1
|
Yang Y, McCullough CG, Seninge L, Guo L, Kwon WJ, Zhang Y, Li NY, Gaddam S, Pan C, Zhen H, Torkelson J, Glass IA, Charville GW, Que J, Stuart JM, Ding H, Oro AE. A spatiotemporal and machine-learning platform facilitates the manufacturing of hPSC-derived esophageal mucosa. Dev Cell 2025; 60:1359-1376.e10. [PMID: 39798574 PMCID: PMC12055484 DOI: 10.1016/j.devcel.2024.12.030] [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] [Revised: 11/30/2023] [Accepted: 12/12/2024] [Indexed: 01/15/2025]
Abstract
Human pluripotent stem cell-derived tissue engineering offers great promise for designer cell-based personalized therapeutics, but harnessing such potential requires a deeper understanding of tissue-level interactions. We previously developed a cell replacement manufacturing method for ectoderm-derived skin epithelium. However, it remains challenging to manufacture the endoderm-derived esophageal epithelium despite possessing a similar stratified epithelial structure. Here, we employ single-cell and spatial technologies to generate a spatiotemporal multi-omics cell census for human esophageal development. We identify the cellular diversity, dynamics, and signal communications for the developing esophageal epithelium and stroma. Using Manatee, a machine-learning algorithm, we prioritize the combinations of candidate human developmental signals for in vitro derivation of esophageal basal cells. Functional validation of Manatee predictions leads to a clinically compatible system for manufacturing human esophageal mucosa.
Collapse
Affiliation(s)
- Ying Yang
- Program in Epithelial Biology and Center for Definitive and Curative Medicine, Stanford University, Stanford, CA, USA
| | - Carmel Grace McCullough
- Program in Epithelial Biology and Center for Definitive and Curative Medicine, Stanford University, Stanford, CA, USA
| | - Lucas Seninge
- Department of Biomolecular Engineering and Genomics Institute, University of California, Santa Cruz, Santa Cruz, CA, USA
| | - Lihao Guo
- Department of Pharmacy Practice and Science, University of Arizona, Tucson, AZ, USA
| | - Woo-Joo Kwon
- Program in Epithelial Biology and Center for Definitive and Curative Medicine, Stanford University, Stanford, CA, USA
| | - Yongchun Zhang
- State Key Laboratory of Microbial Metabolism & Joint International Research Laboratory of Metabolic and Developmental Sciences, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, China
| | - Nancy Yanzhe Li
- Program in Epithelial Biology and Center for Definitive and Curative Medicine, Stanford University, Stanford, CA, USA
| | - Sadhana Gaddam
- Program in Epithelial Biology and Center for Definitive and Curative Medicine, Stanford University, Stanford, CA, USA
| | - Cory Pan
- Program in Epithelial Biology and Center for Definitive and Curative Medicine, Stanford University, Stanford, CA, USA
| | - Hanson Zhen
- Program in Epithelial Biology and Center for Definitive and Curative Medicine, Stanford University, Stanford, CA, USA
| | - Jessica Torkelson
- Program in Epithelial Biology and Center for Definitive and Curative Medicine, Stanford University, Stanford, CA, USA
| | - Ian A Glass
- Birth Defect Research Laboratory Department of Pediatrics, University of Washington, Seattle, WA, USA
| | | | - Jianwen Que
- Department of Medicine, Columbia University, New York, NY, USA
| | - Joshua M Stuart
- Department of Biomolecular Engineering and Genomics Institute, University of California, Santa Cruz, Santa Cruz, CA, USA
| | - Hongxu Ding
- Department of Pharmacy Practice and Science, University of Arizona, Tucson, AZ, USA.
| | - Anthony E Oro
- Program in Epithelial Biology and Center for Definitive and Curative Medicine, Stanford University, Stanford, CA, USA.
| |
Collapse
|
2
|
Wang L, Liu Y, Zhang Y, Sun Q, Wang X, Li Q, Li X. Dual-crosslinkable alginate hydrogel with dynamic viscoelasticity for chondrogenic and osteogenic differentiation of mesenchymal stem cells. Int J Biol Macromol 2025; 307:142346. [PMID: 40118412 DOI: 10.1016/j.ijbiomac.2025.142346] [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: 12/23/2024] [Revised: 03/08/2025] [Accepted: 03/19/2025] [Indexed: 03/23/2025]
Abstract
Tissue engineering presents an advanced approach for cartilage and bone tissue repair, with cells serving as a crucial component of the treatment process. The viscoelasticity, a defining fundamental mechanical property, significantly influences cellular behavior. The majority of current research has primarily focused on comparing static elastic and viscoelastic hydrogels with varying stress relaxation rates, while neglecting the inherent dynamic viscoelastic properties of native tissues. Herein, we developed a dynamic viscoelastic hydrogel system employing modified sodium alginate hydrogels to explore the impact of the transfer of viscoelasticity and elastic mechanical properties on the behavior and fate of mesenchymal stem cells (MSCs). The results demonstrated that a viscoelastic environment facilitates greater cell proliferation and spreading. Moreover, extended exposure to the viscoelastic environment resulted in significantly enhanced secretion of osteogenic/chondrogenic extracellular matrix (ECM), upregulates differentiation-specific gene expression, and supports nuclear localization of Yes-associated protein (YAP) and transcriptional co-activator with PDZ-binding motif (TAZ). This study elucidates the mechanical microenvironment required for MSC differentiation, enriching the theoretical foundation for the design of optimized scaffold in cartilage and bone tissue engineering applications.
Collapse
Affiliation(s)
- Litao Wang
- School of Mechanics and Safety Engineering, National Center for International Joint Research of Micro-nano Molding Technology, Zhengzhou University, Zhengzhou 450001, China
| | - Yajing Liu
- School of Mechanics and Safety Engineering, National Center for International Joint Research of Micro-nano Molding Technology, Zhengzhou University, Zhengzhou 450001, China
| | - Yuhang Zhang
- School of Mechanics and Safety Engineering, National Center for International Joint Research of Micro-nano Molding Technology, Zhengzhou University, Zhengzhou 450001, China
| | - Qingqing Sun
- School of Materials Science and Engineering, Zhengzhou Key Laboratory of Flexible Electronic Materials and Thin-Film Technologies, Zhengzhou University, Zhengzhou 450001, China
| | - Xiaofeng Wang
- School of Mechanics and Safety Engineering, National Center for International Joint Research of Micro-nano Molding Technology, Zhengzhou University, Zhengzhou 450001, China
| | - Qian Li
- School of Mechanics and Safety Engineering, National Center for International Joint Research of Micro-nano Molding Technology, Zhengzhou University, Zhengzhou 450001, China.
| | - Xiaomeng Li
- School of Mechanics and Safety Engineering, National Center for International Joint Research of Micro-nano Molding Technology, Zhengzhou University, Zhengzhou 450001, China.
| |
Collapse
|
3
|
Xu Q, Halle L, Hediyeh-Zadeh S, Kuijs M, Riedweg R, Kilik U, Recaldin T, Yu Q, Rall I, Frum T, Adam L, Parikh S, Kfuri-Rubens R, Gander M, Klein D, Curion F, He Z, Fleck JS, Oost K, Kahnwald M, Barbiero S, Mitrofanova O, Maciag GJ, Jensen KB, Lutolf M, Liberali P, Spence JR, Gjorevski N, Beumer J, Treutlein B, Theis FJ, Camp JG. An integrated transcriptomic cell atlas of human endoderm-derived organoids. Nat Genet 2025; 57:1201-1212. [PMID: 40355592 DOI: 10.1038/s41588-025-02182-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Accepted: 03/27/2025] [Indexed: 05/14/2025]
Abstract
Human pluripotent stem cells and tissue-resident fetal and adult stem cells can generate epithelial tissues of endodermal origin in vitro that recapitulate aspects of developing and adult human physiology. Here, we integrate single-cell transcriptomes from 218 samples covering organoids and other models of diverse endoderm-derived tissues to establish an initial version of a human endoderm-derived organoid cell atlas. The integration includes nearly one million cells across diverse conditions, data sources and protocols. We compare cell types and states between organoid models and harmonize cell annotations through mapping to primary tissue counterparts. Focusing on the intestine and lung, we provide examples of mapping data from new protocols and show how the atlas can be used as a diverse cohort to assess perturbations and disease models. The human endoderm-derived organoid cell atlas makes diverse datasets centrally available and will be valuable to assess fidelity, characterize perturbed and diseased states, and streamline protocol development.
Collapse
Affiliation(s)
- Quan Xu
- Institute of Human Biology (IHB), Roche Pharma Research and Early Development, Roche Innovation Center, Basel, Switzerland.
| | - Lennard Halle
- Department of Computational Health, Institute of Computational Biology, Helmholtz Center Munich, Munich, Germany
| | - Soroor Hediyeh-Zadeh
- Department of Computational Health, Institute of Computational Biology, Helmholtz Center Munich, Munich, Germany
- School of Life Sciences, Technical University of Munich, Munich, Germany
| | - Merel Kuijs
- Department of Computational Health, Institute of Computational Biology, Helmholtz Center Munich, Munich, Germany
| | - Rya Riedweg
- Institute of Human Biology (IHB), Roche Pharma Research and Early Development, Roche Innovation Center, Basel, Switzerland
| | - Umut Kilik
- Institute of Human Biology (IHB), Roche Pharma Research and Early Development, Roche Innovation Center, Basel, Switzerland
- Biozentrum, University of Basel, Basel, Switzerland
| | - Timothy Recaldin
- Roche Innovation Center Basel, Roche Pharma Research and Early Development, Basel, Switzerland
| | - Qianhui Yu
- Institute of Human Biology (IHB), Roche Pharma Research and Early Development, Roche Innovation Center, Basel, Switzerland
| | - Isabell Rall
- Institute of Human Biology (IHB), Roche Pharma Research and Early Development, Roche Innovation Center, Basel, Switzerland
| | - Tristan Frum
- Department of Internal Medicine, Division of Gastroenterology and Hepatology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Lukas Adam
- Institute of Human Biology (IHB), Roche Pharma Research and Early Development, Roche Innovation Center, Basel, Switzerland
| | - Shrey Parikh
- Department of Computational Health, Institute of Computational Biology, Helmholtz Center Munich, Munich, Germany
- School of Life Sciences, Technical University of Munich, Munich, Germany
| | - Raphael Kfuri-Rubens
- Department of Computational Health, Institute of Computational Biology, Helmholtz Center Munich, Munich, Germany
- IIIrd Medical Department, Klinikum rechts der Isar, Munich, Germany
- School of Medicine, Technical University of Munich, Munich, Germany
| | - Manuel Gander
- Department of Computational Health, Institute of Computational Biology, Helmholtz Center Munich, Munich, Germany
| | - Dominik Klein
- Department of Computational Health, Institute of Computational Biology, Helmholtz Center Munich, Munich, Germany
| | - Fabiola Curion
- Department of Computational Health, Institute of Computational Biology, Helmholtz Center Munich, Munich, Germany
- School of Computation, Information and Technology, Technical University of Munich, Munich, Germany
| | - Zhisong He
- Department of Biosystems Science and Engineering, ETH Zürich, Basel, Switzerland
| | - Jonas Simon Fleck
- Institute of Human Biology (IHB), Roche Pharma Research and Early Development, Roche Innovation Center, Basel, Switzerland
| | - Koen Oost
- Friedrich Miescher Institute for Biomedical Research (FMI), Basel, Switzerland
| | - Maurice Kahnwald
- Friedrich Miescher Institute for Biomedical Research (FMI), Basel, Switzerland
| | - Silvia Barbiero
- Friedrich Miescher Institute for Biomedical Research (FMI), Basel, Switzerland
| | - Olga Mitrofanova
- Institute of Human Biology (IHB), Roche Pharma Research and Early Development, Roche Innovation Center, Basel, Switzerland
| | - Grzegorz Jerzy Maciag
- Novo Nordisk Foundation Center for Stem Cell Medicine, reNEW, University of Copenhagen, Copenhagen, Denmark
| | - Kim B Jensen
- Novo Nordisk Foundation Center for Stem Cell Medicine, reNEW, University of Copenhagen, Copenhagen, Denmark
| | - Matthias Lutolf
- Institute of Human Biology (IHB), Roche Pharma Research and Early Development, Roche Innovation Center, Basel, Switzerland
- Laboratory of Stem Cell Bioengineering, École Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | - Prisca Liberali
- Biozentrum, University of Basel, Basel, Switzerland
- Friedrich Miescher Institute for Biomedical Research (FMI), Basel, Switzerland
| | - Jason R Spence
- Department of Internal Medicine, Division of Gastroenterology and Hepatology, University of Michigan Medical School, Ann Arbor, MI, USA
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI, USA
- Department of Biomedical Engineering, University of Michigan College of Engineering, Ann Arbor, MI, USA
| | - Nikolche Gjorevski
- Institute of Human Biology (IHB), Roche Pharma Research and Early Development, Roche Innovation Center, Basel, Switzerland
| | - Joep Beumer
- Institute of Human Biology (IHB), Roche Pharma Research and Early Development, Roche Innovation Center, Basel, Switzerland
| | - Barbara Treutlein
- Department of Biosystems Science and Engineering, ETH Zürich, Basel, Switzerland.
| | - Fabian J Theis
- Department of Computational Health, Institute of Computational Biology, Helmholtz Center Munich, Munich, Germany.
- School of Life Sciences, Technical University of Munich, Munich, Germany.
- School of Computation, Information and Technology, Technical University of Munich, Munich, Germany.
| | - J Gray Camp
- Institute of Human Biology (IHB), Roche Pharma Research and Early Development, Roche Innovation Center, Basel, Switzerland.
- Biozentrum, University of Basel, Basel, Switzerland.
| |
Collapse
|
4
|
Kim H, Choi S, Heo H, Cho SH, Lee Y, Kim D, Jung KO, Rhee S. Applications of Single-Cell Omics Technologies for Induced Pluripotent Stem Cell-Based Cardiovascular Research. Int J Stem Cells 2025; 18:37-48. [PMID: 39129179 PMCID: PMC11867907 DOI: 10.15283/ijsc23183] [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: 11/13/2023] [Revised: 04/30/2024] [Accepted: 04/30/2024] [Indexed: 08/13/2024] Open
Abstract
Single-cell omics technologies have transformed our investigation of genomic, transcriptomic, and proteomic landscapes at the individual cell level. In particular, the application of single-cell RNA sequencing has unveiled the complex transcriptional variations inherent in cardiac cells, offering valuable perspectives into their dynamics. This review focuses on the integration of single-cell omics with induced pluripotent stem cells (iPSCs) in the context of cardiovascular research, offering a unique avenue to deepen our understanding of cardiac biology. By synthesizing insights from various single-cell technologies, we aim to elucidate the molecular intricacies of heart health and diseases. Beyond current methodologies, we explore the potential of emerging paradigms such as single-cell/spatial omics, delving into their capacity to reveal the spatial organization of cellular components within cardiac tissues. Furthermore, we anticipate their transformative role in shaping the future of cardiovascular research. This review aims to contribute to the advancement of knowledge in the field, offering a comprehensive perspective on the synergistic potential of transcriptomic analyses, iPSC applications, and the evolving frontier of spatial omics.
Collapse
Affiliation(s)
- Hyunjoon Kim
- Department of Molecular and Cell Biology, University of California, Berkeley, CA, USA
- K-BioX, Palo Alto, CA, USA
| | - Sohee Choi
- K-BioX, Palo Alto, CA, USA
- Department of Biological Sciences, Sookmyung Women’s University, Seoul, Korea
| | - HyoJung Heo
- Department of Molecular and Cell Biology, University of California, Berkeley, CA, USA
- K-BioX, Palo Alto, CA, USA
| | - Su Han Cho
- K-BioX, Palo Alto, CA, USA
- Department of Biology, Kyung Hee University, Seoul, Korea
| | - Yuna Lee
- K-BioX, Palo Alto, CA, USA
- Department of Systems Biotechnology, Konkuk University, Seoul, Korea
| | - Dohyup Kim
- K-BioX, Palo Alto, CA, USA
- Asthma Research Division, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, USA
| | - Kyung Oh Jung
- K-BioX, Palo Alto, CA, USA
- Department of Anatomy, College of Medicine, Chung-Ang University, Seoul, Korea
| | - Siyeon Rhee
- K-BioX, Palo Alto, CA, USA
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford University, Palo Alto, CA, USA
| |
Collapse
|
5
|
Artegiani B, Hendriks D. Organoids from pluripotent stem cells and human tissues: When two cultures meet each other. Dev Cell 2025; 60:493-511. [PMID: 39999776 DOI: 10.1016/j.devcel.2025.01.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Revised: 06/13/2024] [Accepted: 01/10/2025] [Indexed: 02/27/2025]
Abstract
Human organoids are a widely used tool in cell biology to study homeostatic processes, disease, and development. The term organoids covers a plethora of model systems from different cellular origins that each have unique features and applications but bring their own challenges. This review discusses the basic principles underlying organoids generated from pluripotent stem cells (PSCs) as well as those derived from tissue stem cells (TSCs). We consider how well PSC- and TSC-organoids mimic the different intended organs in terms of cellular complexity, maturity, functionality, and the ongoing efforts to constitute predictive complex models of in vivo situations. We discuss the advantages and limitations associated with each system to answer different biological questions including in the field of cancer and developmental biology, and with respect to implementing emerging advanced technologies, such as (spatial) -omics analyses, CRISPR screens, and high-content imaging screens. We postulate how the two fields may move forward together, integrating advantages of one to the other.
Collapse
Affiliation(s)
| | - Delilah Hendriks
- Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands.
| |
Collapse
|
6
|
Golchin A, Shams F, Moradi F, Sadrabadi AE, Parviz S, Alipour S, Ranjbarvan P, Hemmati Y, Rahnama M, Rasmi Y, Aziz SGG. Single-cell Technology in Stem Cell Research. Curr Stem Cell Res Ther 2025; 20:9-32. [PMID: 38243989 DOI: 10.2174/011574888x265479231127065541] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Revised: 09/23/2023] [Accepted: 10/04/2023] [Indexed: 01/22/2024]
Abstract
Single-cell technology (SCT), which enables the examination of the fundamental units comprising biological organs, tissues, and cells, has emerged as a powerful tool, particularly in the field of biology, with a profound impact on stem cell research. This innovative technology opens new pathways for acquiring cell-specific data and gaining insights into the molecular pathways governing organ function and biology. SCT is not only frequently used to explore rare and diverse cell types, including stem cells, but it also unveils the intricacies of cellular diversity and dynamics. This perspective, crucial for advancing stem cell research, facilitates non-invasive analyses of molecular dynamics and cellular functions over time. Despite numerous investigations into potential stem cell therapies for genetic disorders, degenerative conditions, and severe injuries, the number of approved stem cell-based treatments remains limited. This limitation is attributed to the various heterogeneities present among stem cell sources, hindering their widespread clinical utilization. Furthermore, stem cell research is intimately connected with cutting-edge technologies, such as microfluidic organoids, CRISPR technology, and cell/tissue engineering. Each strategy developed to overcome the constraints of stem cell research has the potential to significantly impact advanced stem cell therapies. Drawing on the advantages and progress achieved through SCT-based approaches, this study aims to provide an overview of the advancements and concepts associated with the utilization of SCT in stem cell research and its related fields.
Collapse
Affiliation(s)
- Ali Golchin
- Cellular and Molecular Research Center, Cellular and Molecular Medicine Institute, Urmia University of Medical Sciences, Urmia, Iran
- Department of Clinical Biochemistry and Applied Cell Sciences, School of Medicine, Urmia University of Medical Sciences, Urmia, Iran
| | - Forough Shams
- Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Shahid, Beheshti University of Medical Sciences, Tehran, Iran
| | - Faezeh Moradi
- Department of Tissue Engineering, School of Medicine, Tarbiat Modares University, Tehran, Iran
| | - Amin Ebrahimi Sadrabadi
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR , Tehran, Iran
| | - Shima Parviz
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Medical Sciences and Technologies, Shiraz, University of Medical Sciences, Shiraz, Iran
| | - Shahriar Alipour
- Cellular and Molecular Research Center, Cellular and Molecular Medicine Institute, Urmia University of Medical Sciences, Urmia, Iran
- Department of Clinical Biochemistry and Applied Cell Sciences, School of Medicine, Urmia University of Medical Sciences, Urmia, Iran
| | - Parviz Ranjbarvan
- Cellular and Molecular Research Center, Cellular and Molecular Medicine Institute, Urmia University of Medical Sciences, Urmia, Iran
- Department of Clinical Biochemistry and Applied Cell Sciences, School of Medicine, Urmia University of Medical Sciences, Urmia, Iran
| | - Yaser Hemmati
- Department of Prosthodontics, Dental Faculty, Urmia University of Medical Science, Urmia, Iran
| | - Maryam Rahnama
- Department of Clinical Biochemistry and Applied Cell Sciences, School of Medicine, Urmia University of Medical Sciences, Urmia, Iran
| | - Yousef Rasmi
- Department of Clinical Biochemistry and Applied Cell Sciences, School of Medicine, Urmia University of Medical Sciences, Urmia, Iran
| | - Shiva Gholizadeh-Ghaleh Aziz
- Department of Clinical Biochemistry and Applied Cell Sciences, School of Medicine, Urmia University of Medical Sciences, Urmia, Iran
| |
Collapse
|
7
|
Crawford AJ, Forjaz A, Bons J, Bhorkar I, Roy T, Schell D, Queiroga V, Ren K, Kramer D, Huang W, Russo GC, Lee MH, Wu PH, Shih IM, Wang TL, Atkinson MA, Schilling B, Kiemen AL, Wirtz D. Combined assembloid modeling and 3D whole-organ mapping captures the microanatomy and function of the human fallopian tube. SCIENCE ADVANCES 2024; 10:eadp6285. [PMID: 39331707 PMCID: PMC11430475 DOI: 10.1126/sciadv.adp6285] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Accepted: 08/22/2024] [Indexed: 09/29/2024]
Abstract
The fallopian tubes play key roles in processes from pregnancy to ovarian cancer where three-dimensional (3D) cellular and extracellular interactions are important to their pathophysiology. Here, we develop a 3D multicompartment assembloid model of the fallopian tube that molecularly, functionally, and architecturally resembles the organ. Global label-free proteomics, innovative assays capturing physiological functions of the fallopian tube (i.e., oocyte transport), and whole-organ single-cell resolution mapping are used to validate these assembloids through a multifaceted platform with direct comparisons to fallopian tube tissue. These techniques converge at a unique combination of assembloid parameters with the highest similarity to the reference fallopian tube. This work establishes (i) an optimized model of the human fallopian tubes for in vitro studies of their pathophysiology and (ii) an iterative platform for customized 3D in vitro models of human organs that are molecularly, functionally, and microanatomically accurate by combining tunable assembloid and tissue mapping methods.
Collapse
Affiliation(s)
- Ashleigh J Crawford
- Johns Hopkins Institute for Nanobiotechnology, The Johns Hopkins University, Baltimore, MD 21218, USA
- Department of Chemical and Biomolecular Engineering, Johns Hopkins Physical Sciences-Oncology Center, The Johns Hopkins University, Baltimore, MD 21218, USA
| | - André Forjaz
- Johns Hopkins Institute for Nanobiotechnology, The Johns Hopkins University, Baltimore, MD 21218, USA
- Department of Chemical and Biomolecular Engineering, Johns Hopkins Physical Sciences-Oncology Center, The Johns Hopkins University, Baltimore, MD 21218, USA
| | - Joanna Bons
- Buck Institute for Research on Aging, Novato, CA 94945, USA
| | - Isha Bhorkar
- Johns Hopkins Institute for Nanobiotechnology, The Johns Hopkins University, Baltimore, MD 21218, USA
- Department of Biomedical Engineering, The Johns Hopkins University, Baltimore, MD 21218, USA
| | - Triya Roy
- Johns Hopkins Institute for Nanobiotechnology, The Johns Hopkins University, Baltimore, MD 21218, USA
- Department of Chemical and Biomolecular Engineering, Johns Hopkins Physical Sciences-Oncology Center, The Johns Hopkins University, Baltimore, MD 21218, USA
| | - David Schell
- Johns Hopkins Institute for Nanobiotechnology, The Johns Hopkins University, Baltimore, MD 21218, USA
- Department of Chemical and Biomolecular Engineering, Johns Hopkins Physical Sciences-Oncology Center, The Johns Hopkins University, Baltimore, MD 21218, USA
| | - Vasco Queiroga
- Johns Hopkins Institute for Nanobiotechnology, The Johns Hopkins University, Baltimore, MD 21218, USA
- Department of Chemical and Biomolecular Engineering, Johns Hopkins Physical Sciences-Oncology Center, The Johns Hopkins University, Baltimore, MD 21218, USA
| | - Kehan Ren
- Johns Hopkins Institute for Nanobiotechnology, The Johns Hopkins University, Baltimore, MD 21218, USA
- Department of Biomedical Engineering, The Johns Hopkins University, Baltimore, MD 21218, USA
| | - Donald Kramer
- Johns Hopkins Institute for Nanobiotechnology, The Johns Hopkins University, Baltimore, MD 21218, USA
- Department of Biotechnology, Johns Hopkins Advanced Academic Programs, The Johns Hopkins University, Baltimore, MD 21218, USA
| | - Wilson Huang
- Johns Hopkins Institute for Nanobiotechnology, The Johns Hopkins University, Baltimore, MD 21218, USA
- Department of Biology, The Johns Hopkins University, Baltimore, MD 21218, USA
| | - Gabriella C Russo
- Johns Hopkins Institute for Nanobiotechnology, The Johns Hopkins University, Baltimore, MD 21218, USA
- Department of Chemical and Biomolecular Engineering, Johns Hopkins Physical Sciences-Oncology Center, The Johns Hopkins University, Baltimore, MD 21218, USA
| | - Meng-Horng Lee
- Johns Hopkins Institute for Nanobiotechnology, The Johns Hopkins University, Baltimore, MD 21218, USA
- Department of Chemical and Biomolecular Engineering, Johns Hopkins Physical Sciences-Oncology Center, The Johns Hopkins University, Baltimore, MD 21218, USA
| | - Pei-Hsun Wu
- Johns Hopkins Institute for Nanobiotechnology, The Johns Hopkins University, Baltimore, MD 21218, USA
- Department of Chemical and Biomolecular Engineering, Johns Hopkins Physical Sciences-Oncology Center, The Johns Hopkins University, Baltimore, MD 21218, USA
| | - Ie-Ming Shih
- Department of Gynecology and Obstetrics, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Department of Pathology, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Department of Oncology, The Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Tian-Li Wang
- Department of Gynecology and Obstetrics, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Department of Pathology, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Department of Oncology, The Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Mark A Atkinson
- Departments of Pathology, Immunology, and Laboratory Medicine, College of Medicine, University of Florida Diabetes Institute, Gainesville, FL 32610, USA
- Departments of Pediatrics, College of Medicine, University of Florida Diabetes Institute, Gainesville, FL 32610, USA
| | | | - Ashley L Kiemen
- Johns Hopkins Institute for Nanobiotechnology, The Johns Hopkins University, Baltimore, MD 21218, USA
- Department of Chemical and Biomolecular Engineering, Johns Hopkins Physical Sciences-Oncology Center, The Johns Hopkins University, Baltimore, MD 21218, USA
- Department of Pathology, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Department of Oncology, The Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Department of Functional Anatomy and Evolution, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Denis Wirtz
- Johns Hopkins Institute for Nanobiotechnology, The Johns Hopkins University, Baltimore, MD 21218, USA
- Department of Chemical and Biomolecular Engineering, Johns Hopkins Physical Sciences-Oncology Center, The Johns Hopkins University, Baltimore, MD 21218, USA
- Department of Biomedical Engineering, The Johns Hopkins University, Baltimore, MD 21218, USA
- Department of Pathology, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| |
Collapse
|
8
|
Yi R, Chen S, Guan M, Liao C, Zhu Y, Ip JPK, Ye T, Chen Y. A single-cell transcriptomic dataset of pluripotent stem cell-derived astrocytes via NFIB/SOX9 overexpression. Sci Data 2024; 11:987. [PMID: 39256463 PMCID: PMC11387634 DOI: 10.1038/s41597-024-03823-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: 11/27/2023] [Accepted: 08/27/2024] [Indexed: 09/12/2024] Open
Abstract
Astrocytes, the predominant glial cells in the central nervous system, play essential roles in maintaining brain function. Reprogramming induced pluripotent stem cells (iPSCs) to become astrocytes through overexpression of the transcription factors, NFIB and SOX9, is a rapid and efficient approach for studying human neurological diseases and identifying therapeutic targets. However, the precise differentiation path and molecular signatures of induced astrocytes remain incompletely understood. Accordingly, we performed single-cell RNA sequencing analysis on 64,736 cells to establish a comprehensive atlas of NFIB/SOX9-directed astrocyte differentiation from human iPSCs. Our dataset provides detailed information about the path of astrocyte differentiation, highlighting the stepwise molecular changes that occur throughout the differentiation process. This dataset serves as a valuable reference for dissecting uncharacterized transcriptomic features of NFIB/SOX9-induced astrocytes and investigating lineage progression during astrocyte differentiation. Moreover, these findings pave the way for future studies on neurological diseases using the NFIB/SOX9-induced astrocyte model.
Collapse
Affiliation(s)
- Ran Yi
- Chinese Academy of Sciences Key Laboratory of Brain Connectome and Manipulation, Shenzhen Key Laboratory of Translational Research for Brain Diseases, the Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen, China
- SIAT-HKUST Joint Laboratory for Brain Science, Chinese Academy of Sciences, Shenzhen, China
| | - Shuai Chen
- Chinese Academy of Sciences Key Laboratory of Brain Connectome and Manipulation, Shenzhen Key Laboratory of Translational Research for Brain Diseases, the Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen, China
- SIAT-HKUST Joint Laboratory for Brain Science, Chinese Academy of Sciences, Shenzhen, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Mingfeng Guan
- Chinese Academy of Sciences Key Laboratory of Brain Connectome and Manipulation, Shenzhen Key Laboratory of Translational Research for Brain Diseases, the Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen, China
- SIAT-HKUST Joint Laboratory for Brain Science, Chinese Academy of Sciences, Shenzhen, China
- Guangdong Provincial Key Laboratory of Brain Science, Disease and Drug Development, HKUST Shenzhen Research Institute, Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen, China
| | - Chunyan Liao
- Chinese Academy of Sciences Key Laboratory of Brain Connectome and Manipulation, Shenzhen Key Laboratory of Translational Research for Brain Diseases, the Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen, China
- SIAT-HKUST Joint Laboratory for Brain Science, Chinese Academy of Sciences, Shenzhen, China
- Guangdong Provincial Key Laboratory of Brain Science, Disease and Drug Development, HKUST Shenzhen Research Institute, Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen, China
| | - Yao Zhu
- School of Biomedical Sciences, the Chinese University of Hong Kong, Hong Kong, China
| | - Jacque Pak Kan Ip
- School of Biomedical Sciences, the Chinese University of Hong Kong, Hong Kong, China
- Gerald Choa Neuroscience Institute, the Chinese University of Hong Kong, Hong Kong, China
- CUHK Shenzhen Research Institute, the Chinese University of Hong Kong, Shenzhen, China
| | - Tao Ye
- Chinese Academy of Sciences Key Laboratory of Brain Connectome and Manipulation, Shenzhen Key Laboratory of Translational Research for Brain Diseases, the Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen, China.
- SIAT-HKUST Joint Laboratory for Brain Science, Chinese Academy of Sciences, Shenzhen, China.
- University of Chinese Academy of Sciences, Beijing, China.
- Guangdong Provincial Key Laboratory of Brain Science, Disease and Drug Development, HKUST Shenzhen Research Institute, Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen, China.
| | - Yu Chen
- Chinese Academy of Sciences Key Laboratory of Brain Connectome and Manipulation, Shenzhen Key Laboratory of Translational Research for Brain Diseases, the Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen, China.
- SIAT-HKUST Joint Laboratory for Brain Science, Chinese Academy of Sciences, Shenzhen, China.
- University of Chinese Academy of Sciences, Beijing, China.
- Guangdong Provincial Key Laboratory of Brain Science, Disease and Drug Development, HKUST Shenzhen Research Institute, Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen, China.
| |
Collapse
|
9
|
Tukker AM, Bowman AB. Application of Single Cell Gene Expression Technologies to Neurotoxicology. CURRENT OPINION IN TOXICOLOGY 2024; 37:100458. [PMID: 38617035 PMCID: PMC11008280 DOI: 10.1016/j.cotox.2023.100458] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/16/2024]
Abstract
Neurotoxicological research faces the challenge of linking biological changes resulting from exposures to neuronal function. An additional challenge is understanding cell-type specific differences and selective vulnerabilities of distinct neuronal populations to toxic insults. Single cell RNA-sequencing (scRNA-seq) allows for measurement of the transcriptome of individual cells. This makes it a valuable tool for validating and characterizing cell types present in multicell type samples in complex tissue or cell culture models, but also for understanding how different cell types respond to toxic insults. Pathway analysis of differentially expressed genes can provide in depth insights into underlying cell type-specific mechanisms of neurotoxicity. Toxicological data often has to be translated to outcomes for human health which requires an understanding of inter-species differences. Transcriptomic data aids in understanding these differences, including understanding developmental timelines of different species. We believe that scRNA-seq holds exciting promises for future neurotoxicological research.
Collapse
Affiliation(s)
- Anke M Tukker
- School of Health Sciences, Purdue University, West Lafayette, IN, USA
| | - Aaron B Bowman
- School of Health Sciences, Purdue University, West Lafayette, IN, USA
| |
Collapse
|
10
|
Yang Y, McCullough CG, Seninge L, Guo L, Kwon WJ, Zhang Y, Li NY, Gaddam S, Pan C, Zhen H, Torkelson J, Glass IA, Charville G, Que J, Stuart J, Ding H, Oro A. A Spatiotemporal and Machine-Learning Platform Accelerates the Manufacturing of hPSC-derived Esophageal Mucosa. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.10.24.563664. [PMID: 37961271 PMCID: PMC10634774 DOI: 10.1101/2023.10.24.563664] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/15/2023]
Abstract
Human pluripotent stem cell-derived tissue engineering offers great promise in designer cell-based personalized therapeutics. To harness such potential, a broader approach requires a deeper understanding of tissue-level interactions. We previously developed a manufacturing system for the ectoderm-derived skin epithelium for cell replacement therapy. However, it remains challenging to manufacture the endoderm-derived esophageal epithelium, despite both possessing similar stratified structure. Here we employ single cell and spatial technologies to generate a spatiotemporal multi-omics cell atlas for human esophageal development. We illuminate the cellular diversity, dynamics and signal communications for the developing esophageal epithelium and stroma. Using the machine-learning based Manatee, we prioritize the combinations of candidate human developmental signals for in vitro derivation of esophageal basal cells. Functional validation of the Manatee predictions leads to a clinically-compatible system for manufacturing human esophageal mucosa. Our approach creates a versatile platform to accelerate human tissue manufacturing for future cell replacement therapies to treat human genetic defects and wounds.
Collapse
|
11
|
Fang Q, Wei Y, Zhang Y, Cao W, Yan L, Kong M, Zhu Y, Xu Y, Guo L, Zhang L, Wang W, Yu Y, Sun J, Yang J. Stem cells as potential therapeutics for hearing loss. Front Neurosci 2023; 17:1259889. [PMID: 37746148 PMCID: PMC10512725 DOI: 10.3389/fnins.2023.1259889] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Accepted: 08/23/2023] [Indexed: 09/26/2023] Open
Abstract
Hearing impairment is a global health problem. Stem cell therapy has become a cutting-edge approach to tissue regeneration. In this review, the recent advances in stem cell therapy for hearing loss have been discussed. Nanomaterials can modulate the stem cell microenvironment to augment the therapeutic effects further. The potential of combining nanomaterials with stem cells for repairing and regenerating damaged inner ear hair cells (HCs) and spiral ganglion neurons (SGNs) has also been discussed. Stem cell-derived exosomes can contribute to the repair and regeneration of damaged tissue, and the research progress on exosome-based hearing loss treatment has been summarized as well. Despite stem cell therapy's technical and practical limitations, the findings reported so far are promising and warrant further investigation for eventual clinical translation.
Collapse
Affiliation(s)
- Qiaojun Fang
- Department of Otolaryngology-Head and Neck Surgery, the Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
- School of Life Sciences and Technology, Southeast University, Nanjing, China
| | - Yongjie Wei
- Department of Otolaryngology-Head and Neck Surgery, the Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Yuhua Zhang
- Department of Otolaryngology-Head and Neck Surgery, the Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Wei Cao
- Department of Otolaryngology-Head and Neck Surgery, the Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Lin Yan
- Department of Otolaryngology-Head and Neck Surgery, the Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Mengdie Kong
- School of Life Sciences and Technology, Southeast University, Nanjing, China
| | - Yongjun Zhu
- Department of Otolaryngology-Head and Neck Surgery, the Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Yan Xu
- Department of Otolaryngology-Head and Neck Surgery, the Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Lingna Guo
- Department of Otolaryngology-Head and Neck Surgery, the Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Lei Zhang
- Department of Otolaryngology-Head and Neck Surgery, the Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Weiqing Wang
- Department of Otolaryngology-Head and Neck Surgery, the Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Yafeng Yu
- Department of Otolaryngology-Head and Neck Surgery, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Jingwu Sun
- Department of Otolaryngology-Head and Neck Surgery, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
| | - Jianming Yang
- Department of Otolaryngology-Head and Neck Surgery, the Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| |
Collapse
|
12
|
Zhu F, Nie G, Liu C. Engineered biomaterials in stem cell-based regenerative medicine. LIFE MEDICINE 2023; 2:lnad027. [PMID: 39872549 PMCID: PMC11749850 DOI: 10.1093/lifemedi/lnad027] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/18/2023] [Accepted: 07/17/2023] [Indexed: 01/30/2025]
Abstract
Stem cell-based regenerative therapies, which harness the self-renewal and differentiation properties of stem cells, have been in the spotlight due to their widespread applications in treating degenerative, aging, and other, generally intractable diseases. Therapeutically effective hematopoietic stem cells, mesenchymal stem cells, embryonic stem cells, and induced pluripotent stem cells have been used in numerous basic and translational studies with exciting results. However, pre-/post-transplantation issues of poor cell survival and retention, uncontrolled differentiation, and insufficient numbers of cells engrafted into host tissues are the major challenges in stem cell-based regenerative therapies. Engineered biomaterials have adjustable biochemical and biophysical properties that significantly affect cell behaviors, such as cell engraftment, survival, migration, and differentiation outcomes, thereby enhancing the engraftment of implanted stem cells and guiding tissue regeneration. Therefore, the combination of stem cell biology with bioengineered materials is a promising strategy to improve the therapeutic outcomes of stem cell-based regenerative therapy. In this review, we summarize the advances in the modulation of behaviors of stem cells via engineered biomaterials. We then present different approaches to harnessing bioengineered materials to enhance the transplantation of stem cells. Finally, we will provide future directions in regenerative therapy using stem cells.
Collapse
Affiliation(s)
- Fei Zhu
- CAS Key Laboratory for Biomedical Effects of Nanomaterials & Nanosafety, CAS Centre for Excellence in Nanoscience, National Centre for Nanoscience and Technology, Beijing 100190, China
| | - Guangjun Nie
- CAS Key Laboratory for Biomedical Effects of Nanomaterials & Nanosafety, CAS Centre for Excellence in Nanoscience, National Centre for Nanoscience and Technology, Beijing 100190, China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Changsheng Liu
- Key Laboratory for Ultrafine Materials of Ministry of Education, East China University of Science and Technology, Shanghai 200237, China
| |
Collapse
|
13
|
Crawford AJ, Forjaz A, Bhorkar I, Roy T, Schell D, Queiroga V, Ren K, Kramer D, Bons J, Huang W, Russo GC, Lee MH, Schilling B, Wu PH, Shih IM, Wang TL, Kiemen A, Wirtz D. Precision-engineered biomimetics: the human fallopian tube. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.06.06.543923. [PMID: 37333379 PMCID: PMC10274705 DOI: 10.1101/2023.06.06.543923] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/20/2023]
Abstract
The fallopian tube has an essential role in several physiological and pathological processes from pregnancy to ovarian cancer. However, there are no biologically relevant models to study its pathophysiology. The state-of-the-art organoid model has been compared to two-dimensional tissue sections and molecularly assessed providing only cursory analyses of the model's accuracy. We developed a novel multi-compartment organoid model of the human fallopian tube that was meticulously tuned to reflect the compartmentalization and heterogeneity of the tissue's composition. We validated this organoid's molecular expression patterns, cilia-driven transport function, and structural accuracy through a highly iterative platform wherein organoids are compared to a three-dimensional, single-cell resolution reference map of a healthy, transplantation-quality human fallopian tube. This organoid model was precision-engineered to match the human microanatomy. One sentence summary Tunable organoid modeling and CODA architectural quantification in tandem help design a tissue-validated organoid model.
Collapse
|
14
|
Cell–scaffold interactions in tissue engineering for oral and craniofacial reconstruction. Bioact Mater 2023; 23:16-44. [DOI: 10.1016/j.bioactmat.2022.10.029] [Citation(s) in RCA: 31] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 10/22/2022] [Accepted: 10/30/2022] [Indexed: 11/09/2022] Open
|
15
|
Discovery of endosomalytic cell-penetrating peptides based on bacterial membrane-targeting sequences. Bioorg Chem 2023; 134:106424. [PMID: 36868126 DOI: 10.1016/j.bioorg.2023.106424] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Revised: 02/05/2023] [Accepted: 02/12/2023] [Indexed: 02/17/2023]
Abstract
Cell-penetrating peptides (CPPs) are prominent scaffolds for drug developments and related research, particularly the endocytic delivery of biomacromolecules. Effective cargo release from endosomes prior to lysosomal degradation is a crucial step, where the rational design and selection of CPPs remains a challenge and calls for deeper mechanistic understandings. Here, we have investigated a strategy of designing CPPs that selectively disrupt endosomal membranes based on bacterial membrane targeting sequences (MTSs). Six synthesized MTS peptides all exhibit cell-penetrating abilities, among which two d-peptides (d-EcMTS and d-TpMTS) are able to escape from endosomes and localize at ER after entering the cell. The utility of this strategy has been demonstrated by the intracellular delivery of green fluorescent protein (GFP). Together, these results suggest that the large pool of bacterial MTSs may be a rich source for the development of novel CPPs.
Collapse
|
16
|
Demonstration of intracellular real-time molecular quantification via FRET-enhanced optical microcavity. Nat Commun 2022; 13:6685. [PMID: 36335126 PMCID: PMC9637138 DOI: 10.1038/s41467-022-34547-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2020] [Accepted: 10/24/2022] [Indexed: 11/08/2022] Open
Abstract
Single cell analysis is crucial for elucidating cellular diversity and heterogeneity as well as for medical diagnostics operating at the ultimate detection limit. Although superbly sensitive biosensors have been developed using the strongly enhanced evanescent fields provided by optical microcavities, real-time quantification of intracellular molecules remains challenging due to the extreme low quantity and limitations of the current techniques. Here, we introduce an active-mode optical microcavity sensing stage with enhanced sensitivity that operates via Förster resonant energy transferring (FRET) mechanism. The mutual effects of optical microcavity and FRET greatly enhances the sensing performance by four orders of magnitude compared to pure Whispering gallery mode (WGM) microcavity sensing system. We demonstrate distinct sensing mechanism of FRET-WGM from pure WGM. Predicted lasing wavelengths of both donor and acceptor by theoretical calculations are in perfect agreement with the experimental data. The proposed sensor enables quantitative molecular analysis at single cell resolution, and real-time monitoring of intracellular molecules over extended periods while maintaining the cell viability. By achieving high sensitivity at single cell level, our approach provides a path toward FRET-enhanced real-time quantitative analysis of intracellular molecules.
Collapse
|
17
|
Habibey R, Rojo Arias JE, Striebel J, Busskamp V. Microfluidics for Neuronal Cell and Circuit Engineering. Chem Rev 2022; 122:14842-14880. [PMID: 36070858 PMCID: PMC9523714 DOI: 10.1021/acs.chemrev.2c00212] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Indexed: 02/07/2023]
Abstract
The widespread adoption of microfluidic devices among the neuroscience and neurobiology communities has enabled addressing a broad range of questions at the molecular, cellular, circuit, and system levels. Here, we review biomedical engineering approaches that harness the power of microfluidics for bottom-up generation of neuronal cell types and for the assembly and analysis of neural circuits. Microfluidics-based approaches are instrumental to generate the knowledge necessary for the derivation of diverse neuronal cell types from human pluripotent stem cells, as they enable the isolation and subsequent examination of individual neurons of interest. Moreover, microfluidic devices allow to engineer neural circuits with specific orientations and directionality by providing control over neuronal cell polarity and permitting the isolation of axons in individual microchannels. Similarly, the use of microfluidic chips enables the construction not only of 2D but also of 3D brain, retinal, and peripheral nervous system model circuits. Such brain-on-a-chip and organoid-on-a-chip technologies are promising platforms for studying these organs as they closely recapitulate some aspects of in vivo biological processes. Microfluidic 3D neuronal models, together with 2D in vitro systems, are widely used in many applications ranging from drug development and toxicology studies to neurological disease modeling and personalized medicine. Altogether, microfluidics provide researchers with powerful systems that complement and partially replace animal models.
Collapse
Affiliation(s)
- Rouhollah Habibey
- Department
of Ophthalmology, Universitäts-Augenklinik
Bonn, University of Bonn, Ernst-Abbe-Straße 2, D-53127 Bonn, Germany
| | - Jesús Eduardo Rojo Arias
- Wellcome—MRC
Cambridge Stem Cell Institute, Jeffrey Cheah Biomedical Centre, Cambridge
Biomedical Campus, University of Cambridge, Cambridge CB2 0AW, United Kingdom
| | - Johannes Striebel
- Department
of Ophthalmology, Universitäts-Augenklinik
Bonn, University of Bonn, Ernst-Abbe-Straße 2, D-53127 Bonn, Germany
| | - Volker Busskamp
- Department
of Ophthalmology, Universitäts-Augenklinik
Bonn, University of Bonn, Ernst-Abbe-Straße 2, D-53127 Bonn, Germany
| |
Collapse
|
18
|
Tang W, Wang H, He H. Protocol to photoactivate adipose-derived stem cell differentiation using a tightly-focused femtosecond laser. STAR Protoc 2022; 3:101574. [PMID: 35880123 PMCID: PMC9307679 DOI: 10.1016/j.xpro.2022.101574] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
The technology to induce stem cell differentiation is of great importance in life science, stem cell therapy, and regenerative medicine. Here, we detail steps to noninvasively activate stem cell differentiation in vitro and in vivo using a tightly focused femtosecond laser. We describe how a single-time transient photoactivation can initiate differentiation without any gene engineering, exogenous substances, or physical contact. This protocol enables the differentiation of adipose-derived stem cells to osteoblasts in vitro and cerebellar granule neuron progenitors to granule neurons in vivo. For complete details on the use and execution of this protocol, please refer to Tang et al. (2022). An easy-to-build optical system to provide transient noninvasive photoactivation All-optical noninvasive approach without exogenous substances or physical contact Differentiation of several stem cell models in vitro and in vivo
Publisher’s note: Undertaking any experimental protocol requires adherence to local institutional guidelines for laboratory safety and ethics.
Collapse
Affiliation(s)
- Wanyi Tang
- School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200030, P.R. China.
| | - Haipeng Wang
- School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200030, P.R. China
| | - Hao He
- School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200030, P.R. China.
| |
Collapse
|
19
|
Kim W, Park E, Yoo HS, Park J, Jung YM, Park JH. Recent Advances in Monitoring Stem Cell Status and Differentiation Using Nano-Biosensing Technologies. NANOMATERIALS (BASEL, SWITZERLAND) 2022; 12:2934. [PMID: 36079970 PMCID: PMC9457759 DOI: 10.3390/nano12172934] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Revised: 08/22/2022] [Accepted: 08/23/2022] [Indexed: 05/14/2023]
Abstract
In regenerative medicine, cell therapies using various stem cells have received attention as an alternative to overcome the limitations of existing therapeutic methods. Clinical applications of stem cells require the identification of characteristics at the single-cell level and continuous monitoring during expansion and differentiation. In this review, we recapitulate the application of various stem cells used in regenerative medicine and the latest technological advances in monitoring the differentiation process of stem cells. Single-cell RNA sequencing capable of profiling the expression of many genes at the single-cell level provides a new opportunity to analyze stem cell heterogeneity and to specify molecular markers related to the branching of differentiation lineages. However, this method is destructive and distorted. In addition, the differentiation process of a particular cell cannot be continuously tracked. Therefore, several spectroscopic methods have been developed to overcome these limitations. In particular, the application of Raman spectroscopy to measure the intrinsic vibration spectrum of molecules has been proposed as a powerful method that enables continuous monitoring of biochemical changes in the process of the differentiation of stem cells. This review provides a comprehensive overview of current analytical methods employed for stem cell engineering and future perspectives of nano-biosensing technologies as a platform for the in situ monitoring of stem cell status and differentiation.
Collapse
Affiliation(s)
- Wijin Kim
- Department of Biomedical Science, Kangwon National University, Chuncheon 24341, Gangwon-do, Korea
| | - Eungyeong Park
- Department of Chemistry, Kangwon National University, Chuncheon 24341, Gangwon-do, Korea
| | - Hyuk Sang Yoo
- Department of Biomedical Science, Kangwon National University, Chuncheon 24341, Gangwon-do, Korea
- Kangwon Radiation Convergence Research Support Center, Kangwon National University, Chuncheon 24341, Gangwon-do, Korea
| | - Jongmin Park
- Department of Chemistry, Kangwon National University, Chuncheon 24341, Gangwon-do, Korea
| | - Young Mee Jung
- Department of Chemistry, Kangwon National University, Chuncheon 24341, Gangwon-do, Korea
- Kangwon Radiation Convergence Research Support Center, Kangwon National University, Chuncheon 24341, Gangwon-do, Korea
| | - Ju Hyun Park
- Department of Biomedical Science, Kangwon National University, Chuncheon 24341, Gangwon-do, Korea
| |
Collapse
|
20
|
Alsinet C, Primo MN, Lorenzi V, Bello E, Kelava I, Jones CP, Vilarrasa-Blasi R, Sancho-Serra C, Knights AJ, Park JE, Wyspianska BS, Trynka G, Tough DF, Bassett A, Gaffney DJ, Alvarez-Errico D, Vento-Tormo R. Robust temporal map of human in vitro myelopoiesis using single-cell genomics. Nat Commun 2022; 13:2885. [PMID: 35610203 PMCID: PMC9130280 DOI: 10.1038/s41467-022-30557-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Accepted: 05/06/2022] [Indexed: 11/09/2022] Open
Abstract
Myeloid cells are central to homeostasis and immunity. Characterising in vitro myelopoiesis protocols is imperative for their use in research, immunotherapies, and understanding human myelopoiesis. Here, we generate a >470K cells molecular map of human induced pluripotent stem cells (iPSC) differentiation into macrophages. Integration with in vivo single-cell atlases shows in vitro differentiation recapitulates features of yolk sac hematopoiesis, before definitive hematopoietic stem cells (HSC) emerge. The diversity of myeloid cells generated, including mast cells and monocytes, suggests that HSC-independent hematopoiesis can produce multiple myeloid lineages. We uncover poorly described myeloid progenitors and conservation between in vivo and in vitro regulatory programs. Additionally, we develop a protocol to produce iPSC-derived dendritic cells (DC) resembling cDC2. Using CRISPR/Cas9 knock-outs, we validate the effects of key transcription factors in macrophage and DC ontogeny. This roadmap of myeloid differentiation is an important resource for investigating human fetal hematopoiesis and new therapeutic opportunities.
Collapse
Affiliation(s)
- Clara Alsinet
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, CB10 1SA, UK. .,Open Targets, Wellcome Genome Campus, Hinxton, Cambridge, CB10 1SA, UK.
| | - Maria Nascimento Primo
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, CB10 1SA, UK.,Open Targets, Wellcome Genome Campus, Hinxton, Cambridge, CB10 1SA, UK
| | - Valentina Lorenzi
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, CB10 1SA, UK
| | - Erica Bello
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, CB10 1SA, UK.,Open Targets, Wellcome Genome Campus, Hinxton, Cambridge, CB10 1SA, UK
| | - Iva Kelava
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, CB10 1SA, UK
| | - Carla P Jones
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, CB10 1SA, UK
| | | | - Carmen Sancho-Serra
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, CB10 1SA, UK
| | - Andrew J Knights
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, CB10 1SA, UK
| | - Jong-Eun Park
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, 34141, Korea
| | - Beata S Wyspianska
- Open Targets, Wellcome Genome Campus, Hinxton, Cambridge, CB10 1SA, UK.,Immunology Research Unit, Medicines Research Centre, GlaxoSmithKline, Stevenage, SG1 2NY, UK
| | - Gosia Trynka
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, CB10 1SA, UK.,Open Targets, Wellcome Genome Campus, Hinxton, Cambridge, CB10 1SA, UK
| | - David F Tough
- Immunology Research Unit, Medicines Research Centre, GlaxoSmithKline, Stevenage, SG1 2NY, UK
| | - Andrew Bassett
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, CB10 1SA, UK.,Open Targets, Wellcome Genome Campus, Hinxton, Cambridge, CB10 1SA, UK
| | - Daniel J Gaffney
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, CB10 1SA, UK.
| | - Damiana Alvarez-Errico
- Josep Carreras Leukaemia Research Institute (IJC), Badalona, 08916, Barcelona, Catalonia, Spain.
| | - Roser Vento-Tormo
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, CB10 1SA, UK.
| |
Collapse
|
21
|
Marečková M, Massalha H, Lorenzi V, Vento-Tormo R. Mapping Human Reproduction with Single-Cell Genomics. Annu Rev Genomics Hum Genet 2022; 23:523-547. [PMID: 35567278 DOI: 10.1146/annurev-genom-120121-114415] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
The trillions of cells in the human body develop as a result of the fusion of two extremely specialized cells: an oocyte and a sperm. This process is essential for the continuation of our species, as it ensures that parental genetic information is mixed and passed on from generation to generation. In addition to producing oocytes, the female reproductive system must provide the environment for the appropriate development of the fetus until birth. New genomic and computational tools offer unique opportunities to study the tight spatiotemporal regulatory mechanisms that are required for the cycle of human reproduction. This review explores how single-cell technologies have been used to build cellular atlases of the human reproductive system across the life span and how these maps have proven useful to better understand reproductive pathologies and dissect the heterogeneity of in vitro model systems. Expected final online publication date for the Annual Review of Genomics and Human Genetics, Volume 23 is October 2022. Please see http://www.annualreviews.org/page/journal/pubdates for revised estimates.
Collapse
Affiliation(s)
- Magda Marečková
- Wellcome Sanger Institute, Cambridge, United Kingdom; .,Nuffield Department of Women's and Reproductive Health, University of Oxford, Oxford, United Kingdom;
| | - Hassan Massalha
- Wellcome Sanger Institute, Cambridge, United Kingdom; .,Theory of Condensed Matter Group, Cavendish Laboratory, University of Cambridge, Cambridge, United Kingdom
| | | | | |
Collapse
|
22
|
Tang W, Wang H, Zhao X, Liu S, Kong SK, Ho A, Chen T, Feng H, He H. Stem cell differentiation with consistent lineage commitment induced by a flash of ultrafast-laser activation in vitro and in vivo. Cell Rep 2022; 38:110486. [PMID: 35263591 DOI: 10.1016/j.celrep.2022.110486] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Revised: 12/06/2021] [Accepted: 02/14/2022] [Indexed: 11/03/2022] Open
Abstract
Recent technological advancements on stem cell differentiation induction have been making great progress in stem cell research, regenerative medicine, and therapeutic applications. However, the risk of off-target differentiation limits the wide application of stem cell therapy strategies. Here, we report a non-invasive all-optical strategy to induce stem cell differentiation in vitro and in vivo that activates individual target stem cells in situ by delivering a transient 100-ms irradiation of a tightly focused femtosecond laser to a submicron cytoplasmic region of primary adipose-derived stem cells (ADSCs). The ADSCs differentiate to osteoblasts with stable lineage commitment that cannot further transdifferentiate because of simultaneous initiation of multiple signaling pathways through specific Ca2+ kinetic patterns. This method can work in vivo to direct mouse cerebellar granule neuron progenitors to granule neurons in intact mouse cerebellums through the skull. Hence, this optical method without any genetic manipulations or exogenous biomaterials holds promising potential in biomedical research and cell-based therapies.
Collapse
Affiliation(s)
- Wanyi Tang
- School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200030, P.R. China
| | - Haipeng Wang
- School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200030, P.R. China
| | - Xiaohui Zhao
- School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200030, P.R. China
| | - Shiyue Liu
- School of Life Sciences, The Chinese University of Hong Kong, Hong Kong 999077, P.R. China
| | - Siu Kai Kong
- School of Life Sciences, The Chinese University of Hong Kong, Hong Kong 999077, P.R. China
| | - Aaron Ho
- Department of Biomedical Engineering, The Chinese University of Hong Kong, Hong Kong 999077, P.R. China
| | - Tunan Chen
- Institute of Neurosurgery, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing 400038, P.R. China
| | - Hua Feng
- Institute of Neurosurgery, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing 400038, P.R. China
| | - Hao He
- School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200030, P.R. China.
| |
Collapse
|
23
|
Barhouse PS, Andrade MJ, Smith Q. Home Away From Home: Bioengineering Advancements to Mimic the Developmental and Adult Stem Cell Niche. FRONTIERS IN CHEMICAL ENGINEERING 2022. [DOI: 10.3389/fceng.2022.832754] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
The inherent self-organizing capacity of pluripotent and adult stem cell populations has advanced our fundamental understanding of processes that drive human development, homeostasis, regeneration, and disease progression. Translating these principles into in vitro model systems has been achieved with the advent of organoid technology, driving innovation to harness patient-specific, cell-laden regenerative constructs that can be engineered to augment or replace diseased tissue. While developmental organization and regenerative adult stem cell niches are tightly regulated in vivo, in vitro analogs lack defined architecture and presentation of physicochemical cues, leading to the unhindered arrangement of mini-tissues that lack complete physiological mimicry. This review aims to highlight the recent integrative engineering approaches that elicit spatio-temporal control of the extracellular niche to direct the structural and functional maturation of pluripotent and adult stem cell derivatives. While the advances presented here leverage multi-pronged strategies ranging from synthetic biology to microfabrication technologies, the methods converge on recreating the biochemical and biophysical milieu of the native tissue to be modeled or regenerated.
Collapse
|
24
|
Abstract
Induced pluripotent stem cell (iPSC)-derived organoids provide models to study human organ development. Single-cell transcriptomics enable highly resolved descriptions of cell states within these systems; however, approaches are needed to directly measure lineage relationships. Here we establish iTracer, a lineage recorder that combines reporter barcodes with inducible CRISPR-Cas9 scarring and is compatible with single-cell and spatial transcriptomics. We apply iTracer to explore clonality and lineage dynamics during cerebral organoid development and identify a time window of fate restriction as well as variation in neurogenic dynamics between progenitor neuron families. We also establish long-term four-dimensional light-sheet microscopy for spatial lineage recording in cerebral organoids and confirm regional clonality in the developing neuroepithelium. We incorporate gene perturbation (iTracer-perturb) and assess the effect of mosaic TSC2 mutations on cerebral organoid development. Our data shed light on how lineages and fates are established during cerebral organoid formation. More broadly, our techniques can be adapted in any iPSC-derived culture system to dissect lineage alterations during normal or perturbed development.
Collapse
|
25
|
Sekine K. Human Organoid and Supporting Technologies for Cancer and Toxicological Research. Front Genet 2021; 12:759366. [PMID: 34745227 PMCID: PMC8569236 DOI: 10.3389/fgene.2021.759366] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Accepted: 10/06/2021] [Indexed: 11/16/2022] Open
Abstract
Recent progress in the field of organoid-based cell culture systems has enabled the use of patient-derived cells in conditions that resemble those in cancer tissue, which are better than two-dimensional (2D) cultured cell lines. In particular, organoids allow human cancer cells to be handled in conditions that resemble those in cancer tissue, resulting in more efficient establishment of cells compared with 2D cultured cell lines, thus enabling the use of multiple patient-derived cells with cells from different genetic background, in keeping with the heterogeneity of the cells. One of the most valuable points of using organoids is that human cells from either healthy or cancerous tissue can be used. Using genome editing technology such as clustered regularly interspaced short palindromic repeats (CRISPR)/CRISPR-associated protein, organoid genomes can be modified to, for example, cancer-prone genomes. The normal, cancer, or genome-modified organoids can be used to evaluate whether chemicals have genotoxic or non-genotoxic carcinogenic activity by evaluating the cancer incidence, cancer progression, and cancer metastasis. In this review, the organoid technology and the accompanying technologies were summarized and the advantages of organoid-based toxicology and its application to pancreatic cancer study were discussed.
Collapse
Affiliation(s)
- Keisuke Sekine
- Laboratory of Cancer Cell Systems, National Cancer Center Research Institute, Tokyo, Japan
| |
Collapse
|
26
|
Lafuente-Gracia L, Borgiani E, Nasello G, Geris L. Towards in silico Models of the Inflammatory Response in Bone Fracture Healing. Front Bioeng Biotechnol 2021; 9:703725. [PMID: 34660547 PMCID: PMC8514728 DOI: 10.3389/fbioe.2021.703725] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Accepted: 09/07/2021] [Indexed: 12/21/2022] Open
Abstract
In silico modeling is a powerful strategy to investigate the biological events occurring at tissue, cellular and subcellular level during bone fracture healing. However, most current models do not consider the impact of the inflammatory response on the later stages of bone repair. Indeed, as initiator of the healing process, this early phase can alter the regenerative outcome: if the inflammatory response is too strongly down- or upregulated, the fracture can result in a non-union. This review covers the fundamental information on fracture healing, in silico modeling and experimental validation. It starts with a description of the biology of fracture healing, paying particular attention to the inflammatory phase and its cellular and subcellular components. We then discuss the current state-of-the-art regarding in silico models of the immune response in different tissues as well as the bone regeneration process at the later stages of fracture healing. Combining the aforementioned biological and computational state-of-the-art, continuous, discrete and hybrid modeling technologies are discussed in light of their suitability to capture adequately the multiscale course of the inflammatory phase and its overall role in the healing outcome. Both in the establishment of models as in their validation step, experimental data is required. Hence, this review provides an overview of the different in vitro and in vivo set-ups that can be used to quantify cell- and tissue-scale properties and provide necessary input for model credibility assessment. In conclusion, this review aims to provide hands-on guidance for scientists interested in building in silico models as an additional tool to investigate the critical role of the inflammatory phase in bone regeneration.
Collapse
Affiliation(s)
- Laura Lafuente-Gracia
- Biomechanics Section, Department of Mechanical Engineering, KU Leuven, Leuven, Belgium.,Prometheus: Division of Skeletal Tissue Engineering, KU Leuven, Leuven, Belgium
| | - Edoardo Borgiani
- Biomechanics Section, Department of Mechanical Engineering, KU Leuven, Leuven, Belgium.,Prometheus: Division of Skeletal Tissue Engineering, KU Leuven, Leuven, Belgium.,Biomechanics Research Unit, GIGA in silico Medicine, University of Liège, Liège, Belgium
| | - Gabriele Nasello
- Biomechanics Section, Department of Mechanical Engineering, KU Leuven, Leuven, Belgium.,Prometheus: Division of Skeletal Tissue Engineering, KU Leuven, Leuven, Belgium.,Skeletal Biology and Engineering Research Center, KU Leuven, Leuven, Belgium
| | - Liesbet Geris
- Biomechanics Section, Department of Mechanical Engineering, KU Leuven, Leuven, Belgium.,Prometheus: Division of Skeletal Tissue Engineering, KU Leuven, Leuven, Belgium.,Biomechanics Research Unit, GIGA in silico Medicine, University of Liège, Liège, Belgium.,Skeletal Biology and Engineering Research Center, KU Leuven, Leuven, Belgium
| |
Collapse
|
27
|
Lin HC, He Z, Ebert S, Schörnig M, Santel M, Nikolova MT, Weigert A, Hevers W, Kasri NN, Taverna E, Camp JG, Treutlein B. NGN2 induces diverse neuron types from human pluripotency. Stem Cell Reports 2021; 16:2118-2127. [PMID: 34358451 PMCID: PMC8452516 DOI: 10.1016/j.stemcr.2021.07.006] [Citation(s) in RCA: 57] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Revised: 07/05/2021] [Accepted: 07/06/2021] [Indexed: 01/22/2023] Open
Abstract
Human neurons engineered from induced pluripotent stem cells (iPSCs) through neurogenin 2 (NGN2) overexpression are widely used to study neuronal differentiation mechanisms and to model neurological diseases. However, the differentiation paths and heterogeneity of emerged neurons have not been fully explored. Here, we used single-cell transcriptomics to dissect the cell states that emerge during NGN2 overexpression across a time course from pluripotency to neuron functional maturation. We find a substantial molecular heterogeneity in the neuron types generated, with at least two populations that express genes associated with neurons of the peripheral nervous system. Neuron heterogeneity is observed across multiple iPSC clones and lines from different individuals. We find that neuron fate acquisition is sensitive to NGN2 expression level and the duration of NGN2-forced expression. Our data reveal that NGN2 dosage can regulate neuron fate acquisition, and that NGN2-iN heterogeneity can confound results that are sensitive to neuron type.
Collapse
Affiliation(s)
- Hsiu-Chuan Lin
- Institute of Molecular and Clinical Ophthalmology Basel, Basel, Switzerland; Department of Ophthalmology, University of Basel, Basel, Switzerland
| | - Zhisong He
- Department of Biosystems Science and Engineering, ETH Zürich, Basel, Switzerland
| | - Sebastian Ebert
- Department of Biosystems Science and Engineering, ETH Zürich, Basel, Switzerland; Max Planck Institute for Evolutionary Anthropology, Leipzig, Germany
| | - Maria Schörnig
- Max Planck Institute for Evolutionary Anthropology, Leipzig, Germany
| | - Malgorzata Santel
- Department of Biosystems Science and Engineering, ETH Zürich, Basel, Switzerland
| | - Marina T Nikolova
- Department of Biosystems Science and Engineering, ETH Zürich, Basel, Switzerland
| | - Anne Weigert
- Max Planck Institute for Evolutionary Anthropology, Leipzig, Germany
| | - Wulf Hevers
- Max Planck Institute for Evolutionary Anthropology, Leipzig, Germany
| | - Nael Nadif Kasri
- Department of Human Genetics, Donders Institute for Brain, Cognition, and Behaviour, Radboudumc, Nijmegen, the Netherlands; Department of Cognitive Neuroscience, Donders Institute for Brain, Cognition, and Behaviour, Radboudumc, Nijmegen, the Netherlands
| | - Elena Taverna
- Max Planck Institute for Evolutionary Anthropology, Leipzig, Germany; Human Technopole, Milan, Italy
| | - J Gray Camp
- Institute of Molecular and Clinical Ophthalmology Basel, Basel, Switzerland; Department of Ophthalmology, University of Basel, Basel, Switzerland; Max Planck Institute for Evolutionary Anthropology, Leipzig, Germany.
| | - Barbara Treutlein
- Department of Biosystems Science and Engineering, ETH Zürich, Basel, Switzerland; Max Planck Institute for Evolutionary Anthropology, Leipzig, Germany; Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany.
| |
Collapse
|
28
|
Haniffa M, Taylor D, Linnarsson S, Aronow BJ, Bader GD, Barker RA, Camara PG, Camp JG, Chédotal A, Copp A, Etchevers HC, Giacobini P, Göttgens B, Guo G, Hupalowska A, James KR, Kirby E, Kriegstein A, Lundeberg J, Marioni JC, Meyer KB, Niakan KK, Nilsson M, Olabi B, Pe'er D, Regev A, Rood J, Rozenblatt-Rosen O, Satija R, Teichmann SA, Treutlein B, Vento-Tormo R, Webb S. A roadmap for the Human Developmental Cell Atlas. Nature 2021; 597:196-205. [PMID: 34497388 PMCID: PMC10337595 DOI: 10.1038/s41586-021-03620-1] [Citation(s) in RCA: 107] [Impact Index Per Article: 26.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Accepted: 05/07/2021] [Indexed: 12/12/2022]
Abstract
The Human Developmental Cell Atlas (HDCA) initiative, which is part of the Human Cell Atlas, aims to create a comprehensive reference map of cells during development. This will be critical to understanding normal organogenesis, the effect of mutations, environmental factors and infectious agents on human development, congenital and childhood disorders, and the cellular basis of ageing, cancer and regenerative medicine. Here we outline the HDCA initiative and the challenges of mapping and modelling human development using state-of-the-art technologies to create a reference atlas across gestation. Similar to the Human Genome Project, the HDCA will integrate the output from a growing community of scientists who are mapping human development into a unified atlas. We describe the early milestones that have been achieved and the use of human stem-cell-derived cultures, organoids and animal models to inform the HDCA, especially for prenatal tissues that are hard to acquire. Finally, we provide a roadmap towards a complete atlas of human development.
Collapse
Affiliation(s)
- Muzlifah Haniffa
- Biosciences Institute, Newcastle University, Newcastle upon Tyne, UK.
- Wellcome Sanger Institute, Hinxton, UK.
- Department of Dermatology and NIHR Newcastle Biomedical Research Centre, Newcastle Hospitals NHS Foundation Trust, Newcastle upon Tyne, UK.
| | - Deanne Taylor
- Department of Biomedical and Health Informatics (DBHi), The Children's Hospital of Philadelphia, Philadelphia, PA, USA
- Department of Pediatrics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Sten Linnarsson
- Division of Molecular Neurobiology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Bruce J Aronow
- Division of Developmental Biology and Biomedical Informatics, Cincinnati Children's Hospital Medical Centre, Cincinnati, OH, USA
| | - Gary D Bader
- The Donnelly Centre, University of Toronto, Toronto, Ontario, Canada
| | - Roger A Barker
- Wellcome and MRC Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
| | - Pablo G Camara
- Department of Genetics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - J Gray Camp
- Institute of Molecular and Clinical Ophthalmology Basel (IOB), University of Basel, Basel, Switzerland
| | - Alain Chédotal
- INSERM, CNRS, Institut de la Vision, Sorbonne Université, Paris, France
| | - Andrew Copp
- Developmental Biology and Cancer Programme, UCL Great Ormond Street Institute of Child Health, London, UK
| | | | - Paolo Giacobini
- Laboratory of Development and Plasticity of the Neuroendocrine Brain, Inserm, CHU Lille, Lille Neuroscience and Cognition, UMR-S 1172, Université Lille, Lille, France
| | - Berthold Göttgens
- Wellcome and MRC Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK
- Department of Haematology, University of Cambridge, Cambridge, UK
| | - Guoji Guo
- Center for Stem Cell and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou, China
| | - Ania Hupalowska
- Klarman Cell Observatory, Broad Institute of Harvard and MIT, Cambridge, MA, USA
| | | | - Emily Kirby
- Centre of Genomics and Policy, McGill University, Montreal, Quebec, Canada
| | - Arnold Kriegstein
- Department of Neurology, University of California San Francisco (UCSF), San Francisco, CA, USA
| | - Joakim Lundeberg
- Science for Life Laboratory, KTH Royal Institute of Technology, Solna, Sweden
| | - John C Marioni
- Cancer Research Institute UK Cambridge Institute, University of Cambridge, Cambridge, UK
| | | | - Kathy K Niakan
- Francis Crick Institute, London, UK
- Centre for Trophoblast Research, Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
| | - Mats Nilsson
- Science for Life Laboratory, Department of Biochemistry and Biophysics, Stockholm University, Stockholm, Sweden
| | - Bayanne Olabi
- Biosciences Institute, Newcastle University, Newcastle upon Tyne, UK
| | - Dana Pe'er
- Computational and Systems Biology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Aviv Regev
- Klarman Cell Observatory, Broad Institute of Harvard and MIT, Cambridge, MA, USA
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA
- Genentech, South San Francisco, CA, USA
| | - Jennifer Rood
- Klarman Cell Observatory, Broad Institute of Harvard and MIT, Cambridge, MA, USA
| | - Orit Rozenblatt-Rosen
- Klarman Cell Observatory, Broad Institute of Harvard and MIT, Cambridge, MA, USA
- Genentech, South San Francisco, CA, USA
| | - Rahul Satija
- New York Genome Center, New York University, New York, NY, USA
| | - Sarah A Teichmann
- Wellcome Sanger Institute, Hinxton, UK
- Cavendish Laboratory, Department of Physics, University of Cambridge, Cambridge, UK
| | - Barbara Treutlein
- Department of Biosystems Science and Engineering, Eidgenössische Technische Hochschule (ETH) Zurich, Basel, Switzerland
| | | | - Simone Webb
- Biosciences Institute, Newcastle University, Newcastle upon Tyne, UK
| |
Collapse
|
29
|
Lee H, Kim N, Rheem HB, Kim BJ, Park JH, Choi IS. A Decade of Advances in Single-Cell Nanocoating for Mammalian Cells. Adv Healthc Mater 2021; 10:e2100347. [PMID: 33890422 DOI: 10.1002/adhm.202100347] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Revised: 04/06/2021] [Indexed: 12/14/2022]
Abstract
Strategic advances in the single-cell nanocoating of mammalian cells have noticeably been made during the last decade, and many potential applications have been demonstrated. Various cell-coating strategies have been proposed via adaptation of reported methods in the surface sciences and/or materials identification that ensure the sustainability of labile mammalian cells during chemical manipulation. Here an overview of the methodological development and potential applications to the healthcare sector in the nanocoating of mammalian cells made during the last decade is provided. The materials used for the nanocoating are categorized into polymers, hydrogels, polyphenolic compounds, nanoparticles, and minerals, and the corresponding strategies are described under the given set of materials. It also suggests, as a future direction, the creation of the cytospace system that is hierarchically composed of the physically separated but mutually interacting cellular hybrids.
Collapse
Affiliation(s)
- Hojae Lee
- Center for Cell‐Encapsulation Research Department of Chemistry KAIST Daejeon 34141 Korea
| | - Nayoung Kim
- Center for Cell‐Encapsulation Research Department of Chemistry KAIST Daejeon 34141 Korea
| | - Hyeong Bin Rheem
- Center for Cell‐Encapsulation Research Department of Chemistry KAIST Daejeon 34141 Korea
| | - Beom Jin Kim
- Department of Chemistry University of Ulsan Ulsan 44610 Korea
| | - Ji Hun Park
- Department of Science Education Ewha Womans University Seoul 03760 Korea
| | - Insung S. Choi
- Center for Cell‐Encapsulation Research Department of Chemistry KAIST Daejeon 34141 Korea
| |
Collapse
|
30
|
Kandhari N, Kraupner-Taylor CA, Harrison PF, Powell DR, Beilharz TH. The Detection and Bioinformatic Analysis of Alternative 3 ' UTR Isoforms as Potential Cancer Biomarkers. Int J Mol Sci 2021; 22:5322. [PMID: 34070203 PMCID: PMC8158509 DOI: 10.3390/ijms22105322] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2021] [Revised: 05/06/2021] [Accepted: 05/06/2021] [Indexed: 12/17/2022] Open
Abstract
Alternative transcript cleavage and polyadenylation is linked to cancer cell transformation, proliferation and outcome. This has led researchers to develop methods to detect and bioinformatically analyse alternative polyadenylation as potential cancer biomarkers. If incorporated into standard prognostic measures such as gene expression and clinical parameters, these could advance cancer prognostic testing and possibly guide therapy. In this review, we focus on the existing methodologies, both experimental and computational, that have been applied to support the use of alternative polyadenylation as cancer biomarkers.
Collapse
Affiliation(s)
- Nitika Kandhari
- Development and Stem Cells Program, Department of Biochemistry and Molecular Biology, Monash Biomedicine Discovery Institute, Monash University, Melbourne, VIC 3800, Australia; (N.K.); (C.A.K.-T.); (P.F.H.)
| | - Calvin A. Kraupner-Taylor
- Development and Stem Cells Program, Department of Biochemistry and Molecular Biology, Monash Biomedicine Discovery Institute, Monash University, Melbourne, VIC 3800, Australia; (N.K.); (C.A.K.-T.); (P.F.H.)
| | - Paul F. Harrison
- Development and Stem Cells Program, Department of Biochemistry and Molecular Biology, Monash Biomedicine Discovery Institute, Monash University, Melbourne, VIC 3800, Australia; (N.K.); (C.A.K.-T.); (P.F.H.)
- Monash Bioinformatics Platform, Monash University, Melbourne, VIC 3800, Australia;
| | - David R. Powell
- Monash Bioinformatics Platform, Monash University, Melbourne, VIC 3800, Australia;
| | - Traude H. Beilharz
- Development and Stem Cells Program, Department of Biochemistry and Molecular Biology, Monash Biomedicine Discovery Institute, Monash University, Melbourne, VIC 3800, Australia; (N.K.); (C.A.K.-T.); (P.F.H.)
| |
Collapse
|
31
|
Chumduri C, Turco MY. Organoids of the female reproductive tract. J Mol Med (Berl) 2021; 99:531-553. [PMID: 33580825 PMCID: PMC8026429 DOI: 10.1007/s00109-020-02028-0] [Citation(s) in RCA: 50] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Revised: 12/09/2020] [Accepted: 12/14/2020] [Indexed: 02/08/2023]
Abstract
Healthy functioning of the female reproductive tract (FRT) depends on balanced and dynamic regulation by hormones during the menstrual cycle, pregnancy and childbirth. The mucosal epithelial lining of different regions of the FRT-ovaries, fallopian tubes, uterus, cervix and vagina-facilitates the selective transport of gametes and successful transfer of the zygote to the uterus where it implants and pregnancy takes place. It also prevents pathogen entry. Recent developments in three-dimensional (3D) organoid systems from the FRT now provide crucial experimental models that recapitulate the cellular heterogeneity and physiological, anatomical and functional properties of the organ in vitro. In this review, we summarise the state of the art on organoids generated from different regions of the FRT. We discuss the potential applications of these powerful in vitro models to study normal physiology, fertility, infections, diseases, drug discovery and personalised medicine.
Collapse
Affiliation(s)
- Cindrilla Chumduri
- Department of Microbiology, University of Würzburg, Biocenter, Würzburg, Germany.
- Max Planck Institute for Infection Biology, Berlin, Germany.
| | - Margherita Y Turco
- Department of Pathology, University of Cambridge, Cambridge, UK.
- Centre for Trophoblast Research, Cambridge, UK.
| |
Collapse
|
32
|
Lin X, Fang F, Wang C, Kankala RK, Zhou S. Inkjet printing-assisted single-cell microarray on a hydrophobic surface chip for real-time monitoring of enzyme kinetics at single-cell level. Talanta 2021; 225:122019. [DOI: 10.1016/j.talanta.2020.122019] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2020] [Revised: 11/25/2020] [Accepted: 12/12/2020] [Indexed: 12/12/2022]
|
33
|
Papantoniou I, Nilsson Hall G, Loverdou N, Lesage R, Herpelinck T, Mendes L, Geris L. Turning Nature's own processes into design strategies for living bone implant biomanufacturing: a decade of Developmental Engineering. Adv Drug Deliv Rev 2021; 169:22-39. [PMID: 33290762 PMCID: PMC7839840 DOI: 10.1016/j.addr.2020.11.012] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2020] [Revised: 11/20/2020] [Accepted: 11/29/2020] [Indexed: 12/14/2022]
Abstract
A decade after the term developmental engineering (DE) was coined to indicate the use of developmental processes as blueprints for the design and development of engineered living implants, a myriad of proof-of-concept studies demonstrate the potential of this approach in small animal models. This review provides an overview of DE work, focusing on applications in bone regeneration. Enabling technologies allow to quantify the distance between in vitro processes and their developmental counterpart, as well as to design strategies to reduce that distance. By embedding Nature's robust mechanisms of action in engineered constructs, predictive large animal data and subsequent positive clinical outcomes can be gradually achieved. To this end, the development of next generation biofabrication technologies should provide the necessary scale and precision for robust living bone implant biomanufacturing.
Collapse
Affiliation(s)
- Ioannis Papantoniou
- Institute of Chemical Engineering Sciences, Foundation for Research and Technology - Hellas (FORTH), Stadiou street, 26504 Patras, Greece; Skeletal Biology & Engineering Research Center, KU Leuven, Herestraat 49 (813), 3000 Leuven, Belgium; Prometheus, The KU Leuven R&D Division for Skeletal Tissue Engineering, Herestraat 49 (813), 3000 Leuven, Belgium.
| | - Gabriella Nilsson Hall
- Skeletal Biology & Engineering Research Center, KU Leuven, Herestraat 49 (813), 3000 Leuven, Belgium; Prometheus, The KU Leuven R&D Division for Skeletal Tissue Engineering, Herestraat 49 (813), 3000 Leuven, Belgium.
| | - Niki Loverdou
- Prometheus, The KU Leuven R&D Division for Skeletal Tissue Engineering, Herestraat 49 (813), 3000 Leuven, Belgium; GIGA in silico medicine, University of Liège, Avenue de l'Hôpital 11 (B34), 4000 Liège, Belgium; Biomechanics Section, KU Leuven, Celestijnenlaan 300C (2419), 3001 Leuven, Belgium.
| | - Raphaelle Lesage
- Prometheus, The KU Leuven R&D Division for Skeletal Tissue Engineering, Herestraat 49 (813), 3000 Leuven, Belgium; Biomechanics Section, KU Leuven, Celestijnenlaan 300C (2419), 3001 Leuven, Belgium.
| | - Tim Herpelinck
- Skeletal Biology & Engineering Research Center, KU Leuven, Herestraat 49 (813), 3000 Leuven, Belgium; Prometheus, The KU Leuven R&D Division for Skeletal Tissue Engineering, Herestraat 49 (813), 3000 Leuven, Belgium.
| | - Luis Mendes
- Skeletal Biology & Engineering Research Center, KU Leuven, Herestraat 49 (813), 3000 Leuven, Belgium; Prometheus, The KU Leuven R&D Division for Skeletal Tissue Engineering, Herestraat 49 (813), 3000 Leuven, Belgium.
| | - Liesbet Geris
- Skeletal Biology & Engineering Research Center, KU Leuven, Herestraat 49 (813), 3000 Leuven, Belgium; GIGA in silico medicine, University of Liège, Avenue de l'Hôpital 11 (B34), 4000 Liège, Belgium; Prometheus, The KU Leuven R&D Division for Skeletal Tissue Engineering, Herestraat 49 (813), 3000 Leuven, Belgium; Biomechanics Section, KU Leuven, Celestijnenlaan 300C (2419), 3001 Leuven, Belgium.
| |
Collapse
|
34
|
Kim YS, Mikos AG. Emerging strategies in reprogramming and enhancing the fate of mesenchymal stem cells for bone and cartilage tissue engineering. J Control Release 2021; 330:565-574. [DOI: 10.1016/j.jconrel.2020.12.055] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Revised: 12/21/2020] [Accepted: 12/29/2020] [Indexed: 02/06/2023]
|
35
|
Longitudinal single-cell RNA-seq of hESCs-derived retinal organoids. SCIENCE CHINA-LIFE SCIENCES 2021; 64:1661-1676. [PMID: 33521856 DOI: 10.1007/s11427-020-1836-7] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Accepted: 10/13/2020] [Indexed: 12/26/2022]
Abstract
Human retina development involves multiple well-studied signaling pathways that promote the genesis of a wide arrange of different cell types in a complex architectural structure. Human embryonic stem cells (hESCs)-derived retinal organoids could recapitulate the human retinal development. We performed single-cell RNA-seq of retinal organoids from 5 time points (D36, D66, D96, D126, D186) and identified 9 distinct populations of cells. In addition, we analyzed the molecular characteristics of each main population and followed them from genesis to maturity by pseudotime analysis and characterized the cell-cell interactions between different cell types. Interestingly, we identified insulin receptor (INSR) as a specifically expressed receptor involved in the genesis of photoreceptors, and pleiothropin (PTN)-protein tyrosine phosphatase receptor type Z1 (PTPRZ1) as a mediator of a previously unknown interaction between Müller and retinal progenitor cells. Taken together, these findings provide a rich transcriptome-based lineage map for studying human retinal development and modeling developmental disorders in retinal organoids.
Collapse
|
36
|
Resolving Neurodevelopmental and Vision Disorders Using Organoid Single-Cell Multi-omics. Neuron 2020; 107:1000-1013. [PMID: 32970995 DOI: 10.1016/j.neuron.2020.09.001] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Revised: 08/26/2020] [Accepted: 08/31/2020] [Indexed: 12/20/2022]
Abstract
Human organoid models of the central nervous system, including the neural retina, are providing unprecedented opportunities to explore human neurodevelopment and neurodegeneration in controlled culture environments. In this Perspective, we discuss how the single-cell multi-omic toolkit has been used to identify features and limitations of brain and retina organoids and how these tools can be deployed to study congenital brain malformations and vision disorders in organoids. We also address how to improve brain and retina organoid protocols to revolutionize in vitro disease modeling.
Collapse
|
37
|
Abstract
Organoids form through self-organization processes in which initially homogeneous populations of stem cells spontaneously break symmetry and undergo in-vivo-like pattern formation and morphogenesis, though the processes controlling this are poorly characterized. While these in vitro self-organized tissues far exceed the microscopic and functional complexity obtained by current tissue engineering technologies, they are non-physiological in shape and size and have limited function and lifespan. Here, we discuss how engineering efforts for guiding stem-cell-based development at multiple stages can form the basis for the assembly of highly complex and rationally designed self-organizing multicellular systems with increased robustness and physiological relevance.
Collapse
|
38
|
Kucinski I, Gottgens B. Advancing Stem Cell Research through Multimodal Single-Cell Analysis. Cold Spring Harb Perspect Biol 2020; 12:a035725. [PMID: 31932320 PMCID: PMC7328456 DOI: 10.1101/cshperspect.a035725] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Technological advances play a key role in furthering our understanding of stem cell biology, and advancing the prospects of regenerative therapies. Highly parallelized methods, developed in the last decade, can profile DNA, RNA, or proteins in thousands of cells and even capture data across two or more modalities (multiomics). This allows unbiased and precise definition of molecular cell states, thus allowing classification of cell types, tracking of differentiation trajectories, and discovery of underlying mechanisms. Despite being based on destructive techniques, novel experimental and bioinformatic approaches enable embedding and extraction of temporal information, which is essential for deconvolution of complex data and establishing cause and effect relationships. Here, we provide an overview of recent studies pertinent to stem cell biology, followed by an outlook on how further advances in single-cell molecular profiling and computational analysis have the potential to shape the future of both basic and translational research.
Collapse
Affiliation(s)
- Iwo Kucinski
- Wellcome-MRC Cambridge Stem Cell Institute and Department of Haematology, University of Cambridge, Jeffrey Cheah Biomedical Centre, Cambridge Biomedical Campus, Cambridge CB2 0AW, United Kingdom
| | - Berthold Gottgens
- Wellcome-MRC Cambridge Stem Cell Institute and Department of Haematology, University of Cambridge, Jeffrey Cheah Biomedical Centre, Cambridge Biomedical Campus, Cambridge CB2 0AW, United Kingdom
| |
Collapse
|
39
|
Alzamil L, Nikolakopoulou K, Turco MY. Organoid systems to study the human female reproductive tract and pregnancy. Cell Death Differ 2020; 28:35-51. [PMID: 32494027 PMCID: PMC7852529 DOI: 10.1038/s41418-020-0565-5] [Citation(s) in RCA: 73] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2020] [Revised: 04/24/2020] [Accepted: 05/15/2020] [Indexed: 12/21/2022] Open
Abstract
Both the proper functioning of the female reproductive tract (FRT) and normal placental development are essential for women’s health, wellbeing, and pregnancy outcome. The study of the FRT in humans has been challenging due to limitations in the in vitro and in vivo tools available. Recent developments in 3D organoid technology that model the different regions of the FRT include organoids of the ovaries, fallopian tubes, endometrium and cervix, as well as placental trophoblast. These models are opening up new avenues to investigate the normal biology and pathology of the FRT. In this review, we discuss the advances, potential, and limitations of organoid cultures of the human FRT. ■. ![]()
Collapse
Affiliation(s)
- Lama Alzamil
- Department of Pathology, University of Cambridge, Tennis Court Road, Cambridge, CB2 1QP, UK
| | | | - Margherita Y Turco
- Department of Pathology, University of Cambridge, Tennis Court Road, Cambridge, CB2 1QP, UK. .,Centre for Trophoblast Research, Downing Street, Cambridge, CB2 3EG, UK.
| |
Collapse
|
40
|
Kamies R, Martinez-Jimenez CP. Advances of single-cell genomics and epigenomics in human disease: where are we now? Mamm Genome 2020; 31:170-180. [PMID: 32270277 PMCID: PMC7368869 DOI: 10.1007/s00335-020-09834-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2020] [Accepted: 03/28/2020] [Indexed: 02/07/2023]
Abstract
Cellular heterogeneity is revolutionizing the way to study, monitor and dissect complex diseases. This has been possible with the technological and computational advances associated to single-cell genomics and epigenomics. Deeper understanding of cell-to-cell variation and its impact on tissue function will open new avenues for early disease detection, accurate diagnosis and personalized treatments, all together leading to the next generation of health care. This review focuses on the recent discoveries that single-cell genomics and epigenomics have facilitated in the context of human health. It highlights the potential of single-cell omics to further advance the development of personalized treatments and precision medicine in cancer, diabetes and chronic age-related diseases. The promise of single-cell technologies to generate new insights about the differences in function between individual cells is just emerging, and it is paving the way for identifying biomarkers and novel therapeutic targets to tackle age, complex diseases and understand the effect of life style interventions and environmental factors.
Collapse
Affiliation(s)
- Rizqah Kamies
- Helmholtz Pioneer Campus, Helmholtz Zentrum München, 85764 Neuherberg, Germany
| | | |
Collapse
|
41
|
Kanton S, Treutlein B, Camp JG. Single-cell genomic analysis of human cerebral organoids. Methods Cell Biol 2020; 159:229-256. [PMID: 32586444 DOI: 10.1016/bs.mcb.2020.03.013] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Investigating early brain development has previously relied on using primary developing brain tissue or two-dimensional cell culture models. Recently, stem cell-derived three-dimensional cell culture systems, collectively called brain organoids, have been developed that can faithfully recapitulate many aspects of early brain development. Together with the ability to reprogram fibroblast or blood cells into induced pluripotent stem cells from humans with neurodevelopmental disorders, this opens new inroads to study patient-specific brain development in a personalized cell culture model. Studying the transcriptomes and regulatory landscape of single cells within brain organoids presents a major advance to understand cell-type specific features and transient states during development, and to link these states to their underlying regulatory logic at high resolution. In this protocol, we describe how to generate single-cell RNA-seq and ATAC-seq (Assay for Transposase-Accessible Chromatin using sequencing) data from the same suspension of organoid cells and focus on reducing batch effects by multiplexing multiple individuals in one experiment. Moreover, we outline basic data processing, analysis, and strategies to correct for batch effects, to account for variability in organoids and for integrating gene expression and open chromatin data.
Collapse
Affiliation(s)
- Sabina Kanton
- Department of Evolutionary Genetics, Max Planck Institute for Evolutionary Anthropology, Leipzig, Germany
| | - Barbara Treutlein
- Department of Evolutionary Genetics, Max Planck Institute for Evolutionary Anthropology, Leipzig, Germany; Department of Biosystems Science and Engineering, ETH Zürich, Basel, Switzerland
| | - J Gray Camp
- Institute of Clinical Ophthalmology (IOB), University of Basel, Basel, Switzerland; Department of Ophthalmology, University of Basel, Basel, Switzerland.
| |
Collapse
|
42
|
Camp JG, Platt R, Treutlein B. Mapping human cell phenotypes to genotypes with single-cell genomics. Science 2020; 365:1401-1405. [PMID: 31604266 DOI: 10.1126/science.aax6648] [Citation(s) in RCA: 66] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The cumulative activity of all of the body's cells, with their myriad interactions, life histories, and environmental experiences, gives rise to a condition that is distinctly human and specific to each individual. It is an enduring goal to catalog our human cell types, to understand how they develop, how they vary between individuals, and how they fail in disease. Single-cell genomics has revolutionized this endeavor because sequencing-based methods provide a means to quantitatively annotate cell states on the basis of high-information content and high-throughput measurements. Together with advances in stem cell biology and gene editing, we are in the midst of a fascinating journey to understand the cellular phenotypes that compose human bodies and how the human genome is used to build and maintain each cell. Here, we will review recent advances into how single-cell genomics is being used to develop personalized phenotyping strategies that cross subcellular, cellular, and tissue scales to link our genome to our cumulative cellular phenotypes.
Collapse
Affiliation(s)
- J Gray Camp
- Institute of Molecular and Clinical Ophthalmology Basel, Basel, Switzerland
| | - Randall Platt
- Department of Biosystems Science and Engineering, ETH Zürich, Basel, Switzerland
| | - Barbara Treutlein
- Department of Biosystems Science and Engineering, ETH Zürich, Basel, Switzerland.
| |
Collapse
|
43
|
Differentiation of human pluripotent stem cells toward pharyngeal endoderm derivatives: Current status and potential. Curr Top Dev Biol 2020; 138:175-208. [PMID: 32220297 DOI: 10.1016/bs.ctdb.2020.01.004] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/04/2022]
Abstract
The pharyngeal apparatus, a transient embryological structure, includes diverse cells from all three germ layers that ultimately contribute to a variety of adult tissues. In particular, pharyngeal endoderm produces cells of the inner ear, palatine tonsils, the thymus, parathyroid and thyroid glands, and ultimobranchial bodies. Each of these structures and organs contribute to vital human physiological processes, including central immune tolerance (thymus) and metabolic homeostasis (parathyroid and thyroid glands, and ultimobranchial bodies). Thus, improper development or damage to pharyngeal endoderm derivatives leads to complicated and severe human maladies, such as autoimmunity, immunodeficiency, hypothyroidism, and/or hypoparathyroidism. To study and treat such diseases, we can utilize human pluripotent stem cells (hPSCs), which differentiate into functionally mature cells in vitro given the proper developmental signals. Here, we discuss current efforts regarding the directed differentiation of hPSCs toward pharyngeal endoderm derivatives. We further discuss model system and therapeutic applications of pharyngeal endoderm cell types produced from hPSCs. Finally, we provide suggestions for improving hPSC differentiation approaches to pharyngeal endoderm derivatives with emphasis on current single cell-omics and 3D culture system technologies.
Collapse
|
44
|
Lange C, Rost F, Machate A, Reinhardt S, Lesche M, Weber A, Kuscha V, Dahl A, Rulands S, Brand M. Single cell sequencing of radial glia progeny reveals the diversity of newborn neurons in the adult zebrafish brain. Development 2020; 147:dev.185595. [PMID: 31908317 PMCID: PMC6983714 DOI: 10.1242/dev.185595] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2019] [Accepted: 11/11/2019] [Indexed: 01/16/2023]
Abstract
Zebrafish display widespread and pronounced adult neurogenesis, which is fundamental for their regeneration capability after central nervous system injury. However, the cellular identity and the biological properties of adult newborn neurons are elusive for most brain areas. Here, we have used short-term lineage tracing of radial glia progeny to prospectively isolate newborn neurons from the her4.1+ radial glia lineage in the homeostatic adult forebrain. Transcriptome analysis of radial glia, newborn neurons and mature neurons using single cell sequencing identified distinct transcriptional profiles, including novel markers for each population. Specifically, we detected two separate newborn neuron types, which showed diversity of cell fate commitment and location. Further analyses showed that these cell types are homologous to neurogenic cells in the mammalian brain, identified neurogenic commitment in proliferating radial glia and indicated that glutamatergic projection neurons are generated in the adult zebrafish telencephalon. Thus, we prospectively isolated adult newborn neurons from the adult zebrafish forebrain, identified markers for newborn and mature neurons in the adult brain, and revealed intrinsic heterogeneity among adult newborn neurons and their homology with mammalian adult neurogenic cell types.
Collapse
Affiliation(s)
- Christian Lange
- Center for Regenerative Therapies Dresden (CRTD), CMCB, Technische Universität Dresden, Fetscherstrasse 105, 01307 Dresden, Germany
| | - Fabian Rost
- Max Planck Institute for the Physics of Complex Systems, Noethnitzer Strasse 38, 01187 Dresden, Germany.,Center for Systems Biology Dresden (CSBD), Pfotenhauer Strasse 108, 01307 Dresden, Germany
| | - Anja Machate
- Center for Regenerative Therapies Dresden (CRTD), CMCB, Technische Universität Dresden, Fetscherstrasse 105, 01307 Dresden, Germany
| | - Susanne Reinhardt
- Center for Regenerative Therapies Dresden (CRTD), CMCB, Technische Universität Dresden, Fetscherstrasse 105, 01307 Dresden, Germany.,DRESDEN-concept Genome Center, c/o Center for Molecular and Cellular Bioengineering (CMCB), Technische Universität Dresden, Fetscherstrasse 105, 01307, Dresden, Germany
| | - Matthias Lesche
- Center for Regenerative Therapies Dresden (CRTD), CMCB, Technische Universität Dresden, Fetscherstrasse 105, 01307 Dresden, Germany.,DRESDEN-concept Genome Center, c/o Center for Molecular and Cellular Bioengineering (CMCB), Technische Universität Dresden, Fetscherstrasse 105, 01307, Dresden, Germany
| | - Anke Weber
- Center for Regenerative Therapies Dresden (CRTD), CMCB, Technische Universität Dresden, Fetscherstrasse 105, 01307 Dresden, Germany
| | - Veronika Kuscha
- Center for Regenerative Therapies Dresden (CRTD), CMCB, Technische Universität Dresden, Fetscherstrasse 105, 01307 Dresden, Germany
| | - Andreas Dahl
- Center for Regenerative Therapies Dresden (CRTD), CMCB, Technische Universität Dresden, Fetscherstrasse 105, 01307 Dresden, Germany.,DRESDEN-concept Genome Center, c/o Center for Molecular and Cellular Bioengineering (CMCB), Technische Universität Dresden, Fetscherstrasse 105, 01307, Dresden, Germany
| | - Steffen Rulands
- Max Planck Institute for the Physics of Complex Systems, Noethnitzer Strasse 38, 01187 Dresden, Germany.,Center for Systems Biology Dresden (CSBD), Pfotenhauer Strasse 108, 01307 Dresden, Germany
| | - Michael Brand
- Center for Regenerative Therapies Dresden (CRTD), CMCB, Technische Universität Dresden, Fetscherstrasse 105, 01307 Dresden, Germany
| |
Collapse
|
45
|
Fischer J, Heide M, Huttner WB. Genetic Modification of Brain Organoids. Front Cell Neurosci 2019; 13:558. [PMID: 31920558 PMCID: PMC6928125 DOI: 10.3389/fncel.2019.00558] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2019] [Accepted: 12/04/2019] [Indexed: 12/12/2022] Open
Abstract
Brain organoids have become increasingly used systems allowing 3D-modeling of human brain development, evolution, and disease. To be able to make full use of these modeling systems, researchers have developed a growing toolkit of genetic modification techniques. These techniques can be applied to mature brain organoids or to the preceding embryoid bodies (EBs) and founding cells. This review will describe techniques used for transient and stable genetic modification of brain organoids and discuss their current use and respective advantages and disadvantages. Transient approaches include adeno-associated virus (AAV) and electroporation-based techniques, whereas stable genetic modification approaches make use of lentivirus (including viral stamping), transposon and CRISPR/Cas9 systems. Finally, an outlook as to likely future developments and applications regarding genetic modifications of brain organoids will be presented.
Collapse
Affiliation(s)
- Jan Fischer
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany
| | - Michael Heide
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany
| | - Wieland B Huttner
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany
| |
Collapse
|
46
|
Fowler JL, Ang LT, Loh KM. A critical look: Challenges in differentiating human pluripotent stem cells into desired cell types and organoids. WILEY INTERDISCIPLINARY REVIEWS-DEVELOPMENTAL BIOLOGY 2019; 9:e368. [PMID: 31746148 DOI: 10.1002/wdev.368] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/30/2019] [Revised: 09/17/2019] [Accepted: 10/21/2019] [Indexed: 12/11/2022]
Abstract
Too many choices can be problematic. This is certainly the case for human pluripotent stem cells (hPSCs): they harbor the potential to differentiate into hundreds of cell types; yet it is highly challenging to exclusively differentiate hPSCs into a single desired cell type. This review focuses on unresolved and fundamental questions regarding hPSC differentiation and critiquing the identity and purity of the resultant cell populations. These are timely issues in view of the fact that hPSC-derived cell populations have or are being transplanted into patients in over 30 ongoing clinical trials. While many in vitro differentiation protocols purport to "mimic development," the exact number and identity of intermediate steps that a pluripotent cell takes to differentiate into a given cell type in vivo remains largely unknown. Consequently, most differentiation efforts inevitably generate a heterogeneous cellular population, as revealed by single-cell RNA-sequencing and other analyses. The presence of unwanted cell types in differentiated hPSC populations does not portend well for transplantation therapies. This provides an impetus to precisely control differentiation to desired ends-for instance, by logically blocking the formation of unwanted cell types or by overexpressing lineage-specifying transcription factors-or by harnessing technologies to selectively purify desired cell types. Conversely, approaches to differentiate three-dimensional "organoids" from hPSCs intentionally generate heterogeneous cell populations. While this is intended to mimic the rich cellular diversity of developing tissues, whether all such organoids are spatially organized in a manner akin to native organs (and thus, whether they fully qualify as organoids) remains to be fully resolved. This article is categorized under: Adult Stem Cells > Tissue Renewal > Regeneration: Stem Cell Differentiation and Reversion Gene Expression > Transcriptional Hierarchies: Cellular Differentiation Early Embryonic Development: Gastrulation and Neurulation.
Collapse
Affiliation(s)
- Jonas L Fowler
- Stanford Institute for Stem Cell Biology & Regenerative Medicine, Stanford-UC Berkeley Siebel Stem Cell Institute, Stanford University School of Medicine, Stanford, California.,Department of Developmental Biology, Bio-X, Cancer Institute, Cardiovascular Institute, ChEM-H, Diabetes Research Center, Maternal & Child Health Research Institute, Wu Tsai Neurosciences Institute, Stanford University School of Medicine, Stanford, California
| | - Lay Teng Ang
- Stanford Institute for Stem Cell Biology & Regenerative Medicine, Stanford-UC Berkeley Siebel Stem Cell Institute, Stanford University School of Medicine, Stanford, California
| | - Kyle M Loh
- Stanford Institute for Stem Cell Biology & Regenerative Medicine, Stanford-UC Berkeley Siebel Stem Cell Institute, Stanford University School of Medicine, Stanford, California.,Department of Developmental Biology, Bio-X, Cancer Institute, Cardiovascular Institute, ChEM-H, Diabetes Research Center, Maternal & Child Health Research Institute, Wu Tsai Neurosciences Institute, Stanford University School of Medicine, Stanford, California
| |
Collapse
|
47
|
Shulman ED, Elkon R. Cell-type-specific analysis of alternative polyadenylation using single-cell transcriptomics data. Nucleic Acids Res 2019; 47:10027-10039. [PMID: 31501864 PMCID: PMC6821429 DOI: 10.1093/nar/gkz781] [Citation(s) in RCA: 60] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2019] [Revised: 08/27/2019] [Accepted: 09/01/2019] [Indexed: 12/22/2022] Open
Abstract
Alternative polyadenylation (APA) is emerging as an important layer of gene regulation because the majority of mammalian protein-coding genes contain multiple polyadenylation (pA) sites in their 3' UTR. By alteration of 3' UTR length, APA can considerably affect post-transcriptional gene regulation. Yet, our understanding of APA remains rudimentary. Novel single-cell RNA sequencing (scRNA-seq) techniques allow molecular characterization of different cell types to an unprecedented degree. Notably, the most popular scRNA-seq protocols specifically sequence the 3' end of transcripts. Building on this property, we implemented a method for analysing patterns of APA regulation from such data. Analyzing multiple datasets from diverse tissues, we identified widespread modulation of APA in different cell types resulting in global 3' UTR shortening/lengthening and enhanced cleavage at intronic pA sites. Our results provide a proof-of-concept demonstration that the huge volume of scRNA-seq data that accumulates in the public domain offers a unique resource for the exploration of APA based on a very broad collection of cell types and biological conditions.
Collapse
Affiliation(s)
- Eldad David Shulman
- Department of Human Molecular Genetics and Biochemistry, Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Ran Elkon
- Department of Human Molecular Genetics and Biochemistry, Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| |
Collapse
|
48
|
Cell diversity in the human cerebral cortex: from the embryo to brain organoids. Curr Opin Neurobiol 2019; 56:194-198. [PMID: 31051421 DOI: 10.1016/j.conb.2019.03.001] [Citation(s) in RCA: 69] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2019] [Accepted: 03/01/2019] [Indexed: 12/21/2022]
Abstract
The development and wiring of the central nervous system is a remarkable biological process that starts with the generation of and interaction between a large diversity of cell types. Our understanding of the developmental logic that drives cellular diversification in the mammalian brain comes, to a large extent, from studies in rodents. However, identifying the unique cellular processes underlying primate corticogenesis has been slow, due to the challenges associated with directly observing and manipulating brain tissue from these species. Recent technological advances in two areas hold promise to accelerate discovery of the mechanisms that govern human brain development, evolution, and pathophysiology of disease. Molecular profiling of large numbers of single cells can now capture cell identity and cell states within a complex tissue. Furthermore, modeling aspects of human organogenesis in vitro, even for tissues as complex as the brain, has been advanced by the use of three-dimensional organoid systems. Here, we describe how these approaches have been applied to date and how they promise to uncover the principles of cell diversification in the developing human brain.
Collapse
|
49
|
Xu SJ, Heller EA. Recent advances in neuroepigenetic editing. Curr Opin Neurobiol 2019; 59:26-33. [PMID: 31015104 DOI: 10.1016/j.conb.2019.03.010] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2018] [Revised: 02/28/2019] [Accepted: 03/18/2019] [Indexed: 02/09/2023]
Abstract
A wealth of studies in the mammalian nervous system indicate the role of epigenetic gene regulation in both basic neurobiological function and disease. However, the relationship between epigenetic regulation and neuropathology is largely correlational due to the presence of mixed cell populations within brain regions and the genome-wide effects of classical approaches to manipulate the epigenome. Locus-specific epigenetic editing allows direct epigenetic regulation of specific genes to elucidate the direct causal relationship between epigenetic modifications and transcription. This review discusses some of the latest innovations in the efficacy and flexibility in this approach that hold promise for neurobiological application.
Collapse
Affiliation(s)
- Song-Jun Xu
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, The University of Pennsylvania, Philadelphia, PA, USA
| | - Elizabeth A Heller
- Department of Systems Pharmacology and Translational Therapeutics and Penn Epigenetics Institute, Perelman School of Medicine, The University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
50
|
Srinivasaiah S, Musumeci G, Mohan T, Castrogiovanni P, Absenger-Novak M, Zefferer U, Mostofi S, Bonyadi Rad E, Grün NG, Weinberg AM, Schäfer U. A 300 μm Organotypic Bone Slice Culture Model for Temporal Investigation of Endochondral Osteogenesis. Tissue Eng Part C Methods 2019; 25:197-212. [DOI: 10.1089/ten.tec.2018.0368] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Affiliation(s)
- Sriveena Srinivasaiah
- Department of Orthopedics and Trauma Surgery, Medical University of Graz, Graz, Austria
- Research Unit for Experimental Neurotraumatology, Department of Neurosurgery, Medical University of Graz, Graz, Austria
| | - Giuseppe Musumeci
- Human Anatomy and Histology Section, Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
| | - Tamilselvan Mohan
- Institute of Chemistry, University of Graz, Graz, Austria
- Laboratory for Characterization and Processing, Faculty of Mechanical Engineering, University of Maribor, Maribor, Slovenia
| | - Paola Castrogiovanni
- Human Anatomy and Histology Section, Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
| | | | - Ulrike Zefferer
- Research Unit for Experimental Neurotraumatology, Department of Neurosurgery, Medical University of Graz, Graz, Austria
| | - Sepideh Mostofi
- Department of Orthopedics and Trauma Surgery, Medical University of Graz, Graz, Austria
| | - Ehsan Bonyadi Rad
- Department of Orthopedics and Trauma Surgery, Medical University of Graz, Graz, Austria
- Department of Internal Medicine, Medical University of Graz, Graz, Austria
| | - Nicole Gabriele Grün
- Department of Orthopedics and Trauma Surgery, Medical University of Graz, Graz, Austria
| | | | - Ute Schäfer
- Research Unit for Experimental Neurotraumatology, Department of Neurosurgery, Medical University of Graz, Graz, Austria
| |
Collapse
|