451
|
|
452
|
|
453
|
Sakaguchi H, Ozaki Y, Ashida T, Matsubara T, Oishi N, Kihara S, Takahashi J. Self-Organized Synchronous Calcium Transients in a Cultured Human Neural Network Derived from Cerebral Organoids. Stem Cell Reports 2019; 13:458-473. [PMID: 31257131 PMCID: PMC6739638 DOI: 10.1016/j.stemcr.2019.05.029] [Citation(s) in RCA: 83] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2018] [Revised: 05/30/2019] [Accepted: 05/30/2019] [Indexed: 12/19/2022] Open
Abstract
The cerebrum is a major center for brain function, and its activity is derived from the assembly of activated cells in neural networks. It is currently difficult to study complex human cerebral neuronal network activity. Here, using cerebral organoids, we report self-organized and complex human neural network activities that include synchronized and non-synchronized patterns. Self-organized neuronal network formation was observed following a dissociation culture of human embryonic stem cell-derived cerebral organoids. The spontaneous individual and synchronized activity of the network was measured via calcium imaging, and subsequent analysis enabled the examination of detailed cell activity patterns, providing simultaneous raster plots, cluster analyses, and cell distribution data. Finally, we demonstrated the feasibility of our system to assess drug-inducible dynamic changes of the network activity. The comprehensive functional analysis of human neuronal networks using this system may offer a powerful tool to access human brain function. Cerebral organoids can recapitulate cerebral characters in 3D order A functional neural network was efficiently formed after dissociation of organoids Calcium activity patterns were examined with clustering and the cell distribution A powerful system for the functional analysis of human neuronal networks
Collapse
Affiliation(s)
- Hideya Sakaguchi
- Department of Clinical Application, Center for iPS Cell Research and Application, Kyoto University, Kyoto 606-8507, Japan.
| | - Yuki Ozaki
- Department of Clinical Application, Center for iPS Cell Research and Application, Kyoto University, Kyoto 606-8507, Japan
| | - Tomoka Ashida
- Department of Clinical Application, Center for iPS Cell Research and Application, Kyoto University, Kyoto 606-8507, Japan
| | - Takayoshi Matsubara
- Life Science Center, MB HQ, Yokogawa Electric Corporation, Ishikawa 920-0177, Japan
| | - Naotaka Oishi
- Informatics Japan, PerkinElmer Japan Co., Ltd., Tokyo 101-0024, Japan
| | - Shunsuke Kihara
- Department of Fundamental Cell Technology, Center for iPS Cell Research and Application, Kyoto University, Kyoto 606-8507, Japan
| | - Jun Takahashi
- Department of Clinical Application, Center for iPS Cell Research and Application, Kyoto University, Kyoto 606-8507, Japan.
| |
Collapse
|
454
|
Ndyabawe K, Kisaalita WS. Engineering microsystems to recapitulate brain physiology on a chip. Drug Discov Today 2019; 24:1725-1730. [PMID: 31226433 DOI: 10.1016/j.drudis.2019.06.008] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2018] [Revised: 05/17/2019] [Accepted: 06/12/2019] [Indexed: 12/17/2022]
Abstract
The structural and functional organization of the human brain consists of 52 regions with distinct cellular organization. In vitro models for normal and pathological states using isolated brain-region-specific 3D engineered tissues fail to recapitulate information integration and/or transfer that arises from connectivity among neuroanatomical structures. Therefore, development of brain-on-a-chip microsystems must shift to multiple region neuron network designs to be relevant in brain functionality and deficit modeling. However, in vitro formation of multiregional networks on microdevices presents several challenges that we illustrate using a few neurological disorders; and we offer guidance, depending on objectives (HTS, disease modeling, etc.) for rational design of microfluidic systems and better emulation of in vivo conditions.
Collapse
Affiliation(s)
- Kenneth Ndyabawe
- School of Chemical, Materials and Biomedical Engineering, College of Engineering, Driftmier Engineering Center, University of Georgia, Athens, GA 30602, USA
| | - William S Kisaalita
- School of Chemical, Materials and Biomedical Engineering, College of Engineering, Driftmier Engineering Center, University of Georgia, Athens, GA 30602, USA.
| |
Collapse
|
455
|
Abstract
Recent studies have demonstrated an array of stem cell-derived, self-organizing miniature organs, termed organoids, that replicate the key structural and functional characteristics of their in vivo counterparts. As organoid technology opens up new frontiers of research in biomedicine, there is an emerging need for innovative engineering approaches for the production, control, and analysis of organoids and their microenvironment. In this Review, we explore organ-on-a-chip technology as a platform to fulfill this need and examine how this technology may be leveraged to address major technical challenges in organoid research. We also discuss emerging opportunities and future obstacles for the development and application of organoid-on-a-chip technology.
Collapse
Affiliation(s)
- Sunghee Estelle Park
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Andrei Georgescu
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Dongeun Huh
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA 19104, USA.
- Institute for Regenerative Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- NSF Science and Technology Center for Engineering Mechanobiology, University of Pennsylvania, Philadelphia, PA 19104, USA
| |
Collapse
|
456
|
Gill E, Willis S, Gerigk M, Cohen P, Zhang D, Li X, Huang YYS. Fabrication of Designable and Suspended Microfibers via Low-Voltage 3D Micropatterning. ACS APPLIED MATERIALS & INTERFACES 2019; 11:19679-19690. [PMID: 31081331 PMCID: PMC6613729 DOI: 10.1021/acsami.9b01258] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/20/2019] [Accepted: 05/13/2019] [Indexed: 05/02/2023]
Abstract
Building two-dimensional (2D) and three-dimensional (3D) fibrous structures in the micro- and nanoscale will offer exciting prospects for numerous applications spanning from sensors to energy storage and tissue engineering scaffolds. Electrospinning is a well-suited technique for drawing micro- to nanoscale fibers, but current methods of building electrospun fibers in 3D are restrictive in terms of printed height, design of macroscopic fiber networks, and choice of polymer. Here, we combine low-voltage electrospinning and additive manufacturing as a method to pattern layers of suspended mesofibers. Layers of fibers are suspended between 3D-printed supports in situ in multiple fiber layers and designable orientations. We examine the key working parameters to attain a threshold for fiber suspension, use those behavioral observations to establish a "fiber suspension indicator", and demonstrate its utility through design of intricate suspended fiber architectures. Individual fibers produced by this method approach the micrometer/submicrometer scale, while the overall suspended 3D fiber architecture can span over a centimeter in height. We demonstrate an application of suspended fiber architectures in 3D cell culture, utilizing patterned fiber topography to guide the assembly of suspended high-cellular-density structures. The solution-based fiber suspension patterning process we report offers a unique competence in patterning soft polymers, including extracellular matrix-like materials, in a high resolution and aspect ratio. The platform could thus offer new design and manufacturing capabilities of devices and functional products by incorporating functional fibrous elements.
Collapse
Affiliation(s)
- Elisabeth
L. Gill
- Department
of Engineering, University of Cambridge, Trumpington Street, Cambridge CB2 1PZ, U.K.
- The
Nanoscience Centre, University of Cambridge, 11 JJ Thomson Avenue, Cambridge CB3 0FF, U.K.
| | - Samuel Willis
- Department
of Engineering, University of Cambridge, Trumpington Street, Cambridge CB2 1PZ, U.K.
| | - Magda Gerigk
- Department
of Engineering, University of Cambridge, Trumpington Street, Cambridge CB2 1PZ, U.K.
- The
Nanoscience Centre, University of Cambridge, 11 JJ Thomson Avenue, Cambridge CB3 0FF, U.K.
| | - Paul Cohen
- Department
of Engineering, University of Cambridge, Trumpington Street, Cambridge CB2 1PZ, U.K.
| | - Duo Zhang
- Department
of Engineering, University of Cambridge, Trumpington Street, Cambridge CB2 1PZ, U.K.
- The
Nanoscience Centre, University of Cambridge, 11 JJ Thomson Avenue, Cambridge CB3 0FF, U.K.
| | - Xia Li
- Department
of Engineering, University of Cambridge, Trumpington Street, Cambridge CB2 1PZ, U.K.
| | - Yan Yan Shery Huang
- Department
of Engineering, University of Cambridge, Trumpington Street, Cambridge CB2 1PZ, U.K.
- The
Nanoscience Centre, University of Cambridge, 11 JJ Thomson Avenue, Cambridge CB3 0FF, U.K.
| |
Collapse
|
457
|
Abstract
Cerebral organoids are an emerging cutting-edge technology to model human brain
development and neurodevelopmental disorders, for which mouse models exhibit significant
limitations. In the human brain, synaptic connections define neural circuits, and synaptic
deficits account for various neurodevelopmental disorders. Thus, harnessing the full power
of cerebral organoids for human brain modeling requires the ability to visualize and
analyze synapses in cerebral organoids. Previously, we devised an optimized method to
generate human cerebral organoids, and showed that optimal organoids express mature-neuron
markers, including synaptic proteins and neurotransmitter receptors and transporters.
Here, we give evidence for synaptogenesis in cerebral organoids, via microscopical
visualization of synapses. We also describe multiple approaches to quantitatively analyze
synapses in cerebral organoids. Collectively, our work provides sufficient evidence for
the possibility of modeling synaptogenesis and synaptic disorders in cerebral organoids,
and may help advance the use of cerebral organoids in molecular neuroscience and studies
of neurodevelopmental disorders such as autism.
Collapse
Affiliation(s)
- Abraam M Yakoub
- Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, CA, USA
| | - Mark Sadek
- Department of Pharmaceutical Biotechnology, University of Illinois College of Pharmacy, Chicago, IL, USA.,Department of Research and Development, Akorn Pharmaceuticals, Vernon Hills, IL, USA
| |
Collapse
|
458
|
Experimental study of rhBMP-2 chitosan nano-sustained release carrier-loaded PLGA/nHA scaffolds to construct mandibular tissue-engineered bone. Arch Oral Biol 2019; 102:16-25. [DOI: 10.1016/j.archoralbio.2019.03.023] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2019] [Revised: 03/09/2019] [Accepted: 03/27/2019] [Indexed: 11/22/2022]
|
459
|
Velasco S, Kedaigle AJ, Simmons SK, Nash A, Rocha M, Quadrato G, Paulsen B, Nguyen L, Adiconis X, Regev A, Levin JZ, Arlotta P. Individual brain organoids reproducibly form cell diversity of the human cerebral cortex. Nature 2019; 570:523-527. [PMID: 31168097 PMCID: PMC6906116 DOI: 10.1038/s41586-019-1289-x] [Citation(s) in RCA: 647] [Impact Index Per Article: 107.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2018] [Accepted: 05/14/2019] [Indexed: 01/08/2023]
Abstract
Experimental models of the human brain are needed for basic understanding of its development and disease1. Human brain organoids hold unprecedented promise for this purpose; however, they are plagued by high organoid-to-organoid variability2,3. This has raised doubts as to whether developmental processes of the human brain can occur outside the context of embryogenesis with a degree of reproducibility that is comparable to the endogenous tissue. Here we show that an organoid model of the dorsal forebrain can reliably generate a rich diversity of cell types appropriate for the human cerebral cortex. We performed single-cell RNA-sequencing analysis of 166,242 cells isolated from 21 individual organoids, finding that 95% of the organoids generate a virtually indistinguishable compendium of cell types, following similar developmental trajectories and with a degree of organoid-to-organoid variability comparable to that of individual endogenous brains. Furthermore, organoids derived from different stem cell lines show consistent reproducibility in the cell types produced. The data demonstrate that reproducible development of the complex cellular diversity of the central nervous system does not require the context of the embryo, and that establishment of terminal cell identity is a highly constrained process that can emerge from diverse stem cell origins and growth environments.
Collapse
Affiliation(s)
- Silvia Velasco
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Amanda J Kedaigle
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Klarman Cell Observatory, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Sean K Simmons
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Klarman Cell Observatory, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Allison Nash
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Marina Rocha
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Giorgia Quadrato
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Eli and Edythe Broad CIRM Center for Regenerative Medicine and Stem Cell Research at USC, Los Angeles, CA, USA
| | - Bruna Paulsen
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Lan Nguyen
- Klarman Cell Observatory, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Xian Adiconis
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Klarman Cell Observatory, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Aviv Regev
- Klarman Cell Observatory, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Howard Hughes Medical Institute, Koch Institute of Integrative Cancer Research, Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Joshua Z Levin
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Klarman Cell Observatory, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Paola Arlotta
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, USA.
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA.
| |
Collapse
|
460
|
Ebrahimkhani MR, Ebisuya M. Synthetic developmental biology: build and control multicellular systems. Curr Opin Chem Biol 2019; 52:9-15. [PMID: 31102790 DOI: 10.1016/j.cbpa.2019.04.006] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2019] [Revised: 04/03/2019] [Accepted: 04/09/2019] [Indexed: 02/08/2023]
Abstract
Synthetic biology offers a bottom-up engineering approach that intends to understand complex systems via design-build-test cycles. Embryonic development comprises complex processes that originate at the level of gene regulatory networks in a cell and emerge into collective cellular behaviors with multicellular forms and functions. Here, we review synthetic biology approaches to development that involve building de novo developmental trajectories or engineering control in stem cell-derived multicellular systems. The field of synthetic developmental biology is rapidly growing with the help of recent advances in artificial gene circuits, self-organizing organoids, and controllable tissue microenvironments. The outcome will be a blueprint to decode principles of morphogenesis and to create programmable organoids with novel designs or improved functions.
Collapse
Affiliation(s)
- Mo R Ebrahimkhani
- Biodesign Institute, Arizona State Tempe, AZ, USA; School of Biological and Health Systems Engineering, Arizona State Tempe, AZ, USA; Mayo Clinic College of Medicine and Science, Phoenix, AZ, USA.
| | - Miki Ebisuya
- European Molecular Biology Laboratory (EMBL) Barcelona, Dr. Aiguader, 88, 08003, Barcelona, Spain.
| |
Collapse
|
461
|
Arlotta P. Organoids required! A new path to understanding human brain development and disease. Nat Methods 2019; 15:27-29. [PMID: 29298289 DOI: 10.1038/nmeth.4557] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Our ability to study the developing human brain has recently been dramatically advanced by the development of human 'brain organoids', three-dimensional culture systems that recapitulate selected aspects of human brain development in reductionist, yet complex, tissues in vitro. Here I discuss the promises and challenges this new model system presents.
Collapse
Affiliation(s)
- Paola Arlotta
- Department of Stem Cell and Regenerative Biology, Harvard University
| |
Collapse
|
462
|
Kyrousi C, Cappello S. Using brain organoids to study human neurodevelopment, evolution and disease. WILEY INTERDISCIPLINARY REVIEWS-DEVELOPMENTAL BIOLOGY 2019; 9:e347. [PMID: 31071759 DOI: 10.1002/wdev.347] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/19/2018] [Revised: 02/18/2019] [Accepted: 04/07/2019] [Indexed: 01/12/2023]
Abstract
The brain is one of the most complex organs, responsible for the advanced intellectual and cognitive ability of humans. Although primates are to some extent capable of performing cognitive tasks, their abilities are less evolved. One of the reasons for this is the vast differences in the brain of humans compared to other mammals, in terms of shape, size and complexity. Such differences make the study of human brain development fascinating. Interestingly, the cerebral cortex is by far the most complex brain region resulting from its selective evolution within mammals over millions of years. Unraveling the molecular and cellular mechanisms regulating brain development, as well as the evolutionary differences seen across species and the need to understand human brain disorders, are some of the reasons why scientists are interested in improving their current knowledge on human corticogenesis. Toward this end, several animal models including primates have been used, however, these models are limited in their extent to recapitulate human-specific features. Recent technological achievements in the field of stem cell research, which have enabled the generation of human models of corticogenesis, called brain or cerebral organoids, are of great importance. This review focuses on the main cellular and molecular features of human corticogenesis and the use of brain organoids to study it. We will discuss the key differences between cortical development in human and nonhuman mammals, the technological applications of brain organoids and the different aspects of cortical development in normal and pathological conditions, which can be modeled using brain organoids. This article is categorized under: Comparative Development and Evolution > Regulation of Organ Diversity Nervous System Development > Vertebrates: General Principles.
Collapse
Affiliation(s)
- Christina Kyrousi
- Department of Developmental Neurobiology, Max Planck Institute of Psychiatry, Munich, Germany
| | - Silvia Cappello
- Department of Developmental Neurobiology, Max Planck Institute of Psychiatry, Munich, Germany
| |
Collapse
|
463
|
Holloway EM, Capeling MM, Spence JR. Biologically inspired approaches to enhance human organoid complexity. Development 2019; 146:dev166173. [PMID: 30992275 PMCID: PMC6503984 DOI: 10.1242/dev.166173] [Citation(s) in RCA: 57] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Organoids are complex three-dimensional in vitro organ-like model systems. Human organoids, which are derived from human pluripotent stem cells or primary human donor tissue, have been used to address fundamental questions about human development, stem cell biology and organ regeneration. Focus has now shifted towards implementation of organoids for biological discovery and advancing existing systems to more faithfully recapitulate the native organ. This work has highlighted significant unknowns in human biology and has invigorated new exploration into the cellular makeup of human organs during development and in the adult - work that is crucial for providing appropriate benchmarks for organoid systems. In this Review, we discuss efforts to characterize human organ cellular complexity and attempts to make organoid models more realistic through co-culture, transplantation and bioengineering approaches.
Collapse
Affiliation(s)
- Emily M Holloway
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Meghan M Capeling
- Department of Biomedical Engineering, University of Michigan College of Engineering, Ann Arbor, MI 48109, USA
| | - Jason R Spence
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
- Department of Biomedical Engineering, University of Michigan College of Engineering, Ann Arbor, MI 48109, USA
- Department of Internal Medicine, Division of Gastroenterology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
- Center for Organogenesis, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| |
Collapse
|
464
|
Abstract
Brain organoids are self-assembled three-dimensional aggregates generated from pluripotent stem cells with cell types and cytoarchitectures that resemble the embryonic human brain. As such, they have emerged as novel model systems that can be used to investigate human brain development and disorders. Although brain organoids mimic many key features of early human brain development at molecular, cellular, structural and functional levels, some aspects of brain development, such as the formation of distinct cortical neuronal layers, gyrification, and the establishment of complex neuronal circuitry, are not fully recapitulated. Here, we summarize recent advances in the development of brain organoid methodologies and discuss their applications in disease modeling. In addition, we compare current organoid systems to the embryonic human brain, highlighting features that currently can and cannot be recapitulated, and discuss perspectives for advancing current brain organoid technologies to expand their applications.
Collapse
Affiliation(s)
- Xuyu Qian
- Department of Neuroscience and Mahoney Institute for Neurosciences, Perelman School for Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Biomedical Engineering Graduate Program, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Hongjun Song
- Department of Neuroscience and Mahoney Institute for Neurosciences, Perelman School for Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Department of Cell and Developmental Biology, Perelman School for Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Institute for Regenerative Medicine, Perelman School for Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- The Epigenetics Institute, Perelman School for Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Guo-Li Ming
- Department of Neuroscience and Mahoney Institute for Neurosciences, Perelman School for Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Department of Cell and Developmental Biology, Perelman School for Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Institute for Regenerative Medicine, Perelman School for Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Department of Psychiatry, Perelman School for Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| |
Collapse
|
465
|
Song L, Yuan X, Jones Z, Griffin K, Zhou Y, Ma T, Li Y. Assembly of Human Stem Cell-Derived Cortical Spheroids and Vascular Spheroids to Model 3-D Brain-like Tissues. Sci Rep 2019; 9:5977. [PMID: 30979929 PMCID: PMC6461701 DOI: 10.1038/s41598-019-42439-9] [Citation(s) in RCA: 103] [Impact Index Per Article: 17.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2018] [Accepted: 04/01/2019] [Indexed: 02/07/2023] Open
Abstract
Human cerebral organoids derived from induced pluripotent stem cells (iPSCs) provide novel tools for recapitulating the cytoarchitecture of human brain and for studying biological mechanisms of neurological disorders. However, the heterotypic interactions of neurovascular units, composed of neurons, pericytes, astrocytes, and brain microvascular endothelial cells, in brain-like tissues are less investigated. The objective of this study is to investigate the impacts of neural spheroids and vascular spheroids interactions on the regional brain-like tissue patterning in cortical spheroids derived from human iPSCs. Hybrid neurovascular spheroids were constructed by fusion of human iPSC-derived cortical neural progenitor cell (iNPC) spheroids, endothelial cell (iEC) spheroids, and the supporting human mesenchymal stem cells (MSCs). Single hybrid spheroids were constructed at different iNPC: iEC: MSC ratios of 4:2:0, 3:2:1 2:2:2, and 1:2:3 in low-attachment 96-well plates. The incorporation of MSCs upregulated the secretion levels of cytokines VEGF-A, PGE2, and TGF-β1 in hybrid spheroid system. In addition, tri-cultured spheroids had high levels of TBR1 (deep cortical layer VI) and Nkx2.1 (ventral cells), and matrix remodeling genes, MMP2 and MMP3, as well as Notch-1, indicating the crucial role of matrix remodeling and cell-cell communications on cortical spheroid and organoid patterning. Moreover, tri-culture system elevated blood-brain barrier gene expression (e.g., GLUT-1), CD31, and tight junction protein ZO1 expression. Treatment with AMD3100, a CXCR4 antagonist, showed the immobilization of MSCs during spheroid fusion, indicating a CXCR4-dependent manner of hMSC migration and homing. This forebrain-like model has potential applications in understanding heterotypic cell-cell interactions and novel drug screening in diseased human brain.
Collapse
Affiliation(s)
- Liqing Song
- Department of Chemical and Biomedical Engineering, FAMU-FSU College of Engineering, Florida State University, Tallahassee, FL, USA
| | - Xuegang Yuan
- Department of Chemical and Biomedical Engineering, FAMU-FSU College of Engineering, Florida State University, Tallahassee, FL, USA
| | - Zachary Jones
- Department of Biomedical Sciences, College of Medicine, Florida State University, Tallahassee, Florida, USA
| | - Kyle Griffin
- Department of Chemical and Biomedical Engineering, FAMU-FSU College of Engineering, Florida State University, Tallahassee, FL, USA
| | - Yi Zhou
- Department of Biomedical Sciences, College of Medicine, Florida State University, Tallahassee, Florida, USA
| | - Teng Ma
- Department of Chemical and Biomedical Engineering, FAMU-FSU College of Engineering, Florida State University, Tallahassee, FL, USA.
| | - Yan Li
- Department of Chemical and Biomedical Engineering, FAMU-FSU College of Engineering, Florida State University, Tallahassee, FL, USA.
| |
Collapse
|
466
|
Sharf T, Hansma PK, Hari MA, Kosik KS. Non-contact monitoring of extra-cellular field potentials with a multi-electrode array. LAB ON A CHIP 2019; 19:1448-1457. [PMID: 30887972 DOI: 10.1039/c8lc00984h] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
Developing tools to enable non-invasive, high-throughput electrophysiology measurements of large functional-networks of electrogenic cells used as in vitro disease models for the heart and brain remains an outstanding challenge for preclinical drug discovery, where failures are costly and can prove to be fatal during clinical trials. Here we demonstrate, for the first time, that it is possible to perform non-contact monitoring of extra-cellular field potentials with a multi-electrode array (MEA). To do this preliminary demonstration we built a prototype with a custom mechanical stage to micro-position cells grown on conventional glass coverslips over the recording surface of a MEA sensor. The prototype can monitor extra-cellular fields generated by multi-cellular networks in a non-contact configuration, enabling a single MEA sensor to probe different cultures in succession, without fouling or degrading its sensitive electronic surface. This first demonstration with easy to culture cardiomyocyte cells and a prototype device points to the exciting possibility for instrument development leading to more efficient and cost-effective drug screening paradigms for cardiovascular and neurological diseases.
Collapse
Affiliation(s)
- Tal Sharf
- Neuroscience Research Institute, University of California Santa Barbara, Santa Barbara, CA 93106, USA.
| | | | | | | |
Collapse
|
467
|
Pacitti D, Privolizzi R, Bax BE. Organs to Cells and Cells to Organoids: The Evolution of in vitro Central Nervous System Modelling. Front Cell Neurosci 2019; 13:129. [PMID: 31024259 PMCID: PMC6465581 DOI: 10.3389/fncel.2019.00129] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2018] [Accepted: 03/14/2019] [Indexed: 02/05/2023] Open
Abstract
With 100 billion neurons and 100 trillion synapses, the human brain is not just the most complex organ in the human body, but has also been described as "the most complex thing in the universe." The limited availability of human living brain tissue for the study of neurogenesis, neural processes and neurological disorders has resulted in more than a century-long strive from researchers worldwide to model the central nervous system (CNS) and dissect both its striking physiology and enigmatic pathophysiology. The invaluable knowledge gained with the use of animal models and post mortem human tissue remains limited to cross-species similarities and structural features, respectively. The advent of human induced pluripotent stem cell (hiPSC) and 3-D organoid technologies has revolutionised the approach to the study of human brain and CNS in vitro, presenting great potential for disease modelling and translational adoption in drug screening and regenerative medicine, also contributing beneficially to clinical research. We have surveyed more than 100 years of research in CNS modelling and provide in this review an historical excursus of its evolution, from early neural tissue explants and organotypic cultures, to 2-D patient-derived cell monolayers, to the latest development of 3-D cerebral organoids. We have generated a comprehensive summary of CNS modelling techniques and approaches, protocol refinements throughout the course of decades and developments in the study of specific neuropathologies. Current limitations and caveats such as clonal variation, developmental stage, validation of pluripotency and chromosomal stability, functional assessment, reproducibility, accuracy and scalability of these models are also discussed.
Collapse
Affiliation(s)
- Dario Pacitti
- Molecular and Clinical Sciences Research Institute, St George’s, University of London, London, United Kingdom
- College of Medicine and Health, St Luke’s Campus, University of Exeter, Exeter, United Kingdom
| | - Riccardo Privolizzi
- Gene Transfer Technology Group, Institute for Women’s Health, University College London, London, United Kingdom
| | - Bridget E. Bax
- Molecular and Clinical Sciences Research Institute, St George’s, University of London, London, United Kingdom
| |
Collapse
|
468
|
Werner C. Matrix-mediated modulation of neuron identity. Nat Biomed Eng 2019; 2:473-474. [PMID: 30948829 DOI: 10.1038/s41551-018-0270-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
- Carsten Werner
- Max Bergmann Center of Biomaterials, Leibniz Institute of Polymer Research Dresden and Center for Regenerative Therapies Dresden, TU Dresden, Dresden, Germany.
| |
Collapse
|
469
|
TCW J. Human iPSC application in Alzheimer’s disease and Tau-related neurodegenerative diseases. Neurosci Lett 2019; 699:31-40. [DOI: 10.1016/j.neulet.2019.01.043] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2018] [Revised: 11/23/2018] [Accepted: 01/23/2019] [Indexed: 12/11/2022]
|
470
|
Specification of positional identity in forebrain organoids. Nat Biotechnol 2019; 37:436-444. [PMID: 30936566 PMCID: PMC6447454 DOI: 10.1038/s41587-019-0085-3] [Citation(s) in RCA: 219] [Impact Index Per Article: 36.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2017] [Accepted: 02/22/2019] [Indexed: 01/28/2023]
Abstract
Human brain organoids generated with current technologies recapitulate histological features of the human brain, but they lack a reproducible topographic organization. During development, spatial topography is determined by gradients of signaling molecules released from discrete signaling centers. We hypothesized that introduction of a signaling center into forebrain organoids would specify the positional identity of neural tissue in a distance-dependent manner. Here, we present a system to trigger a sonic hedgehog (SHH) protein gradient in developing forebrain organoids that enables ordered self-organization along dorso-ventral and antero-posterior positional axes. SHH-patterned forebrain organoids establish major forebrain subdivisions that are positioned with in vivo-like topography. Consistent with its behavior in vivo, SHH exhibits long-range signaling activity in organoids. Finally, we use SHH-patterned cerebral organoids as a tool to study the role of cholesterol metabolism in SHH signaling. Together, this work identifies inductive signaling as an effective organizing strategy to recapitulate in vivo-like topography in human brain organoids.
Collapse
|
471
|
Giandomenico SL, Mierau SB, Gibbons GM, Wenger LMD, Masullo L, Sit T, Sutcliffe M, Boulanger J, Tripodi M, Derivery E, Paulsen O, Lakatos A, Lancaster MA. Cerebral organoids at the air-liquid interface generate diverse nerve tracts with functional output. Nat Neurosci 2019; 22:669-679. [PMID: 30886407 PMCID: PMC6436729 DOI: 10.1038/s41593-019-0350-2] [Citation(s) in RCA: 380] [Impact Index Per Article: 63.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2018] [Accepted: 01/28/2019] [Indexed: 12/17/2022]
Abstract
Neural organoids have the potential to improve our understanding of human brain development and neurological disorders. However, it remains to be seen whether these tissues can model circuit formation with functional neuronal output. Here we have adapted air-liquid interface culture to cerebral organoids, leading to improved neuronal survival and axon outgrowth. The resulting thick axon tracts display various morphologies, including long-range projection within and away from the organoid, growth-cone turning, and decussation. Single-cell RNA sequencing reveals various cortical neuronal identities, and retrograde tracing demonstrates tract morphologies that match proper molecular identities. These cultures exhibit active neuronal networks, and subcortical projecting tracts can innervate mouse spinal cord explants and evoke contractions of adjacent muscle in a manner dependent on intact organoid-derived innervating tracts. Overall, these results reveal a remarkable self-organization of corticofugal and callosal tracts with a functional output, providing new opportunities to examine relevant aspects of human CNS development and disease.
Collapse
Affiliation(s)
| | - Susanna B Mierau
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
| | - George M Gibbons
- John van Geest Centre for Brain Repair and Division of Stem Cell Neurobiology, Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
| | - Lea M D Wenger
- John van Geest Centre for Brain Repair and Division of Stem Cell Neurobiology, Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
| | - Laura Masullo
- MRC Laboratory of Molecular Biology, Cambridge Biomedical Campus, Cambridge, UK
| | - Timothy Sit
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
| | - Magdalena Sutcliffe
- MRC Laboratory of Molecular Biology, Cambridge Biomedical Campus, Cambridge, UK
| | - Jerome Boulanger
- MRC Laboratory of Molecular Biology, Cambridge Biomedical Campus, Cambridge, UK
| | - Marco Tripodi
- MRC Laboratory of Molecular Biology, Cambridge Biomedical Campus, Cambridge, UK
| | - Emmanuel Derivery
- MRC Laboratory of Molecular Biology, Cambridge Biomedical Campus, Cambridge, UK
| | - Ole Paulsen
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
| | - András Lakatos
- John van Geest Centre for Brain Repair and Division of Stem Cell Neurobiology, Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
- Wellcome Trust-MRC Cambridge Stem Cell Institute, Cambridge Biomedical Campus, Cambridge, UK
| | - Madeline A Lancaster
- MRC Laboratory of Molecular Biology, Cambridge Biomedical Campus, Cambridge, UK.
| |
Collapse
|
472
|
Studying Heterotypic Cell⁻Cell Interactions in the Human Brain Using Pluripotent Stem Cell Models for Neurodegeneration. Cells 2019; 8:cells8040299. [PMID: 30939814 PMCID: PMC6523455 DOI: 10.3390/cells8040299] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2019] [Revised: 03/26/2019] [Accepted: 03/29/2019] [Indexed: 02/08/2023] Open
Abstract
Human cerebral organoids derived from induced pluripotent stem cells (iPSCs) provide novel tools for recapitulating the cytoarchitecture of the human brain and for studying biological mechanisms of neurological disorders. However, the heterotypic interactions of neurovascular units, composed of neurons, pericytes (i.e., the tissue resident mesenchymal stromal cells), astrocytes, and brain microvascular endothelial cells, in brain-like tissues are less investigated. In addition, most cortical organoids lack a microglia component, the resident immune cells in the brain. Impairment of the blood-brain barrier caused by improper crosstalk between neural cells and vascular cells is associated with many neurodegenerative disorders. Mesenchymal stem cells (MSCs), with a phenotype overlapping with pericytes, have promotion effects on neurogenesis and angiogenesis, which are mainly attributed to secreted growth factors and extracellular matrices. As the innate macrophages of the central nervous system, microglia regulate neuronal activities and promote neuronal differentiation by secreting neurotrophic factors and pro-/anti-inflammatory molecules. Neuronal-microglia interactions mediated by chemokines signaling can be modulated in vitro for recapitulating microglial activities during neurodegenerative disease progression. In this review, we discussed the cellular interactions and the physiological roles of neural cells with other cell types including endothelial cells and microglia based on iPSC models. The therapeutic roles of MSCs in treating neural degeneration and pathological roles of microglia in neurodegenerative disease progression were also discussed.
Collapse
|
473
|
George J, Hsu CC, Nguyen LTB, Ye H, Cui Z. Neural tissue engineering with structured hydrogels in CNS models and therapies. Biotechnol Adv 2019; 42:107370. [PMID: 30902729 DOI: 10.1016/j.biotechadv.2019.03.009] [Citation(s) in RCA: 79] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2018] [Revised: 02/25/2019] [Accepted: 03/11/2019] [Indexed: 01/27/2023]
Abstract
The development of techniques to create and use multiphase microstructured hydrogels (granular hydrogels or microgels) has enabled the generation of cultures with more biologically relevant architecture and use of structured hydrogels is especially pertinent to the development of new types of central nervous system (CNS) culture models and therapies. We review material choice and the customisation of hydrogel structure, as well as the use of hydrogels in developmental models. Combining the use of structured hydrogel techniques with developmentally relevant tissue culture approaches will enable the generation of more relevant models and treatments to repair damaged CNS tissue architecture.
Collapse
Affiliation(s)
- Julian George
- Institute of Biomedical Engineering, Department of Engineering Science, University of Oxford, Oxford, United Kingdom
| | - Chia-Chen Hsu
- Institute of Biomedical Engineering, Department of Engineering Science, University of Oxford, Oxford, United Kingdom
| | - Linh Thuy Ba Nguyen
- Institute of Biomedical Engineering, Department of Engineering Science, University of Oxford, Oxford, United Kingdom
| | - Hua Ye
- Institute of Biomedical Engineering, Department of Engineering Science, University of Oxford, Oxford, United Kingdom.
| | - Zhanfeng Cui
- Institute of Biomedical Engineering, Department of Engineering Science, University of Oxford, Oxford, United Kingdom.
| |
Collapse
|
474
|
Cell migration promotes dynamic cellular interactions to control cerebral cortex morphogenesis. Nat Rev Neurosci 2019; 20:318-329. [DOI: 10.1038/s41583-019-0148-y] [Citation(s) in RCA: 57] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
475
|
Logan S, Arzua T, Canfield SG, Seminary ER, Sison SL, Ebert AD, Bai X. Studying Human Neurological Disorders Using Induced Pluripotent Stem Cells: From 2D Monolayer to 3D Organoid and Blood Brain Barrier Models. Compr Physiol 2019; 9:565-611. [PMID: 30873582 PMCID: PMC6705133 DOI: 10.1002/cphy.c180025] [Citation(s) in RCA: 85] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Neurological disorders have emerged as a predominant healthcare concern in recent years due to their severe consequences on quality of life and prevalence throughout the world. Understanding the underlying mechanisms of these diseases and the interactions between different brain cell types is essential for the development of new therapeutics. Induced pluripotent stem cells (iPSCs) are invaluable tools for neurological disease modeling, as they have unlimited self-renewal and differentiation capacity. Mounting evidence shows: (i) various brain cells can be generated from iPSCs in two-dimensional (2D) monolayer cultures; and (ii) further advances in 3D culture systems have led to the differentiation of iPSCs into organoids with multiple brain cell types and specific brain regions. These 3D organoids have gained widespread attention as in vitro tools to recapitulate complex features of the brain, and (iii) complex interactions between iPSC-derived brain cell types can recapitulate physiological and pathological conditions of blood-brain barrier (BBB). As iPSCs can be generated from diverse patient populations, researchers have effectively applied 2D, 3D, and BBB models to recapitulate genetically complex neurological disorders and reveal novel insights into molecular and genetic mechanisms of neurological disorders. In this review, we describe recent progress in the generation of 2D, 3D, and BBB models from iPSCs and further discuss their limitations, advantages, and future ventures. This review also covers the current status of applications of 2D, 3D, and BBB models in drug screening, precision medicine, and modeling a wide range of neurological diseases (e.g., neurodegenerative diseases, neurodevelopmental disorders, brain injury, and neuropsychiatric disorders). © 2019 American Physiological Society. Compr Physiol 9:565-611, 2019.
Collapse
Affiliation(s)
- Sarah Logan
- Department of Cell Biology, Neurobiology & Anatomy, Medical College of Wisconsin, Milwaukee, WI, USA
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Thiago Arzua
- Department of Cell Biology, Neurobiology & Anatomy, Medical College of Wisconsin, Milwaukee, WI, USA
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Scott G. Canfield
- Department of Cellular & Integrative Physiology, IU School of Medicine-Terre Haute, Terre Haute, IN, USA
| | - Emily R. Seminary
- Department of Cell Biology, Neurobiology & Anatomy, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Samantha L. Sison
- Department of Cell Biology, Neurobiology & Anatomy, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Allison D. Ebert
- Department of Cell Biology, Neurobiology & Anatomy, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Xiaowen Bai
- Department of Cell Biology, Neurobiology & Anatomy, Medical College of Wisconsin, Milwaukee, WI, USA
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI, USA
| |
Collapse
|
476
|
Goto-Silva L, Ayad NME, Herzog IL, Silva NP, Lamien B, Orlande HRB, da Costa Souza A, Ribeiro S, Martins M, Domont GB, Junqueira M, Tovar-Moll F, Rehen SK. Computational fluid dynamic analysis of physical forces playing a role in brain organoid cultures in two different multiplex platforms. BMC DEVELOPMENTAL BIOLOGY 2019; 19:3. [PMID: 30841924 PMCID: PMC6404276 DOI: 10.1186/s12861-019-0183-y] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/14/2018] [Accepted: 01/31/2019] [Indexed: 12/21/2022]
Abstract
Background Organoid cultivation in suspension culture requires agitation at low shear stress to allow for nutrient diffusion, which preserves tissue structure. Multiplex systems for organoid cultivation have been proposed, but whether they meet similar shear stress parameters as the regularly used spinner flask and its correlation with the successful generation of brain organoids has not been determined. Results Here we used computational fluid dynamics (CFD) to simulate two multiplex culture conditions: steering plates on an orbital shaker and the use of a previously described bioreactor. The bioreactor had low speed and high shear stress regions that may affect cell aggregate growth, depending on volume, whereas the computed variables of the steering plates were closer to those of the spinning flask. Conclusion Our protocol improves the initial steps of the standard brain organoid formation, and the produced organoids displayed regionalized brain structures, including retinal pigmented cells. Overall, we conclude that suspension culture on orbital steering plates is a cost-effective practical alternative to previously described platforms for the cultivation of brain organoids for research and multiplex testing. Electronic supplementary material The online version of this article (10.1186/s12861-019-0183-y) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Livia Goto-Silva
- D'Or Institute for Research and Education (IDOR), Rua Diniz Cordeiro, 30 - Botafogo, Rio de Janeiro, RJ, 22281-100, Brazil
| | - Nadia M E Ayad
- D'Or Institute for Research and Education (IDOR), Rua Diniz Cordeiro, 30 - Botafogo, Rio de Janeiro, RJ, 22281-100, Brazil
| | - Iasmin L Herzog
- Department of Mechanical Engineering, Politecnica/COPPE - Federal University of Rio de Janeiro, UFRJ, Av. Horácio Macedo, 2030, Cidade Universitária, Rio de Janeiro, RJ, 21941-914, Brazil
| | - Nilton P Silva
- Department of Mechanical Engineering, Politecnica/COPPE - Federal University of Rio de Janeiro, UFRJ, Av. Horácio Macedo, 2030, Cidade Universitária, Rio de Janeiro, RJ, 21941-914, Brazil
| | - Bernard Lamien
- Department of Mechanical Engineering, Politecnica/COPPE - Federal University of Rio de Janeiro, UFRJ, Av. Horácio Macedo, 2030, Cidade Universitária, Rio de Janeiro, RJ, 21941-914, Brazil
| | - Helcio R B Orlande
- Department of Mechanical Engineering, Politecnica/COPPE - Federal University of Rio de Janeiro, UFRJ, Av. Horácio Macedo, 2030, Cidade Universitária, Rio de Janeiro, RJ, 21941-914, Brazil
| | - Annie da Costa Souza
- Brain Institute, Federal University of Rio Grande do Norte, Av. Nascimento de Castro, 2155, Natal, RN, 59056-450, Brazil
| | - Sidarta Ribeiro
- Brain Institute, Federal University of Rio Grande do Norte, Av. Nascimento de Castro, 2155, Natal, RN, 59056-450, Brazil
| | - Michele Martins
- Proteomics Unit, Institute of Chemistry, Federal University of Rio de Janeiro, UFRJ, Av. Athos da Silveira Ramos 149, Rio de Janeiro, 21941-909, Brazil
| | - Gilberto B Domont
- Proteomics Unit, Institute of Chemistry, Federal University of Rio de Janeiro, UFRJ, Av. Athos da Silveira Ramos 149, Rio de Janeiro, 21941-909, Brazil
| | - Magno Junqueira
- Proteomics Unit, Institute of Chemistry, Federal University of Rio de Janeiro, UFRJ, Av. Athos da Silveira Ramos 149, Rio de Janeiro, 21941-909, Brazil
| | - Fernanda Tovar-Moll
- D'Or Institute for Research and Education (IDOR), Rua Diniz Cordeiro, 30 - Botafogo, Rio de Janeiro, RJ, 22281-100, Brazil.,Institute of Biomedical Sciences, Federal University of Rio de Janeiro, UFRJ, Av. Carlos Chagas Filho 373, Bloco K, Cidade Universitária, Rio de Janeiro, RJ, 21941-902, Brazil
| | - Stevens K Rehen
- D'Or Institute for Research and Education (IDOR), Rua Diniz Cordeiro, 30 - Botafogo, Rio de Janeiro, RJ, 22281-100, Brazil. .,Institute of Biomedical Sciences, Federal University of Rio de Janeiro, UFRJ, Av. Carlos Chagas Filho 373, Bloco K, Cidade Universitária, Rio de Janeiro, RJ, 21941-902, Brazil.
| |
Collapse
|
477
|
Varrault A, Journot L, Bouschet T. Cerebral Cortex Generated from Pluripotent Stem Cells to Model Corticogenesis and Rebuild Cortical Circuits: In Vitro Veritas? Stem Cells Dev 2019; 28:361-369. [PMID: 30661489 DOI: 10.1089/scd.2018.0233] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Organoids and cells generated in vitro from pluripotent stem cells (PSCs) are considered to be robust models of development and a conceivable source of transplants for putative cell therapy. However, a fundamental question about organoids and cells generated from PSCs is as follows: do they faithfully reproduce the in vivo tissue they are supposed to mimic and replace? This question is particularly relevant to complex tissues such as the cerebral cortex. In this review, we have tackled this issue by comparing cerebral cortices generated in vitro from PSCs to the in vivo cortex, with a particular focus on their respective cellular composition, molecular and epigenetic signatures, and brain connectivity. In short, in vitro cortex generated from PSCs reproduces most of the cardinal features of the in vivo cortex, including temporal corticogenesis and connectivity when PSC-derived cortical cells are grafted in recipient mouse cortex. However, compared to in vivo cortex, in vitro cortex lacks microglia and blood vessels and is less mature. Recent experiments show that the brain of the transplanted host provides these missing cell types together with an environment that promotes the synaptic maturation of the cortical transplant. Taken together, these data suggest that corticogenesis is largely intrinsic and well recapitulated in vitro, while the full maturation of cortical cells requires additional environmental clues. Finally, we propose some lines of work to improve corticogenesis from PSCs as a tool to model corticogenesis and rebuild cortical circuits.
Collapse
Affiliation(s)
- Annie Varrault
- Institut de Génomique Fonctionnelle, IGF, CNRS, INSERM, Université de Montpellier, Montpellier, France
| | - Laurent Journot
- Institut de Génomique Fonctionnelle, IGF, CNRS, INSERM, Université de Montpellier, Montpellier, France
| | - Tristan Bouschet
- Institut de Génomique Fonctionnelle, IGF, CNRS, INSERM, Université de Montpellier, Montpellier, France
| |
Collapse
|
478
|
P Rothenbücher TS, Martínez-Serrano A. Human cerebral organoids and neural 3D tissues in basic research, and their application to study neurological diseases. FUTURE NEUROLOGY 2019. [DOI: 10.2217/fnl-2018-0043] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Affiliation(s)
- Theresa S P Rothenbücher
- Department of Molecular Biology, Univ. Autónoma de Madrid; & Department of Molecular Neuropathology, Center of Molecular Biology Severo Ochoa (CBMSO, UAM-CSIC), Nicolás Cabrera 1, 28049 Madrid, Spain
| | - Alberto Martínez-Serrano
- Department of Molecular Biology, Univ. Autónoma de Madrid; & Department of Molecular Neuropathology, Center of Molecular Biology Severo Ochoa (CBMSO, UAM-CSIC), Nicolás Cabrera 1, 28049 Madrid, Spain
| |
Collapse
|
479
|
Daniel E, Cleaver O. Vascularizing organogenesis: Lessons from developmental biology and implications for regenerative medicine. Curr Top Dev Biol 2019; 132:177-220. [PMID: 30797509 DOI: 10.1016/bs.ctdb.2018.12.012] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Organogenesis requires tightly coordinated and patterned growth of numerous cell types to form a fully mature and vascularized organ. Endothelial cells (ECs) that line blood vessels develop alongside the growing organ, but only recently has their role in directing epithelial and stromal growth been appreciated. Endothelial, epithelial, and stromal cells in embryonic organs actively communicate with one another throughout development to ensure that the organ forms appropriately. What signals tell blood vessel progenitors where to go? How are tissues influenced by the vasculature that pervades it? In this chapter, we review the ways in which crosstalk between ECs and epithelial or stromal cells during development leads to a fully patterned pancreas, lung, or kidney. ECs in all of these organs are necessary for proper epithelial and stromal growth, but how they direct this process is organ- and time-specific, highlighting the concept of dynamic EC heterogeneity. We end with a discussion on how understanding cell-cell crosstalk during development can be applied therapeutically through the generation of transplantable miniature organ-like tissues called "organoids." We will discuss the current state of organoid technology and highlight the major challenges in forming a properly patterned vascular network that will be critical in transforming them into a viable therapeutic option.
Collapse
Affiliation(s)
- Edward Daniel
- Department of Molecular Biology and Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Ondine Cleaver
- Department of Molecular Biology and Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, TX, United States.
| |
Collapse
|
480
|
Buchsbaum IY, Cappello S. Neuronal migration in the CNS during development and disease: insights from in vivo and in vitro models. Development 2019; 146:146/1/dev163766. [DOI: 10.1242/dev.163766] [Citation(s) in RCA: 78] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
ABSTRACT
Neuronal migration is a fundamental process that governs embryonic brain development. As such, mutations that affect essential neuronal migration processes lead to severe brain malformations, which can cause complex and heterogeneous developmental and neuronal migration disorders. Our fragmented knowledge about the aetiology of these disorders raises numerous issues. However, many of these can now be addressed through studies of in vivo and in vitro models that attempt to recapitulate human-specific mechanisms of cortical development. In this Review, we discuss the advantages and limitations of these model systems and suggest that a complementary approach, using combinations of in vivo and in vitro models, will broaden our knowledge of the molecular and cellular mechanisms that underlie defective neuronal positioning in the human cerebral cortex.
Collapse
Affiliation(s)
- Isabel Yasmin Buchsbaum
- Developmental Neurobiology, Max Planck Institute of Psychiatry, 80804 Munich, Germany
- Graduate School of Systemic Neurosciences, Ludwig-Maximilians-University Munich, 82152 Planegg, Germany
| | - Silvia Cappello
- Developmental Neurobiology, Max Planck Institute of Psychiatry, 80804 Munich, Germany
| |
Collapse
|
481
|
St Clair D, Johnstone M. Using mouse transgenic and human stem cell technologies to model genetic mutations associated with schizophrenia and autism. Philos Trans R Soc Lond B Biol Sci 2019; 373:rstb.2017.0037. [PMID: 29352035 PMCID: PMC5790834 DOI: 10.1098/rstb.2017.0037] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/02/2017] [Indexed: 12/22/2022] Open
Abstract
Solid progress has occurred over the last decade in our understanding of the molecular genetic basis of neurodevelopmental disorders, and of schizophrenia and autism in particular. Although the genetic architecture of both disorders is far more complex than previously imagined, many key loci have at last been identified. This has allowed in vivo and in vitro technologies to be refined to model specific high-penetrant genetic loci involved in both disorders. Using the DISC1/NDE1 and CYFIP1/EIF4E loci as exemplars, we explore the opportunities and challenges of using animal models and human-induced pluripotent stem cell technologies to further understand/treat and potentially reverse the worst consequences of these debilitating disorders. This article is part of a discussion meeting issue ‘Of mice and mental health: facilitating dialogue between basic and clinical neuroscientists’.
Collapse
Affiliation(s)
- David St Clair
- Institute of Medical Sciences, University of Aberdeen, Foresterhill, Aberdeen, UK
| | - Mandy Johnstone
- Division of Psychiatry, University of Edinburgh, Royal Edinburgh Hospital, Edinburgh, UK.,Centre for Genomic and Experimental Medicine, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, UK.,Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, UK
| |
Collapse
|
482
|
Chen HI, Song H, Ming GL. Applications of Human Brain Organoids to Clinical Problems. Dev Dyn 2019; 248:53-64. [PMID: 30091290 PMCID: PMC6312736 DOI: 10.1002/dvdy.24662] [Citation(s) in RCA: 79] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2018] [Revised: 07/30/2018] [Accepted: 08/01/2018] [Indexed: 12/13/2022] Open
Abstract
Brain organoids are an exciting new technology with the potential to significantly change how diseases of the brain are understood and treated. These three-dimensional neural tissues are derived from the self-organization of pluripotent stem cells, and they recapitulate the developmental process of the human brain, including progenitor zones and rudimentary cortical layers. Brain organoids have been valuable in investigating different aspects of developmental neurobiology and comparative biology. Several characteristics of organoids also make them attractive as models of brain disorders. Data generated from human organoids are more generalizable to patients because of the match in species background. Personalized organoids also can be generated from patient-derived induced pluripotent stem cells. Furthermore, the three-dimensionality of brain organoids supports cellular, mechanical, and topographical cues that are lacking in planar systems. In this review, we discuss the translational potential of brain organoids, using the examples of Zika virus, autism-spectrum disorder, and glioblastoma multiforme to consider how they could contribute to disease modeling, personalized medicine, and testing of therapeutics. We then discuss areas of improvement in organoid technology that will enhance the translational potential of brain organoids, as well as the possibility of their use as substrates for repairing cerebral circuitry after injury. Developmental Dynamics 248:53-64, 2019. © 2018 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- H. Isaac Chen
- Department of Neurosurgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
- Corporal Michael J. Crescenz Veterans Affairs Medical Center, Philadelphia, PA
- Institute for Regenerative Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Hongjun Song
- Department of Neuroscience, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
- Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
- Institute for Regenerative Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
- The Epigenetics Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Guo-li Ming
- Department of Neuroscience, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
- Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
- Institute for Regenerative Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| |
Collapse
|
483
|
Vaez Ghaemi R, Co IL, McFee MC, Yadav VG. Brain Organoids: A New, Transformative Investigational Tool for Neuroscience Research. ACTA ACUST UNITED AC 2019; 3:e1800174. [PMID: 32627343 DOI: 10.1002/adbi.201800174] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2018] [Revised: 08/27/2018] [Indexed: 12/22/2022]
Abstract
Brain organoids are self-assembled, three-dimensionally structured tissues that are typically derived from pluripotent stem cells. They are multicellular aggregates that more accurately recapitulate the tissue microenvironment compared to the other cell culture systems and can also reproduce organ function. They are promising models for evaluating drug leads, particularly those that target neurodegeneration, since they are genetically and phenotypically stable over prolonged durations of culturing and they reasonably reproduce critical physiological phenomena such as biochemical gradients and responses by the native tissue to stimuli. Beyond drug discovery, the use of brain organoids could also be extended to investigating early brain development and identifying the mechanisms that elicit neurodegeneration. Herein, the current state of the fabrication and use of brain organoids in drug development and medical research is summarized. Although the use of brain organoids represents a quantum leap over existing investigational tools used by the pharmaceutical industry, they are nonetheless imperfect systems that could be greatly improved through bioengineering. To this end, some key scientific challenges that would need to be addressed in order to enhance the relevance of brain organoids as model tissue are listed. Potential solutions to these challenges, including the use of bioprinting, are highlighted thereafter.
Collapse
Affiliation(s)
- Roza Vaez Ghaemi
- Department of Chemical & Biological Engineering & School of Biomedical Engineering, The University of British Columbia, Vancouver, British Columbia, V6T 1Z4, Canada
| | - Ileana L Co
- Department of Chemical & Biological Engineering & School of Biomedical Engineering, The University of British Columbia, Vancouver, British Columbia, V6T 1Z4, Canada
| | - Matthew C McFee
- Department of Chemical & Biological Engineering & School of Biomedical Engineering, The University of British Columbia, Vancouver, British Columbia, V6T 1Z4, Canada
| | - Vikramaditya G Yadav
- Department of Chemical & Biological Engineering & School of Biomedical Engineering, The University of British Columbia, Vancouver, British Columbia, V6T 1Z4, Canada
| |
Collapse
|
484
|
Liu F, Huang J, Zhang L, Chen J, Zeng Y, Tang Y, Liu Z. Advances in Cerebral Organoid Systems and their Application in Disease Modeling. Neuroscience 2018; 399:28-38. [PMID: 30578974 DOI: 10.1016/j.neuroscience.2018.12.013] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2018] [Revised: 12/08/2018] [Accepted: 12/12/2018] [Indexed: 01/09/2023]
Abstract
Processes associated with human brain development and function are exceedingly complex, limiting our capacity to investigate disease status and potential treatment strategies in vitro. Recent advancements in human cerebral organoid systems-which replicate early stage neural tube formation, neuroepithelium differentiation, and whole-brain regional differentiation-have allowed researchers to generate more accurate models of brain development and disease. The generation of region-specific cerebral organoids also allows for the direct investigation of the etiology and pathological processes associated with inherited and acquired brain diseases, drug discovery, and drug toxicity. In this review, we provide an overview of various neural differentiation technologies, as well as a critical analysis of their strengths and limitations. We primarily focus on the generation of three-dimensional brain organoid systems and their application in infectious disease modeling, high-throughput compound screening, and neurodevelopmental disease modeling.
Collapse
Affiliation(s)
- Fangkun Liu
- Department of Neurosurgery, Xiangya Hospital, Central South University (CSU), Changsha, China
| | - Jing Huang
- Department of Psychiatry, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China; Mental Health Institute of the Second Xiangya Hospital, Central South University, Chinese National Clinical Research Center on Mental Disorders (xiangya), Chinese National Technology Institute on Mental Disorders, Hunan Key Laboratory of Psychiatry and Mental Health, Changsha, Hunan 410011, China
| | - Liyang Zhang
- Department of Neurosurgery, Xiangya Hospital, Central South University (CSU), Changsha, China
| | - Jindong Chen
- Department of Psychiatry, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China; Mental Health Institute of the Second Xiangya Hospital, Central South University, Chinese National Clinical Research Center on Mental Disorders (xiangya), Chinese National Technology Institute on Mental Disorders, Hunan Key Laboratory of Psychiatry and Mental Health, Changsha, Hunan 410011, China
| | - Yu Zeng
- Department of Neurosurgery, Xiangya Hospital, Central South University (CSU), Changsha, China
| | - Yongjian Tang
- Department of Neurosurgery, Xiangya Hospital, Central South University (CSU), Changsha, China
| | - Zhixiong Liu
- Department of Neurosurgery, Xiangya Hospital, Central South University (CSU), Changsha, China.
| |
Collapse
|
485
|
McCaughey-Chapman A, Connor B. Human Cortical Neuron Generation Using Cell Reprogramming: A Review of Recent Advances. Stem Cells Dev 2018; 27:1674-1692. [DOI: 10.1089/scd.2018.0122] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Affiliation(s)
- Amy McCaughey-Chapman
- Department of Pharmacology and Clinical Pharmacology, Centre for Brain Research, School of Medical Science, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | - Bronwen Connor
- Department of Pharmacology and Clinical Pharmacology, Centre for Brain Research, School of Medical Science, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| |
Collapse
|
486
|
Xu H, Jiao Y, Qin S, Zhao W, Chu Q, Wu K. Organoid technology in disease modelling, drug development, personalized treatment and regeneration medicine. Exp Hematol Oncol 2018; 7:30. [PMID: 30534474 PMCID: PMC6282260 DOI: 10.1186/s40164-018-0122-9] [Citation(s) in RCA: 118] [Impact Index Per Article: 16.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2018] [Accepted: 11/29/2018] [Indexed: 12/13/2022] Open
Abstract
Organoid technology bridges the gap between conventional two-dimensional cell line culture and in vivo models. The near-physiological technology can virtually recapitulates organ development and human diseases, such as infectious diseases, genetic abnormality and even cancers. In addition, organoids can more accurately predict drug responses, and serve as an excellent platform for drug development, including efficacy evaluation, toxicity testing and pharmacokinetics analysis. Furthermore, organoids can also be exploited to explore the possible optimized treatment strategies for each individual patient. Besides, organoid technology is a promising strategy for regeneration medicine and transplantation use, which can overcome the deficiency in the supply of healthy donor tissues and inherent immunological rejection through establishing isogenic organoids from minuscule amounts of patient biopsies. Collectively, organoids hold enormous potential for clinical applications and bring basic research closer to clinical practice. In this review, we described common organoid lines, summarized the potential clinical applications, and outlined the current limitations.
Collapse
Affiliation(s)
- Hanxiao Xu
- Department of Oncology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan, 430030 China
| | - Ying Jiao
- Department of Oncology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan, 430030 China
| | - Shuang Qin
- Department of Oncology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan, 430030 China
| | - Weiheng Zhao
- Department of Oncology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan, 430030 China
| | - Qian Chu
- Department of Oncology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan, 430030 China
| | - Kongming Wu
- Department of Oncology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan, 430030 China
| |
Collapse
|
487
|
Gardner E, Ellington A. Reprogramming the brain with synthetic neurobiology. Curr Opin Biotechnol 2018; 58:37-44. [PMID: 30458406 DOI: 10.1016/j.copbio.2018.10.013] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2018] [Accepted: 10/26/2018] [Indexed: 12/28/2022]
Abstract
The mammalian brain is among the most complex organs known in biology. Historically, neuroscience techniques have consisted primarily of low-throughput microscopy and electrophysiological approaches. While these methods will continue to serve the community, the emerging field of synthetic neurobiology may be better equipped to scale with systems neuroscience. By using genetic techniques to achieve cell-type specificity, a map of the connectome, neural activation and recording, and ultimately to program neural development itself, we can begin to build a better framework with which to understand the brain's mechanisms.
Collapse
Affiliation(s)
- Elizabeth Gardner
- Center for Systems and Synthetic Biology, Institute for Cellular and Molecular Biology, Department of Molecular Biosciences, University of Texas, 2500 Speedway, Austin, TX 78712, USA
| | - Andrew Ellington
- Center for Systems and Synthetic Biology, Institute for Cellular and Molecular Biology, Department of Molecular Biosciences, University of Texas, 2500 Speedway, Austin, TX 78712, USA.
| |
Collapse
|
488
|
Jabaudon D, Lancaster M. Exploring landscapes of brain morphogenesis with organoids. Development 2018; 145:145/22/dev172049. [PMID: 30455367 DOI: 10.1242/dev.172049] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
The field of developmental neuroscience is benefitting from recent technological advances that allow access to organogenesis in vitro via organoid preparations. These methods have been applied to better understanding neural identity, and have opened up a window into the early events that occur during development of the human brain. However, current approaches are not without their limitations, and although brain organoids and other in vitro paradigms recapitulate many processes with remarkable fidelity, there are clear differences between brain organoid development in vitro and brain development in vivo These topics were discussed extensively at a recent workshop organized by The Company of Biologists entitled 'Thinking beyond the dish: taking in vitro neural differentiation to the next level'. Here, we summarize the common themes that emerged from the workshop and highlight some of the limitations and the potential of this emerging technology. In particular, we discuss how organoids can help us understand not only healthy and diseased brain, but also explore new arrays of cellular behaviors.
Collapse
Affiliation(s)
- Denis Jabaudon
- Department of Basic Neurosciences, University of Geneva, 1211 Geneva, Switzerland .,Clinic of Neurology, Geneva University Hospital, Geneva, Switzerland
| | - Madeline Lancaster
- MRC Laboratory of Molecular Biology, Cambridge Biomedical Campus, Cambridge CB2 0QH, UK
| |
Collapse
|
489
|
Althammer F, Jirikowski G, Grinevich V. The oxytocin system of mice and men-Similarities and discrepancies of oxytocinergic modulation in rodents and primates. Peptides 2018; 109:1-8. [PMID: 30261208 DOI: 10.1016/j.peptides.2018.09.003] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/30/2018] [Revised: 09/18/2018] [Accepted: 09/18/2018] [Indexed: 01/21/2023]
Abstract
Nonapeptides and their respective receptors have been conserved throughout evolution and display astonishing similarities among the animal kingdom. They can be found in worms, birds, fish, amphibians, reptiles and mammals, including rodents, non-human primates and humans. In particular, the neuropeptide oxytocin (OT) has attracted the attention of scientists due to its profound effects on social behavior. However, although both the neuropeptide and its receptor are identical in rodents and primates, the effects of OT vary greatly in the two species. Here, we provide a brief overview about OT's role in the evolution of mammals and provide reasons for the manifold effects of OT within the brain with a particular focus on the discrepancy of OT's effects in rodents and primates. In addition, we suggest new approaches towards improvement of translatability of scientific studies and highlight the most recent advances in animal models for autism spectrum disorder, a disease, in which the normal function of the OT system seems to be impaired.
Collapse
Affiliation(s)
- Ferdinand Althammer
- Schaller Research Group on Neuropeptides at German Cancer Research Center (DKFZ) and Cell Network Cluster of Excellence at the University of Heidelberg, Heidelberg, Germany.
| | | | - Valery Grinevich
- Schaller Research Group on Neuropeptides at German Cancer Research Center (DKFZ) and Cell Network Cluster of Excellence at the University of Heidelberg, Heidelberg, Germany; Central Institute of Mental Health (ZI), Mannheim, Germany
| |
Collapse
|
490
|
De Gregorio V, Imparato G, Urciuolo F, Netti PA. Micro-patterned endogenous stroma equivalent induces polarized crypt-villus architecture of human small intestinal epithelium. Acta Biomater 2018; 81:43-59. [PMID: 30282052 DOI: 10.1016/j.actbio.2018.09.061] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2018] [Revised: 08/29/2018] [Accepted: 09/28/2018] [Indexed: 12/14/2022]
Abstract
The small intestine is the major site for digestion, drug and nutrient absorption, as well as a primary site for many diseases. Current in vitro gut models fail in reproducing the complex intestinal extracellular matrix (ECM) network of the lamina propria and the peculiar architecture of the crypt-villus axis. Here we proposed a novel in vitro human intestine model that mimics the intestinal stromal topography and composition and strictly reproduces the tissue polarity with the crypt-villus architecture. First we developed a 3D human intestinal stromal equivalent (3D-ISE) composed of human intestinal subepithelial myofibroblasts (ISEMFs) embedded in their own extracellular matrix. Then, we seeded human colon carcinoma-derived cells (Caco-2) onto flat or patterned cell-synthetized stromal equivalent structure and cultured them until the formation of a well-oriented epithelium. We demonstrated that the patterned stroma increases the absorbing surface area, the epithelial proliferation rate, and the density of microvilli. In addition it induces changes in the biological functions of the epithelial cells such as enzymes and mucus production, polarization and tightness showing a physiological cell-lineage compartmentalization along the crypt/villi axes with the undifferentiated phenotypes at the base. At last, we reproduced an inflamed intestinal tissue model in which we identified the contribution of the stromal microenvironment by molecular (cytokines release and MMPs production) and immunofluorescence analyses and the effects of the epithelial-stromal cross-talk in the intestinal innate immunity by multiphoton investigation that revealed differences in the collagen network architecture. STATEMENT OF SIGNIFICANCE: The intestinal stroma morphology and composition has a fundamental role in crypt-villus development and appropriate epithelial cell-lineage compartmentalization. On this base, here we develop an engineered organotypic model of human intestine equivalent in which a functional epithelial/ECM crosstalk is recapitulated. Due to its accessible luminal surface it provides a new platform for preclinical studies of mucosal immunology and bowel inflammation as well as the assessment of pharmaco-toxicity studies.
Collapse
Affiliation(s)
- Vincenza De Gregorio
- Center for Advanced Biomaterials for HealthCare@CRIB, Istituto Italiano di Tecnologia, Largo Barsanti e Matteucci 53, 80125 Naples, Italy
| | - Giorgia Imparato
- Center for Advanced Biomaterials for HealthCare@CRIB, Istituto Italiano di Tecnologia, Largo Barsanti e Matteucci 53, 80125 Naples, Italy.
| | - Francesco Urciuolo
- Department of Chemical Materials and Industrial Production (DICMAPI) University of Naples Federico II, P.le Tecchio 80, 80125 Naples, Italy
| | - Paolo A Netti
- Center for Advanced Biomaterials for HealthCare@CRIB, Istituto Italiano di Tecnologia, Largo Barsanti e Matteucci 53, 80125 Naples, Italy; Department of Chemical Materials and Industrial Production (DICMAPI) University of Naples Federico II, P.le Tecchio 80, 80125 Naples, Italy; Interdisciplinary Research Centre on Biomaterials (CRIB), University of Naples Federico II, P.le Tecchio 80, 80125 Naples, Italy
| |
Collapse
|
491
|
Knight GT, Lundin BF, Iyer N, Ashton LM, Sethares WA, Willett RM, Ashton RS. Engineering induction of singular neural rosette emergence within hPSC-derived tissues. eLife 2018; 7:37549. [PMID: 30371350 PMCID: PMC6205811 DOI: 10.7554/elife.37549] [Citation(s) in RCA: 77] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2018] [Accepted: 10/15/2018] [Indexed: 12/13/2022] Open
Abstract
Human pluripotent stem cell (hPSC)-derived neural organoids display unprecedented emergent properties. Yet in contrast to the singular neuroepithelial tube from which the entire central nervous system (CNS) develops in vivo, current organoid protocols yield tissues with multiple neuroepithelial units, a.k.a. neural rosettes, each acting as independent morphogenesis centers and thereby confounding coordinated, reproducible tissue development. Here, we discover that controlling initial tissue morphology can effectively (>80%) induce single neural rosette emergence within hPSC-derived forebrain and spinal tissues. Notably, the optimal tissue morphology for observing singular rosette emergence was distinct for forebrain versus spinal tissues due to previously unknown differences in ROCK-mediated cell contractility. Following release of geometric confinement, the tissues displayed radial outgrowth with maintenance of a singular neuroepithelium and peripheral neuronal differentiation. Thus, we have identified neural tissue morphology as a critical biophysical parameter for controlling in vitro neural tissue morphogenesis furthering advancement towards biomanufacture of CNS tissues with biomimetic anatomy and physiology.
Collapse
Affiliation(s)
- Gavin T Knight
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, United States.,Wisconsin Institute for Discovery, University of Wisconsin-Madison, Madison, United States
| | - Brady F Lundin
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, United States.,Wisconsin Institute for Discovery, University of Wisconsin-Madison, Madison, United States
| | - Nisha Iyer
- Wisconsin Institute for Discovery, University of Wisconsin-Madison, Madison, United States
| | - Lydia Mt Ashton
- Department of Consumer Science, University of Wisconsin-Madison, Madison, United States
| | - William A Sethares
- Department of Electrical and Computer Engineering, University of Wisconsin-Madison, Madison, United States
| | - Rebecca M Willett
- Wisconsin Institute for Discovery, University of Wisconsin-Madison, Madison, United States.,Department of Electrical and Computer Engineering, University of Wisconsin-Madison, Madison, United States
| | - Randolph Scott Ashton
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, United States.,Wisconsin Institute for Discovery, University of Wisconsin-Madison, Madison, United States
| |
Collapse
|
492
|
Soldner F, Jaenisch R. Stem Cells, Genome Editing, and the Path to Translational Medicine. Cell 2018; 175:615-632. [PMID: 30340033 PMCID: PMC6461399 DOI: 10.1016/j.cell.2018.09.010] [Citation(s) in RCA: 98] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2018] [Revised: 07/31/2018] [Accepted: 09/05/2018] [Indexed: 12/22/2022]
Abstract
The derivation of human embryonic stem cells (hESCs) and the stunning discovery that somatic cells can be reprogrammed into human induced pluripotent stem cells (hiPSCs) holds the promise to revolutionize biomedical research and regenerative medicine. In this Review, we focus on disorders of the central nervous system and explore how advances in human pluripotent stem cells (hPSCs) coincide with evolutions in genome engineering and genomic technologies to provide realistic opportunities to tackle some of the most devastating complex disorders.
Collapse
Affiliation(s)
- Frank Soldner
- The Whitehead Institute for Biomedical Research, 455 Main Street, Cambridge, MA 02142, USA
| | - Rudolf Jaenisch
- The Whitehead Institute for Biomedical Research, 455 Main Street, Cambridge, MA 02142, USA; Department of Biology, Massachusetts Institute of Technology, 31 Ames Street, Cambridge, MA 02139, USA.
| |
Collapse
|
493
|
Lancaster MA, Corsini NS, Wolfinger S, Gustafson EH, Phillips AW, Burkard TR, Otani T, Livesey FJ, Knoblich JA. Publisher Correction: Guided self-organization and cortical plate formation in human brain organoids. Nat Biotechnol 2018; 36:1016. [PMID: 30307926 DOI: 10.1038/nbt1018-1016a] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
This corrects the article DOI: 10.1038/nbt.3906.
Collapse
|
494
|
Kim S, Cho AN, Min S, Kim S, Cho SW. Organoids for Advanced Therapeutics and Disease Models. ADVANCED THERAPEUTICS 2018. [DOI: 10.1002/adtp.201800087] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Affiliation(s)
- Suran Kim
- Department of Biotechnology; Yonsei University; Seoul 03722 Republic of Korea
| | - Ann-Na Cho
- Department of Biotechnology; Yonsei University; Seoul 03722 Republic of Korea
| | - Sungjin Min
- Department of Biotechnology; Yonsei University; Seoul 03722 Republic of Korea
| | - Sooyeon Kim
- Department of Biotechnology; Yonsei University; Seoul 03722 Republic of Korea
| | - Seung-Woo Cho
- Department of Biotechnology; Yonsei University; Seoul 03722 Republic of Korea
| |
Collapse
|
495
|
Nayler SP, Becker EBE. The Use of Stem Cell-Derived Neurons for Understanding Development and Disease of the Cerebellum. Front Neurosci 2018; 12:646. [PMID: 30319335 PMCID: PMC6168705 DOI: 10.3389/fnins.2018.00646] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2018] [Accepted: 08/29/2018] [Indexed: 11/16/2022] Open
Abstract
The cerebellum is a fascinating brain structure, containing more neurons than the rest of the brain combined. The cerebellum develops according to a highly orchestrated program into a well-organized laminar structure. Much has been learned about the underlying genetic networks controlling cerebellar development through the study of various animal models. Cerebellar development in humans however, is significantly protracted and more complex. Given that the cerebellum regulates a number of motor and non-motor functions and is affected in a wide variety of neurodevelopmental and neurodegenerative disorders, a better understanding of human cerebellar development is highly desirable. Pluripotent stem cells offer an exciting new tool to unravel human cerebellar development and disease by providing a dynamic and malleable platform, which is amenable to genetic manipulation and temporally unrestricted sampling. It remains to be seen, however, whether in vitro neuronal cultures derived from pluripotent stem cells fully recapitulate the formation and organization of the developing nervous system, with many reports detailing the functionally immature nature of these cultures. Nevertheless, recent advances in differentiation protocols, cell-sampling methodologies, and access to informatics resources mean that the field is poised for remarkable discoveries. In this review, we provide a general overview of the field of neuronal differentiation, focusing on the cerebellum and highlighting conceptual advances in understanding neuronal maturity, including a discussion of both current and emerging methods to classify, and influence neuroanatomical identity and maturation status.
Collapse
Affiliation(s)
- Samuel P Nayler
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, United Kingdom
| | - Esther B E Becker
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
496
|
|
497
|
Xiang Y, Yoshiaki T, Patterson B, Cakir B, Kim KY, Cho YS, Park IH. Generation and Fusion of Human Cortical and Medial Ganglionic Eminence Brain Organoids. ACTA ACUST UNITED AC 2018; 47. [PMID: 30854156 DOI: 10.1002/cpsc.61] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Three-dimensional (3D) brain organoid culture has become an essential tool for investigating human brain development and modeling neurological disorders during the past few years. Given the specific regionalization during brain development, it is important to produce distinct brain organoids that reproduce different brain regions and their interaction. The authors' laboratory recently established the platform to generate brain organoids resembling the medial ganglionic eminence (MGE), a specific brain region responsible for interneurogenesis, and found when fusing with organoid resembling the cortex, the fused organoids enabled modeling of interneuron migration in the brain. This unit describes four basic protocols that have been successfully applied in the authors' laboratory, covering the generation of embryonic body (EB) with neuroectodermal fate, the production of MGE organoids (hMGEOs) and cortical organoids (hCOs), and the fusion of the two organoids.
Collapse
Affiliation(s)
- Yangfei Xiang
- Department of Genetics, Yale Stem Cell Center, Yale School of Medicine, New Haven, CT 06520, USA
| | - Tanaka Yoshiaki
- Department of Genetics, Yale Stem Cell Center, Yale School of Medicine, New Haven, CT 06520, USA
| | - Benjamin Patterson
- Department of Genetics, Yale Stem Cell Center, Yale School of Medicine, New Haven, CT 06520, USA
| | - Bilal Cakir
- Department of Genetics, Yale Stem Cell Center, Yale School of Medicine, New Haven, CT 06520, USA
| | - Kun-Yong Kim
- Department of Genetics, Yale Stem Cell Center, Yale School of Medicine, New Haven, CT 06520, USA
| | - Yee Sook Cho
- Regenerative Medicine Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, 305-806, ROK
| | - In-Hyun Park
- Department of Genetics, Yale Stem Cell Center, Yale School of Medicine, New Haven, CT 06520, USA
| |
Collapse
|
498
|
Patel R, Muir M, Cvetkovic C, Krencik R. Concepts toward directing human astroplasticity to promote neuroregeneration. Dev Dyn 2018; 248:21-33. [DOI: 10.1002/dvdy.24655] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2018] [Revised: 06/19/2018] [Accepted: 06/19/2018] [Indexed: 12/20/2022] Open
Affiliation(s)
| | | | - Caroline Cvetkovic
- Center for Neuroregeneration, Department of Neurosurgery; Houston Methodist Research Institute; Houston Texas
| | - Robert Krencik
- Center for Neuroregeneration, Department of Neurosurgery; Houston Methodist Research Institute; Houston Texas
| |
Collapse
|
499
|
Cholley PE, Moehlin J, Rohmer A, Zilliox V, Nicaise S, Gronemeyer H, Mendoza-Parra MA. Modeling gene-regulatory networks to describe cell fate transitions and predict master regulators. NPJ Syst Biol Appl 2018; 4:29. [PMID: 30083390 PMCID: PMC6070484 DOI: 10.1038/s41540-018-0066-z] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2018] [Revised: 06/13/2018] [Accepted: 06/21/2018] [Indexed: 01/10/2023] Open
Abstract
Complex organisms originate from and are maintained by the information encoded in the genome. A major challenge of systems biology is to develop algorithms that describe the dynamic regulation of genome functions from large omics datasets. Here, we describe TETRAMER, which reconstructs gene-regulatory networks from temporal transcriptome data during cell fate transitions to predict “master” regulators by simulating cascades of temporal transcription-regulatory events.
Collapse
Affiliation(s)
- Pierre-Etienne Cholley
- 1Equipe Labellisée Ligue Contre le Cancer, Department of Functional Genomics and Cancer, Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Centre National de la Recherche Scientifique UMR 7104, Institut National de la Santé et de la Recherche Médicale U964, University of Strasbourg, Illkirch, France.,2Present Address: Computational Systems Biology Infrastructure, Chalmers University of Technology, Kemivägen 10, 41296 Gothenburg, Sweden
| | - Julien Moehlin
- 1Equipe Labellisée Ligue Contre le Cancer, Department of Functional Genomics and Cancer, Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Centre National de la Recherche Scientifique UMR 7104, Institut National de la Santé et de la Recherche Médicale U964, University of Strasbourg, Illkirch, France
| | - Alexia Rohmer
- 1Equipe Labellisée Ligue Contre le Cancer, Department of Functional Genomics and Cancer, Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Centre National de la Recherche Scientifique UMR 7104, Institut National de la Santé et de la Recherche Médicale U964, University of Strasbourg, Illkirch, France
| | - Vincent Zilliox
- 1Equipe Labellisée Ligue Contre le Cancer, Department of Functional Genomics and Cancer, Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Centre National de la Recherche Scientifique UMR 7104, Institut National de la Santé et de la Recherche Médicale U964, University of Strasbourg, Illkirch, France
| | - Samuel Nicaise
- 1Equipe Labellisée Ligue Contre le Cancer, Department of Functional Genomics and Cancer, Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Centre National de la Recherche Scientifique UMR 7104, Institut National de la Santé et de la Recherche Médicale U964, University of Strasbourg, Illkirch, France
| | - Hinrich Gronemeyer
- 1Equipe Labellisée Ligue Contre le Cancer, Department of Functional Genomics and Cancer, Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Centre National de la Recherche Scientifique UMR 7104, Institut National de la Santé et de la Recherche Médicale U964, University of Strasbourg, Illkirch, France
| | - Marco Antonio Mendoza-Parra
- 1Equipe Labellisée Ligue Contre le Cancer, Department of Functional Genomics and Cancer, Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Centre National de la Recherche Scientifique UMR 7104, Institut National de la Santé et de la Recherche Médicale U964, University of Strasbourg, Illkirch, France.,3Present Address: UMR 8030 Génomique Métabolique, Genoscope, Institut François Jacob, CEA, CNRS, University of Evry-val-d'Essonne, University Paris-Saclay, 91057 Évry, France
| |
Collapse
|
500
|
Induction of myelinating oligodendrocytes in human cortical spheroids. Nat Methods 2018; 15:700-706. [PMID: 30046099 PMCID: PMC6508550 DOI: 10.1038/s41592-018-0081-4] [Citation(s) in RCA: 231] [Impact Index Per Article: 33.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2018] [Accepted: 07/02/2018] [Indexed: 01/07/2023]
Abstract
Organoid technologies provide an accessible system to examine cellular composition, interactions, and organization in the developing human brain, but previously have lacked oligodendrocytes, the myelinating glia of the central nervous system. Here we reproducibly generate oligodendrocytes and myelin in human pluripotent stem cell-derived “oligocortical spheroids”. Transcriptional, immunohistochemical, and electron microscopy analyses demonstrate molecular features consistent with maturing oligodendrocytes by 20 weeks in culture, including expression of MYRF, PLP1, and MBP proteins and initial myelin wrapping of axons, with maturation to longitudinal wrapping and compact myelin by 30 weeks. Promyelinating drugs enhance the rate and extent of oligodendrocyte generation and myelination, while oligocortical spheroids generated from patients with a genetic myelin disorder recapitulate human disease phenotypes. Oligocortical spheroids provide a versatile platform to observe and dissect the complex interactions required for myelination of the developing central nervous system and offer new opportunities for disease modeling and therapeutic development in human tissue.
Collapse
|