Institute for Regenerative Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Nash Family Department of Neuroscience, Department of Neurology, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Ronald M. Loeb Center for Alzheimer's Disease, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Vivian Lee
- Department of Ophthalmology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, 19104, USA
- Department of Dermatology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - John T Seykora
- Department of Dermatology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Sunny Y Wong
- Department of Dermatology, University of Michigan Medical School, Ann Arbor, MI, 48109, USA
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI, 48109, USA
| | - Carlo Iomini
- Department of Ophthalmology, Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD, 21231, USA.
- Department of Cell Biology, Johns Hopkins University School of Medicine, Baltimore, MD, 21231, USA.
| | - Sarah E Millar
- Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA.
- Institute for Regenerative Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA.
- Department of Cell, Developmental and Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA.
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA.
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA.
- Department of Dermatology, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA.
| |
Collapse
|
10
|
Yang S, Seo J, Choi J, Kim SH, Kuk Y, Park KC, Kang M, Byun S, Joo JY. Towards understanding cancer dormancy over strategic hitching up mechanisms to technologies. Mol Cancer 2025; 24:47. [PMID: 39953555 PMCID: PMC11829473 DOI: 10.1186/s12943-025-02250-9] [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: 12/26/2024] [Accepted: 01/28/2025] [Indexed: 02/17/2025] Open
Abstract
Delving into cancer dormancy has been an inherent task that may drive the lethal recurrence of cancer after primary tumor relief. Cells in quiescence can survive for a short or long term in silence, may undergo genetic or epigenetic changes, and can initiate relapse through certain contextual cues. The state of dormancy can be induced by multiple conditions including cancer drug treatment, in turn, undergoes a life cycle that generally occurs through dissemination, invasion, intravasation, circulation, immune evasion, extravasation, and colonization. Throughout this cascade, a cellular machinery governs the fate of individual cells, largely affected by gene regulation. Despite its significance, a precise view of cancer dormancy is yet hampered. Revolutionizing advanced single cell and long read sequencing through analysis methodologies and artificial intelligence, the most recent stage in the research tool progress, is expected to provide a holistic view of the diverse aspects of cancer dormancy.
Collapse
Affiliation(s)
- Sumin Yang
- Department of Pharmacy, College of Pharmacy, Hanyang University, Ansan, Gyeonggi-do, 15588, Korea
| | - Jieun Seo
- Genomic Medicine Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, 34141, Korea
- Department of Functional Genomics, University of Science and Technology, Daejeon, 34113, Korea
| | - Jeonghyeon Choi
- Department of Pharmacy, College of Pharmacy, Hanyang University, Ansan, Gyeonggi-do, 15588, Korea
| | - Sung-Hyun Kim
- Department of Pharmacy, College of Pharmacy, Hanyang University, Ansan, Gyeonggi-do, 15588, Korea
| | - Yunmin Kuk
- Genomic Medicine Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, 34141, Korea
- Department of Functional Genomics, University of Science and Technology, Daejeon, 34113, Korea
| | - Kyung Chan Park
- Genomic Medicine Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, 34141, Korea
- Department of Functional Genomics, University of Science and Technology, Daejeon, 34113, Korea
| | - Mingon Kang
- Department of Computer Science, University of Nevada, Las Vegas, NV, 89154, USA
| | - Sangwon Byun
- Genomic Medicine Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, 34141, Korea.
- Department of Functional Genomics, University of Science and Technology, Daejeon, 34113, Korea.
| | - Jae-Yeol Joo
- Department of Pharmacy, College of Pharmacy, Hanyang University, Ansan, Gyeonggi-do, 15588, Korea.
- Department of Pharmacy, College of Pharmacy, Hanyang University, Rm 407, Bldg.42, 55 Hanyangdaehak-ro, Sangnok-gu Ansan, Gyeonggi-do, 15588, Republic of Korea.
| |
Collapse
|
11
|
Muñoz-Blat I, Pérez-Moraga R, Castillo-Marco N, Cordero T, Ochando A, Ortega-Sanchís S, Parras-Moltó M, Monfort-Ortiz R, Satorres-Perez E, Novillo B, Perales A, Gormley M, Granados-Aparici S, Noguera R, Roson B, Fisher SJ, Simón C, Garrido-Gómez T. Multi-omics-based mapping of decidualization resistance in patients with a history of severe preeclampsia. Nat Med 2025; 31:502-513. [PMID: 39775038 PMCID: PMC11835751 DOI: 10.1038/s41591-024-03407-7] [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: 04/26/2024] [Accepted: 11/08/2024] [Indexed: 01/11/2025]
Abstract
Endometrial decidualization resistance (DR) is implicated in various gynecological and obstetric conditions. Here, using a multi-omic strategy, we unraveled the cellular and molecular characteristics of DR in patients who have suffered severe preeclampsia (sPE). Morphological analysis unveiled significant glandular anatomical abnormalities, confirmed histologically and quantified by the digitization of hematoxylin and eosin-stained tissue sections. Single-cell RNA sequencing (scRNA-seq) of endometrial samples from patients with sPE (n = 11) and controls (n = 12) revealed sPE-associated shifts in cell composition, manifesting as a stromal mosaic state characterized by proliferative stromal cells (MMP11 and SFRP4) alongside IGFBP1+ decidualized cells, with concurrent epithelial mosaicism and a dearth of epithelial-stromal transition associated with decidualization. Cell-cell communication network mapping underscored aberrant crosstalk among specific cell types, implicating crucial pathways such as endoglin, WNT and SPP1. Spatial transcriptomics in a replication cohort validated DR-associated features. Laser capture microdissection/mass spectrometry in a second replication cohort corroborated several scRNA-seq findings, notably the absence of stromal to epithelial transition at a pathway level, indicating a disrupted response to steroid hormones, particularly estrogens. These insights shed light on potential molecular mechanisms underpinning DR pathogenesis in the context of sPE.
Collapse
Affiliation(s)
- Irene Muñoz-Blat
- Carlos Simon Foundation, Valencia, Spain
- INCLIVA Health Research Institute, Valencia, Spain
| | | | | | | | | | | | | | - Rogelio Monfort-Ortiz
- Department of Obstetrics and Gynecology, Hospital Universitario y Politécnico La Fe, Valencia, Spain
| | - Elena Satorres-Perez
- Department of Obstetrics and Gynecology, Hospital Universitario y Politécnico La Fe, Valencia, Spain
| | - Blanca Novillo
- Department of Obstetrics and Gynecology, Hospital Universitario y Politécnico La Fe, Valencia, Spain
| | - Alfredo Perales
- Department of Obstetrics and Gynecology, Hospital Universitario y Politécnico La Fe, Valencia, Spain
| | - Matthew Gormley
- Department of Obstetrics, Gynecology and Reproductive Sciences, Center of Reproductive Science, University of California San Francisco, San Francisco, CA, USA
| | - Sofia Granados-Aparici
- INCLIVA Health Research Institute, Valencia, Spain
- Department of Pathology, Medical School, University of Valencia, Valencia, Spain
- Centro de Investigación Biomédica en Red de Cáncer, Instituto de Salud Carlos III, Madrid, Spain
| | - Rosa Noguera
- INCLIVA Health Research Institute, Valencia, Spain
- Department of Pathology, Medical School, University of Valencia, Valencia, Spain
- Centro de Investigación Biomédica en Red de Cáncer, Instituto de Salud Carlos III, Madrid, Spain
| | | | - Susan J Fisher
- Department of Obstetrics, Gynecology and Reproductive Sciences, Center of Reproductive Science, University of California San Francisco, San Francisco, CA, USA
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California San Francisco, San Francisco, CA, USA
- Sandler-Moore Mass Spectrometry Core Facility, University of California San Francisco, San Francisco, CA, USA
| | - Carlos Simón
- Carlos Simon Foundation, Valencia, Spain.
- INCLIVA Health Research Institute, Valencia, Spain.
- Department of Pediatrics, Obstetrics and Gynecology, University of Valencia, Valencia, Spain.
- Department of Obstetrics and Gynecology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA.
| | - Tamara Garrido-Gómez
- Carlos Simon Foundation, Valencia, Spain.
- INCLIVA Health Research Institute, Valencia, Spain.
| |
Collapse
|
12
|
Chen Y, Zhang Y, Gan J, Ni K, Chen M, Bahar I, Xing J. GraphVelo allows for accurate inference of multimodal omics velocities and molecular mechanisms for single cells. RESEARCH SQUARE 2025:rs.3.rs-5613372. [PMID: 39877092 PMCID: PMC11774466 DOI: 10.21203/rs.3.rs-5613372/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/31/2025]
Abstract
RNA velocities and generalizations emerge as powerful approaches for extracting time-resolved information from high-throughput snapshot single-cell data. Yet, several inherent limitations restrict applying the approaches to genes not suitable for RNA velocity inference due to complex transcriptional dynamics, low expression, or lacking splicing dynamics, or data of non-transcriptomic modality. Here, we present GraphVelo, a graph-based machine learning procedure that uses as input the RNA velocities inferred from existing methods and infers velocity vectors lying in the tangent space of the low-dimensional manifold formed by the single cell data. GraphVelo preserves vector magnitude and direction information during transformations across different data representations. Tests on multiple synthetic and experimental scRNA-seq data including viral-host interactome and multi-omics datasets demonstrate that GraphVelo, together with downstream generalized dynamo analyses, extends RNA velocities to multi-modal data and reveals quantitative nonlinear regulation relations between genes, virus and host cells, and different layers of gene regulation.
Collapse
Affiliation(s)
- Yuhao Chen
- Department of Bioinformatics, College of Life Sciences, Zhejiang University, Hangzhou, 310058, China
- Department of Computational and Systems Biology, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Yan Zhang
- Department of Computational and Systems Biology, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Jiaqi Gan
- Department of Computational and Systems Biology, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Ke Ni
- Department of Computational and Systems Biology, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Ming Chen
- Department of Bioinformatics, College of Life Sciences, Zhejiang University, Hangzhou, 310058, China
- Zhejiang Key Laboratory of Multi-omics Precision Diagnosis and Treatment of Liver Diseases, Department of General Surgery, Sir Run-Run Shaw Hospital, Zhejiang University School of Medicine, 310016, Hangzhou, China
| | - Ivet Bahar
- Laufer Center for Physical and Quantitative Biology, and Department of Biochemistry and Cell Biology, Renaissance School of Medicine, Stony Brook University, Stony Brook, NY, USA
| | - Jianhua Xing
- Department of Computational and Systems Biology, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
- University of Pittsburgh Hillman Cancer Center, Pittsburgh, PA, USA
- Department of Physics and Astronomy, University of Pittsburgh, Pittsburgh, PA, USA
| |
Collapse
|
13
|
Chen Y, Zhang Y, Gan J, Ni K, Chen M, Bahar I, Xing J. GraphVelo allows for accurate inference of multimodal velocities and molecular mechanisms for single cells. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.12.03.626638. [PMID: 39677753 PMCID: PMC11642879 DOI: 10.1101/2024.12.03.626638] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2024]
Abstract
RNA velocities and generalizations emerge as powerful approaches for extracting time-resolved information from high-throughput snapshot single-cell data. Yet, several inherent limitations restrict applying the approaches to genes not suitable for RNA velocity inference due to complex transcriptional dynamics, low expression, or lacking splicing dynamics, or data of non-transcriptomic modality. Here, we present GraphVelo, a graph-based machine learning procedure that uses as input the RNA velocities inferred from existing methods and infers velocity vectors lying in the tangent space of the low-dimensional manifold formed by the single cell data. GraphVelo preserves vector magnitude and direction information during transformations across different data representations. Tests on multiple synthetic and experimental scRNA-seq data including viral-host interactome and multi-omics datasets demonstrate that GraphVelo, together with downstream generalized dynamo analyses, extends RNA velocities to multi-modal data and reveals quantitative nonlinear regulation relations between genes, virus and host cells, and different layers of gene regulation.
Collapse
Affiliation(s)
- Yuhao Chen
- Department of Bioinformatics, College of Life Sciences, Zhejiang University, Hangzhou, 310058, China
- Department of Computational and Systems Biology, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Yan Zhang
- Department of Computational and Systems Biology, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Jiaqi Gan
- Department of Computational and Systems Biology, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Ke Ni
- Department of Computational and Systems Biology, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Ming Chen
- Department of Bioinformatics, College of Life Sciences, Zhejiang University, Hangzhou, 310058, China
- Zhejiang Key Laboratory of Multi-omics Precision Diagnosis and Treatment of Liver Diseases, Department of General Surgery, Sir Run-Run Shaw Hospital, Zhejiang University School of Medicine, 310016, Hangzhou, China
| | - Ivet Bahar
- Laufer Center for Physical and Quantitative Biology, and Department of Biochemistry and Cell Biology, Renaissance School of Medicine, Stony Brook University, Stony Brook, NY, USA
| | - Jianhua Xing
- Department of Computational and Systems Biology, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
- University of Pittsburgh Hillman Cancer Center, Pittsburgh, PA, USA
- Department of Physics and Astronomy, University of Pittsburgh, Pittsburgh, PA, USA
| |
Collapse
|
14
|
Gong J, Lee C, Kim H, Kim J, Jeon J, Park S, Cho K. Control of Cellular Differentiation Trajectories for Cancer Reversion. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2402132. [PMID: 39661721 PMCID: PMC11744559 DOI: 10.1002/advs.202402132] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Revised: 11/08/2024] [Indexed: 12/13/2024]
Abstract
Cellular differentiation is controlled by intricate layers of gene regulation, involving the modulation of gene expression by various transcriptional regulators. Due to the complexity of gene regulation, identifying master regulators across the differentiation trajectory has been a longstanding challenge. To tackle this problem, a computational framework, single-cell Boolean network inference and control (BENEIN), is presented. Applying BENEIN to human large intestinal single-cell transcriptome data, MYB, HDAC2, and FOXA2 are identified as the master regulators whose inhibition induces enterocyte differentiation. It is found that simultaneous knockdown of these master regulators can revert colorectal cancer cells into normal-like enterocytes by synergistically inducing differentiation and suppressing malignancy, which is validated by in vitro and in vivo experiments.
Collapse
Affiliation(s)
- Jeong‐Ryeol Gong
- Department of Bio and Brain EngineeringKorea Advanced Institute of Science and TechnologyDaejeon34141Republic of Korea
| | - Chun‐Kyung Lee
- Department of Bio and Brain EngineeringKorea Advanced Institute of Science and TechnologyDaejeon34141Republic of Korea
| | - Hoon‐Min Kim
- Department of Bio and Brain EngineeringKorea Advanced Institute of Science and TechnologyDaejeon34141Republic of Korea
| | - Juhee Kim
- Department of Bio and Brain EngineeringKorea Advanced Institute of Science and TechnologyDaejeon34141Republic of Korea
| | - Jaeog Jeon
- Department of Bio and Brain EngineeringKorea Advanced Institute of Science and TechnologyDaejeon34141Republic of Korea
| | - Sunmin Park
- Department of Bio and Brain EngineeringKorea Advanced Institute of Science and TechnologyDaejeon34141Republic of Korea
| | - Kwang‐Hyun Cho
- Department of Bio and Brain EngineeringKorea Advanced Institute of Science and TechnologyDaejeon34141Republic of Korea
| |
Collapse
|
15
|
Lu L, Ono N, Welch JD. Linking transcriptome and morphology in bone cells at cellular resolution with generative AI. J Bone Miner Res 2024; 40:20-26. [PMID: 39303095 DOI: 10.1093/jbmr/zjae151] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Revised: 08/26/2024] [Accepted: 09/17/2024] [Indexed: 09/22/2024]
Abstract
Recent advancements in deep learning (DL) have revolutionized the capability of artificial intelligence (AI) by enabling the analysis of large-scale, complex datasets that are difficult for humans to interpret. However, large amounts of high-quality data are required to train such generative AI models successfully. With the rapid commercialization of single-cell sequencing and spatial transcriptomics platforms, the field is increasingly producing large-scale datasets such as histological images, single-cell molecular data, and spatial transcriptomic data. These molecular and morphological datasets parallel the multimodal text and image data used to train highly successful generative AI models for natural language processing and computer vision. Thus, these emerging data types offer great potential to train generative AI models that uncover intricate biological processes of bone cells at a cellular level. In this Perspective, we summarize the progress and prospects of generative AI applied to these datasets and their potential applications to bone research. In particular, we highlight three AI applications: predicting cell differentiation dynamics, linking molecular and morphological features, and predicting cellular responses to perturbations. To make generative AI models beneficial for bone research, important issues, such as technical biases in bone single-cell datasets, lack of profiling of important bone cell types, and lack of spatial information, needs to be addressed. Realizing the potential of generative AI for bone biology will also likely require generating large-scale, high-quality cellular-resolution spatial transcriptomics datasets, improving the sensitivity of current spatial transcriptomics datasets, and thorough experimental validation of model predictions.
Collapse
Affiliation(s)
- Lu Lu
- Department of Computational Medicine and Bioinformatics, University of Michigan, 100 Washtenaw Ave. Ann Arbor, MI 48109, United States
| | - Noriaki Ono
- Department of Diagnostic and Biomedical Sciences, University of Texas Health Science Center at Houston School of Dentistry, 1941 East Road, Houston, TX 77054, United States
| | - Joshua D Welch
- Department of Computational Medicine and Bioinformatics, University of Michigan, 100 Washtenaw Ave. Ann Arbor, MI 48109, United States
- Department of Electrical Engineering and Computer Science, University of Michigan, 1301 Beal Ave. Ann Arbor, MI 48109, United States
| |
Collapse
|
16
|
Liao X, Kang L, Peng Y, Chai X, Xie P, Lin C, Ji H, Jiao Y, Liu J. Multivariate stochastic modeling for transcriptional dynamics with cell-specific latent time using SDEvelo. Nat Commun 2024; 15:10849. [PMID: 39738101 DOI: 10.1038/s41467-024-55146-5] [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] [Accepted: 11/28/2024] [Indexed: 01/01/2025] Open
Abstract
Recently, RNA velocity has driven a paradigmatic change in single-cell RNA sequencing (scRNA-seq) studies, allowing the reconstruction and prediction of directed trajectories in cell differentiation and state transitions. Most existing methods of dynamic modeling use ordinary differential equations (ODE) for individual genes without applying multivariate approaches. However, this modeling strategy inadequately captures the intrinsically stochastic nature of transcriptional dynamics governed by a cell-specific latent time across multiple genes, potentially leading to erroneous results. Here, we present SDEvelo, a generative approach to inferring RNA velocity by modeling the dynamics of unspliced and spliced RNAs via multivariate stochastic differential equations (SDE). Uniquely, SDEvelo explicitly models inherent uncertainty in transcriptional dynamics while estimating a cell-specific latent time across genes. Using both simulated and four scRNA-seq and spatial transcriptomics datasets, we show that SDEvelo can model the random dynamic patterns of mature-state cells while accurately detecting carcinogenesis. Additionally, the estimated gene-shared latent time can facilitate many downstream analyses for biological discovery. We demonstrate that SDEvelo is computationally scalable and applicable to both scRNA-seq and sequencing-based spatial transcriptomics data.
Collapse
Affiliation(s)
- Xu Liao
- School of Data Science, The Chinese University of Hong Kong-Shenzhen, Shenzhen, China
- Centre for Quantitative Medicine, Duke-NUS Medical School, Singapore, Singapore
| | - Lican Kang
- Institute for Math and AI, Wuhan University, Wuhan, China
- School of Mathematics and Statistics, Wuhan University, Wuhan, China
| | - Yihao Peng
- School of Data Science, The Chinese University of Hong Kong-Shenzhen, Shenzhen, China
| | - Xiaoran Chai
- Cancer and Stem Cell Biology Program, Duke-NUS Medical School, Singapore, Singapore
| | - Peng Xie
- School of Biological Science & Medical Engineering, Southeast University, Nanjing, China
| | - Chengqi Lin
- Key Laboratory of Developmental Genes and Human Disease, School of Life Science and Technology, Southeast University, Nanjing, China
| | - Hongkai Ji
- Department of Biostatistics, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - Yuling Jiao
- School of Artificial Intelligence, Wuhan University, Wuhan, China.
- Hubei Key Laboratory of Computational Science, Wuhan University, Wuhan, China.
| | - Jin Liu
- School of Data Science, The Chinese University of Hong Kong-Shenzhen, Shenzhen, China.
| |
Collapse
|
17
|
Wang Y, Dede M, Mohanty V, Dou J, Li Z, Chen K. A statistical approach for systematic identification of transition cells from scRNA-seq data. CELL REPORTS METHODS 2024; 4:100913. [PMID: 39644902 PMCID: PMC11704623 DOI: 10.1016/j.crmeth.2024.100913] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Revised: 09/01/2024] [Accepted: 11/13/2024] [Indexed: 12/09/2024]
Abstract
Decoding cellular state transitions is crucial for understanding complex biological processes in development and disease. While recent advancements in single-cell RNA sequencing (scRNA-seq) offer insights into cellular trajectories, existing tools primarily study expressional rather than regulatory state shifts. We present CellTran, a statistical approach utilizing paired-gene expression correlations to detect transition cells from scRNA-seq data without explicitly resolving gene regulatory networks. Applying our approach to various contexts, including tissue regeneration, embryonic development, preinvasive lesions, and humoral responses post-vaccination, reveals transition cells and their distinct gene expression profiles. Our study sheds light on the underlying molecular mechanisms driving cellular state transitions, enhancing our ability to identify therapeutic targets for disease interventions.
Collapse
Affiliation(s)
- Yuanxin Wang
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Merve Dede
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Vakul Mohanty
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Jinzhuang Dou
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Ziyi Li
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Ken Chen
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| |
Collapse
|
18
|
Yin H, Duo H, Li S, Qin D, Xie L, Xiao Y, Sun J, Tao J, Zhang X, Li Y, Zou Y, Yang Q, Yang X, Hao Y, Li B. Unlocking biological insights from differentially expressed genes: Concepts, methods, and future perspectives. J Adv Res 2024:S2090-1232(24)00560-5. [PMID: 39647635 DOI: 10.1016/j.jare.2024.12.004] [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: 07/28/2024] [Revised: 10/12/2024] [Accepted: 12/03/2024] [Indexed: 12/10/2024] Open
Abstract
BACKGROUND Identifying differentially expressed genes (DEGs) is a core task of transcriptome analysis, as DEGs can reveal the molecular mechanisms underlying biological processes. However, interpreting the biological significance of large DEG lists is challenging. Currently, gene ontology, pathway enrichment and protein-protein interaction analysis are common strategies employed by biologists. Additionally, emerging analytical strategies/approaches (such as network module analysis, knowledge graph, drug repurposing, cell marker discovery, trajectory analysis, and cell communication analysis) have been proposed. Despite these advances, comprehensive guidelines for systematically and thoroughly mining the biological information within DEGs remain lacking. AIM OF REVIEW This review aims to provide an overview of essential concepts and methodologies for the biological interpretation of DEGs, enhancing the contextual understanding. It also addresses the current limitations and future perspectives of these approaches, highlighting their broad applications in deciphering the molecular mechanism of complex diseases and phenotypes. To assist users in extracting insights from extensive datasets, especially various DEG lists, we developed DEGMiner (https://www.ciblab.net/DEGMiner/), which integrates over 300 easily accessible databases and tools. KEY SCIENTIFIC CONCEPTS OF REVIEW This review offers strong support and guidance for exploring DEGs, and also will accelerate the discovery of hidden biological insights within genomes.
Collapse
Affiliation(s)
- Huachun Yin
- College of Life Sciences, Chongqing Normal University, Chongqing 401331, PR China; Department of Neurosurgery, Xinqiao Hospital, The Army Medical University, Chongqing 400037, PR China; Department of Neurobiology, Chongqing Key Laboratory of Neurobiology, The Army Medical University, Chongqing 400038, PR China
| | - Hongrui Duo
- College of Life Sciences, Chongqing Normal University, Chongqing 401331, PR China
| | - Song Li
- Department of Neurosurgery, Xinqiao Hospital, The Army Medical University, Chongqing 400037, PR China
| | - Dan Qin
- Department of Biology, College of Science, Northeastern University, Boston, MA 02115, USA
| | - Lingling Xie
- College of Life Sciences, Chongqing Normal University, Chongqing 401331, PR China
| | - Yingxue Xiao
- College of Life Sciences, Chongqing Normal University, Chongqing 401331, PR China
| | - Jing Sun
- College of Life Sciences, Chongqing Normal University, Chongqing 401331, PR China
| | - Jingxin Tao
- College of Life Sciences, Chongqing Normal University, Chongqing 401331, PR China
| | - Xiaoxi Zhang
- College of Life Sciences, Chongqing Normal University, Chongqing 401331, PR China
| | - Yinghong Li
- Chongqing Key Laboratory of Big Data for Bio Intelligence, Chongqing University of Posts and Telecommunications, Chongqing 400065, PR China
| | - Yue Zou
- College of Life Sciences, Chongqing Normal University, Chongqing 401331, PR China
| | - Qingxia Yang
- Zhejiang Provincial Key Laboratory of Precision Diagnosis and Therapy for Major Gynecological Diseases, Women's Hospital, Zhejiang University School of Medicine, Hangzhou 310058, PR China
| | - Xian Yang
- College of Life Sciences, Chongqing Normal University, Chongqing 401331, PR China
| | - Youjin Hao
- College of Life Sciences, Chongqing Normal University, Chongqing 401331, PR China.
| | - Bo Li
- College of Life Sciences, Chongqing Normal University, Chongqing 401331, PR China.
| |
Collapse
|
19
|
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; 21:2271-2286. [PMID: 39482463 PMCID: PMC11621032 DOI: 10.1038/s41592-024-02471-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [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
|
20
|
Zhang P, Wang G. Uncovering cell cycle speed modulations with statistical inference. Nat Methods 2024; 21:2231-2232. [PMID: 39482462 DOI: 10.1038/s41592-024-02484-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2024]
Affiliation(s)
- 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
| | - 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
|
21
|
Sun R, Cao W, Li S, Jiang J, Shi Y, Zhang B. scGRN-Entropy: Inferring cell differentiation trajectories using single-cell data and gene regulation network-based transfer entropy. PLoS Comput Biol 2024; 20:e1012638. [PMID: 39585902 PMCID: PMC11627384 DOI: 10.1371/journal.pcbi.1012638] [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: 07/08/2024] [Revised: 12/09/2024] [Accepted: 11/12/2024] [Indexed: 11/27/2024] Open
Abstract
Research on cell differentiation facilitates a deeper understanding of the fundamental processes of life, elucidates the intrinsic mechanisms underlying diseases such as cancer, and advances the development of therapeutics and precision medicine. Existing methods for inferring cell differentiation trajectories from single-cell RNA sequencing (scRNA-seq) data primarily rely on static gene expression data to measure distances between cells and subsequently infer pseudotime trajectories. In this work, we introduce a novel method, scGRN-Entropy, for inferring cell differentiation trajectories and pseudotime from scRNA-seq data. Unlike existing approaches, scGRN-Entropy improves inference accuracy by incorporating dynamic changes in gene regulatory networks (GRN). In scGRN-Entropy, an undirected graph representing state transitions between cells is constructed by integrating both static relationships in gene expression space and dynamic relationships in the GRN space. The edges of the undirected graph are then refined using pseudotime inferred based on cell entropy in the GRN space. Finally, the Minimum Spanning Tree (MST) algorithm is applied to derive the cell differentiation trajectory. We validate the accuracy of scGRN-Entropy on eight different real scRNA-seq datasets, demonstrating its superior performance in inferring cell differentiation trajectories through comparative analysis with existing state-of-the-art methods.
Collapse
Affiliation(s)
- Rui Sun
- School of Mathematical & Physical Sciences, Wuhan Textile University, Wuhan, Hubei, China
- Center for Applied Mathematics and Interdisciplinary Studies, Wuhan Textile University, Wuhan, Hubei, China
| | - Wenjie Cao
- School of Mathematics, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - ShengXuan Li
- School of Mathematical & Physical Sciences, Wuhan Textile University, Wuhan, Hubei, China
- Center for Applied Mathematics and Interdisciplinary Studies, Wuhan Textile University, Wuhan, Hubei, China
| | - Jian Jiang
- School of Mathematical & Physical Sciences, Wuhan Textile University, Wuhan, Hubei, China
- Center for Applied Mathematics and Interdisciplinary Studies, Wuhan Textile University, Wuhan, Hubei, China
| | - Yazhou Shi
- School of Mathematical & Physical Sciences, Wuhan Textile University, Wuhan, Hubei, China
- Center for Applied Mathematics and Interdisciplinary Studies, Wuhan Textile University, Wuhan, Hubei, China
| | - Bengong Zhang
- School of Mathematical & Physical Sciences, Wuhan Textile University, Wuhan, Hubei, China
- Center for Applied Mathematics and Interdisciplinary Studies, Wuhan Textile University, Wuhan, Hubei, China
| |
Collapse
|
22
|
Mizukoshi C, Kojima Y, Nomura S, Hayashi S, Abe K, Shimamura T. DeepKINET: a deep generative model for estimating single-cell RNA splicing and degradation rates. Genome Biol 2024; 25:229. [PMID: 39237934 PMCID: PMC11378460 DOI: 10.1186/s13059-024-03367-8] [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: 12/14/2023] [Accepted: 08/04/2024] [Indexed: 09/07/2024] Open
Abstract
Messenger RNA splicing and degradation are critical for gene expression regulation, the abnormality of which leads to diseases. Previous methods for estimating kinetic rates have limitations, assuming uniform rates across cells. DeepKINET is a deep generative model that estimates splicing and degradation rates at single-cell resolution from scRNA-seq data. DeepKINET outperforms existing methods on simulated and metabolic labeling datasets. Applied to forebrain and breast cancer data, it identifies RNA-binding proteins responsible for kinetic rate diversity. DeepKINET also analyzes the effects of splicing factor mutations on target genes in erythroid lineage cells. DeepKINET effectively reveals cellular heterogeneity in post-transcriptional regulation.
Collapse
Affiliation(s)
- Chikara Mizukoshi
- Division of Systems Biology, Graduate School of Medicine, Nagoya University, Aichi, Japan.
- Nagoya University Hospital, Aichi, Japan.
| | - Yasuhiro Kojima
- Laboratory of Computational Life Science, National Cancer Center Research Institute, Tokyo, Japan.
- Department of Computational and Systems Biology, Medical Research Institute, Tokyo Medical and Dental University, Tokyo, Japan.
| | - Satoshi Nomura
- Division of Systems Biology, Graduate School of Medicine, Nagoya University, Aichi, Japan
| | - Shuto Hayashi
- Department of Computational and Systems Biology, Medical Research Institute, Tokyo Medical and Dental University, Tokyo, Japan
| | - Ko Abe
- Department of Computational and Systems Biology, Medical Research Institute, Tokyo Medical and Dental University, Tokyo, Japan
| | - Teppei Shimamura
- Division of Systems Biology, Graduate School of Medicine, Nagoya University, Aichi, Japan.
- Department of Computational and Systems Biology, Medical Research Institute, Tokyo Medical and Dental University, Tokyo, Japan.
| |
Collapse
|
23
|
Zhu X, Xu M, Portal C, Lin Y, Ferdinand A, Peng T, Morrisey EE, Dlugosz AA, Castellano JM, Lee V, Seykora JT, Iomini C, Millar SE. Identification of Meibomian gland stem cell populations and mechanisms of aging. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.09.607015. [PMID: 39149265 PMCID: PMC11326261 DOI: 10.1101/2024.08.09.607015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 08/17/2024]
Abstract
Meibomian glands secrete lipid-rich meibum, which prevents tear evaporation. Aging-related Meibomian gland shrinkage may result in part from stem cell exhaustion and is associated with evaporative dry eye disease, a common condition lacking effective treatment. The identities and niche of Meibomian gland stem cells and the signals controlling their activity are poorly defined. Using snRNA-seq, in vivo lineage tracing, ex vivo live imaging, and genetic studies in mice, we identified markers for stem cell populations that maintain distinct regions of the gland and uncovered Hh signaling as a key regulator of stem cell proliferation. Consistent with this, human Meibomian gland carcinoma exhibited increased Hh signaling. Aged glands displayed decreased Hh and EGF signaling, deficient innervation, and loss of collagen I in niche fibroblasts, indicating that alterations in both glandular epithelial cells and their surrounding microenvironment contribute to age-related degeneration. These findings suggest new approaches to treat aging-associated Meibomian gland loss.
Collapse
Affiliation(s)
- Xuming Zhu
- Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Institute for Regenerative Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Department of Cell, Developmental and Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Mingang Xu
- Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Institute for Regenerative Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Department of Cell, Developmental and Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Celine Portal
- Department of Ophthalmology, Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD, 21231, USA
- Department of Cell Biology, Johns Hopkins University School of Medicine, Baltimore, MD, 21231, USA
| | - Yvonne Lin
- Department of Ophthalmology, Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD, 21231, USA
- Department of Cell Biology, Johns Hopkins University School of Medicine, Baltimore, MD, 21231, USA
| | - Alyssa Ferdinand
- Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Institute for Regenerative Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Department of Cell, Developmental and Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Tien Peng
- Department of Medicine, University of California San Francisco, San Francisco, CA 94143, USA
| | - Edward E. Morrisey
- Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Andrzej A. Dlugosz
- Department of Dermatology and the Rogel Cancer Center, University of Michigan Medical School, Ann Arbor, MI 48109, USA
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Joseph M. Castellano
- Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Institute for Regenerative Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Nash Family Department of Neuroscience, Department of Neurology, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Ronald M. Loeb Center for Alzheimer’s Disease, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Vivian Lee
- Department of Ophthalmology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
- Department of Dermatology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - John T. Seykora
- Department of Dermatology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Carlo Iomini
- Department of Ophthalmology, Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD, 21231, USA
- Department of Cell Biology, Johns Hopkins University School of Medicine, Baltimore, MD, 21231, USA
| | - Sarah E Millar
- Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Institute for Regenerative Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Department of Cell, Developmental and Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| |
Collapse
|
24
|
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
|
25
|
He D, Gao Y, Chan SS, Quintana-Parrilla N, Patro R. Forseti: a mechanistic and predictive model of the splicing status of scRNA-seq reads. Bioinformatics 2024; 40:i297-i306. [PMID: 38940130 PMCID: PMC11256924 DOI: 10.1093/bioinformatics/btae207] [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] [Indexed: 06/29/2024] Open
Abstract
MOTIVATION Short-read single-cell RNA-sequencing (scRNA-seq) has been used to study cellular heterogeneity, cellular fate, and transcriptional dynamics. Modeling splicing dynamics in scRNA-seq data is challenging, with inherent difficulty in even the seemingly straightforward task of elucidating the splicing status of the molecules from which sequenced fragments are drawn. This difficulty arises, in part, from the limited read length and positional biases, which substantially reduce the specificity of the sequenced fragments. As a result, the splicing status of many reads in scRNA-seq is ambiguous because of a lack of definitive evidence. We are therefore in need of methods that can recover the splicing status of ambiguous reads which, in turn, can lead to more accuracy and confidence in downstream analyses. RESULTS We develop Forseti, a predictive model to probabilistically assign a splicing status to scRNA-seq reads. Our model has two key components. First, we train a binding affinity model to assign a probability that a given transcriptomic site is used in fragment generation. Second, we fit a robust fragment length distribution model that generalizes well across datasets deriving from different species and tissue types. Forseti combines these two trained models to predict the splicing status of the molecule of origin of reads by scoring putative fragments that associate each alignment of sequenced reads with proximate potential priming sites. Using both simulated and experimental data, we show that our model can precisely predict the splicing status of many reads and identify the true gene origin of multi-gene mapped reads. AVAILABILITY AND IMPLEMENTATION Forseti and the code used for producing the results are available at https://github.com/COMBINE-lab/forseti under a BSD 3-clause license.
Collapse
Affiliation(s)
- Dongze He
- Center for Bioinformatics and Computational Biology, University of Maryland, College Park, MD 20742, United States
- Program in Computational Biology, Bioinformatics and Genomices, University of Maryland, College Park, MD 20742, United States
| | - Yuan Gao
- Center for Bioinformatics and Computational Biology, University of Maryland, College Park, MD 20742, United States
- Program in Computational Biology, Bioinformatics and Genomices, University of Maryland, College Park, MD 20742, United States
| | - Spencer Skylar Chan
- Department of Computer Science, University of Maryland, College Park, MD 20742, United States
| | | | - Rob Patro
- Center for Bioinformatics and Computational Biology, University of Maryland, College Park, MD 20742, United States
- Department of Computer Science, University of Maryland, College Park, MD 20742, United States
| |
Collapse
|
26
|
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
|
27
|
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
|
28
|
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: 15] [Impact Index Per Article: 15.0] [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
|
29
|
Angelozzi M, Karvande A, Lefebvre V. SOXC are critical regulators of adult bone mass. Nat Commun 2024; 15:2956. [PMID: 38580651 PMCID: PMC10997656 DOI: 10.1038/s41467-024-47413-2] [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: 09/08/2023] [Accepted: 03/28/2024] [Indexed: 04/07/2024] Open
Abstract
Pivotal in many ways for human health, the control of adult bone mass is governed by complex, incompletely understood crosstalk namely between mesenchymal stem cells, osteoblasts and osteoclasts. The SOX4, SOX11 and SOX12 (SOXC) transcription factors were previously shown to control many developmental processes, including skeletogenesis, and SOX4 was linked to osteoporosis, but how SOXC control adult bone mass remains unknown. Using SOXC loss- and gain-of-function mouse models, we show here that SOXC redundantly promote prepubertal cortical bone mass strengthening whereas only SOX4 mitigates adult trabecular bone mass accrual in early adulthood and subsequent maintenance. SOX4 favors bone resorption over formation by lowering osteoblastogenesis and increasing osteoclastogenesis. Single-cell transcriptomics reveals its prevalent expression in Lepr+ mesenchymal cells and ability to upregulate genes for prominent anti-osteoblastogenic and pro-osteoclastogenic factors, including interferon signaling-related chemokines, contributing to these adult stem cells' secretome. SOXC, with SOX4 predominantly, are thus key regulators of adult bone mass.
Collapse
Affiliation(s)
- Marco Angelozzi
- Department of Surgery, Division of Orthopaedics, Children's Hospital of Philadelphia, Philadelphia, PA, USA.
| | - Anirudha Karvande
- Department of Surgery, Division of Orthopaedics, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Véronique Lefebvre
- Department of Surgery, Division of Orthopaedics, Children's Hospital of Philadelphia, Philadelphia, PA, USA.
| |
Collapse
|
30
|
He D, Gao Y, Chan SS, Quintana-Parrilla N, Patro R. Forseti: A mechanistic and predictive model of the splicing status of scRNA-seq reads. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.02.01.577813. [PMID: 38370848 PMCID: PMC10871212 DOI: 10.1101/2024.02.01.577813] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/20/2024]
Abstract
Motivation Short-read single-cell RNA-sequencing (scRNA-seq) has been used to study cellular heterogeneity, cellular fate, and transcriptional dynamics. Modeling splicing dynamics in scRNA-seq data is challenging, with inherent difficulty in even the seemingly straightforward task of elucidating the splicing status of the molecules from which sequenced fragments are drawn. This difficulty arises, in part, from the limited read length and positional biases, which substantially reduce the specificity of the sequenced fragments. As a result, the splicing status of many reads in scRNA-seq is ambiguous because of a lack of definitive evidence. We are therefore in need of methods that can recover the splicing status of ambiguous reads which, in turn, can lead to more accuracy and confidence in downstream analyses. Results We develop Forseti, a predictive model to probabilistically assign a splicing status to scRNA-seq reads. Our model has two key components. First, we train a binding affinity model to assign a probability that a given transcriptomic site is used in fragment generation. Second, we fit a robust fragment length distribution model that generalizes well across datasets deriving from different species and tissue types. Forseti combines these two trained models to predict the splicing status of the molecule of origin of reads by scoring putative fragments that associate each alignment of sequenced reads with proximate potential priming sites. Using both simulated and experimental data, we show that our model can precisely predict the splicing status of reads and identify the true gene origin of multi-gene mapped reads. Availability Forseti and the code used for producing the results are available at https://github.com/COMBINE-lab/forseti under a BSD 3-clause license.
Collapse
Affiliation(s)
- Dongze He
- Center for Bioinformatics and Computational Biology, University of Maryland, College Park, MD 20742, USA
- Program in Computational Biology, Bioinformatics and Genomices, University of Maryland, College Park, MD 20742, USA
| | - Yuan Gao
- Center for Bioinformatics and Computational Biology, University of Maryland, College Park, MD 20742, USA
- Program in Computational Biology, Bioinformatics and Genomices, University of Maryland, College Park, MD 20742, USA
| | - Spencer Skylar Chan
- Department of Computer Science, University of Maryland, College Park, MD 20742, USA
| | | | - Rob Patro
- Center for Bioinformatics and Computational Biology, University of Maryland, College Park, MD 20742, USA
- Department of Computer Science, University of Maryland, College Park, MD 20742, USA
| |
Collapse
|
31
|
Liu Y, Huang K, Chen W. Resolving cellular dynamics using single-cell temporal transcriptomics. Curr Opin Biotechnol 2024; 85:103060. [PMID: 38194753 DOI: 10.1016/j.copbio.2023.103060] [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: 10/01/2023] [Revised: 12/04/2023] [Accepted: 12/10/2023] [Indexed: 01/11/2024]
Abstract
Cellular dynamics, the transition of a cell from one state to another, is central to understanding developmental processes and disease progression. Single-cell transcriptomics has been pushing the frontiers of cellular dynamics studies into a genome-wide and single-cell level. While most single-cell RNA sequencing approaches are disruptive and only provide a snapshot of cell states, the dynamics of a cell could be reconstructed by either exploiting temporal information hiding in the transcriptomics data or integrating additional information. In this review, we describe these approaches, highlighting their underlying principles, key assumptions, and the rationality to interpret the results as models. We also discuss the recently emerging nondisruptive live-cell transcriptomics methods, which are highly complementary to the computational models for their assumption-free nature.
Collapse
Affiliation(s)
- Yifei Liu
- Key Laboratory of Quantitative Synthetic Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Kai Huang
- Key Laboratory of Quantitative Synthetic Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Wanze Chen
- Key Laboratory of Quantitative Synthetic Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China.
| |
Collapse
|
32
|
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: 3] [Impact Index Per Article: 3.0] [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
|
33
|
Cui H, Maan H, Vladoiu MC, Zhang J, Taylor MD, Wang B. DeepVelo: deep learning extends RNA velocity to multi-lineage systems with cell-specific kinetics. Genome Biol 2024; 25:27. [PMID: 38243313 PMCID: PMC10799431 DOI: 10.1186/s13059-023-03148-9] [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: 09/26/2023] [Accepted: 12/18/2023] [Indexed: 01/21/2024] Open
Abstract
Existing RNA velocity estimation methods strongly rely on predefined dynamics and cell-agnostic constant transcriptional kinetic rates, assumptions often violated in complex and heterogeneous single-cell RNA sequencing (scRNA-seq) data. Using a graph convolution network, DeepVelo overcomes these limitations by generalizing RNA velocity to cell populations containing time-dependent kinetics and multiple lineages. DeepVelo infers time-varying cellular rates of transcription, splicing, and degradation, recovers each cell's stage in the differentiation process, and detects functionally relevant driver genes regulating these processes. Application to various developmental and pathogenic processes demonstrates DeepVelo's capacity to study complex differentiation and lineage decision events in heterogeneous scRNA-seq data.
Collapse
Affiliation(s)
- Haotian Cui
- Peter Munk Cardiac Center, University Health Network, Toronto, Ontario, Canada
- Department of Computer Science, University of Toronto, Toronto, Ontario, Canada
- Vector Institute, Toronto, Ontario, Canada
| | - Hassaan Maan
- Peter Munk Cardiac Center, University Health Network, Toronto, Ontario, Canada
- Vector Institute, Toronto, Ontario, Canada
- Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada
| | - Maria C Vladoiu
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Jiao Zhang
- The Arthur and Sonia Labatt Brain Tumor Research Centre, The Hospital for Sick Children, Toronto, Ontario, Canada
- Developmental and Stem Cell Biology Program, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Michael D Taylor
- The Arthur and Sonia Labatt Brain Tumor Research Centre, The Hospital for Sick Children, Toronto, Ontario, Canada
- Developmental and Stem Cell Biology Program, The Hospital for Sick Children, Toronto, Ontario, Canada
- Baylor College of Medicine, Houston, TX, USA
- Texas Children's Hospital, Houston, TX, USA
| | - Bo Wang
- Peter Munk Cardiac Center, University Health Network, Toronto, Ontario, Canada.
- Department of Computer Science, University of Toronto, Toronto, Ontario, Canada.
- Vector Institute, Toronto, Ontario, Canada.
- Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada.
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada.
| |
Collapse
|
34
|
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
|
35
|
Cooke JP, Lai L. Transflammation in tissue regeneration and response to injury: How cell-autonomous inflammatory signaling mediates cell plasticity. Adv Drug Deliv Rev 2023; 203:115118. [PMID: 37884127 PMCID: PMC10842620 DOI: 10.1016/j.addr.2023.115118] [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: 10/18/2022] [Revised: 08/01/2023] [Accepted: 10/23/2023] [Indexed: 10/28/2023]
Abstract
Inflammation is a first responder against injury and infection and is also critical for the regeneration and repair of tissue after injury. The role of professional immune cells in tissue healing is well characterized. Professional immune cells respond to pathogens with humoral and cytotoxic responses; remove cellular debris through efferocytosis; secrete angiogenic cytokines and growth factors to repair the microvasculature and parenchyma. However, non-immune cells are also capable of responding to damage or pathogens. Non-immune somatic cells express pattern recognition receptors (PRRs) to detect pathogen-associated molecular patterns (PAMPs) and damage-associated molecular patterns (DAMPs). The PRRs activation leads to the release of inflammatory cytokines required for tissue defense and repair. Notably, the activation of PRRs also triggers epigenetic changes that promote DNA accessibility and cellular plasticity. Thus, non-immune cells directly respond to the local inflammatory cues and can undergo phenotypic modifications or even cell lineage transitions to facilitate tissue regeneration. This review will focus on the novel role of cell-autonomous inflammatory signaling in mediating cell plasticity, a process which is termed transflammation. We will discuss the regulation of this process by changes in the functions and expression levels of epigenetic modifiers, as well as metabolic and ROS/RNS-mediated epigenetic modulation of DNA accessibility during cell fate transition. We will highlight the recent technological developments in detecting cell plasticity and potential therapeutic applications of transflammation in tissue regeneration.
Collapse
Affiliation(s)
- John P Cooke
- Department of Cardiovascular Sciences, Houston Methodist Research Institute, Houston, TX, United States
| | - Li Lai
- Department of Cardiovascular Sciences, Houston Methodist Research Institute, Houston, TX, United States.
| |
Collapse
|
36
|
Song J, Zaidi SAA, He L, Zhang S, Zhou G. Integrative Analysis of Machine Learning and Molecule Docking Simulations for Ischemic Stroke Diagnosis and Therapy. Molecules 2023; 28:7704. [PMID: 38067435 PMCID: PMC10707570 DOI: 10.3390/molecules28237704] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2023] [Revised: 11/13/2023] [Accepted: 11/17/2023] [Indexed: 12/18/2023] Open
Abstract
Due to the narrow therapeutic window and high mortality of ischemic stroke, it is of great significance to investigate its diagnosis and therapy. We employed weighted gene coexpression network analysis (WGCNA) to ascertain gene modules related to stroke and used the maSigPro R package to seek the time-dependent genes in the progression of stroke. Three machine learning algorithms were further employed to identify the feature genes of stroke. A nomogram model was built and applied to evaluate the stroke patients. We analyzed single-cell RNA sequencing (scRNA-seq) data to discern microglia subclusters in ischemic stroke. The RNA velocity, pseudo time, and gene set enrichment analysis (GSEA) were performed to investigate the relationship of microglia subclusters. Connectivity map (CMap) analysis and molecule docking were used to screen a therapeutic agent for stroke. A nomogram model based on the feature genes showed a clinical net benefit and enabled an accurate evaluation of stroke patients. The RNA velocity and pseudo time analysis showed that microglia subcluster 0 would develop toward subcluster 2 within 24 h from stroke onset. The GSEA showed that the function of microglia subcluster 0 was opposite to that of subcluster 2. AZ_628, which screened from CMap analysis, was found to have lower binding energy with Mmp12, Lgals3, Fam20c, Capg, Pkm2, Sdc4, and Itga5 in microglia subcluster 2 and maybe a therapeutic agent for the poor development of microglia subcluster 2 after stroke. Our study presents a nomogram model for stroke diagnosis and provides a potential molecule agent for stroke therapy.
Collapse
Affiliation(s)
- Jingwei Song
- Department of Medical Cell Biology and Genetics, Guangdong Key Laboratory of Genomic Stability and Disease Prevention, Shenzhen Key Laboratory of Anti-Aging and Regenerative Medicine, and Shenzhen Engineering Laboratory of Regenerative Technologies for Orthopaedic Diseases, Health Sciences Center, Shenzhen University, Shenzhen 518060, China; (J.S.); (S.A.A.Z.); (L.H.)
| | - Syed Aqib Ali Zaidi
- Department of Medical Cell Biology and Genetics, Guangdong Key Laboratory of Genomic Stability and Disease Prevention, Shenzhen Key Laboratory of Anti-Aging and Regenerative Medicine, and Shenzhen Engineering Laboratory of Regenerative Technologies for Orthopaedic Diseases, Health Sciences Center, Shenzhen University, Shenzhen 518060, China; (J.S.); (S.A.A.Z.); (L.H.)
| | - Liangge He
- Department of Medical Cell Biology and Genetics, Guangdong Key Laboratory of Genomic Stability and Disease Prevention, Shenzhen Key Laboratory of Anti-Aging and Regenerative Medicine, and Shenzhen Engineering Laboratory of Regenerative Technologies for Orthopaedic Diseases, Health Sciences Center, Shenzhen University, Shenzhen 518060, China; (J.S.); (S.A.A.Z.); (L.H.)
| | - Shuai Zhang
- Brain Research Centre, Department of Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen 518055, China
| | - Guangqian Zhou
- Department of Medical Cell Biology and Genetics, Guangdong Key Laboratory of Genomic Stability and Disease Prevention, Shenzhen Key Laboratory of Anti-Aging and Regenerative Medicine, and Shenzhen Engineering Laboratory of Regenerative Technologies for Orthopaedic Diseases, Health Sciences Center, Shenzhen University, Shenzhen 518060, China; (J.S.); (S.A.A.Z.); (L.H.)
- Lungene Biotech Ltd., Shenzhen 518060, China
- Senotherapeutics Ltd., Hangzhou 311100, China
| |
Collapse
|
37
|
Chakraborty A, Li Y, Zhang C, Li Y, Rebello KR, Li S, Xu S, Vasquez HG, Zhang L, Luo W, Wang G, Chen K, Coselli JS, LeMaire SA, Shen YH. Epigenetic Induction of Smooth Muscle Cell Phenotypic Alterations in Aortic Aneurysms and Dissections. Circulation 2023; 148:959-977. [PMID: 37555319 PMCID: PMC10529114 DOI: 10.1161/circulationaha.123.063332] [Citation(s) in RCA: 38] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Accepted: 07/17/2023] [Indexed: 08/10/2023]
Abstract
BACKGROUND Smooth muscle cell (SMC) phenotypic switching has been increasingly detected in aortic aneurysm and dissection (AAD) tissues. However, the diverse SMC phenotypes in AAD tissues and the mechanisms driving SMC phenotypic alterations remain to be identified. METHODS We examined the transcriptomic and epigenomic dynamics of aortic SMC phenotypic changes in mice with angiotensin II-induced AAD by using single-cell RNA sequencing and single-cell sequencing assay for transposase-accessible chromatin. SMC phenotypic alteration in aortas from patients with ascending thoracic AAD was examined by using single-cell RNA sequencing analysis. RESULTS Single-cell RNA sequencing analysis revealed that aortic stress induced the transition of SMCs from a primary contractile phenotype to proliferative, extracellular matrix-producing, and inflammatory phenotypes. Lineage tracing showed the complete transformation of SMCs to fibroblasts and macrophages. Single-cell sequencing assay for transposase-accessible chromatin analysis indicated that these phenotypic alterations were controlled by chromatin remodeling marked by the reduced chromatin accessibility of contractile genes and the induced chromatin accessibility of genes involved in proliferation, extracellular matrix, and inflammation. IRF3 (interferon regulatory factor 3), a proinflammatory transcription factor activated by cytosolic DNA, was identified as a key driver of the transition of aortic SMCs from a contractile phenotype to an inflammatory phenotype. In cultured SMCs, cytosolic DNA signaled through its sensor STING (stimulator of interferon genes)-TBK1 (tank-binding kinase 1) to activate IRF3, which bound and recruited EZH2 (enhancer of zeste homolog 2) to contractile genes to induce repressive H3K27me3 modification and gene suppression. In contrast, double-stranded DNA-STING-IRF3 signaling induced inflammatory gene expression in SMCs. In Sting-/- mice, the aortic stress-induced transition of SMCs into an inflammatory phenotype was prevented, and SMC populations were preserved. Finally, profound SMC phenotypic alterations toward diverse directions were detected in human ascending thoracic AAD tissues. CONCLUSIONS Our study reveals the dynamic epigenetic induction of SMC phenotypic alterations in AAD. DNA damage and cytosolic leakage drive SMCs from a contractile phenotype to an inflammatory phenotype.
Collapse
Affiliation(s)
- Abhijit Chakraborty
- Division of Cardiothoracic Surgery, Michael E. DeBakey Department of Surgery (A.C., Y.L., C.Z., K.R.R., Y.L., S.X., W.L., H.G.V., L.Z., J.S.C., S.A.L., Y.H.S.), Baylor College of Medicine, Houston, TX
- Department of Cardiovascular Surgery, The Texas Heart Institute, Houston (A.C., Y.L., C.Z., K.R.R., Y.L., W.L., H.G.V., L.Z., J.S.C., S.A.L.)
| | - Yanming Li
- Division of Cardiothoracic Surgery, Michael E. DeBakey Department of Surgery (A.C., Y.L., C.Z., K.R.R., Y.L., S.X., W.L., H.G.V., L.Z., J.S.C., S.A.L., Y.H.S.), Baylor College of Medicine, Houston, TX
- Department of Cardiovascular Surgery, The Texas Heart Institute, Houston (A.C., Y.L., C.Z., K.R.R., Y.L., W.L., H.G.V., L.Z., J.S.C., S.A.L.)
| | - Chen Zhang
- Division of Cardiothoracic Surgery, Michael E. DeBakey Department of Surgery (A.C., Y.L., C.Z., K.R.R., Y.L., S.X., W.L., H.G.V., L.Z., J.S.C., S.A.L., Y.H.S.), Baylor College of Medicine, Houston, TX
- Department of Cardiovascular Surgery, The Texas Heart Institute, Houston (A.C., Y.L., C.Z., K.R.R., Y.L., W.L., H.G.V., L.Z., J.S.C., S.A.L.)
| | - Yang Li
- Division of Cardiothoracic Surgery, Michael E. DeBakey Department of Surgery (A.C., Y.L., C.Z., K.R.R., Y.L., S.X., W.L., H.G.V., L.Z., J.S.C., S.A.L., Y.H.S.), Baylor College of Medicine, Houston, TX
- Department of Cardiovascular Surgery, The Texas Heart Institute, Houston (A.C., Y.L., C.Z., K.R.R., Y.L., W.L., H.G.V., L.Z., J.S.C., S.A.L.)
| | - Kimberly R Rebello
- Division of Cardiothoracic Surgery, Michael E. DeBakey Department of Surgery (A.C., Y.L., C.Z., K.R.R., Y.L., S.X., W.L., H.G.V., L.Z., J.S.C., S.A.L., Y.H.S.), Baylor College of Medicine, Houston, TX
- Department of Cardiovascular Surgery, The Texas Heart Institute, Houston (A.C., Y.L., C.Z., K.R.R., Y.L., W.L., H.G.V., L.Z., J.S.C., S.A.L.)
| | - Shengyu Li
- Center for Bioinformatics and Computational Biology, Houston Methodist Research Institute, TX (S.L., G.W.)
| | - Samantha Xu
- Division of Cardiothoracic Surgery, Michael E. DeBakey Department of Surgery (A.C., Y.L., C.Z., K.R.R., Y.L., S.X., W.L., H.G.V., L.Z., J.S.C., S.A.L., Y.H.S.), Baylor College of Medicine, Houston, TX
| | - Hernan G Vasquez
- Division of Cardiothoracic Surgery, Michael E. DeBakey Department of Surgery (A.C., Y.L., C.Z., K.R.R., Y.L., S.X., W.L., H.G.V., L.Z., J.S.C., S.A.L., Y.H.S.), Baylor College of Medicine, Houston, TX
- Department of Cardiovascular Surgery, The Texas Heart Institute, Houston (A.C., Y.L., C.Z., K.R.R., Y.L., W.L., H.G.V., L.Z., J.S.C., S.A.L.)
| | - Lin Zhang
- Division of Cardiothoracic Surgery, Michael E. DeBakey Department of Surgery (A.C., Y.L., C.Z., K.R.R., Y.L., S.X., W.L., H.G.V., L.Z., J.S.C., S.A.L., Y.H.S.), Baylor College of Medicine, Houston, TX
- Department of Cardiovascular Surgery, The Texas Heart Institute, Houston (A.C., Y.L., C.Z., K.R.R., Y.L., W.L., H.G.V., L.Z., J.S.C., S.A.L.)
| | - Wei Luo
- Division of Cardiothoracic Surgery, Michael E. DeBakey Department of Surgery (A.C., Y.L., C.Z., K.R.R., Y.L., S.X., W.L., H.G.V., L.Z., J.S.C., S.A.L., Y.H.S.), Baylor College of Medicine, Houston, TX
- Department of Cardiovascular Surgery, The Texas Heart Institute, Houston (A.C., Y.L., C.Z., K.R.R., Y.L., W.L., H.G.V., L.Z., J.S.C., S.A.L.)
| | - Guangyu Wang
- Center for Bioinformatics and Computational Biology, Houston Methodist Research Institute, TX (S.L., G.W.)
| | - Kaifu Chen
- Department of Pediatrics, Harvard Medical School, Boston, MA (K.C.)
| | - Joseph S Coselli
- Division of Cardiothoracic Surgery, Michael E. DeBakey Department of Surgery (A.C., Y.L., C.Z., K.R.R., Y.L., S.X., W.L., H.G.V., L.Z., J.S.C., S.A.L., Y.H.S.), Baylor College of Medicine, Houston, TX
- Cardiovascular Research Institute (J.S.C., S.A.L., Y.H.S.), Baylor College of Medicine, Houston, TX
- Department of Cardiovascular Surgery, The Texas Heart Institute, Houston (A.C., Y.L., C.Z., K.R.R., Y.L., W.L., H.G.V., L.Z., J.S.C., S.A.L.)
| | - Scott A LeMaire
- Division of Cardiothoracic Surgery, Michael E. DeBakey Department of Surgery (A.C., Y.L., C.Z., K.R.R., Y.L., S.X., W.L., H.G.V., L.Z., J.S.C., S.A.L., Y.H.S.), Baylor College of Medicine, Houston, TX
- Cardiovascular Research Institute (J.S.C., S.A.L., Y.H.S.), Baylor College of Medicine, Houston, TX
- Department of Cardiovascular Surgery, The Texas Heart Institute, Houston (A.C., Y.L., C.Z., K.R.R., Y.L., W.L., H.G.V., L.Z., J.S.C., S.A.L.)
| | - Ying H Shen
- Division of Cardiothoracic Surgery, Michael E. DeBakey Department of Surgery (A.C., Y.L., C.Z., K.R.R., Y.L., S.X., W.L., H.G.V., L.Z., J.S.C., S.A.L., Y.H.S.), Baylor College of Medicine, Houston, TX
- Cardiovascular Research Institute (J.S.C., S.A.L., Y.H.S.), Baylor College of Medicine, Houston, TX
| |
Collapse
|
38
|
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
|